Skip to main content
Signal Transduction and Targeted Therapy logoLink to Signal Transduction and Targeted Therapy
. 2025 Apr 25;10:132. doi: 10.1038/s41392-025-02175-1

Diabetic neuropathy: cutting-edge research and future directions

Yang Yang 1,2,✉,#, Bing Zhao 1,2,#, Yuanzhe Wang 1,2, Hongli Lan 1,2, Xinyu Liu 1,2, Yue Hu 1,2, Peng Cao 1,2,
PMCID: PMC12022100  PMID: 40274830

Abstract

Diabetic neuropathy (DN) is a prevalent and debilitating complication of diabetes mellitus, significantly impacting patient quality of life and contributing to morbidity and mortality. Affecting approximately 50% of patients with diabetes, DN is predominantly characterized by distal symmetric polyneuropathy, leading to sensory loss, pain, and motor dysfunction, often resulting in diabetic foot ulcers and lower-limb amputations. The pathogenesis of DN is multifaceted, involving hyperglycemia, dyslipidemia, oxidative stress, mitochondrial dysfunction, and inflammation, which collectively damage peripheral nerves. Despite extensive research, disease-modifying treatments remain elusive, with current management primarily focusing on symptom control. This review explores the complex mechanisms underlying DN and highlights recent advances in diagnostic and therapeutic strategies. Emerging insights into the molecular and cellular pathways have unveiled potential targets for intervention, including neuroprotective agents, gene and stem cell therapies, and innovative pharmacological approaches. Additionally, novel diagnostic tools, such as corneal confocal microscopy and biomarker-based tests, have improved early detection and intervention. Lifestyle modifications and multidisciplinary care strategies can enhance patient outcomes. While significant progress has been made, further research is required to develop therapies that can effectively halt or reverse disease progression, ultimately improving the lives of individuals with DN. This review provides a comprehensive overview of current understanding and future directions in DN research and management.

Subject terms: Neurodevelopmental disorders, Diseases of the nervous system

Introduction

Diabetic neuropathy (DN) is among the most prevalent chronic complications of types 1 and 2 diabetes mellitus (T1DM and T2DM), significantly compromising patients’ quality of life and contributing to elevated morbidity and mortality rates.1,2 The number of patients with diabetes globally is expected to reach 783 million by 2045.3 The diabetes population could reach 1.31 billion by 2050.4,5 Diabetic peripheral neuropathy (DPN), the most common form of DN, is characterized by distal symmetric polyneuropathy, leading to sensory loss, pain, and motor dysfunction in the extremities and is a primary cause of foot ulcers and lower-limb amputations in diabetes patients.1,4,6 DPN imposes significant economic burdens on healthcare systems.7,8 With the rising global prevalence of diabetes driven by aging populations and lifestyle changes, the impact of DN is expected to escalate.9 Despite its severity, DN has received comparatively less attention in clinical research and therapeutic development than other diabetic complications, such as nephropathy and retinopathy, necessitating further research.6,1012

The pathogenesis of DN is multifactorial and remains poorly understood.1,4 Hyperglycemia, dyslipidemia, and insulin resistance activate various metabolic pathways, resulting in oxidative stress, mitochondrial dysfunction, inflammation, and microvascular damage, culminating in neuronal injury, Schwann cell damage, and myelin sheath degeneration.1315 Additionally, impaired neurotrophic support and autoimmune mechanisms may further exacerbate neuronal degeneration.16 Despite extensive research, effective treatments capable of halting or reversing DN’s progression remain elusive. Current management strategies primarily focus on alleviating symptoms and controlling pain.11 Antidepressants, anticonvulsants, and opioids have shown limited efficacy and are associated with various side effects, such as dizziness and nausea.17,18 Furthermore, no novel therapies have received approval in recent years.4,11

Recent research has shifted toward elucidating the molecular pathways involved in DN to identify potential therapeutic targets.6,11 Areas of interest include oxidative stress, chronic low-grade inflammation, mitochondrial dysfunction, and central sensitization mechanisms that contribute to neuropathic pain. Early diagnostic markers and advanced imaging techniques, such as corneal confocal microscopy and skin biopsy, enable timely intervention.1,11,19 Novel pharmacological agents, including sodium-glucose cotransporter 2 (SGLT2) inhibitors, glucagon-like peptide-1 (GLP-1) receptor agonists, and ion channel blockers, have shown promising neuroprotective and pain management effects.11,20,21 Additionally, non-pharmacological approaches, including lifestyle modifications, are being explored to enhance patient outcomes and quality of life.22,23

The profound impact of DN on individuals and healthcare systems, combined with the limitations of current treatment options, necessitates further research.6,15 This review summarizes recent advances in the pathophysiology of DN, discusses current diagnostic and therapeutic strategies, and highlights potential future directions in managing this debilitating condition.24

Epidemiology of DN

Global prevalence and trends

Current estimates suggest that approximately 50% of diabetes patients will develop some form of neuropathy.4 However, the reported prevalence of DN varies significantly across studies, ranging from 6% to over 60%.4,25 The prevalence of DPN is significantly higher in developing countries, often exceeding 50% among diabetes patients.26 In a comprehensive study conducted in China, the prevalence of DN among diabetes patients was 37%.27 The prevalence of DN is also influenced by age, sex, and ethnicity, with males exhibiting higher rates than females.5 Prediabetes is a significant risk factor for DN, with the prevalence of DPN among prediabetes individuals ranging from 6 to 25%.11 Compared with those with T1DM, patients with T2DM may present with DN at diagnosis or during the prediabetic state.28 In the MONICA/KORA Augsburg surveys, the prevalence of peripheral neuropathy increased from 7.4% in individuals with normal glucose tolerance to 28.0% in those with T2DM as glucose tolerance worsened.28 Additionally, the method used to diagnose DN affects the reported prevalence. For example, when using questionnaires like MNSIQ, the prevalence of DN in T1DM patients is estimated at 11–13%;29,30 however, when structured clinical examinations and vibration perception thresholds are used, the prevalence increases to 28%.31,32 With more sensitive tests, the prevalence exceeds 50%.33,34 The number of young individuals diagnosed with T2DM has been increasing.35 Geographic and cultural factors also influence the prevalence of DN.36 Recent studies have adopted widely recognized definitions of neuropathic pain.37 Most reports estimate the prevalence of painful DPN (PDPN) among diabetes patients to be between 13 and 35%.38,39 In the MONICA/KORA study, the overall prevalence of neuropathic pain was 13.3% among diabetes patients, 8.7% in those with impaired glucose tolerance, 4.2% in those with impaired fasting glucose, and 1.2% in individuals with normal glucose tolerance.40

The increase in diabetes prevalence likely increases the incidence of DN. DN imposes substantial costs due to increased healthcare utilization, lost productivity, and decreased quality of life.41 Despite its clinical significance, epidemiological research concerning DN is less extensive than other diabetic complications.42 Addressing the gaps in DN epidemiology requires coordinated efforts to improve research methodologies and promote international collaboration to develop effective prevention and treatment strategies.1

Risk factors

The development and progression of DN are influenced by various factors, including the duration of diabetes, glycemic control, age, obesity, dyslipidemia, insulin resistance, chronic low-grade inflammation, lifestyle choices, cardiovascular health, and genetic predispositions.5,11,43 Prolonged diabetes leads to cumulative nerve damage.44,45 Elevated glycated hemoglobin (HbA1c) levels are associated with increased neuropathy risk.13,46 Intensive glycemic control significantly reduces the incidence and progression of neuropathy in T1DM and T2DM patients.47,48 Age-related degenerative changes make older adults more susceptible to nerve damage, with DN prevalence significantly increasing in individuals over 50 years old.1,49

Obesity and dyslipidemia are major metabolic risk factors; a higher body mass index (BMI) and dyslipidemia promote insulin resistance and metabolic dysfunction, contributing to neuropathy development.1,5052 Insulin resistance, even without overt hyperglycemia, independently increases DN risk by causing peripheral nerve damage.53,54 Chronic low-grade systemic inflammation elevates levels of C-reactive protein (CRP) and proinflammatory cytokines, leading to endothelial dysfunction and nerve injury.55 Lifestyle factors such as smoking, excessive alcohol consumption, and physical inactivity are linked to DN development, exacerbating oxidative stress and vascular dysfunction.22,56,57 Cardiovascular diseases and related risk factors (e.g., hypertension, atherosclerosis) impair blood flow to peripheral nerves, worsening neuropathic symptoms.33,58 Genome-wide association studies (GWAS) have identified genetic variants related to metabolic regulation, nerve repair, and vascular function.59,60 Specific genes implicated include vascular endothelial growth factor (VEGF), neurotrophic receptor tyrosine kinase 1, sodium voltage-gated channel alpha subunit 2 (SCN2A), angiotensin-converting enzyme, and methylenetetrahydrofolate reductase, with polymorphisms associated with increased DN susceptibility.6164 Additionally, emerging evidence suggests that virus infections, such as COVID-19, exacerbate neuropathies in diabetes patients through inflammatory and immune-mediated mechanisms.6567

Although the risk factors for PDPN are poorly understood, female sex, longer diabetes duration, increased neuropathy severity, renal dysfunction, higher HbA1c levels, and higher BMI have been suggested as contributing factors.6874 Genetic factors have also been suggested; gain-of-function mutations in voltage-gated sodium channel genes increase neuronal excitability and are associated with PDPN.75,76 GWAS have identified single nucleotide polymorphisms near genes GFRA2, HMGB1P46, and ZSCAN20-TLR12P in patients with PDPN (Fig. 1).77,78 Comprehensive research on the risk factors for DN is limited, particularly concerning the genetic and viral infection influences on PDPN. Future studies should explore these complex factors to develop more effective prevention and treatment strategies for better disease management goals.

Fig. 1.

Fig. 1

Risk factors for diabetic neuropathy. The development and progression of DN are influenced by multiple factors, including diabetes duration, poor glycemic control, advanced age, and MetS, which encompasses obesity, hypertension, and dyslipidemia. Other contributing factors include chronic low-grade inflammation, lifestyle choices such as smoking and alcohol abuse, and genetic predisposition. Risk factors for painful diabetic neuropathy are less well-defined, but overlap with those for diabetic nephropathy. These include poor blood glucose control, susceptibility genes, and independent factors such as female sex, increased neuropathy severity, renal dysfunction, and higher BMI. BMI body mass index, DN diabetic neuropathy, MetS metabolic syndrome

Classification of DN

DN encompasses a heterogeneous group of disorders affecting various components of the peripheral nervous system (PNS) in patients with diabetes.1,4,11,41,79 Classification is essential for understanding the diverse manifestations and guiding appropriate management strategies (Fig. 2).

Fig. 2.

Fig. 2

Classification of diabetic neuropathy. Diabetic neuropathies comprise a diverse group of clinical syndromes, typically classified by the pattern of neurological involvement. Distal symmetric polyneuropathy (DSPN) is the most common form, which affects approximately 50% of patients with diabetes, characterized by sensory loss, pain, and motor dysfunction in the extremities. Autonomic neuropathy, affecting 20–40% of patients with diabetes, primarily involves the autonomic nervous system, impacting organs such as the heart, gastrointestinal tract, and urogenital system. Mononeuropathy, with an incidence of < 1%, usually involves isolated cranial nerves (e.g., oculomotor, facial) or peripheral nerves (e.g., facial). Radiculoplexus neuropathy, also occurring in <1% of diabetes cases, predominantly affects the lumbar or cervical plexus. DSPN distal symmetric polyneuropathy, DM Diabetes mellitus

DPN

Diffuse neuropathies are the most common forms of DN, primarily represented by distal symmetric polyneuropathy (DSPN), also known as DPN.4 DPN affects approximately 50% of individuals with diabetes during their lifetime and can be classified based on the type of nerve fibers affected.4,44

Small fiber neuropathy (SFN)

SFN primarily affects small unmyelinated C fibers and thinly myelinated Aδ fibers, leading to disturbances in pain and temperature sensation.80,81 The overall prevalence of SFN in diabetes patients ranges from 20 to 30%, depending on the diagnostic criteria and population studied.1,4 Painful DN, often associated with SFN, occurs in approximately 25% of diabetes patients and significantly impacts the quality of life due to chronic pain and sensory disturbances.1,4 Early symptoms often include painful sensations, such as burning pain, electric shock-like sensations, sharp pains, and hyperalgesia.82,83 Patients may also experience paresthesia and allodynia, where non-painful stimuli are perceived as painful.84,85 Recent studies highlight the importance of SFN in DN and its potential role as an early indicator of neuropathic complications.1,86

Large fiber neuropathy

Large fiber neuropathy affects larger myelinated Aβ fibers, resulting in sensory and motor deficits.1 The prevalence of large fiber neuropathy in diabetes patients is 40–60%.87,88 Sensory deficits include reduced proprioception and vibration sense, leading to numbness and balance difficulties.1,89 Motor deficits manifest as muscle weakness and atrophy in severe cases, as well as loss of deep tendon reflexes, particularly the ankle reflex.90,91 These impairments increase the risk of falls and foot ulcers due to unnoticed injuries.92,93 Advances in neurophysiological assessments, such as nerve conduction studies (NCS), have improved the detection of large fiber involvement in DN.89,94

Mixed fiber neuropathy

In many patients, small and large fibers are affected, leading to a combination of sensory deficits and neuropathic pain.1,72 The prevalence of mixed fiber neuropathy is substantial, with up to 50%.26,95 Clinical manifestations include symmetrical sensory loss typically initiated in the toes and progresses proximally in a “stocking-glove” distribution.1 Neuropathic pain is present in a significant subset of patients.83 Involvement of autonomic fibers can lead to sweating abnormalities, orthostatic hypotension, and gastrointestinal disturbances.1 Early detection and comprehensive management are crucial to prevent complications.26,95

Diagnosis and screening recommendations

Early detection of DPN is crucial to prevent foot ulcers and amputations, among other complications.5,96 Effective diagnosis and screening involve a combination of clinical assessments, sensory testing methods, electrophysiological studies, and advanced diagnostic methods tailored to the specific neuropathy subtype.80,97101 A detailed history taking identifies symptoms related to sensory, motor, and autonomic dysfunction, while a physical examination focuses on sensory and motor deficits, and reflex abnormalities.44,102 Sensory testing methods include the 10 g monofilament test for pressure perception,103 the 128 Hz tuning fork for vibration sense,35 pinprick and temperature sensation tests for small fiber function,104 and loss or reduction of ankle reflexes indicating large fiber involvement.91 NCS is recommended for atypical presentations or to exclude other causes of neuropathy.79,105 Advanced diagnostic methods involve skin biopsy for intraepidermal nerve fiber density (IENFD),87,104 quantitative sensory testing (QST) for temperature and pain thresholds, corneal confocal microscopy for early small fiber damage,19,106 and MRN for detailed imaging of nerve structures.107

Screening guidelines and components

For T2DM, screening for DPN should begin at diagnosis and occur annually;79,108 conversely, for T1DM, screening should start five years after diagnosis and continue annually;79,108 individuals with prediabetes exhibiting neuropathic symptoms should be screened;109 high-risk patients should be prioritized for screening.110,111 Screening components include routine foot examinations for ulcers and abnormalities, patient education on foot care and early signs of neuropathy, and management of modifiable risk factors such as hypertension, dyslipidemia, and smoking. Regular screening and education are essential components of comprehensive care.110,111

Diabetic autonomic neuropathy (DAN)

DAN is a serious and prevalent complication of diabetes originating from damage to the autonomic nervous system (ANS), which governs involuntary bodily functions. The overall prevalence of DAN varies, ranging from 20 to 40% among diabetes patients.112 DAN encompasses multiple subtypes, including cardiovascular autonomic neuropathy (CAN), gastrointestinal autonomic neuropathy, genitourinary autonomic neuropathy, sudomotor dysfunction, hypoglycemia unawareness, and pupillary dysfunction, each affecting different organ systems and presenting a spectrum of clinical manifestations.

CAN

CAN is one of the most clinically significant forms of DAN due to its strong association with increased morbidity and mortality. The prevalence of CAN among diabetes patients ranges from 20 to 60%.113,114 Early stages of CAN may be asymptomatic and detectable only through reduced heart rate variability (HRV) during deep breathing.115 As CAN progresses, patients may experience resting tachycardia, exercise intolerance, orthostatic hypotension, and silent myocardial ischemia.116

Diagnostic approaches for CAN include HRV tests such as the deep breathing test, valsalva maneuver, and orthostatic testing, along with ambulatory blood pressure monitoring to identify irregular circadian blood pressure patterns.117120 Recent advancements have introduced novel biomarkers and advanced imaging techniques to enhance early detection.114,121

Gastrointestinal autonomic neuropathy

Gastrointestinal autonomic neuropathy presents with various symptoms, such as esophageal dysmotility, gastroparesis, and enteropathy, including dysphagia, gastroesophageal reflux, delayed gastric emptying, diarrhea, constipation, and fecal incontinence.122124 The prevalence of gastrointestinal autonomic neuropathy in diabetes patients is estimated to be between 30 and 50%.124,125

Key diagnostic approaches include gastric emptying scintigraphy, the 13C-octanoic acid breath test, wireless motility capsule for gastroparesis, and esophageal manometry with pH monitoring for esophageal dysmotility.126130 Recent innovations, such as Gastric Per-Oral Endoscopic Myotomy (G-POEM) for refractory gastroparesis and advancements in enteric nervous system research, aim for targeted therapies.125,131,132

Genitourinary autonomic neuropathy

Genitourinary autonomic neuropathy involves bladder dysfunction, such as decreased bladder sensation, urinary retention, recurrent urinary tract infections, and incontinence, alongside sexual dysfunction.133137 The prevalence of genitourinary autonomic neuropathy is up to 40% in diabetes patients, with erectile dysfunction (ED) affecting up to 75% of diabetic males due to vascular and neuropathic factors.135,136 In females, symptoms include decreased libido, vaginal dryness, dyspareunia, and reduced sexual arousal, with prevalence of 30–50%.138

Diagnostic approaches encompass post-void residual volume measurement, urodynamic studies, comprehensive medical and sexual history assessments, nocturnal penile tumescence, penile Doppler ultrasonography for males, and detailed sexual function histories and hormonal evaluations for females.139141 Recent advancements have aided in restoring autonomic nerve function.142,143

Sudomotor dysfunction

Sudomotor dysfunction is characterized by anhidrosis or hyperhidrosis, leading to dry skin or compensatory excessive sweating, respectively, which can increase infection risks.144 The prevalence of sudomotor dysfunction in diabetes patients is approximately 30%.145

Diagnostic approaches include the Quantitative Sudomotor Axon Reflex Test, Electrochemical Skin Conductance (ESC) using devices like Sudoscan, and the Neuropad Test.146148 Recent developments feature Sudoscan as a reliable early detection method and advanced skin biopsy techniques to assess IENFD.145,149,150

Hypoglycemia unawareness

Hypoglycemia unawareness impairs counterregulatory hormonal responses during hypoglycemia, which reduces symptom awareness and heightens the risk of severe hypoglycemic episodes.151,152 The prevalence of hypoglycemia unawareness among individuals with diabetes on intensive insulin therapy can be as high as 30–40%.114,152 Clinically, patients may not recognize hypoglycemia until neuroglycopenic symptoms manifest, such as confusion or seizures.153

Diagnostic approaches focus on identifying impaired counterregulatory responses and may include continuous glucose monitoring systems to detect unrecognized hypoglycemic events.154 Recent advancements involve Hybrid Closed-Loop Systems for automated insulin delivery and structured education programs for restoring hypoglycemia awareness.114,155,156

Pupillary dysfunction

Pupillary dysfunction manifests as miosis, reduced light reflex, and difficulties in adapting to lighting changes.157,158 The prevalence of pupillary dysfunction in diabetes patients is around 25%.125,157 Diagnostic approaches include pupillometry for objectively measuring pupil size and reactivity, pharmacological testing to assess responses to agents like pilocarpine, and dynamic pupillometry as a non-invasive biomarker for autonomic dysfunction.125,157,159 Recent progress encompasses the application of Artificial Intelligence (AI) algorithms to analyze pupillary responses for early detection.125,157,159

Recent studies on DAN have advanced diagnostic techniques and subtype classification; however, the comprehension of its pathophysiological mechanisms and effective treatment strategies remains inadequate. It is necessary to strengthen studies on various types of autonomic neuropathy, develop more precise diagnostic tools, and explore personalized treatment plans to improve patients’ quality of life.

Mononeuropathy

Mononeuropathy refers to the damage of a single or a group of nerves, resulting in localized symptoms such as pain, weakness, or sensory loss in the affected area. Diabetes patients are more susceptible to mononeuropathies compared to non-diabetes individuals.24,41 Although mononeuropathies are less common than other forms of DN, with a prevalence estimated to be below 1% among diabetes patients,44,160 and are more frequently observed in older individuals and those with a longer duration of diabetes.42 Commonly affected nerves include the cranial nerves.161 The facial nerve (cranial nerve VII) may present with facial paralysis or weakness, and less commonly, the trochlear (CN IV), trigeminal (CN V), and abducens (CN VI) nerves can be involved.41,162 Peripheral nerves commonly affected include the median nerve, the ulnar nerve, the radial nerve, and the common peroneal (fibular) nerve.163165 Multiple mononeuropathy, or mononeuritis multiplex, involves two or more non-contiguous nerve trunks and presents with asymmetric, stepwise progression of symptoms, necessitating differentiation from more symmetrical polyneuropathy.166,167 The pathophysiology of diabetic mononeuropathy includes ischemic mechanisms, such as the occlusion of the vasa nervorum leading to nerve infarction, metabolic factors, including hyperglycemia-induced oxidative stress, and the formation of advanced glycation end products, contributing to nerve damage.13,168

Diagnosis involves a comprehensive clinical evaluation, including detailed neurological examination, electrophysiological studies like NCS and electromyography (EMG) to assess motor and sensory fiber function and localize lesions, as well as imaging studies such as magnetic resonance imaging (MRI) to exclude compressive or structural causes.41,169,170 Differential diagnoses include entrapment neuropathies, infectious neuropathies, and inflammatory conditions.167,171,172 Management strategies focus on tight glycemic control to prevent further nerve damage, symptomatic treatment with analgesics for pain management, physical therapy to maintain muscle strength and prevent contractures, and surgical interventions for entrapment neuropathies when necessary.17,173,174 The prognosis for diabetic mononeuropathy is generally favorable, which can significantly improve patient outcomes.175 Recent advancements include the development of neuroprotective agents114,176 with potentially profound implications for patient care. These agents may help to delay or prevent nerve damage progression, potentially improving quality of life by reducing pain and preserving motor functions in affected patients. Integrating these neuroprotective treatments into standard care protocols could reduce the frequency of interventions, such as surgeries or advanced physical therapy, thus improving the efficacy and efficiency of patient care in clinical settings.

Radiculoplexus neuropathy

Radiculoplexus neuropathy is a rare disabling complication of diabetes mellitus characterized by damage to nerve roots and plexuses, most commonly affecting the lumbosacral plexus and, less frequently, the cervical plexus.24,41 Although DRPN has an estimated prevalence of < 1% among diabetes populations,177179 it predominantly occurs in patients with T2DM, especially in middle-aged and older adults.178 Clinically, DRPN presents with a sudden or subacute onset of unilateral, severe pain in the hip, thigh, or buttock, often preceding motor weakness by weeks.180,181 Motor symptoms include progressive weakness and atrophy of proximal lower-limb muscles, leading to difficulty rising from a seated position or climbing stairs, accompanied by muscle fasciculations and hyporeflexia.182,183 Systemic features, such as significant weight loss, are reported in up to 60% of patients.184,185

The pathophysiology of DRPN involves ischemic injury due to microvasculitis leading to nerve fiber ischemia and immune-mediated mechanisms, as evidenced by inflammatory infiltrates in nerve biopsies.15,186 Diagnosis is primarily based on clinical evaluation supported by electrophysiological studies.187,188 Laboratory tests may reveal elevated inflammatory markers, including erythrocyte sedimentation rate or CRP, and nerve biopsy, though rarely performed, can demonstrate microvasculitis and perivascular inflammatory infiltrates.189

Management of DRPN focuses on optimizing glycemic control to prevent further nerve damage.17,173,184,190 The prognosis for DRPN is generally favorable, with many patients experiencing significant recovery over months to years, although some may have residual weakness, particularly with early diagnosis and appropriate management.191 There have been several advancements in DRPN research, including exploring neuroprotective agents.192,193 Current research on DRPN is limited due to its rarity; therefore, data on its pathophysiological mechanisms are scarce. Diagnostic and treatment methods warrant further optimization. Research on this condition remains necessary to develop more effective diagnostic tools and treatment strategies to improve patient outcomes.

Diabetic myelopathy is a rare condition characterized by spinal cord dysfunction in diabetes patients without other identifiable causes, presenting with progressive spastic paraparesis, sensory disturbances below the lesion level, and potential bladder and bowel dysfunction.194 The pathophysiology is thought to involve ischemia due to microangiopathy and metabolic disturbances.195 Diagnosis typically relies on an MRI of the spine, which may reveal spinal cord atrophy or signal changes and the exclusion of compressive lesions.114 Evoked potentials may also be utilized to detect delayed conduction times.196 Management primarily focuses on optimal glycemic control and symptomatic treatment.5

Additionally, central nervous system (CNS) complications are more prevalent in individuals with diabetes, with up to 50% experiencing some degree of cognitive decline.197,198 Cognitive impairment in diabetes is attributed to chronic hyperglycemia leading to cerebrovascular disease, neurodegeneration, and inflammation.196,199 Management strategies include intensive glycemic control to potentially slow cognitive decline, as well as lifestyle interventions to support cognitive health.200,201 Furthermore, mood disorders are significantly more common in diabetes patients, adversely affecting glycemic control and increasing the risk of complications.202 Management of these mood disorders involves psychological interventions such as cognitive behavioral therapy (CBT) and pharmacotherapy with antidepressants, considering their metabolic effects.173,203 Understanding and addressing spinal and CNS involvement in DN is crucial for comprehensive patient care.5

Pathophysiology of DN

DN is a multifaceted neurodegenerative disorder resulting from the interplay of metabolic, vascular, and neurodegenerative processes primarily driven by chronic hyperglycemia. Chronic high blood sugar levels initiate a cascade of metabolic abnormalities, which ultimately induce oxidative stress and inflammation, exacerbating nerve injury through excessive production of reactive oxygen species (ROS) and promoting neuronal apoptosis.4,11,15 Inflammatory responses mediated by proinflammatory cytokines and pathways, such as nuclear factor kappa B (NF-κB) and Toll-like receptor 4 (TLR4), worsen the condition by amplifying oxidative stress and hindering neuronal repair.13,53,204206 Disturbances in lipid metabolism contribute to neuronal apoptosis, while insulin resistance and deficiencies in neurotrophic factors impede nerve regeneration.4,207,208 The resultant neuronal hyperexcitability and microglial activation lead to chronic neuropathic pain.206,209212

DN preferentially targets sensory and autonomic axons, with motor axons affected to a lesser extent. The condition is characterized by the “dying back” of distal sensory axons in a length-dependent manner, presenting as a “stocking and glove” pattern of sensory loss.4,44 Schwann cells are also targeted by chronic hyperglycemia, leading to demyelination features in severe cases.210,213 Damage to Schwann cells affects axonal support, including cytoskeletal properties and axon-Schwann cell interactions essential for intra-axonal mRNA translation.214217 Additionally, sensory neurons in the dorsal root ganglia (DRG) exhibit altered phenotypes, such as reduced neurofilament synthesis, endoplasmic reticulum stress, and changes in key plasticity molecules, such as growth-associated protein 43 (GAP43) and heat shock proteins, leading to mitochondrial dysfunction and loss of peripheral nerve function.13,15,218,219 Recent studies have demonstrated alterations in mRNA and microRNA expression in DRG neurons exposed to chronic diabetes, highlighting the upregulation of pathways involved in inflammation, bioenergetics, and lipid processing.33,220222 These insights facilitated studies on targeted therapies.33,206,211,222,223

DPN pathogenesis

DPN is a complex neurodegenerative disorder resulting from the interplay of hyperglycemia-induced metabolic pathways, oxidative stress, inflammation, and dyslipidemia. These metabolic disturbances activate several pathogenic pathways that contribute to neuronal dysfunction and death, ultimately manifesting as DPN.1,15

Hyperglycemia-induced metabolic pathways

Polyol pathway activation

In a hyperglycemic state, excess glucose is shunted into the polyol pathway, where aldose reductase catalyzes its reduction to sorbitol, which in turn is oxidized to fructose by sorbitol dehydrogenase. This diversion leads to the accumulation of sorbitol and fructose within cells, increasing intracellular osmolarity and causing osmotic stress.224,225 The consumption of NADPH in this pathway reduces its availability for regenerating glutathione, thereby increasing oxidative stress.13,225 The resultant oxidative damage impairs nerve conduction and leads to neuronal apoptosis. This excess pathway activity causes oxidative stress and Na+/K+-ATPase dysfunction. These effects result in edema, demyelination, and peripheral nerve necrosis, impairing nerve conduction.15,226,227

Recent therapeutic approaches focus on aldose reductase inhibitors (ARIs), such as epalrestat and ranirestat, which have shown promise in reducing sorbitol accumulation and ameliorating neuropathic symptoms in clinical trials.228,229 Genetic studies suggest that polymorphisms in the aldose reductase gene influence individual susceptibility to DPN,230 highlighting the potential for personalized therapies.

Hexosamine pathway

Excess glucose not metabolized via glycolysis can enter the hexosamine pathway, which leads to increased production of uridine diphosphate N-acetylglucosamine (UDP-GlcNAc), which is used for O-linked N-acetylglucosamine (O-GlcNAc) modification of proteins.231,232 These modifications alter gene expression and protein function, causing vascular dysfunction, inflammation, and oxidative stress, all of which contribute to the pathogenesis of DPN.224,233 Benfotiamine, a lipid-soluble thiamine derivative, has been shown to activate transketolase activity, thereby redirecting glycolytic intermediates away from the hexosamine pathway.234 Benfotiamine may improve neuropathic symptoms by reducing UDP-GlcNAc production; however, its long-term efficacy remains uncertain.224,235 Currently, no specific inhibitors of the hexosamine pathway have been clinically validated to slow DPN progression, underscoring the need for novel therapeutic agents.236

Advanced glycation end products (AGEs) formation

Chronic hyperglycemia promotes the non-enzymatic glycation of proteins, lipids, and nucleic acids, leading to the formation of AGEs.237,238 AGEs accumulate in peripheral nerves and interact with the receptor for AGEs (RAGE) on cell surfaces, activating intracellular signaling pathways involving NF-κB.239,240 This activation increases the expression of proinflammatory cytokines and adhesion molecules, contributing to oxidative stress, inflammation, and vascular dysfunction.238,241 Therapeutic strategies targeting AGEs include using AGE inhibitors, such as aminoguanidine, which prevents AGE formation.239,241 RAGE antagonists are being explored to block the downstream effects of AGE-RAGE interactions to reduce inflammation and oxidative stress in DPN.236,238

Protein kinase C pathway

Protein kinase C (PKC) is involved in intracellular signaling. PKC activation disrupts normal metabolism, leading to ATPase phosphorylation and metabolic disorders, including changes in VEGF and vasoconstriction, which affect pain perception.242 PKC activation enhances excitatory currents in the dorsal horn and trigeminal neurons, contributing to nociceptive hypersensitivity in DPN.243 PKC also affects guanosine’s regulatory effects on glial cells, reducing their viability and increasing oxidative damage.244 Berberine alleviates diabetes-induced pain by inhibiting transient receptor potential vanilloid 1 (TRPV1) and the PKC pathway, reducing inflammation (Fig. 3).245

Fig. 3.

Fig. 3

Pathogenesis of diabetic neuropathy. The cellular mechanisms underlying DN involve hyperglycemia, dyslipidemia, and altered insulin signaling. In diabetes mellitus, these factors drive excessive activation of the polyol, hexosamine, and PKC pathways, as well as receptors for RAGE activation. In T1DM, insulin deficiency leads to reduced insulin signaling, while in T2DM, insulin resistance results in impaired PI3K-AKT signaling. These disruptions, along with hyperlipidemia and dyslipidemia, either individually or in combination, contribute to pathological changes in neurons, Schwann cells, and vascular cells, leading to nerve dysfunction and neuropathy. Key pathological processes include DNA damage, ER stress, mitochondrial dysfunction, oxidative stress, neurodegeneration, loss of neurotrophic signaling, and microvascular dysfunction. These changes can also trigger inflammatory and immune-mediated neurotoxicity. The relevance of each pathway to neuropathy development varies based on the cell type, disease profile, and stage, as different cells exhibit varying susceptibility to metabolic dysfunction. AGE advanced glycation end-product, DAG diacylglycerol, ER endoplasmic reticulum, FFAs free fatty acids, GLUT glucose transporters, G-3-P glucosamine 3-phosphate, G-6-P glucosamine 6-phosphate, LDL low-density lipoprotein, LOX-1 oxidized LDL receptor-1, PKC protein kinase C, RAGE AGE-specific receptor, ROS reactive oxygen species, TLR4 Toll-like receptor 4, T1DM type 1 diabetes mellitus, T2DM type 2 diabetes mellitus, UDP-GlcNAc uridine diphosphate N-acetylglucosamine

Dyslipidemia

Dyslipidemia significantly contributes to the pathogenesis of DPN, independent of hyperglycemia. Elevated serum triglyceride levels correlate with the progression of DPN and a decline in myelinated fiber density.52,246 Rodent models fed high-fat diets develop peripheral neuropathy predominantly affecting small fibers, mirroring human DPN.247,248

Saturated fatty acids (SFAs) induce mitochondrial depolarization and reduce the number and velocity of motile mitochondria in DRG neurons.53 The detrimental effects are more pronounced with longer-chain SFAs, while monounsaturated fatty acids can mitigate these deficits by enhancing β-oxidation and ATP production.249,250 In vivo studies confirm mitochondrial dysfunction in high-fat diet mice, which develop neuropathy marked by reduced nerve conduction velocities and decreased IENFD.250 These mice exhibit impaired axonal mitochondrial membrane potential and bioenergetic function, indicating a lack of reserve capacity.249,251 Dyslipidemia also affects Schwann cells, leading to mitochondrial dysfunction, endoplasmic reticulum stress, and oxidative stress, ultimately causing cell death.252 Oxidized low-density lipoproteins (oxLDL) are injurious to DRG neurons and vascular endothelial cells, contributing to neuronal damage and microvascular dysfunction.253255 OxLDL interacts with various receptors, such as lectin-like oxidized LDL receptor-1 (LOX-1), TLR4, and RAGE, activating inflammatory pathways and increasing ROS accumulation.256,257 Gene expression profiling reveals altered lipid metabolism in nerves of patients with DPN and rodent models, suggesting that dysregulated lipid metabolism contributes to DPN.220,258,259

While the efficacy of statins in alleviating DPN symptoms remains unclear,260,261 emerging lipid-lowering therapies showed potential in improving nerve function in DPN models.262,263 Additionally, substituting dietary SFAs with monounsaturated fatty acids improves nerve function in HFD mice without significantly altering plasma metabolic profiles, suggesting that dietary fats influence neuropathy development.250 Therefore, targeting dyslipidemia and its downstream effects may offer promising therapeutic avenues for DPN management (Fig. 3).

Oxidative stress and mitochondrial dysfunction

Oxidative stress is a critical factor in the development of DPN.264,265 Chronic hyperglycemia elevates ROS production through multiple pathways, including enhanced glucose oxidation, activation of NADPH oxidases, and dysfunction of the mitochondrial electron transport chain.266,267 Excessive ROS levels lead to oxidative damage to DNA, proteins, and lipids, resulting in neuronal apoptosis and impaired nerve function.55,268

High glucose levels impair mitochondrial oxidative phosphorylation, decreasing ATP production and increasing ROS generation.4 The resulting energy deficit and accumulation of damaged mitochondria contribute to neuronal degeneration and loss of nerve fibers.269,270 An imbalance between mitochondrial biogenesis and fission leads to the formation of small, dysfunctional mitochondria, affecting both neurons and Schwann cells and exacerbating peripheral nerve damage.271,272 Additionally, hyperglycemia-induced oxidative and nitrosative stress activates poly(ADP-ribose) polymerase (PARP), a nuclear enzyme involved in DNA repair.273 Overactivation of PARP depletes NAD+, impairing glycolysis and ATP production, which can lead to neuronal death.274,275

Therapeutic strategies targeting oxidative stress and mitochondrial dysfunction include using antioxidants like alpha-lipoic acid (ALA), which improves neuropathic symptoms and nerve conduction velocities in patients with DPN.276,277 Mitochondria-targeted antioxidants are being investigated for their ability to protect mitochondrial function and reduce neuronal apoptosis.278,279 Lifestyle interventions, particularly aerobic exercise, have also demonstrated benefits in enhancing mitochondrial biogenesis and function, offering a non-pharmacological approach to managing DPN (Fig. 3).280,281

Inflammation

Inflammation is a pivotal contributor to the pathogenesis of DPN. Chronic hyperglycemia induces a proinflammatory state characterized by elevated levels of cytokines such as tumor necrosis factor-alpha (TNF-α), interleukin(IL)-1 beta, and IL-6.282 These cytokines sensitize nociceptive neurons, leading to neuropathic pain, and promote neuronal damage through demyelination and axonal degeneration.283,284 Hyperglycemia activates NF-κB signaling pathways, enhancing the expression of inflammatory genes and further amplifying the inflammatory response.285 Immune cells infiltrate peripheral nerves in diabetes patients, releasing additional inflammatory mediators that exacerbate nerve injury.286,287 PNS-resident macrophages surveil the endoneurial space under normal conditions.288 Upon injury, they are joined by infiltrating macrophages to initiate nerve repair.288,289 In diabetic conditions, these macrophages can transition to a proinflammatory phenotype, accumulate, and contribute to nerve damage.290292 Accumulating macrophages release cytokines and chemokines, including IL-1β, IL-6, chemokine ligand 2 (CCL2), and TNF-α, intensifying the inflammatory environment and aggravating nerve damage.293,294

Depleting macrophages or inhibiting their migration improves mechanical sensation and nerve function in animal models.295,296 Cytokine inhibitors and non-steroidal anti-inflammatory drugs are being explored for their potential to alleviate neuropathic pain.83,284 Immunomodulatory therapies targeting specific components of the immune response offer promising avenues for reducing nerve inflammation and promoting repair (Fig. 3).297,298

Microvascular contributions

Microvascular dysfunction plays a significant role in the pathogenesis of DPN. Diabetes-induced endothelial dysfunction impairs blood, causing vasoconstriction.5,299 This microvascular impairment reduces oxygen and nutrient delivery to peripheral nerves, leading to nerve ischemia and hypoxia.300,301 The hypoxic environment exacerbates oxidative stress, contributing to neuronal degeneration and loss of nerve fibers. Endoneurial microangiopathy, characterized by capillary abnormalities, occurs early in DPN and correlates with clinical, neurophysiological, and morphological abnormalities.302304 Patients with diabetes exhibit increased endoneurial capillary density compared with healthy individuals, suggesting a compensatory response to nerve ischemia.303 However, the thickening of the basement membrane correlates with nerve damage, indicating that these vascular changes are detrimental.302 Moreover, diabetic nerves demonstrate poor vasodilation of epineurial arterioles, impairing the regulation of endoneurial blood flow.305,306 This impaired vasodilatory response appears before decreases in nerve conduction velocity (NCV) and contributes to reduced endoneurial perfusion and ischemia. In experimental models, interventions to improve vascular health enhanced nerve perfusion and promoted nerve regeneration.307309

Endothelial dysfunction is well recognized in DPN.310 Systemic markers of endothelial dysfunction and vascular inflammation are associated with DPN.94,297 Additionally, hyperglycemia-induced activation of protein PKC affects the expression of VEGF, promotes vasoconstriction, and induces hypoxia.311 Diabetes also decreases mediators of blood vessel formation and angiopoietins, contributing to poor nerve health.312 Capillary dysfunction with reduced glucose and oxygen extraction has been hypothesized to contribute to endoneurial hypoxia.313 Endothelial cells are susceptible to excess nutrients through hyperglycemia- and dyslipidemia-induced oxidative stress, further contributing to nerve damage during diabetes.225,314 Administration of VEGF in diabetic models increased NCV and vasa nervorum density, indicating potential therapeutic benefits.309,315 Therapies targeting endothelial function are being explored to prevent or mitigate DPN progression (Fig. 3)316,317.

Impaired insulin signaling

Insulin signaling plays a crucial role in maintaining peripheral nerve health, and its impairment significantly contributes to the pathogenesis of DPN. Insulin receptor signaling stimulates the PI3K/Akt pathway in the PNS, directly related to axon growth, myelin formation, and neuronal survival.318 In T2DM, insulin resistance in metabolically active tissues, such as muscle, fat, and liver, leads to hyperglycemia and dyslipidemia.319,320 Elevated plasma glucose, HbA1c levels, and abnormal lipid profiles correlate with systemic insulin resistance.320 Peripheral nerves develop insulin resistance in T2DM.16,321,322 Both neurons and Schwann cells express insulin receptors (IRs), particularly in sensory neurons of DRG and at nodes of Ranvier.323,324 Insulin promotes neurite outgrowth, supports neuronal survival,323,325 and regulates PNS metabolism.16,326 In conditions of nutrient excess, as seen in T2DM, insulin resistance blunts insulin-mediated nutrient and energy responses, impairing nerve metabolism and bioenergetic failure.327,328 This disruption contributes to nerve injury by affecting mitochondrial function and axonal trafficking. This mechanism mirrors the distal-to-proximal pattern of nerve damage observed clinically in patients with DPN.329,330

Direct insulin signaling in neurons can reverse features of DN independent of glucose levels.331,332 Insulin administration near the nerve or skin enhances nerve regeneration and repairs diabetes-induced abnormalities in animal models.333,334 However, in patients with T2DM, correcting hyperglycemia with insulin therapy has a limited impact on DPN.101,335 This disparity may be attributed to the development of insulin resistance within neurons.321,336 Altered phosphorylation of insulin receptor substrate proteins, particularly IRS-1 and IRS-2, is implicated in neuronal insulin resistance,20,337 which impairs insulin’s neurotrophic effects, leading to reduced neurite outgrowth and increased vulnerability to injury.338,339

In contrast, patients with T1DM often benefit from insulin therapy regarding neuropathic outcomes, as they typically do not exhibit significant insulin resistance.340 Understanding the role of insulin signaling and insulin resistance in peripheral nerves is essential for developing targeted therapies for DPN. Recent therapeutic approaches focus on enhancing insulin signaling pathways or reducing insulin resistance within peripheral nerves to improve neuropathic symptoms (Fig. 3).251

Impaired neurotrophic support

In DPN, levels of neurotrophic factors are significantly reduced due to hyperglycemia-induced metabolic disturbances. The deficiency of neurotrophic support leads to neuronal atrophy, impaired axonal transport, and decreased nerve regeneration capacity.4,312 Dysfunctional retrograde transport of neurotrophins from distal axons to neuronal cell bodies contributes to neuronal degeneration. Exogenous administration of neurotrophic factors can promote nerve regeneration and improve functional outcomes in experimental models of DPN.341 For instance, treatment with nerve growth factor (NGF) or brain-derived neurotrophic factor (BDNF) enhances neurite outgrowth and supports neuronal survival. However, clinical translation has been challenging due to delivery issues and side effects.342 Vitamin B deficiencies, particularly of B12 and B6, can exacerbate neurological dysfunction in DPN.343 Supplements like Metanx®, which contains L-methylfolate, methylcobalamin, and pyridoxal-5′-phosphate, have improved symptoms by enhancing endothelial function and restoring neurotrophic support.264,344 Recent research has explored novel delivery methods to enhance the efficacy and safety of neurotrophic factor-based therapies.345,346 For example, gene therapy delivering NGF or its receptors has shown promise in enhancing nerve regeneration in diabetic models.341

Schwann cell injury

In DPN, hyperglycemia and oxidative stress lead to Schwann cell dysfunction and apoptosis, resulting in demyelination and impaired nerve conduction.347,348 High glucose levels inhibit the PI3K/Akt pathway in Schwann cells, leading to overexpression of DNA methyltransferases (DNMT1 and DNMT3a) and upregulation of thioredoxin-interacting protein (TXNIP). This results in decreased autophagy and increased apoptosis, contributing to Schwann cell dysfunction.349 Activation of the PI3K/Akt pathway promotes Schwann cell survival and differentiation and enhances myelin formation.350 Mitochondrial dysfunction in Schwann cells triggers alterations in lipid metabolism, leading to myelin sheath dysfunction and accumulation of toxic lipid intermediates, which cause axonal degeneration and neuropathy.351,352 Lipotoxicity in Schwann cells results in cell dysfunction and death through mechanisms involving endoplasmic reticulum stress, mitochondrial dysfunction, and increased oxidative stress.252 Therapeutic strategies to enhance Schwann cell survival and function include using antioxidants, neurotrophic factors, and agents that modulate Schwann cell signaling pathways.353,354 For instance, protecting Schwann cells from glucose-induced toxicity improves nerve function and promotes remyelination in experimental models.355 Schwann cells provide metabolic support to axons, and disruption of this coupling contributes to DPN.356,357 Targeting pathways that improve Schwann cell metabolism and support axonal energy requirements may offer new therapeutic avenues (Fig. 3).

CNS involvement

The CNS plays a significant role in the development and maintenance of DPN. Hyperglycemia-induced changes affect the spinal cord and brain regions involved in pain processin.358,359 Structural changes, such as spinal cord atrophy and reduced gray matter volume in the primary somatosensory cortex, have been observed in patients with DPN.359,360 Functional abnormalities also play a critical role in DPN, including altered thalamic activity and disrupted thalamocortical connectivity. These changes lead to central sensitization, where non-painful stimuli are perceived as painful (allodynia), and pain responses are amplified (hyperalgesia).361363 Proton magnetic resonance spectroscopy studies have shown a reduced N-acetylaspartate to creatine ratio in the thalamus and parietal white matter pathways of DPN patients, indicating neuronal dysfunction.364,365 PDPN is associated with increased thalamic vascularity and functional reorganization of the primary somatosensory cortex.362,366 Multimodal therapeutic approaches combining pharmacotherapy, CBTs, and neuromodulation techniques are being explored to alleviate neuropathic pain and improve patient outcomes.84,367 The involvement of the CNS opens new avenues for research and the development of novel therapeutic targets aimed at modulating central pain pathways in DPN (Fig. 4).

Fig. 4.

Fig. 4

Peripheral and central mechanisms in diabetic neuropathy. Various alterations in peripheral and central neurons contribute to the pathophysiology of DN. In neuronal perikarya, hyperglycemia exacerbates endoplasmic reticulum stress, mitochondrial damage, and oxidative stress, while microvascular changes promote infiltration of inflammatory cells and factors at the neuronal and axonal levels, increasing the expression of voltage-gated sodium channels like Nav1.8, leading to hyperexcitability. Hyperglycemia in nerve fibers disrupts Schwann cell autophagy, increases the release of extracellular vesicles containing microRNAs and lipotoxic species, and reduces nerve growth factor secretion, impairing Schwann-axon interactions and myelin repair. These changes accelerate DN progression and contribute to hyperexcitability in myelinated axons by reducing the expression of shaker-type potassium channels, leading to heightened responses to stimuli and ectopic neuronal activity. Peripheral nerve damage triggers nociceptor hypersensitivity through inflammation, altered transducer activity (e.g., TRPV1, TRPM8, and P2X3R), and ion channel expression changes in sodium, potassium, and calcium channels. Methylglyoxal-induced glycation of nociceptor terminal ion channels forms AGEs, resulting in channel hyperfunction and neuronal hyperexcitability, including increased expression of Nav1.8. In DN, genetic variants (e.g., Nav1.7 and Nav1.8), methylglyoxal-modified TRPA1, SUMO-modified TRPV1, and HCN2 overactivation due to elevated cAMP contribute to neuronal hyperexcitability. Central sensitization results from an imbalance between facilitatory and inhibitory modulation of pain signals in the spinal cord and brain. This involves ascending pathways such as the spinothalamic tract (pain perception), the spinoreticular tract, and pathways through the parabrachial nucleus to the hypothalamus and amygdala, which are associated with autonomic function and emotional responses such as fear and anxiety. Descending pathways can inhibit or facilitate nociceptive signal transmission at the spinal level. The gut microbiota also plays a key role in DN, with dysbiosis linked to inflammation, metabolic disturbances, and nerve damage due to increased intestinal permeability and endotoxin translocation. Beneficial metabolites such as SCFAs, including butyrate, support nerve regeneration and reduce neuroinflammation, offering new therapeutic opportunities for DN management. AGEs advanced glycation end products, cAMP cyclic adenosine monophosphate, CNS central nervous system, DN diabetic neuropathy, DPN diabetic peripheral neuropathy, EVs extracellular vesicles, HCN2 hyperpolarization-activated cyclic nucleotide-gated 2, P2X3R P2X receptor subtype 3, SC Schwann cell, SCFAs short-chain fatty acids, TRPV1 transient receptor potential vanilloid 1, TRPA1 transient receptor potential ankyrin 1, TRPM8 transient receptor potential melastatin 8

Cellular autophagy and cell death

In DPN, hyperglycemia impairs autophagy by inhibiting key autophagy-related proteins, resulting in the accumulation of dysfunctional mitochondria and protein aggregates.368,369 Enhancing autophagy can mitigate nerve damage in DPN. Rapamycin, which activates autophagy by inhibiting the mTOR pathway, has shown neuroprotective effects in experimental models.370,371 Ongoing research aims to identify safe and effective autophagy modulators as potential therapeutic strategies for DPN.372,373 The mTOR pathway plays a significant role in DPN development by affecting autophagy and apoptosis in Schwann cells, myelin formation, and lipid metabolism, thereby influencing myelin and axonal integrity.242 Astragaloside IV inhibits the PI3K/Akt/mTOR pathway by upregulating miR-155 expression, thereby preventing apoptosis and enhancing autophagy to alleviate myelin damage in Schwann cells and improve neurological function in DPN rat models.374 Similarly, Lycium barbarum polysaccharides exert a protective effect by inhibiting the mTOR/p70S6K pathway, promoting autophagy in Schwann cells.375 Therefore, the state of the mTOR pathway affects the balance between autophagy and apoptosis in Schwann cells.

Hyperglycemia-induced oxidative stress and inflammation activate intrinsic and extrinsic apoptotic pathways, leading to programmed cell death of neurons and Schwann cells.376,377 Necroptosis has been implicated in the pathogenesis of DN.378 Agents that inhibit apoptotic signaling, such as caspase and necroptosis inhibitors, are currently being explored for their potential neuroprotective effects. Excessive pyroptosis leads to rapid rupture of the plasma membrane and the excessive release of proinflammatory cytokines and chemokines, which results in neuroinflammation that triggers a cascade of secondary injuries culminating in neuronal death.379381 Glucagon-like peptide-1 receptor agonists (GLP-1RAs) may act directly on DRG neurons by inhibiting the activation of inflammatory vesicle signaling pathways.382,383 An in-depth understanding of the underlying mechanisms of cellular pyroptosis is essential to determine its role in the pathogenesis of DPN and develop effective therapeutic agents targeting the pyroptosis signaling pathway to alleviate neuropathic symptoms.

Genetic and epigenetic factors

Genetic predispositions significantly influence the risk of developing DPN. Variations in genes associated with inflammation, oxidative stress response, and neuronal function have been linked to increased susceptibility to neuropathy in diabetes patients.384,385 Additionally, GWAS has identified several novel genetic loci associated with DPN, underscoring the complex genetic architecture underlying this condition.78,386 Epigenetic modifications alter gene expression without changing the DNA sequence and can be influenced by environmental factors, including hyperglycemia.387,388 These epigenetic changes contribute to the activation of pathogenic pathways involved in DPN.389,390 Recent research has identified specific genetic and epigenetic markers for early detection and risk stratification and developed epigenetic therapies to reverse harmful modifications and restore normal gene expression.242,374 Therapeutic approaches targeting epigenetic regulators are being explored to mitigate the progression of DPN.389,390 Moreover, improvements in CRISPR-Cas9 technology present opportunities for accurate genetic and epigenetic alterations to rectify inherited genetic variants and epigenetic markers associated with DPN.391,392 Understanding the interplay between genetic and epigenetic factors is crucial for developing personalized medicine strategies to effectively prevent and treat DPN.

Non-coding RNAs

Non-coding RNAs, including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), are crucial regulators in the pathophysiology of DPN. These molecules modulate gene expression at transcriptional and post-transcriptional levels, influencing inflammation, oxidative stress, and neuronal repair.393,394 For instance, miR-146a regulates inflammatory responses by targeting IRAK1 and TRAF6, and its decreased expression contributes to increased inflammation in DPN.395 Similarly, lncRNAs can act as competing endogenous RNAs (ceRNAs), sequestering miRNAs and affecting their regulatory functions.396 Circulating non-coding RNAs are potential biomarkers for early diagnosis and monitoring of disease progression.397,398

Moreover, Schwann cell-derived extracellular vesicles (EVs) play a significant role in DPN pathogenesis. Hyperglycemia-treated Schwann cells release pathologic EVs that suppress axonal growth in vitro and accelerate DPN development in vivo in db/db T2D mice.399 These EVs contain elevated levels of miR-28, miR-31a, and miR-130a, which reduce the expression of target proteins.399 In contrast, homeostatic Schwann cell-derived EVs increase DRG neurite outgrowth in vitro and restore small and large fiber functions upon injection into db/db mice.400 The underlying neuroprotective mechanisms involve increased levels of miR-21, miR-27a, and miR-146a, along with decreased expression of target proteins.401,402 miR-21 in Schwann cell-derived EVs has beneficial effects on neurite outgrowth in vitro.401 These findings suggest that Schwann cells under impaired metabolic conditions release pathologic EVs that promote nerve degeneration and DPN, whereas homeostatic EVs may facilitate nerve repair.

Gut microbiota

The gut-brain axis is a pivotal area of interest in studying DPN. Dysbiosis is associated with systemic inflammation and metabolic dysfunction in diabetes, both of which can exacerbate DPN.403,404 Alterations in gut microbiota can increase intestinal permeability, leading to the translocation of endotoxins and the activation of immune responses that contribute to peripheral nerve damage.405 Modulating the gut microbiota through dietary interventions, probiotics, and fecal microbiota transplantation can positively influence neuronal health and alleviate neuropathic symptoms.406408 Microbial metabolites, such as short-chain fatty acids (SCFAs), play a significant role in neuroinflammation and nerve regeneration.409,410 This holds promise for developing novel therapeutic strategies targeting the gut microbiota to manage DPN effectively.388,389 Butyrate enhances the integrity of the blood-nerve barrier and reduces inflammatory cytokine production, protecting peripheral nerves from damage.411 Additionally, specific probiotic strains have been identified that can restore gut microbiota balance, reduce systemic inflammation, and improve nerve conduction in diabetic models.412,413 Fecal microbiota transplantation (FMT) has also been explored as a method to reset gut microbiota composition, demonstrating improvements in metabolic parameters and neuropathic pain in preclinical studies.414,415 Metabolites produced by gut bacteria influence host energy metabolism, immune responses, and neuronal function.416 Understanding these interactions is crucial for identifying biomarkers for early detection and for developing targeted microbiota-based therapies.297,417 Ongoing clinical trials are assessing the efficacy of microbiota modulation in patients with DPN to translate these findings into clinical application.418,419

Although research on the pathophysiological mechanisms of DN covers multiple aspects, several pathways remain poorly understood, and the clinical application of treatment strategies remains limited. Future should aim to investigate these complex mechanisms, develop precise therapeutic methods, and advance personalized medicine and emerging fields, such as gut microbiota research to enhance treatment outcomes.

Mechanisms of PDPN

Among patients with DN, 30–50% develop neuropathic pain.38 This pain commonly presents as spontaneous burning sensations in the feet, and patients may also report positive sensory symptoms such as brush-evoked allodynia (pain from normally non-painful stimuli) and paresthesias, which are often accompanied by sensory loss, leading patients to describe the paradox of their feet being continuously painful yet insensate to touch.420 Neuropathic pain arises due to persistent maladaptive structural and functional changes within the somatosensory system following peripheral nerve injury.421 Research, primarily using experimental models of mechanical nerve damage, has explored the mechanisms underlying neuropathic pain, revealing increased excitation and facilitation of pain signals, along with a loss of inhibition in both the PNS and the CNS.421 However, neurophysiological measurements, molecular pathways, and pathological findings do not fully explain the presence of neuropathic pain in diabetes.421423 Structural and functional changes within the somatosensory system, along with maladaptive responses in the CNS and PNS, contribute to the mechanisms underlying PDPN (Fig. 4).422,423

Peripheral mechanisms of PDPN

Peripheral mechanisms in PDPN involve multiple factors. Small fibers are preferentially impaired in PDPN. However, measurements of IENFD and basic QST have not conclusively demonstrated their preferential involvement in PDPN.422 Two large cross-sectional studies using detailed QST profiling found greater thermal insensitivity in painful than painless DPN, suggesting more advanced small fiber dysfunction.69,70 Small nerve fibers may become sensitized following increased distal terminal injury and regeneration in the presence of neurotrophins.87 Alternatively, hyperexcitable small fiber neurons in PDPN might be more susceptible to bioenergetic failure, increasing distal nerve fiber degeneration.424 Compared with painless DPN, PDPN is associated with more severe corneal small fiber damage425427 and increased markers of degeneration (axonal swelling) and regeneration.428,429 However, the reported findings are inconsistent, possibly due to selection bias and small study sizes.421,425,430

Studies have demonstrated greater cardiac autonomic dysfunction in PDPN than in painless DPN,421,427,431 while others found no association.432,433 Profound abnormalities in the arteriovenous anatomy have been described on the epineurial surface in patients with acute painful neuropathy during rapid glycemic control.434 Patients with PDPN have increased epineurial blood flow and intravascular oxygen saturation, perhaps secondary to arteriovenous shunting due to sympathetic denervation.84 The topical application of vasodilators provides pain relief in PDPN, suggesting a role for hypoxia and/or vascular dysfunction in the generation of neuropathic pain in DPN.435,436

Neuronal injury promotes a neuroinflammatory response, leading to the accumulation of immune cells and the release of inflammatory cytokines, contributing to neuronal hypersensitivity. In PDPN, these mechanisms facilitate ectopic neuronal activity and enhance response to sensory stimuli.69 Cross-sectional studies have identified inflammatory mediators associated with PDPN.437,438 Elevated serum levels of soluble intracellular adhesion molecule 1 (sICAM-1), indicative of increased vascular inflammation and/or endothelial dysfunction, have also been reported in PDPN.432,433 Microglia may contribute to neuropathic pain through impaired descending modulation, altering the balance between descending inhibition and excitation toward the latter.432 Minocycline, known to reduce microglial activation, reduces neuropathic pain in Sprague–Dawley rat models of T1DM with DPN.433

Methylglyoxal, a reactive dicarbonyl by-product of glycolysis, induces post-translational modifications of the voltage-gated sodium channel Nav1.8, increasing the excitability of nociceptive neurons.434 Despite showing promise as a biomarker of neuropathic pain, a cross-sectional study found no association between methylglyoxal and PDPN.428 Peripheral nociceptor terminals respond to noxious stimuli via an array of receptors and ion channels to initiate an action potential. The altered expression, modification, and function of these transducers as TRPV1, transient receptor potential ankyrin 1 (TRPA1), and transient receptor potential melastatin 8 (TRPM8), contribute to peripheral sensitization and may play an important role in pain phenotyping.11,84,439446 Painful neuropathies cause alterations in ion channels.435 Gain of function of sodium channels leads to increased neuronal excitability, signal transduction, and neurotransmitter release.436 Preclinical69,434 and human genetic studies69,437,438 in PDPN have shown evidence of enhanced function of voltage-gated sodium channels. Concurrently, there is a downregulation in the expression of components of voltage-gated potassium channels, which normally regulate neuronal excitability, repolarization, and frequency of action potentials.438 Voltage-gated calcium channels (VGCCs) are functionally diverse and involved in peripheral and central nociception.437 T-type (CaV3.2) calcium channels are especially significant in DN.447450 VGCCs, present in the presynaptic terminal, are responsible for neurotransmitter release and the propagation of pain signals to second-order neurons.438 Components of subtypes of these channels are upregulated in rodent DRG cells displaying neuropathic pain; agents targeting these channels provide analgesia.1,11 Ligand (ATP)-gated non-selective cation channels are found on neurons, Schwann cells, and microglia.451 In streptozotocin (STZ)-induced diabetic rat and mouse models, elevated expression of P2X2 and P2X3 receptors in the DRG, along with increased current density and P2X4 receptor expression on satellite glial cells, indicates that P2X4 receptor activation in the DRG significantly contributes to DPN-induced mechanical pain hypersensitivity (Fig. 4)452.

Dysfunction within the CNS

Brain imaging studies have demonstrated an association between PDPN and structural changes in key somatosensory regions, such as the thalamus and somatosensory cortex, and altered functional connectivity within ascending pain pathways and cortical regions involved in pain processing. Disruptions in the descending pain modulatory system can amplify or inhibit pain signals.366,453,454 The relationship between these structural and functional brain changes and metabolic factors remains poorly understood, with potential mechanisms including vascular injury, oxidative stress, osmotic imbalances, and glial and neuronal damage.455 Future research should elucidate how aberrant sensory afferent activity interacts with maladaptive CNS plasticity to initiate and sustain neuropathic pain.456

In the spinal cord, the influx of neuropathic signals to the dorsal horn leads to central sensitization, characterized by increased spontaneous neuronal activity, lowered activation thresholds for peripheral stimuli, heightened responses to sub-threshold stimuli, and expansion of receptive fields.457 Activation of N-methyl-D-aspartate (NMDA) receptors amplifies and prolongs spinal dorsal horn neuron responses, further contributing to pain perception.421,458 Alterations in the balance of descending modulatory pathways are hypothesized to sustain chronic pain states, with a reduction in inhibitory control and an increase in facilitatory activity within both the brain and spinal cord.421 Specifically, dysfunction within the ventrolateral periaqueductal gray (vlPAG)-mediated descending pain modulatory system has been reported in patients with PDPN.454 Moreover, centrally mediated factors, such as cognitive processes (e.g., attention, distraction, catastrophizing), contextual beliefs about pain, and emotional states (e.g., depression, anxiety), significantly influence the pain experience.421,459,460

The thalamus plays a pivotal role in modulating and processing somatosensory inputs before they reach the cerebral cortex and may serve as a central biomarker for neuropathic pain in diabetes mellitus.459 In rat models of T1DM, thalamic ventroposterolateral neurons exhibit hyperexcitability in response to stimuli.461 These neurons also demonstrate increased spontaneous activity independent of ascending afferent input and possess enlarged receptive fields, suggesting that thalamic neurons function as central generators or amplifiers of pain in DPN.462 Magnetic resonance perfusion imaging studies have identified increased thalamic vascularity in patients with PDPN during resting states, likely reflecting heightened neuronal activity.463,464 Additionally, decreased thalamocortical functional connectivity observed through resting-state functional MRI correlates with PDPN.465

The anterior cingulate cortex, essential for emotional pain processing, shows increased blood flow in patients with PDPN, a hyperperfusion state that normalizes following duloxetine treatment.438 Functional MRI studies have reported augmented blood oxygen level-dependent (BOLD) responses in limbic and striatal structures in response to heat pain in PDPN patients.437 Furthermore, structural and functional alterations in the primary SSC are associated with pain phenotypes in DPN.432,438 Notably, the analgesic response to intravenous lidocaine, a sodium channel blocker, in PDPN patients correlates with increased SSC volume and enhanced functional connectivity between insular and corticolimbic circuits.432 These findings highlight the necessity for advanced imaging studies to further clarify the relationship between CNS alterations and individualized pain phenotypes in PDPN (Fig. 4). Current research on the mechanisms underlying PDPN has identified multiple peripheral and central factors; however, the specific interactions between these mechanisms remain nebulous, and existing models cannot fully explain the presence of diabetic neuropathic pain. Future studies should focus on elucidating the complex interactions within the somatosensory system and developing targeted therapies based on advanced imaging and molecular insights.

DAN

DAN is a serious and common complication of diabetes mellitus that affects the ANS, which controls involuntary bodily functions. DAN can impact various organ systems, including the cardiovascular, gastrointestinal, genitourinary, and sudomotor systems, leading to significant morbidity and mortality among diabetes patients.

CAN

CAN is characterized by impaired autonomic control over cardiovascular function, resulting from damage to the sympathetic and parasympathetic fibers that innervate the heart and blood vessels. This imbalance between sympathetic and parasympathetic activity leads to resting tachycardia, reduced HRV, orthostatic hypotension, and an increased risk of silent myocardial ischemia and sudden cardiac death.5,466 Microvascular insufficiency leads to ischemia of autonomic nerves. Inflammation and autoimmune mechanisms also play significant roles in the pathogenesis of CAN.112,467 Recent studies have focused on the early detection of CAN using advanced diagnostic tools like spectral analysis of HRV and cardiac imaging techniques. There is growing interest in the role of inflammatory markers and genetic predispositions in the development of CAN.468,469 Moreover, therapeutic interventions targeting metabolic pathways, oxidative stress, and inflammation are being explored to prevent or slow the progression of CAN.42

Gastrointestinal autonomic neuropathy

Gastrointestinal autonomic neuropathy affects the enteric nervous system, leading to dysregulation of gastrointestinal motility and function. This dysregulation can manifest as esophageal dysmotility, gastroparesis (delayed gastric emptying), intestinal enteropathy, constipation, or diarrhea.470,471 Impaired vagal nerve function exacerbates gastrointestinal symptoms.472,473 Recent advancements in imaging and motility testing have enhanced the early diagnosis of GAN, utilizing tools such as high-resolution manometry and wireless motility capsules. Therapeutic strategies include prokinetic agents, dietary modifications, and interventions targeting the gut microbiota, aiming to restore normal gastrointestinal function and alleviate symptoms.474

Genitourinary dysfunction

Genitourinary dysfunction in DAN encompasses both bladder dysfunction (diabetic cystopathy) and sexual dysfunction. In males, ED is a prevalent complication, while females may experience sexual dysfunction, including decreased libido and arousal disorders.475,476 Chronic hyperglycemia-induced neuronal damage to the autonomic fibers innervating the bladder and genitalia leads to impaired neurotransmission.477 Additionally, vascular changes, hormonal imbalances, and psychological factors contribute to genitourinary dysfunction in diabetes.478 Addressing modifiable risk factors and providing psychosocial support enhance patient outcomes.479,480 These advancements offer promising avenues for managing genitourinary complications in DN.481 Emerging evidence indicates that the gut microbiota plays a significant role in the development of DAN. Dysbiosis, or an imbalance in gut microbial composition, can influence the ANS through metabolic, immune, and neural pathways. Microbial metabolites, such as SCFAs and lipopolysaccharides (LPS), may affect gut permeability, systemic inflammation, and neural function.482 Recent studies have demonstrated alterations in the gut microbiota of patients with diabetic CAN.483 Modulating the gut microbiota through probiotics, prebiotics, and dietary interventions has shown the potential to improve autonomic function and reduce inflammation.41 Understanding the gut-brain axis in the context of DAN opens new avenues for therapeutic interventions.

Current research on DAN has advanced diagnostic tools and treatment strategies, but the understanding of its complex mechanisms and the impact of gut microbiota remains insufficient. Future studies should delve deeper into these mechanisms, develop more effective diagnostic methods, and create personalized treatment plans to improve patient outcomes.

Focal and multifocal neuropathies

The primary pathophysiological mechanism underlying focal and multifocal neuropathies is ischemic injury resulting from the occlusion or compromise of the vasa nervorum—the small blood vessels supplying peripheral nerves. Diabetes-induced microvascular alterations, including the thickening of the capillary basement membrane and endothelial hyperplasia, diminish blood flow to nerves, leading to ischemia and subsequent nerve fiber damage.484 Recent research has highlighted the role of immune dysregulation in diabetic focal neuropathies, with nerve biopsies revealing inflammatory infiltrates that suggest potential benefits from immunosuppressive therapies.171,485 Advances in imaging techniques have enhanced the diagnosis and monitoring of these neuropathies, allowing for earlier and more accurate identification of affected nerves.486 Clinically, early recognition and differentiation of focal and multifocal neuropathies are crucial for effective management, which includes stringent glycemic control, pain management, and addressing underlying vascular or inflammatory processes. In cases where inflammation is prominent, immunosuppressive therapies may be warranted to prevent further nerve damage and improve patient outcomes.187 Ongoing research continues to explore the efficacy of novel therapeutic interventions and the optimization of existing treatments to better manage these complex neuropathic conditions.

Current therapies for DN

Targeted prevention and treatment of DN

DN is a common and serious complication of diabetes mellitus, leading to significant morbidity and a diminished quality of life. Despite its prevalence, there are currently no specific therapeutic strategies that can reverse diabetic nerve damage once it has occurred. Early signs of DN are often subtle and nonspecific, which can lead to delayed diagnosis and progression to irreversible stages of nerve damage.101 Therefore, proactive prevention and early intervention are crucial to mitigate the impact of this condition (Fig. 5).

Fig. 5.

Fig. 5

Management strategies for diabetic neuropathy. Maintaining good glycemic control helps prevent the development of neuropathy in patients with type 1 diabetes, though the effect is less pronounced in those with type 2 diabetes. Lifestyle modifications, such as a balanced diet and regular exercise, are recommended for all patients. Mechanism-based treatments focus on addressing the underlying pathophysiology of diabetic neuropathy to alleviate symptoms and slow disease progression. Current therapeutic strategies include neuroprotective agents (methylcobalamin), antioxidants (alpha-lipoic acid), aldose reductase inhibitors (epalrestat), microcirculation enhancers (prostaglandin E1), and metabolic enhancers (acetyl-L-carnitine). PDPN is a frequent and debilitating complication that severely impacts patients’ quality of life. First-line and second-line treatments for PDPN involve various drug classes, including anticonvulsants (pregabalin and gabapentin), serotonin-norepinephrine reuptake inhibitors (duloxetine and venlafaxine), sodium channel inhibitors (carbamazepine and oxcarbazepine), and tricyclic antidepressants (amitriptyline and nortriptyline). Opioids (tapentadol and tramadol) should be avoided due to their significant adverse effects and high potential for addiction. Non-pharmacological therapies, such as TENS, SCS, and acupuncture, are personalized based on individual patient needs. TENS transcutaneous electrical nerve stimulation, SCS spinal cord stimulation, PDPN painful diabetic peripheral neuropathy

Glycemic control

Intensive glycemic control is vital in reducing the incidence and progression of DN in patients with T1DM. Maintaining blood glucose levels as close to normal as possible significantly reduced the risk of developing DPN and DAN.487,488 Long-term follow-up studies from the Epidemiology of Diabetes Interventions and Complications (EDIC) research have confirmed these benefits.489 In T2DM, the role of glycemic control in preventing neuropathy is more complex. The Action to Control Cardiovascular Risk in Diabetes trial and the Veterans Affairs Diabetes Trial have shown that while intensive glycemic control may slow the progression of neuropathy, it does not significantly prevent its onset.490,491 The UK Prospective Diabetes Study indicated a modest reduction in neuropathy risk with improved glycemic control.492 These findings suggest that hyperglycemia is only one of multiple factors contributing to neuropathy in T2DM, highlighting the need for a multifactorial approach to management. While aiming for optimal glycemic targets, it is essential to balance the benefits of tight glucose control against the risks of hypoglycemia, especially in individuals with long-standing diabetes or comorbidities.493 Personalized glycemic targets should be established based on patient-specific factors, including duration of diabetes, age, comorbid conditions, and risk of hypoglycemia.494

Lifestyle modification

Lifestyle modification plays a pivotal role in the prevention and management of DN. Risk factors, such as obesity, hypertension, dyslipidemia, smoking, and physical inactivity, are associated with an increased incidence and severity of neuropathy.299,495 Adopting a healthy diet that emphasizes whole grains, fruits, vegetables, lean proteins, and healthy fats can improve glycemic control and reduce cardiovascular risk factors.496 Specific dietary patterns, such as the Mediterranean diet, have been associated with reduced inflammation and improved metabolic profiles in diabetes patients.497 Regular physical activity has been shown to improve insulin sensitivity, aid in weight management, and enhance cardiovascular health. Exercise may also have direct neuroprotective effects by promoting nerve regeneration and improving NCV.498,499 Smoking is an independent risk factor for DN and microvascular complications.500 Smoking cessation is strongly recommended to reduce the risk of neuropathy progression and improve overall health outcomes. Health education is vital for patients to make informed lifestyle choices. Structured education programs can improve self-management skills, adherence to treatment plans, and awareness of the importance of regular monitoring for early signs of neuropathy.501,502

Key recommendations:

  • T1DM patients: Early and intensive glycemic control is recommended to prevent or delay the onset and progression of DPN and DAN.

  • T2DM patients: Glycemic control is recommended to slow the progression of neuropathy; however, its effect on prevention is less pronounced than T1DM.

  • Individualized targets: Glycemic targets should be individualized to balance efficacy and safety, particularly in patients with advanced disease or multiple comorbidities.

  • Lifestyle interventions: Implement lifestyle modifications, including a healthy diet, regular physical activity, and smoking cessation, to reduce the risk of neuropathy in patients with prediabetes, metabolic syndrome, and T2DM.

  • Patient-centered approach: Provide individualized education and support to encourage adherence to lifestyle changes and self-care behaviors.

Mechanism-based treatments

Mechanism-based treatments aim to target the underlying pathophysiological processes of DN to alleviate symptoms and prevent disease progression. Current therapies encompass various strategies, including neuroprotective agents, antioxidants, ARIs, microcirculation enhancers, metabolic enhancers, and certain traditional herbal medicines (Fig. 5).

Neuroprotective agents

Methylcobalamin, an active form of vitamin B12, plays a crucial role in nerve regeneration and myelin sheath formation and promotes the synthesis of nucleic acids and proteins within nerve cells, facilitating axonal regeneration and improving NCV. Recent clinical trials have demonstrated that methylcobalamin can significantly improve neuropathic symptoms and nerve function in patients with DPN.503,504

Antioxidants

ALA is a potent antioxidant that mitigates oxidative stress, a key contributor to DN, inhibits lipid peroxidation, improves endoneurial blood flow, and enhances nerve glucose uptake and energy metabolism. Clinical studies have shown that ALA significantly improves neuropathic symptoms and nerve conduction velocities.505507

ARIs

Epalrestat is an aldose reductase inhibitor that prevents the accumulation of sorbitol and fructose within nerve cells, which can cause osmotic stress and neuronal damage. By inhibiting this pathway, epalrestat improves metabolic imbalances associated with DN. Epalrestat also alleviates subjective symptoms and enhances NCV.197,508

Microcirculation enhancers

Prostaglandin E1 (PGE1) and its analogs act as vasodilators, improving microcirculation by relaxing the vascular smooth muscle and reducing platelet aggregation. PGE1 enhances symptoms and nerve conduction velocities in DPN patients, particularly when combined with neuroprotective agents, such as methylcobalamin.509,510 Additionally, Pentoxifylline improves microcirculation by decreasing blood viscosity and enhancing erythrocyte flexibility, which improves NCV and reduces neuropathic symptoms in DPN patients.309 Kallidinogenase, another microcirculation enhancer, increases capillary blood flow by generating bradykinin, leading to vasodilation and enhanced tissue perfusion. Clinical studies indicate positive effects on DPN symptoms and nerve function.511

Metabolic enhancers

Acetyl-L-carnitine (ALC) facilitates fatty acid transport into mitochondria, enhancing cellular energy production, and has neuroprotective properties that alleviate neuropathic pain, improve sensory function, and enhance nerve conduction.512,513 ALC significantly improved pain reduction and nerve fiber regeneration in DPN patients treated with ALC.512,513

Herbal medicines

Traditional Chinese medicine includes various herbal preparations being explored for their potential benefits in managing DN. Mudan Granules, containing herbs such as Salvia miltiorrhiza and Angelica sinensis, are believed to promote blood circulation and relieve pain. Mudan granules may have comparable efficacy to methylcobalamin in treating DPN, improving symptoms and nerve conduction velocities.514 Dan Shen dripping pills, derived from Salvia miltiorrhiza, improve microcirculation, inhibit platelet aggregation, and reduce oxidative stress. These pills can enhance symptoms and nerve conduction velocities in DPN patients, both alone and in combination with methylcobalamin.515,516

Current treatments for DN focus on glycemic control, lifestyle modifications, and mechanism-based therapies but not on reversing nerve damage. Some therapies have limited efficacy. In the future, it is necessary to develop more effective reversal therapies, optimize early diagnostic methods, and advance personalized treatment strategies to improve patient outcomes and quality of life.

Treatment of PDNP

Effective management of PDPN primarily relies on pharmacological interventions, supplemented by non-pharmacological therapies tailored to individual patient needs.

Pharmacological treatments

Recent guidelines recommend the following classes of medications for the treatment of PDPN (Fig. 5):

First-line medications

Anticonvulsants

  • Pregabalin: Demonstrates significant pain reduction and improves sleep and quality of life in several randomized controlled trials (RCTs).17,517 Pregabalin has linear pharmacokinetics, allowing for twice-daily dosing and a faster onset of action.

  • Gabapentin: Effective in reducing neuropathic pain symptoms but requires gradual titration and thrice-daily dosing due to its pharmacokinetic profile.17,518 However, gabapentin is less expensive, which is an important consideration for patients, especially those with high-deductible health plans in the United States.519

Serotonin-norepinephrine reuptake inhibitors (SNRIs)

  • Duloxetine: The most prescribed SNRI for PDPN and has regulatory approval for treating neuropathic pain.367,520 Duloxetine relieves pain by increasing the synaptic availability of serotonin and norepinephrine in descending pathways, enhancing endogenous pain modulation. Duloxetine significantly reduces pain scores and improves functional outcomes.521524 The most common adverse effects are nausea, dry mouth, dizziness, somnolence, fatigue, and constipation.

  • Venlafaxine: An alternative SNRI that has demonstrated efficacy in treating PDPN in several studies.525,526 However, in a randomized controlled trial involving 244 patients with PDPN, 7 treated with venlafaxine developed clinically significant electrocardiogram changes during treatment, making it less preferred in patients with cardiovascular conditions.525

Sodium channel inhibitors

  • Carbamazepine and oxcarbazepine: Recently included as first-line treatments in guidelines, carbamazepine and oxcarbazepine reduce neuronal excitability by blocking voltage-gated sodium channels.37 They are used when α2δ ligands and SNRIs are ineffective or contraindicated.

    Tricyclic antidepressants (TCAs)

  • Amitriptyline and nortriptyline: Effective in managing PDPN by inhibiting monoamine reuptake and blocking sodium channels.421,527 Amitriptyline is the most frequently prescribed TCA and is relatively inexpensive.23 However, their use is limited by anticholinergic side effects, especially in elderly patients and those with cardiovascular disease.527,528

Second-line medications

Opioids and atypical opioids

  • Tapentadol: A centrally acting analgesic with both opioid agonist and norepinephrine reuptake inhibition properties. It may be considered in refractory cases but carries risks of dependence and adverse effects.529

  • Tramadol: should be used cautiously due to similar concerns regarding dependence and side effects.530 While opioids can provide pain relief, their high risk of addiction and other safety concerns limit their use. The most recent ADA position statement does not recommend opioid use as first- or second-line therapies for treating neuropathic pain associated with DN.531,532 When opioids are prescribed, very close monitoring of the patient is advised.533

Topical agents such as the capsaicin 8% patch provide localized pain relief by desensitizing nociceptive fibers, while the lidocaine patch 5% is useful for focal neuropathic pain with a favorable safety profile.534,535 Combination therapy, starting with a single agent and titrating to the maximum tolerated dose, may enhance efficacy when pain control is inadequate, with recent studies suggesting superior pain relief compared to monotherapy.520,536

Non-pharmacological treatments

Non-pharmacological treatments encompass acupuncture, which has gained acceptance as a complementary therapy for PDPN by modulating pain through endogenous opioid release and altering neurotransmitter levels, significantly reducing pain intensity.537,538 Electrical stimulation therapies, including transcutaneous electrical nerve stimulation (TENS) and spinal cord stimulation (SCS), have demonstrated effectiveness in reducing neuropathic pain intensity and improving quality of life.539,540 Lifestyle interventions, such as a healthy diet, regular physical activity, and smoking cessation, are essential in managing PDPN by improving glycemic control, enhancing nerve function, and reducing pain perception.541,542 Physical therapy and rehabilitation, including balance training, gait training, and strength exercises, improve functional mobility and reduce the risk of falls.22 Additionally, CBT addresses the psychological aspects of chronic pain, helping patients to cope with and manage pain, thereby enhancing treatment outcomes when combined with pharmacotherapy (Fig. 5)22. Current treatments for PDPN primarily rely on pharmacological and non-pharmacological interventions; however, existing medications have limited efficacy and potential side effects, and evidence for non-pharmacological therapies remains limited. Future studies should focus on developing more effective and safer treatments and optimizing multimodal therapeutic strategies to improve pain management and patient quality of life.

Management of DPN-related complications

Effective management of complications arising from DPN is essential to reduce morbidity and improve patient outcomes. Foot complications are a significant concern; therefore, regular foot examinations are recommended for patients with diabetes at least annually to identify risk factors for ulceration and amputation.543 Additionally, patient education on proper foot care, including daily inspections, appropriate footwear, and maintaining foot hygiene, is crucial in preventing severe foot-related complications.544 Interventions, such as pressure offloading using therapeutic footwear and insoles, help redistribute pressure and prevent ulcer formation, while establishing multidisciplinary foot care teams involving endocrinologists, podiatrists, surgeons, and nurses reduces amputation rates and healthcare costs.545,546 Patients with DPN are also at an increased risk of falls due to sensory loss, balance impairment, and muscle weakness. Fall prevention strategies are, therefore, vital. Assessment of fall risk through evaluating gait and balance can identify patients who are at high risk.547 Interventions, such as balance and strength training, improve proprioception and reduce the risk of falls by enhancing muscle strength and coordination.548 Additionally, a medication review to adjust or discontinue medications that may impair cognition or balance is recommended to decrease fall risk.549 Chronic pain and disability associated with DPN can lead to significant psychological distress. Psychological support is, therefore, an integral component of comprehensive DPN management. Assessment using validated scales can identify patients in need of mental health support.550 CBT and mindfulness-based stress reduction have been shown to improve coping strategies and reduce pain perception, thereby enhancing overall treatment outcomes.551 Additionally, support groups provide peer support and shared experiences, which can enhance emotional well-being and provide a network of encouragement for patients dealing with chronic pain.552 Current management of DPN-related complications primarily relies on foot examinations, fall prevention, and psychological support; however, there are shortcomings in implementing patient education, multidisciplinary collaboration, and psychological interventions. Future studies should optimize these management strategies and strengthen multimodal interventions to reduce complication rates and improve patient quality of life.

Treatment of DAN

DAN is a serious complication of diabetes mellitus that affects the ANS, leading to dysfunction in multiple organ systems. Currently, there is no effective etiological treatment for DAN, and management primarily focuses on alleviating clinical symptoms and improving patients’ quality of life. Specific abnormal clinical presentations require targeted interventions to mitigate their impact on daily functioning.

CAN

Management strategies for severe CAN, particularly orthostatic hypotension, focus on alleviating symptoms, extending standing time, enhancing physical capacity, and improving overall activity levels rather than solely increasing standing blood pressure. Non-pharmacological interventions include medication adjustment.553 Fluid and salt intake should be encouraged to increase plasma volume, with a high-sodium diet potentially raising standing blood pressure and alleviating symptoms.554 Physical countermeasures encompass the use of compression garments to reduce venous pooling in the lower extremities and abdomen, as well as teaching physical maneuvers to increase venous return when standing.555,556

Lifestyle modifications include head-up tilt sleeping, which elevates the head of the bed by 10–30° to reduce nocturnal diuresis and improve morning orthostatic tolerance, and advising patients to avoid triggers such as prolonged standing, sudden postural changes, high ambient temperatures, and large meals that may exacerbate symptoms.557,558 Dietary adjustments for postprandial hypotension recommend small, frequent meals with low carbohydrate content to minimize splanchnic blood pooling after eating.559 When non-pharmacological measures are insufficient, pharmacological treatments may be considered. These include midodrine, an alpha-1 adrenergic agonist approved by the FDA for symptomatic orthostatic hypotension that induces vasoconstriction to increase peripheral resistance and standing blood pressure;560 droxidopa, a norepinephrine precursor that enhances sympathetic tone by increasing norepinephrine levels and improving orthostatic symptoms;561 fludrocortisone, a mineralocorticoid that promotes sodium and water retention to expand plasma volume, though it must be used cautiously due to the risk of supine hypertension and hypokalemia;562 pyridostigmine, an acetylcholinesterase inhibitor that augments sympathetic ganglion transmission to increase vascular resistance during standing without significantly affecting supine blood pressure.563 Monitoring and considerations include dose titration, initiating medications at low doses and gradually increasing to the maximum tolerated dose while monitoring efficacy and adverse effects;564 careful monitoring for supine hypertension, which occurs in approximately 50% of patients with orthostatic hypotension to balance treatment benefits with potential risks;565 combination therapy, where if monotherapy is ineffective, medications with different mechanisms of action may be combined.566

Gastrointestinal autonomic neuropathy

Gastrointestinal autonomic neuropathy can affect any part of the gastrointestinal tract, leading to symptoms such as gastroparesis, constipation, and diarrhea. Management focuses on symptom relief and improving gastrointestinal motility. Dietary modifications are essential, including encouraging patients to consume small, frequent meals to reduce gastric distension and improve emptying, adopting a low-fiber, low-fat diet to minimize delays in gastric emptying, and incorporating liquid nutrients that are easier to digest.567 A comprehensive medication review is necessary to identify and discontinue medications that may impair gastric motility.568 Pharmacological treatments for gastroparesis include metoclopramide, a prokinetic and antiemetic agent that stimulates gastric emptying by antagonizing dopamine receptors; however, its use is limited by the risk of serious side effects like tardive dyskinesia, necessitating the lowest effective dose for the shortest duration.569 Erythromycin enhances gastric contractions and may be beneficial in the short term, although its long-term use is limited by tachyphylaxis and potential antibiotic resistance.570 Domperidone is not FDA-approved but is available in some countries under investigational protocols.571 Other interventions include gastric electrical stimulation572 and optimizing glycemic control.573

Genitourinary autonomic neuropathy

Genitourinary autonomic neuropathy in diabetes patients can lead to significant dysfunctions, such as ED, neurogenic bladder, and female sexual dysfunction, each requiring targeted management strategies to improve patient outcomes. ED is commonly managed with first-line treatments, including phosphodiesterase type 5 (PDE5) inhibitors (sildenafil, tadalafil, and vardenafil), which enhance nitric oxide-mediated vasodilation to facilitate erections and are effective in many diabetes patients.574 Second-line treatments involve intracavernosal injections of alprostadil for those unresponsive to oral medications and the use of vacuum erection devices, which are non-invasive methods to engorge the penis with blood.575,576 Third-line treatments include penile prosthesis implantation for patients who fail or are intolerant to other therapies.577 Conversely, non-pharmacological interventions, such as bladder training with scheduled voiding and pelvic floor exercises, along with intermittent catheterization, are essential to prevent urinary retention and reduce infection risks.578 Pharmacological treatments include cholinergic agonists, such as bethanechol, to stimulate detrusor muscle contraction, antimuscarinic agents, such as oxybutynin or tolterodine, for overactive bladder symptoms, and beta-3 agonists, such as mirabegron, to relax bladder smooth muscle during the storage phase.579,580 Advanced therapies encompass botulinum toxin injections to reduce bladder overactivity and sacral neuromodulation.581,582 Female sexual dysfunction associated with CAN is managed by optimizing glycemic control to enhance nerve function, promptly treating urinary and genital infections, using lubricants and moisturizers to alleviate vaginal dryness, and implementing pelvic floor therapy to strengthen pelvic muscles.583586 Additionally, psychological support through counseling and sex therapy addresses psychological factors, such as depression, anxiety, and relationship issues, thereby improving sexual function and overall quality of life.550,551

Sudomotor dysfunction

Gustatory sweating, a form of sudomotor dysfunction, is characterized by excessive sweating of the face and neck during eating, especially in response to spicy or acidic foods. Effective management strategies for gustatory sweating include topical anticholinergic agents and botulinum toxin injections. Glycopyrrolate cream at a concentration of 0.5% is topically applied to reduce sweating by blocking acetylcholine receptors in sweat glands, thereby decreasing perspiration.587 When topical treatments are insufficient, Botulinum Toxin Type A injections into the affected areas inhibit acetylcholine release at neuromuscular junctions, resulting in a significant reduction in sweating for several months.588

Current treatments for DAN focus primarily on alleviating symptoms and improving quality of life but lack effective etiological therapies; some treatments have limited efficacy. Future studies should develop more targeted therapies and optimize comprehensive management strategies to enhance patient outcomes and quality of life.

DN and associated complications

Diabetic foot disease is significantly exacerbated in patients with DPN due to reduced sensation and impaired wound healing, elevating the risk of foot ulcers and infections and potentially leading to lower-limb amputations. A recent systematic review and meta-analysis highlighted the high global prevalence of diabetic foot ulceration among DPN patients, emphasizing the critical need for early detection and comprehensive management strategies to prevent severe outcomes.589 DN and DPN share a reciprocal relationship, both arising from prolonged hyperglycemia and common pathophysiological mechanisms, such as oxidative stress, inflammation, and endothelial dysfunction. Patients with DPN are at high risk of developing chronic kidney disease; renal impairment can further exacerbate neuropathic symptoms through the accumulation of uremic toxins that adversely affect nerve function.590 Diabetic retinopathy is also closely associated with DPN, with shared risk factors, such as poor glycemic control, hypertension, and dyslipidemia contributing to the development of both microvascular complications. A cross-sectional study found a significant correlation between the severity of DPN and the progression of diabetic retinopathy, suggesting overlapping pathogenic pathways and the need for integrated management approaches.42 Additionally, DPN is linked to an increased risk of cardiovascular disease (CVD). Autonomic neuropathy, a component of DPN, can lead to abnormalities in HRV and elevate the risk of silent myocardial ischemia, particularly in patients with T2DM. CAN is associated with a higher incidence of cardiovascular events and mortality among diabetes patients.5

The coexistence of DPN with other diabetes-related complications necessitates a comprehensive approach to patient care, especially for elderly patients. Holistic management involves routine screening for DPN and associated complications.543 Multidisciplinary care models involving endocrinologists, neurologists, podiatrists, nephrologists, ophthalmologists, cardiologists, dietitians, and physical therapists improve patient outcomes by addressing all aspects of health comprehensively, thereby reducing hospitalizations and healthcare costs.591 In elderly patients, managing DPN alongside other chronic conditions becomes more complex due to polypharmacy, potential drug interactions, and physical and cognitive limitations. This emphasizes the importance of individualized care plans for elderly diabetes patients, balancing glycemic targets with the risk of hypoglycemia and other adverse effects.592 Furthermore, patient education and support are crucial for effective diabetes management. Indeed, a systematic review highlighted that psychological support and patient education are integral components of effective diabetes management.593 Current research on DN and its associated complications remains limited in terms of comprehensive management and multidisciplinary collaboration, and the interactions between different complications remain poorly understood. It is necessary to enhance the development of integrated treatment strategies, promote interdisciplinary cooperation, and fully investigate the shared pathophysiological mechanisms to improve patient outcomes and quality of life.

Emerging therapies and future drug prospects

DN is a complex and multifactorial complication of diabetes mellitus, often progressing despite optimal glycemic control. Current treatments primarily focus on symptom management, with limited efficacy in halting or reversing disease progression. This underscores the urgent need for novel therapies that target the underlying mechanisms of DN.

Unmet clinical needs

Despite stringent glycemic control, several patients with T2DM continue to experience the progression of DN. Existing cardiovascular treatments, such as aspirin and antioxidants, primarily address comorbid conditions but show limited efficacy in treating DN.5 Additionally, patients often develop tolerance to analgesics, necessitating prolonged high-dose therapies and combination regimens.17 The COMBO-DN study found that drug combinations did not significantly outperform high-dose monotherapy.594 The OPTION-DM study determined which monotherapy and combination therapy of amitriptyline, duloxetine, and pregabalin are the most clinically efficacious; the authors showed that all three treatment pathways and monotherapies had similar analgesic efficacy.595

Therefore, treatments that restore normal neurological function, or at least slow disease progression by targeting specific pathogenic mechanisms, remain warranted. Restoring normal neurological function, or at least slowing disease progression, should be the focus of new therapies to address DN. Furthermore, defining the optimal treatment duration and enhancing analgesic strategies remain necessary. New pain management medications should exhibit higher response rates than existing options and aim to target specific pain pathways while improving understanding of dosing regimens and treatment cycles.5

Novel targeted drugs

Advancements in understanding the pathophysiological mechanisms of DN have led to the development of novel targeted therapies aimed at specific molecular pathways involved in nerve damage and pain transmission (Table 1 and Fig. 6).

Table 1.

Emerging drugs in diabetic neuropathy

Agent Mechanism of action/target Clinical trials’ number Participants (n) Phase Main outcome
Empagliflozin598 SGLT2 inhibitor NCT05977465 50 2 Empagliflozin showed a significant improvement in the electrophysiological studies and a significant decrease in the pain score and the mean serum levels of NSE and MDA.
Canagliflozin601 SGLT2 inhibitor NCT02554126 4401 2 Canagliflozin did not affect the risk of neuropathy events.
Liraglutide602 GLPR agonist NCT02138045 39 2 Neuronal function was unaltered at the central, autonomic or peripheral level.
Exenatide604 GLPR agonist NCT00855439 46 2 There were no statistically significant treatment group differences in the prevalence of CCN, IENFD and nerve conductions.
Dulaglutide605 GLPR agonist UMIN000044264 120 2 Diabetes treatment-related quality of life scores associated with pain and gastrointestinal symptoms were also superior in the dulaglutide group.
Ranirestat606 Aldose reductase inhibitor NCT00101426 549 3 Ranirestat influenced motor nerve function in mild to moderate DSPN.
Benfotiamine608 Anti-AGEs DRKS00014832 57 2 Undergoing.
Ruboxistaurin609 PKC inhibitor NCT00190970 52 2 Ruboxistaurin enhanced skin microvascular blood flow at the distal calf, reduced sensory symptoms, improved the quality of life, and was well tolerated.
Actovegin611 PARP inhibitor NCT00483730 567 3 Actovegin improved neuropathic symptoms, VPT, sensory function, and quality of life.
VX-150 NaV1.8 inhibitor NCT03304522 34 2 VX-150 could reduce pain score caused by small fiber neuropathy.
Vixotrigine(BIIB074)614 Voltage-Gated sodium channel blocker NCT03339336 265 2 Vixotrigine showed significant improvement in diabetes patients with painful small fiber neuropathy.
Mirogabalin616 Calcium channel modulator NCT02318706 834 3 Mirogabalin showed statistically significant pain relief (vs. placebo) in Asian PDPN patients.
ABT-639617 Calcium channel blocker NCT01345045 194 2 There was no difference (p = 0.582) in pain alleviation between ABT-639 and placebo and ABT-639 showed no safety signals.
HSK16149618 Calcium channel blocker NCT04647773 725 3 Efficacy of HSK16149 capsules was superior to placebo in all groups for relieving PDPN and appeared well tolerated.
Eliapixant (BAY 1817080)620 Selective P2X3 antagonist NCT04641273 135 2 Eliapixant in patients with diabetic neuropathic pain (DNP) did not translate to any relevant improvement in different pain intensity outcomes compared with placebo.
MDL-28170621 Calpain inhibitor MDL-28170 reduced mechanical allodynia and thermal hyperalgesia and improved nerve conduction velocity.
ISC 17536622 TRPA1 antagonist NCT01726413 138 2 ISC 17536 had a statistically and clinically significant improvement effects on pain.
Dextromethorphan623 NMDAR antagonist NCT00695565 45 1 Dextromethorphan was effective in a dose-dependent manner in selected patients with painful diabetic neuropathy.
NYX-2925624 NMDAR antagonist NCT04146896 228 2 NYX-2925 did not achieve a statistically significant difference compared with placebo in the primary endpoint, but improved pain scores in patients with DPN in a phase 2 study.
NRD.E1626 Regulation of the phosphorylation of Lyn tyrosine kinase NCT02345291 88 2 NRD.E1 induced a clinically relevant pain reduction and it was well tolerated.
EMA401627 AT2R antagonist

NCT03094195

NCT03297294

45

49

2 EMA401 showed analgesic efficacy and safety in two studies.
ARA 290628 Erythropoietin analog NTR3858 50 2 ARA 290 benefited metabolic control and neuropathic symptoms in patients with T2DM.
SB-509653 PhVEGF165 gene transfer IND577 29 2 This treatment ameliorated neuropathic symptoms.
Buprenorphine629 Partial μ-opioid receptor agonist ACTRN12609000647235 186 2 Transdermal buprenorphine, when tolerated, was an effective therapy for PDPN.
PL37631 Dual enkephalinase inhibitor EudraCT Number:2013-004876-37 120 2 Early termination.
Nebivolol633 Beta-blocker NCT06201611 120 3 Undergoing.
Clonidine gel634 α2-adrenoceptor agonist NCT00695565 180 2 Topical clonidine gel significantly reduced the level of foot pain in painful diabetic neuropathy.
LX-9211636 AAK1 inhibitor NCT04455633 319 2 LX-9211 improved PDPN.
PW507637 σ1R antagonist PW507 demonstrated significant efficacy in alleviating mechanical allodynia and thermal hyperalgesia following both acute and chronic (2-week) administration.
E-52862638 σ1R antagonist E-52862 reversed neuropathic and vascular signs in the ZDF rat.
Fulranumab639 Human monoclonal antibody against NGF NCT00993018 77 2 Fulranumab treatment resulted in dose-dependent efficacy and was generally well tolerated.
Tanezumab640 Humanized monoclonal antibody against NGF

NCT01087203

NCT00568321

73

96

2 Tanezumab provided effective pain reduction in DPN and no new safety concerns were observed.
Salsalate641 NF-κB inhibitor NCT01480297 8 2 Salsalate could improve nerve conduction velocity.
ALC643 Neuroprotective and neurotrophic effects NCT05319275 907 3 ALC significantly improved symptoms and somatosensory symptoms.
Long-acting PEGylated C-peptide645 Improving nerve blood flow and nerve conduction velocity NCT01681290 250 2 A significant reduction in VPT in patients treated with C-peptide compared with placebo.
Ricoliostat647 HDAC6 inhibitor NCT03176472 282 2 Changes in average daily pain were not different between the ricolinostat and placebo groups.
Metanx343 Oxidative nitrosative stress NCT00726713 214 2 Results showed improvements in neuropathic symptoms and quality of life.
VM202651 plasmid DNA encoding HGF isoform NCT02427464 500 3 VM202 demonstrated significant pain reduction and was well tolerated.
Seal oil omega-3 polyunsaturated fatty acids656 Nerve structure and function protection NCT02034266 40 2 ω-3 supplementation was associated with increase in corneal nerve fiber length in T1DM.

AGEs advanced glycation end products, ALC acetyllevocarnitine hydrochloride, AT2R angiotensin II type 2 receptor, CCN confirmed clinical neuropathy, DNP diabetic neuropathic pain, DSPN distal symmetric polyneuropathy, GLPR glucose-dependent insulinotropic polypeptide receptor, HDAC6 histone deacetylase 6, HGF hepatocyte growth factor, IENFD intraepidermal nerve fiber density, MDA malondialdehyde, NF-κb nuclear factor kappa-light-chain-enhancer of activated B cells, NGF nerve growth factor, NMDAR N-methyl-D-aspartate receptor, NSE neuron-specific enolase, PDPN painful diabetic peripheral neuropathy, P2X3R P2X receptor subtype 3, σ1R sigma 1 receptor, SGLT2 sodium-glucose transport protein 2, TRPA1 transient receptor potential ankyrin 1, T1DM types 1 diabetes mellitu, T2DM types 2 diabetes mellitu, VEGF vascular endothelial growth factor, VPT vibration perception threshold, ZDF zucker diabetic fatty

Fig. 6.

Fig. 6

Emerging therapeutic strategies for diabetic neuropathy. A range of repurposed drugs and preclinically tested lead compounds are under investigation for the management of DN. Newer targeted therapies, including SGLT2 inhibitors and GLP receptor agonists, are being integrated into the prevention and treatment of DN. Additionally, novel ion channel modulators, such as voltage-gated sodium and calcium channel blockers, TRPA1 and TRPV1 antagonists, NMDAR antagonists, and P2X3 receptor antagonists, are employed to mitigate pain in patients with PDPN. Innovative approaches such as stem cell therapy, gene therapy, and fecal microbiota transplantation are also being explored to enhance DN management. Concurrently, emerging molecular biomarkers, including non-coding RNAs and neuronal injury markers, are under investigation to improve the clinical diagnosis and prognosis of DN. AGEs advanced glycation end products, AT2R angiotensin II type 2 receptor, BDNF brain-derived neurotrophic factor, DN diabetic neuropathy, FMT fecal microbiota transplantation, EPO erythropoietin, GLPR glucose-dependent insulinotropic polypeptide receptor, HDAC6 histone deacetylase 6, HGF hepatocyte growth factor, IL-1RA interleukin-1 receptor antagonist, iPSC induced pluripotent stem cells, MSCs mesenchymal stem cells, NF-κb nuclear factor kappa-light-chain-enhancer of activated B cells, NGF nerve growth factor, NLR neutrophil-to-lymphocyte ratio, NMDAR N-methyl-D-aspartate receptor, NSE neuron-specific enolase, PDPN painful diabetic peripheral neuropathy, pNF-H phosphorylated neurofilament heavy chain, P2X3R P2X receptor subtype 3, P2X4R P2X receptor subtype 4, SCFA short-chain fatty acids, SGLT2 sodium-glucose transport protein 2, SOD superoxide dismutase, TRPV1 transient receptor potential vanilloid 1, TRPA1 transient receptor potential ankyrin 1, TRPM8 transient receptor potential melastatin 8, VEGF vascular endothelial growth factor

SGLT2 inhibitors

  • Empagliflozin: Empagliflozin is a novel SGLT2 inhibitor, the underlying mechanisms of which include ameliorating hyperglycemia, preventing the development of endothelial dysfunction, reducing oxidative stress, and improving metabolic profiles.596 Empagliflozin can prevent the loss of skin intraepidermal nerve fibers and mesangial matrix expansion597 and a potential therapeutic effect on DPN. In a phase 2 study, comparing the results of the empagliflozin arm to the control arm showed a significant improvement in the electrophysiological studies and a significant decrease in the pain score and the mean serum levels of neuron-specific enolase and malondialdehyde in patients with DPN.598

  • Canagliflozin: Aa an SGLT2 inhibitor for the treatment of T2DM,599 canagliflozin reduces obesity-related inflammation in neural tissue and skeletal muscle.600 In a multicenter, randomized, double-blind, placebo-controlled trial, canagliflozin did not affect the risk of neuropathy events.601 Future large, randomized studies with prespecified neurologic endpoints are warranted to determine the effects of SGLT2 inhibitors on DN.

GLP-1 receptor agonists

  • Liraglutide: Liraglutide has shown anti-inflammatory properties. In a randomized, double-blinded, placebo-controlled trial, compared with placebo, liraglutide reduced IL-6 with concomitant numerical reductions in other proinflammatory cytokines; however, neuronal function was unaltered at the central, autonomic, or peripheral level.602

  • Exenatide: Exenatide may have direct neuroprotective and neurotrophic effects independent of its glycemic effects.603 In an 18-month proof-of-concept open-label randomized study, no statistically significant treatment group differences in the prevalence of confirmed clinical neuropathy (CCN), IENFD, and nerve conductions were observed.604

  • Dulaglutide: In a randomized, parallel-group, multicenter, open-label trial, diabetes treatment-related quality of life scores associated with pain and gastrointestinal symptoms were superior in the dulaglutide group.605

Aldose reductase inhibitor

  • Ranirestat: Ranirestat is a potent aldose reductase inhibitor that reduces nerve sorbitol accumulation. A clinical trial has shown that ranirestat affects motor nerve function in mild to moderate DPN.606 It failed to show a statistically significant difference in sensory nerve function relative to placebo.

Anti-AGEs

  • Benfotiamine: Benfotiamine, a lipid-soluble derivative of thiamine. The ability of benfotiamine to inhibit three major pathways (the hexosamine pathway, the AGE formation pathway, and the diacylglycerol (DAG)-PKC pathway) simultaneously might be clinically useful in preventing the development and progression of diabetic complications.233 In a double-blind, placebo-controlled, phase-3 study, benfotiamine can reduce pain and is well tolerated.607 A randomized, double-blind, placebo-controlled trial over 12 months to assess the effects of benfotiamine on morphometric, neurophysiological, and clinical measures in patients with T2DM with symptomatic polyneuropathy is undergoing.608

PKC inhibitor

  • Ruboxistaurin: Ruboxistaurin targets the PKC-β isoform implicated in diabetic microvascular complications. In a 6-month, randomized, double-masked, placebo-controlled study, ruboxistaurin enhanced skin microvascular blood flow at the distal calf, reduced sensory symptoms, improved the quality of life, and was well tolerated in DPN patients.609

PARP inhibitor

  • Actovegin: Actovegin ameliorated sensory nerve conduction velocity (SNCV) and IENFD of experimental DN via mechanisms involving suppression of PARP activation.610 Actovegin improved neuropathic symptoms, vibration perception threshold (VPT), sensory function, and quality of life in patients with DPN in a multicenter, randomized, double-blind trial.611

Voltage-gated sodium and calcium channel blockers

  • VX-150: A pro-drug of a highly selective NaV1.8 inhibitor. VX-150 induces analgesia without compromising subject safety in a phase 1 study. VX-150 also has a significant effect on cold pressure pain thresholds and heat pain thresholds.612 A phase 2 study showed that VX-150 could reduce pain scores caused by SFN.

  • Vixotrigine (BIIB074): Vixotrigine is a state-dependent sodium channel blocker that targets hyperexcitable neurons involved in pain transmission. A phase 2a trial in patients with trigeminal neuralgia, vixotrigine may provide a potential treatment option, although it did not show a statistically significant advantage.613 In a phase 2 study, vixotrigine was investigated for the treatment of idiopathic or diabetes-associated painful SFN. Vixotrigine showed significant improvement in diabetes patients with painful SFN.614 Its efficacy in painful DN is currently under investigation.

  • Mirogabalin: Mirogabalin selectively binds to the α2δ-1 subunit of VGCCs, inhibiting overactive neurons. Mirogabalin provides effective pain relief with improved tolerability compared to traditional agents like pregabalin.615 In a phase-3 study, mirogabalin showed statistically significant pain relief (vs. placebo) in Asian PDPN patients.616

  • ABT-639: A peripherally acting highly selective T-type Cav3.2 calcium channel blocker. A phase 2 clinical trial reported no difference in pain alleviation in patients with DN pain between ABT-639 and placebo and that ABT-639 showed no safety signals.617

  • HSK16149: HSK16149 is a novel ligand α2-δ subunit of VGCC. A Phase-3 randomized clinical trial showed that the efficacy of HSK16149 capsules was superior to placebo in all groups for relieving PDPN and appeared well tolerated.618 Forty- and 80-mg/d doses of HSK16149 were recommended for treating patients with PDPN in China. On May 20, 2024, HSK16149 was approved by the National Food and Drug Administration (NMPA) of China for treating PDNP in adults.

Selective P2X3 antagonist

  • Eliapixant (BAY 1817080): ATP-dependent P2X3 receptors play a fundamental role in nerve fiber sensitization and pathological pain pathways.619 Eliapixant in patients with DNP did not translate to any relevant improvement in different pain intensity outcomes compared with placebo in phase 2a of the PUCCINI study.620

Calpain inhibitor

  • MDL-28170: Calpain plays an important role in the pathophysiology of neurological complications. In DRG neurons of STZ-induced diabetes rats, intraperitoneal administration of MDL-28170 has shown beneficial effects in alleviating DN via modulation of TTX-R Na (+) channel kinetics and reducing oxidative stress and neuroinflammation in STZ-induced diabetes rats.621

TRPA1 antagonist

  • ISC 17536: ISC 17536 is a potent and selective antagonist targeting TRPA1. In a randomized, double-blind, placebo-controlled trial, patients with preserved small nerve fiber function as defined by QST had a statistically and clinically significant improvement in pain.622

NMDA receptor antagonists

  • Dextromethorphan: NMDA receptor antagonists may mitigate central sensitization and enhance descending inhibitory pathways involved in neuropathic pain. Dextromethorphan and memantine are being explored for their potential benefits in DPN. In efficacy and dose-response trials, no comparison with placebo reached statistical significance in the efficacy trial.623 Dextromethorphan is effective in a dose-related fashion in selected patients with painful DN.

  • NYX-2925: A novel co-agonist to glutamate at the NMDA receptor. A first-in-human, randomized, double-blind study of safety and pharmacokinetics in adults suggested that NYX-2925 was safe and well tolerated.624 NYX-2925 did not achieve a statistically significant difference with placebo in the primary endpoint; however, it improved pain scores in patients with DPN in a phase 2 study.

Regulation of the phosphorylation of Lyn tyrosine kinase

  • NRD.E1: Presumably, the mechanism of action of NRD.E1 involves regulation of the phosphorylation of Lyn tyrosine kinase, a key step in P2X4 receptor upregulation in neuropathic pain induced by nerve injury.625 NRD.E1 is a novel non-opioid therapeutic that is being developed for the treatment of PDPN.626 In phase 2a, randomized, dose-finding, proof-of-concept study, NRD.E1 induced a clinically relevant pain reduction and was well tolerated.

Angiotensin II type 2 receptor antagonists

  • EMA401: EMA401 has been investigated for its potential to reduce inflammation in peripheral tissues and decrease peripheral sensitization. EMA401 showed analgesic efficacy and safety in two randomized, double-blind, phase 2 studies of postherpetic neuralgia and painful DN. Although both studies were terminated early due to long-term preclinical hepatotoxicity.627

Erythropoietin analogs

  • ARA 290 is an 11-amino acid peptide derived from erythropoietin that interacts with the innate repair receptor to promote anti-inflammatory and neuroprotective effects. A double-blind, placebo-controlled, investigator-initiated clinical trial showed that ARA 290 benefited both metabolic control and neuropathic symptoms in patients with T2DM.628

μ-opioid receptor agonis

  • Buprenorphine: Transdermal buprenorphine was evaluated in a randomized, double-blind, parallel-group, placebo-controlled trial. Pain reduction by 30% was more frequent in the buprenorphine vs. placebo group, but many patients withdrew from the study.629 Transdermal buprenorphine, when tolerated, is an effective therapy for PDPN.

Dual enkephalinase inhibitor

  • PL37: An orally administered dual inhibitor of enkephalinases.630 PL37 can increase the concentration of enkephalins peripherally and enhance the endogenous opiodergic system. PL37 showed a significant analgesic effect in animal models that can reduce neuropathic pain without the possible side effects of opiates.631 However, similar results were not found in a 4-week phase 2a study involving patients with PDPN. This trial was prematurely discontinued, and the results have not been published.

β-adrenoceptor antagonist

  • Nebivolol: Nebivolol is a β-adrenoceptor antagonist with nitric oxide enhancement, vasodilation, and antioxidant effects. Nebivolol can alleviate thermal hyperalgesia in diabetes rats by releasing NO activity.632 Nebivolol prevents disease progression in patients with DPN, probably through its dual effects as an antioxidant and NO enhancer.633 A 3-arm, open-label, stratified randomized controlled trial with a blinded endpoint assessment to evaluate a NO generator (Nebivolol) as a disease-modifying medication in DPN is undergoing.

α2-adrenoceptor agonists

  • Clonidine gel: A randomized, double-blind, placebo-controlled, parallel-group, multicenter trial showed that topical clonidine gel significantly reduced the level of pain in subjects with DN in whom there are functional (and possibly sensitized) nociceptors in the affected skin.634

AAK1 inhibitor

  • LX-9211: Junctional protein 2-associated kinase 1 (AAK1) is widely expressed in the brain and spinal cord and is a viable target for the treatment of neuropathic pain, and BMS-986176/LX-9211 is a novel class of bis(iso)aryl ethers that are highly selective, CNS-permeable, and potent AAK1 inhibitors.635 A double-blind, randomized, placebo-controlled trial of LX-9211 for pain relief and safety in DPN initially found that LX-9211 improved PDPN.636

Sigma 1 receptor (σ1R) antagonists

  • PW507: σ1R plays a key role in regulating cellular processes associated with pain modulation. In preclinical rat models of STZ-induced DN, PW507 demonstrated significant efficacy in alleviating mechanical allodynia and thermal hyperalgesia following acute and chronic (2-week) administration without inducing tolerance and visual evidence of toxicity.637 This report offers evidence for the potential use of PW507 as a promising therapeutic option for PDN.

  • E-52862: E-52862 administration reverses neuropathic (behavioral and electrophysiological) and vascular signs in the Zucker diabetic fatty (ZDF) rat.638 Blocking σ1R prevents DN in rats and is a potentially effective treatment for peripheral neuropathies in diabetes patients.

Monoclonal antibody against NGF

  • Fulranumab: Fulranumab is a human monoclonal antibody against NGF. A phase 2, double-blind, placebo-controlled trial randomized patients with moderate-severe PDPN to subcutaneous fulranumab or placebo.639 Despite early study termination (clinical hold) by the US Food and Drug Administration, fulranumab treatment resulted in dose-dependent efficacy and was generally well tolerated.

  • Tanezumab: Tanezumab is a humanized monoclonal antibody against NGF. Two RCTs examined the efficacy of tanezumab vs. placebo in patients with DPN or postherpetic neuralgia.640 In the DPN study, tanezumab provided effective pain reduction in DPN, with no safety concerns observed.

Anti-inflammatory agents

  • Salsalate: NF-κB plays a crucial role in the transcription of proinflammatory cytokines contributing to nerve damage. Salsalate, an NF-κB inhibitor, can improve fasting plasma glucose, reduce HbA1c levels, and reduce proinflammatory markers in T2DM patients.641 A phase 2 study showed that salsalate can improve NCV.

Acetyllevocarnitine hydrochloride (ALC)

  • ALC: ALC has neuroprotective and neurotrophic effects on the PNS.642 In a multicenter, randomized, double-blind, placebo-controlled phase-3 clinical trial conducted in China, acetyllevocarnitine hydrochloride treatment for 24 weeks significantly improved symptoms and somatosensory symptoms in patients with DPN, with a treatment effect of 65% higher than placebo.643

Long-acting PEGylated C-peptide

  • C-peptide has neuroprotective properties in T1DM, improving nerve blood flow and NCV and increasing expression of beneficial neurotrophic factors.644 A long-acting form of C-peptide has been developed to enhance its stability and half-life. A 12-month clinical trial demonstrated a significant reduction in VPT in patients treated with C-peptide compared with placebo.645 However, the primary endpoint of improved SNCV was not met, and further studies remain uncertain.

HDAC6 inhibitor

  • Ricoliostat: HDAC6 inhibitors can promote the degradation of protein aggregates or protect neurons from oxidative stress, hence can be used as possible drugs for treating peripheral neuropathy.646 Ricoliostat, a selective HDAC6 inhibitor, failed to ameliorate neuropathic pain in patients with DPN when administered 120 mg/kg daily of ricolinostat for 12 weeks compared with placebo.647 However, ricoliostat has a favorable safety and tolerability profile and does not exclude the possibility that longer treatment may have a therapeutic effect on DPN or other small fiber neuropathies.

Combination nutraceuticals

  • Metanx: Metanx is a prescription medical food containing L-methylfolate, methylcobalamin, and pyridoxal-5′-phosphate. Metanx ingredients counteract endothelial NO synthase uncoupling and oxidative stress in vascular endothelium and peripheral nerve.344 A randomized trial showed improvements in neuropathic symptoms and quality of life measures in patients with DPN. However, further studies are needed to confirm its efficacy and long-term benefits.343

Stem cell therapy

Stem cell therapy offers a regenerative approach to treating DPN by promoting nerve repair and regeneration.

  • Mesenchymal stem cells (MSCs): MSCs can differentiate into neuronal cells and secrete neurotrophic factors like NGF and BDNF.648 Preclinical studies have shown that MSC transplantation improves nerve function and reduces neuropathic pain in diabetic animal models649.

  • Induced pluripotent stem cells (iPSCs): iPSCs derived from patients’ cells offer a personalized therapy option. Research indicates that chemogenetic stimulation of hiPSC-derived neural cells enhances cell activity and neuron-to-neuron interactions in vitro.650 Clinical trials are ongoing to evaluate the safety and efficacy of stem cell therapies in patients with DPN.

Gene therapy

Gene therapy targets the underlying genetic and molecular mechanisms contributing to DPN.

  • VM202: VM202 is a plasmid DNA encoding human hepatocyte growth factor (HGF) isoform. HGF promotes angiogenesis and neurogenesis, potentially improving nerve regeneration. A phase-3 study was conducted to evaluate the safety and efficacy of VM202 in DPN. VM202 demonstrated significant pain reduction and was well tolerated, although not all endpoints were met.651,652

  • SB-509 (PhVEGF165 gene transfer): VEGF gene therapy aims to enhance angiogenesis and nerve repair. An open-label trial used VEGF165 gene transfer as an intra-muscular injection in the ischemic thigh in patients with critical ischemia with/without DM; the treatment ameliorated neuropathic symptoms.653

Microbiota therapy

Emerging evidence suggests that the gut microbiota plays a role in the pathogenesis of DPN.

  • FMT: FMT involves transferring gut microbiota from healthy donors to patients to restore microbial balance. A randomized, double-blind, and placebo-controlled trial demonstrated that FMT reduced inflammatory markers and improved neuropathic symptoms in patients with DPN.654

  • Probiotic supplementation: A randomized controlled trial has shown that probiotic supplementation among patients with diabetes significantly decreased biomarkers of inflammation and oxidative stress.655

  • Omega-3 supplementation: In a single-arm, open-label trial of seal oil ω-3 PUFA supplementation (composed of eicosapentaenoic acid, docosapentaenoic acid, and docosahexaenoic acid) for 1 year in patients with T1DM and diabetic sensorimotor polyneuropathy, a 29% improvement in the primary endpoint of corneal nerve fiber length was observed. However, there was no change in nerve conduction or sensory function.656

  • Dietary interventions: High-fiber and ketogenic diets have been used to treat chronic pain conditions. High-fiber diets, possibly through gut microbiota modulation, increase short-chain fatty acid production,657 while a ketogenic diet reduces metabolic syndrome-induced allodynia and increases epidermal axon density in mice.658

Advanced drug delivery systems

Innovations in drug delivery aim to enhance the efficacy and specificity of therapeutic agents for DN. Nanometer-sized drug carriers can improve bioavailability, achieve controlled release, enhance the selectivity of therapeutic agents, minimize side effects, and optimize treatment outcomes.659 Nanotechnology-based strategies, such as lipid and polymeric nanoparticles, have been widely investigated for their potential application in improving drug delivery to enhance action to the target area to mitigate diabetic neuropathic pain.660 Nanotechnology-based interventions offer promising prospects in diabetes management. Advanced formulations allow for sustained release of therapeutic agents, reducing dosing frequency and improving patient compliance.661

Biomarkers and diagnostic advances

Identifying reliable biomarkers for DN is crucial for early diagnosis and monitoring of disease progression.

  • Oxidative stress markers: Elevated levels of methylglyoxal (MG) and decreased superoxide dismutase (SOD) activity are associated with DPN, reflecting increased oxidative stress.225

  • Inflammatory markers: Neutrophil-to-lymphocyte ratio (NLR) and IL-1 receptor antagonist (IL-1RA) levels correlate with DPN severity, indicating the role of inflammation.662

  • Non-Coding RNAs: Altered expression of miR-146a has been implicated in DPN pathogenesis by modulating inflammatory pathways.663 Long non-coding RNAs (lncRNAs), such as NONRATT021972, may regulate pain signaling pathways and are potential therapeutic targets.664 The level of serum NONRATT021972 is positively correlated with the degree of neuropathic pain in T2DM patients. Inhibiting NONRATT021972 may relieve the symptoms of neuropathic pain in T2DM patients.665

  • Neuronal damage markers: Elevated serum neuron-specific enolase (NSE) levels reflect neuronal injury and correlate with DPN severity. The serum NSE level in DPN patients is significantly higher than that in diabetes patients without neuropathy, and the serum NSE level is related to the severity or lesion stage of DPN.666 Increased serum phosphorylated neurofilament heavy chain (pNF-H) is associated with SFN in patients with impaired glucose tolerance.667

Current treatments for DN primarily focus on symptom management, lacking effectiveness in reversing or halting disease progression. Emerging gene therapy and cell-based regenerative treatments face considerable challenges in delivery and specificity. Targeted delivery to peripheral nerves remains challenging due to the blood-nerve barrier, and off-target effects can pose safety risks. Future research should focus on vector development for enhanced specificity and safety and optimize CRISPR-based approaches for long-term gene editing in DN-related genes. Moreover, although preclinical studies have yielded promising results, large-scale, double-blinded clinical trials remain necessary to verify the therapeutic efficacy and feasibility of these approaches in DN patients.

Future directions for clinical management and patient care

Personalized medicine

Personalized medicine, leveraging genomics, metabolomics, and other multi-omics approaches, holds significant promise for the early diagnosis and therapeutic monitoring of DN. Integrating biomarkers into clinical practice can tailor interventions, leading to more effective management strategies. Genetic variants influencing drug metabolism can facilitate the selection of pharmacological treatments, improving efficacy and reducing adverse effects. For instance, pharmacogenomic testing can identify patients more likely to respond to specific analgesics or may be at risk for adverse reactions.668 This approach enhances therapeutic outcomes by aligning treatments with the patient’s genetic makeup. Recent studies have highlighted the utility of biomarkers, such as NGF and BDNF, in predicting the progression of neuropathy and response to therapy. Elevated levels of NGF and BDNF have been associated with nerve regeneration and repair mechanisms.669,670 Metabolomics offers insights into metabolic alterations associated with DN. Metabolic profiling can identify specific patterns linked to neuropathy, facilitating early detection and personalized interventions.671

Additionally, catalyzing DPN discoveries through bioinformatics is crucial due to the complex pathophysiology of DPN. Integrative methods combining two or more Omics datasets can identify novel associations in DPN, and computational methods can be leveraged to identify drug targets.672,673 Transcriptomics research analyzes gene expression changes in peripheral nerves of both DPN animal models and humans, consistently revealing immune response and inflammatory pathway involvement in neuropathy. Single-cell RNA sequencing and spatial transcriptomics are novel approaches that provide deeper insights into the cellular heterogeneity in DPN pathogenesis.674676 Integrating the vast knowledge from Omics datasets with machine learning and deep learning will be crucial in DPN research to identify therapeutic targets.671

Multidisciplinary management

A multidisciplinary approach involving endocrinologists, neurologists, podiatrists, pain specialists, and rehabilitation therapists is crucial for the comprehensive management of DN patients. Collaborative care models enhance lifestyle interventions by focusing on diet, exercise, and foot care, essential for preventing complications such as diabetic foot ulcers. Evidence suggests that integrated care pathways lead to improvements, including reduced hospitalizations and enhanced quality of life.591,677 Multidisciplinary teams address various aspects of DN, such as glycemic control, neuropathic pain management, and prevention of secondary complications. For instance, regular foot examinations conducted by podiatrists can prevent ulcerations and amputations by identifying and treating problems early.678 Rehabilitation specialists provide tailored exercise programs to improve nerve function and reduce pain, with physical therapy shown to enhance balance, strength, and overall mobility in DN patients.679 Additionally, dietitians assist in nutritional management, optimizing metabolic control, and potentially slowing disease progression.

Digital health and remote monitoring

Advancements in digital health technologies, such as wearable devices and telemedicine, offer promising avenues for long-term patient management. These technologies facilitate continuous monitoring of patients’ health status, enabling timely interventions and personalized care plans. Wearable devices can monitor parameters like blood glucose levels and physical activity and even detect early signs of foot ulcers through temperature and pressure sensors.680 Smart insoles and socks with sensors can alert patients and healthcare providers to areas of high pressure or temperature changes indicative of tissue damage.681 Telemedicine provides remote access to healthcare services, which is especially beneficial for patients with limited mobility or those living in remote areas. Telehealth interventions have been shown to improve glycemic control and patient satisfaction.682 Mobile Health Applications facilitate self-management by providing educational resources, medication reminders, and tools for tracking symptoms and glucose levels.683,684 Advanced imaging techniques, such as high-resolution MRI and positron emission tomography, offer potential for early DN detection and disease progression monitoring.685,686

AI in DN management

AI and machine learning technologies are increasingly being integrated into healthcare, offering new opportunities for the management of DN. Early Detection and Diagnosis: AI algorithms can analyze large datasets to identify patterns indicative of early neuropathic changes. Machine learning models have been developed to predict the onset of DN by processing clinical variables, genetic information, and imaging data.687 For example, deep learning techniques applied to NCS and skin biopsy images can enhance diagnostic accuracy.688 AI can assist in stratifying patients based on their risk of developing complications. Predictive analytics can consider many factors, including glycemic control, comorbidities, and lifestyle behaviors, to forecast disease progression and personalize intervention strategies.689 Decision support tools utilizing AI can help clinicians select the most effective therapies, adjust dosages, and anticipate potential side effects.689,690 AI-powered applications enhance remote monitoring by analyzing data from wearable devices and alerting healthcare providers to significant changes. Natural language processing enables virtual assistants and chatbots to support patients.690

Despite the potential benefits, challenges include data privacy concerns, the need for large and diverse datasets to train algorithms, and ensuring the transparency and interpretability of AI models remain.691,692 Ethical considerations and regulatory frameworks must evolve alongside technological advancements to ensure patient safety and trust. Bioinformatics and multi-omics approaches are pivotal in uncovering the complex pathophysiological mechanisms of DN, facilitating novel therapeutic targets. However, addressing the associated challenges is essential to fully realize the benefits of AI in DN management.

Conclusions

Despite advancements in our understanding of DN, addressing the growing healthcare burden posed by this complication of diabetes remains challenging. A paradigm shift is occurring, with research focusing on a more comprehensive understanding of nerve metabolism, including the interactions between glucose, lipids, and bioenergetics in the PNS. The focus of preclinical studies has shifted from investigating glucose metabolism alone to exploring more complex mechanisms, such as the metabolic interplay between Schwann cells and axons and the role of insulin resistance in nerve function.

Further research remains warranted to address unanswered questions, such as whether DN results from global metabolic reprogramming of the PNS and how energy transfer or toxic byproducts between nerve cells contribute to the disease. Addressing these questions will aid in developing therapies that target the underlying mechanisms of DN.

The rising prevalence of diabetes and obesity, coupled with DN’s severe impact on quality of life—including pain, disability, and increased risk of amputations—necessitate effective treatment. The next decade will be pivotal in translating accumulating clinical and preclinical findings into meaningful therapies that can prevent or reverse the course of DN, ultimately improving outcomes for millions of patients.

Acknowledgements

This work was supported by the National Natural Science Foundation of China [grant numbers 82125037], Science Foundation for Distinguished Young Scholars of Jiangsu province [grant number BK20231528], Key R&D Program of Jiangsu Province [grant number BE2022815], Jiangsu Provincial Medical Innovation Center [grant number CXZX202225], Young Elite Scientists Sponsorship Program of the China Association for Science and Technology [grant number 2021-QNRC2-B17], and 333 high-level talents project of Jiangsu Province for Yang Yang [grant number BRA202202]. All figures in this article were created using BioRender.com (https://biorender.com).

Author contributions

Y.Y. contributed to the conception and manuscript design. Y.Y., B.Z., Y.W., X.L., H.L., and Y.H. were involved in manuscript preparation, revision, and critique of the manuscript for important intellectual content. Y.Y. and P.C. were involved in funding acquisition. All authors have read and approved the final manuscript.

Competing interests

The authors declare no competing interests.

Footnotes

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

These authors contributed equally: Yang Yang, Bing Zhao

Contributor Information

Yang Yang, Email: young1570@126.com.

Peng Cao, Email: cao_peng@njucm.edu.cn.

References

  • 1.Sloan, G., Selvarajah, D. & Tesfaye, S. Pathogenesis, diagnosis and clinical management of diabetic sensorimotor peripheral neuropathy. Nat. Rev. Endocrinol.17, 400–420 (2021). [DOI] [PubMed] [Google Scholar]
  • 2.Gregory, G. A. et al. Global incidence, prevalence, and mortality of type 1 diabetes in 2021 with projection to 2040: s modelling study. Lancet Diabetes Endocrinol.10, 741–760 (2022). [DOI] [PubMed] [Google Scholar]
  • 3.Sun, H. et al. IDF Diabetes Atlas: global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res. Clin. Pract.183, 109119 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Feldman, E. L. et al. Diabetic neuropathy. Nat. Rev. Dis. Prim.5, 42 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Pop-Busui, R. et al. Diabetic neuropathy: a position statement by the American Diabetes Association. Diabetes Care40, 136–154 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Armstrong, D. G., Boulton, A. J. M. & Bus, S. A. Diabetic foot ulcers and their recurrence. N. Engl. J. Med.376, 2367–2375 (2017). [DOI] [PubMed] [Google Scholar]
  • 7.Ang, L., Jaiswal, M., Martin, C. & Pop-Busui, R. Glucose control and diabetic neuropathy: lessons from recent large clinical trials. Curr. Diabetes Rep.14, 528 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Hicks, C. W. & Selvin, E. Epidemiology of peripheral neuropathy and lower extremity disease in diabetes. Curr. Diabetes Rep.19, 86 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Saeedi, P. et al. Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: Results from the International Diabetes Federation Diabetes Atlas, 9th edition. Diabetes Res. Clin. Pract.157, 107843 (2019). [DOI] [PubMed] [Google Scholar]
  • 10.Dewanjee, S. et al. Molecular mechanism of diabetic neuropathy and its pharmacotherapeutic targets. Eur. J. Pharmacol.833, 472–523 (2018). [DOI] [PubMed] [Google Scholar]
  • 11.Eid, S. A. et al. New perspectives in diabetic neuropathy. Neuron111, 2623–2641 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Bönhof, G. J. et al. Emerging biomarkers, tools, and treatments for diabetic polyneuropathy. Endocr. Rev.40, 153–192 (2019). [DOI] [PubMed] [Google Scholar]
  • 13.Pang, L. et al. Understanding diabetic neuropathy: Focus on oxidative stress. Oxid. Med. Cell. Longev.2020, 9524635 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Ye, S., Cheng, Z. L., Zhuo, D. Y. & Liu, S. M. Different types of cell death in diabetic neuropathy: a focus on mechanisms and therapeutic strategies. Int. J. Mol. Sci.25, 8126 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Feldman, E. L., Nave, K. A., Jensen, T. S. & Bennett, D. L. H. New horizons in diabetic neuropathy: Mechanisms, bioenergetics, and pain. Neuron93, 1296–1313 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Fernyhough, P. Mitochondrial dysfunction in diabetic neuropathy: a series of unfortunate metabolic events. Curr. Diab Rep.15, 89 (2015). [DOI] [PubMed] [Google Scholar]
  • 17.Finnerup, N. B. et al. Pharmacotherapy for neuropathic pain in adults: a systematic review and meta-analysis. Lancet Neurol.14, 162–173 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Rafiullah, M. & Siddiqui, K. Pharmacological treatment of diabetic peripheral neuropathy: an update. CNS Neurol. Disord. Drug Targets21, 884–900 (2022). [DOI] [PubMed] [Google Scholar]
  • 19.Petropoulos, I. N. et al. Corneal confocal microscopy: ready for prime time. Clin. Exp. Optom.103, 265–277 (2020). [DOI] [PubMed] [Google Scholar]
  • 20.Kan, M., Guo, G. F., Singh, B., Singh, V. & Zochodne, D. W. Glucagon-like peptide 1, insulin, sensory neurons, and diabetic neuropathy. J. Neuropathol. Exp. Neurol.71, 494–510 (2012). [DOI] [PubMed] [Google Scholar]
  • 21.Maccari, R. & Ottanà, R. Sodium-glucose cotransporter inhibitors as antidiabetic drugs: current development and future perspectives. J. Med. Chem.65, 10848–10881 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Kluding, P. M. et al. The effect of exercise on neuropathic symptoms, nerve function, and cutaneous innervation in people with diabetic peripheral neuropathy. J. Diabetes Complicat.26, 424–429 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Alam, U., Sloan, G. & Tesfaye, S. Treating pain in diabetic neuropathy: current and developmental drugs. Drugs80, 363–384 (2020). [DOI] [PubMed] [Google Scholar]
  • 24.GBD 2021 Diabetes Collaborators. Global, regional, and national burden of diabetes from 1990 to 2021, with projections of prevalence to 2050: a systematic analysis for the Global Burden of Disease Study 2021. Lancet402, 203–234 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ziegler, D., Papanas, N., Vinik, A. I. & Shaw, J. E. Epidemiology of polyneuropathy in diabetes and prediabetes. Handb. Clin. Neurol.126, 3–22 (2014). [DOI] [PubMed] [Google Scholar]
  • 26.Zhang, Y. Q. et al. Global disability burdens of diabetes-related lower-extremity complications in 1990 and 2016. Diabetes Care43, 964–974 (2020). [DOI] [PubMed] [Google Scholar]
  • 27.Wang, W. M. et al. Prevalence and risk factors of diabetic peripheral neuropathy: a population-based cross-sectional study in China. Diabetes Metab. Res. Rev.39, e3702 (2023). [DOI] [PubMed] [Google Scholar]
  • 28.Ziegler, D., Rathmann, W., Dickhaus, T., Meisinger, C. & Mielck, A. Prevalence of polyneuropathy in pre-diabetes and diabetes is associated with abdominal obesity and macroangiopathy: the MONICA/KORA Augsburg Surveys S2 and S3. Diabetes Care31, 464–469 (2008). [DOI] [PubMed] [Google Scholar]
  • 29.Jeyam, A. et al. Diabetic neuropathy is a substantial burden in people with type 1 diabetes and is strongly associated with socioeconomic disadvantage: a population-representative study from Scotland. Diabetes Care43, 734–742 (2020). [DOI] [PubMed] [Google Scholar]
  • 30.Mizokami-Stout, K. R. et al. The contemporary prevalence of diabetic neuropathy in type 1 diabetes: findings from the T1D exchange. Diabetes Care43, 806–812 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Tabák, A. G., Herder, C., Rathmann, W., Brunner, E. J. & Kivimäki, M. Prediabetes: a high-risk state for diabetes development. Lancet379, 2279–2290 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Tesfaye, S. et al. Prevalence of diabetic peripheral neuropathy and its relation to glycaemic control and potential risk factors: the EURODIAB IDDM Complications Study. Diabetologia39, 1377–1384 (1996). [DOI] [PubMed] [Google Scholar]
  • 33.Braffett, B. H. et al. Risk factors for diabetic peripheral neuropathy and cardiovascular autonomic neuropathy in the Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications (DCCT/EDIC) study. Diabetes69, 1000–1010 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Park, J. H. & Kim, D. S. The necessity of the simple tests for diabetic peripheral neuropathy in type 2 diabetes mellitus patients without neuropathic symptoms in clinical practice. Diabetes Metab. J.42, 442–446 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Jaiswal, M. et al. Prevalence of and risk factors for diabetic peripheral neuropathy in youth with type 1 and type 2 diabetes: search for diabetes in youth study. Diabetes Care40, 1226–1232 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Zilliox, L. A. & Russell, J. W. Physical activity and dietary interventions in diabetic neuropathy: a systematic review. Clin. Auton. Res.29, 443–455 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Baron, R., Dickenson, A. H., Calvo, M., Dib-Hajj, S. D. & Bennett, D. L. Maximizing treatment efficacy through patient stratification in neuropathic pain trials. Nat. Rev. Neurol.19, 53–64 (2023). [DOI] [PubMed] [Google Scholar]
  • 38.Abbott, C. A., Malik, R. A., van Ross, E. R., Kulkarni, J. & Boulton, A. J. Prevalence and characteristics of painful diabetic neuropathy in a large community-based diabetic population in the U.K. Diabetes Care34, 2220–2224 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Truini, A. et al. A cross-sectional study investigating frequency and features of definitely diagnosed diabetic painful polyneuropathy. Pain159, 2658–2666 (2018). [DOI] [PubMed] [Google Scholar]
  • 40.Ziegler, D., Rathmann, W., Dickhaus, T., Meisinger, C. & Mielck, A. Neuropathic pain in diabetes, prediabetes and normal glucose tolerance: the MONICA/KORA Augsburg Surveys S2 and S3. Pain Med.10, 393–400 (2009). [DOI] [PubMed] [Google Scholar]
  • 41.Dillon, B. R., Ang, L. & Pop-Busui, R. Spectrum of diabetic neuropathy: new insights in diagnosis and treatment. Annu. Rev. Med.75, 293–306 (2024). [DOI] [PubMed] [Google Scholar]
  • 42.Tesfaye, S. et al. Diabetic neuropathies: update on definitions, diagnostic criteria, estimation of severity, and treatments. Diabetes Care33, 2285–2293 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Liu, X. X., Xu, Y. Y., An, M. M. & Zeng, Q. B. The risk factors for diabetic peripheral neuropathy: a meta-analysis. PLoS ONE14, e0212574 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Callaghan, B. C., Cheng, H. T., Stables, C. L., Smith, A. L. & Feldman, E. L. Diabetic neuropathy: clinical manifestations and current treatments. Lancet Neurol.11, 521–534 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Gregg, E. W. et al. The burden of diabetes-associated multiple long-term conditions on years of life spent and lost. Nat. Med.30, 2830–2837 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.den Braber, N. et al. Glucose regulation beyond HbA1c in type 2 diabetes treated with insulin: real-world evidence from the DIALECT-2 cohort. Diabetes Care44, 2238–2244 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Zoungas, S. et al. Effects of intensive glucose control on microvascular outcomes in patients with type 2 diabetes: a meta-analysis of individual participant data from randomised controlled trials. Lancet Diabetes Endocrinol.5, 431–437 (2017). [DOI] [PubMed] [Google Scholar]
  • 48.Wang, C. S. et al. Diabetic peripheral neuropathy: age-stratified glycemic control. Front. Endocrinol. (Lausanne)15, 1377923 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Starling, S. Ageing: somatic mutations accumulate in ageing and diseased neurons. Nat. Rev. Neurol.14, 64 (2018). [DOI] [PubMed] [Google Scholar]
  • 50.Calcutt, N. A., Cooper, M. E., Kern, T. S. & Schmidt, A. M. Therapies for hyperglycaemia-induced diabetic complications: from animal models to clinical trials. Nat. Rev. Drug Discov.8, 417–429 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Callaghan, B. C. et al. Association between metabolic syndrome components and polyneuropathy in an obese population. JAMA Neurol.73, 1468–1476 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Smith, A. G. & Singleton, J. R. Obesity and hyperlipidemia are risk factors for early diabetic neuropathy. J. Diabetes Complicat.27, 436–442 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Rumora, A. E. et al. Dyslipidemia impairs mitochondrial trafficking and function in sensory neurons. FASEB J.32, 195–207 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Vincent, A. M., Callaghan, B. C., Smith, A. L. & Feldman, E. L. Diabetic neuropathy: cellular mechanisms as therapeutic targets. Nat. Rev. Neurol.7, 573–583 (2011). [DOI] [PubMed] [Google Scholar]
  • 55.Jin, H. Y. & Park, T. S. Role of inflammatory biomarkers in diabetic peripheral neuropathy. J. Diabetes Investig.9, 1016–1018 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Clair, C., Cohen, M. J., Eichler, F., Selby, K. J. & Rigotti, N. A. The effect of cigarette smoking on diabetic peripheral neuropathy: a systematic review and meta-analysis. J. Gen. Intern. Med.30, 1193–1203 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Gu, Y., Dennis, S. M., Kiernan, M. C. & Harmer, A. R. Aerobic exercise training may improve nerve function in type 2 diabetes and pre-diabetes: a systematic review. Diabetes Metab. Res. Rev.35, e3099 (2019). [DOI] [PubMed] [Google Scholar]
  • 58.Yang, C. P. et al. Cardiovascular risk factors increase the risks of diabetic peripheral neuropathy in patients with type 2 diabetes mellitus: the Taiwan diabetes study. Medicine94, e1783 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Jankovic, M. et al. Genetic and epigenomic modifiers of diabetic neuropathy. Int. J. Mol. Sci.22, 4887 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Mahajan, A. et al. Fine-mapping type 2 diabetes loci to single-variant resolution using high-density imputation and islet-specific epigenome maps. Nat. Genet.50, 1505–1513 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Adki, K. M. & Kulkarni, Y. A. Biomarkers in diabetic neuropathy. Arch. Physiol. Biochem.129, 460–475 (2023). [DOI] [PubMed] [Google Scholar]
  • 62.Kakavand Hamidi, A., Radfar, M. & Amoli, M. M. Association between MTHFR variant and diabetic neuropathy. Pharmacol. Rep.70, 1–5 (2018). [DOI] [PubMed] [Google Scholar]
  • 63.Tordai, D. Z. et al. Genetic factors associated with the development of neuropathy in type 2 diabetes. Int. J. Mol. Sci.25, 1815 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Zitouni, K., Tinworth, L. & Earle, K. A. Ethnic differences in the +405 and -460 vascular endothelial growth factor polymorphisms and peripheral neuropathy in patients with diabetes residing in a North London, community in the United Kingdom. BMC Neurol.17, 125 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Apicella, M. et al. COVID-19 in people with diabetes: understanding the reasons for worse outcomes. Lancet Diabetes Endocrinol.8, 782–792 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Ellul, M. A. et al. Neurological associations of COVID-19. Lancet Neurol.19, 767–783 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Romero-Sánchez, C. M. et al. Neurologic manifestations in hospitalized patients with COVID-19: the ALBACOVID registry. Neurology95, e1060–e1070 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Hébert, H. L., Veluchamy, A., Torrance, N. & Smith, B. H. Risk factors for neuropathic pain in diabetes mellitus. Pain158, 560–568 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Raputova, J. et al. Sensory phenotype and risk factors for painful diabetic neuropathy: a cross-sectional observational study. Pain158, 2340–2353 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Themistocleous, A. C. et al. The Pain in Neuropathy Study (PiNS): a cross-sectional observational study determining the somatosensory phenotype of painful and painless diabetic neuropathy. Pain157, 1132–1145 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Ziegler, D. et al. Painful and painless neuropathies are distinct and largely undiagnosed entities in subjects participating in an educational initiative (PROTECT study). Diabetes Res. Clin. Pract.139, 147–154 (2018). [DOI] [PubMed] [Google Scholar]
  • 72.Worthington, A. et al. Spinal inhibitory dysfunction in patients with painful or painless diabetic neuropathy. Diabetes Care44, 1835–1841 (2021). [DOI] [PubMed] [Google Scholar]
  • 73.Hagedorn, J. M. et al. An overview of painful diabetic peripheral neuropathy: diagnosis and treatment advancements. Diabetes Res. Clin. Pract.188, 109928 (2022). [DOI] [PubMed] [Google Scholar]
  • 74.Ponirakis, G. et al. Prevalence and risk factors for diabetic neuropathy and painful diabetic neuropathy in primary and secondary healthcare in Qatar. J. Diabetes Investig.12, 592–600 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Alsaloum, M. et al. A gain-of-function sodium channel β2-subunit mutation in painful diabetic neuropathy. Mol. Pain15, 1744806919849802 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Blesneac, I. et al. Rare NaV1.7 variants associated with painful diabetic peripheral neuropathy. Pain159, 469–480 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Meng, W. H. et al. A genome-wide association study provides evidence of sex-specific involvement of Chr1p35.1 (ZSCAN20-TLR12P) and Chr8p23.1 (HMGB1P46) with diabetic neuropathic pain. EBioMedicine2, 1386–1393 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Meng, W. et al. A genome-wide association study suggests an association of Chr8p21.3 (GFRA2) with diabetic neuropathic pain. Eur. J. Pain19, 392–399 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.American Diabetes Association. 11. Microvascular complications and foot care: Standards of Medical Care in Diabetes-2019. Diabetes Care42, S124–S138 (2019). [DOI] [PubMed] [Google Scholar]
  • 80.Devigili, G. et al. Diagnostic criteria for small fibre neuropathy in clinical practice and research. Brain142, 3728–3736 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Mulvey, M. R. et al. Neuropathic pain in cancer: systematic review, performance of screening tools and analysis of symptom profiles. Br. J. Anaesth.119, 765–774 (2017). [DOI] [PubMed] [Google Scholar]
  • 82.Finnerup, N. B., Kuner, R. & Jensen, T. S. Neuropathic pain: from mechanisms to treatment. Physiol. Rev.101, 259–301 (2021). [DOI] [PubMed] [Google Scholar]
  • 83.Tesfaye, S. Advances in the management of diabetic peripheral neuropathy. Curr. Opin. Support. Palliat. Care3, 136–143 (2009). [DOI] [PubMed] [Google Scholar]
  • 84.Baron, R., Binder, A. & Wasner, G. Neuropathic pain: diagnosis, pathophysiological mechanisms, and treatment. Lancet Neurol.9, 807–819 (2010). [DOI] [PubMed] [Google Scholar]
  • 85.Hovaguimian, A. & Gibbons, C. H. Diagnosis and treatment of pain in small-fiber neuropathy. Curr. Pain Headache Rep.15, 193–200 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Boruchow, S. A. & Gibbons, C. H. Utility of skin biopsy in management of small fiber neuropathy. Muscle Nerve48, 877–882 (2013). [DOI] [PubMed] [Google Scholar]
  • 87.Lauria, G. et al. European Federation of Neurological Societies/Peripheral Nerve Society Guideline on the use of skin biopsy in the diagnosis of small fiber neuropathy. Report of a joint task force of the European Federation of Neurological Societies and the Peripheral Nerve Society. Eur. J. Neurol.17, 903–912, e944-9 (2010). [DOI] [PubMed] [Google Scholar]
  • 88.Røikjer, J. et al. The histamine-induced axon-reflex response in people with type 1 diabetes with and without peripheral neuropathy and pain: a clinical, observational study. J. Pain23, 1167–1176 (2022). [DOI] [PubMed] [Google Scholar]
  • 89.Yang, Z. R. et al. Simple tests to screen for diabetic peripheral neuropathy. Cochrane Database Syst. Rev.2018, Cd010975 (2018). [Google Scholar]
  • 90.Vinik, A. I., Nevoret, M. L., Casellini, C. & Parson, H. Diabetic neuropathy. Endocrinol. Metab. Clin. North Am.42, 747–787 (2013). [DOI] [PubMed] [Google Scholar]
  • 91.Cai, Q. et al. The physiological significance of A-waves in early diabetic neuropathy: assessment of motor nerve fibers by neurophysiological techniques. Front. Syst. Neurosci.15, 633915 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Monteiro-Soares, M., Boyko, E. J., Ribeiro, J., Ribeiro, I. & Dinis-Ribeiro, M. Predictive factors for diabetic foot ulceration: a systematic review. Diabetes Metab. Res. Rev.28, 574–600 (2012). [DOI] [PubMed] [Google Scholar]
  • 93.Richardson, J. K., Eckner, J. T., Allet, L., Kim, H. & Ashton-Miller, J. A. Complex and simple clinical reaction times are associated with gait, balance, and major fall injury in older subjects with diabetic peripheral neuropathy. Am. J. Phys. Med. Rehabil.96, 8–16 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Jende, J. M. E. et al. Diabetic neuropathy differs between type 1 and type 2 diabetes: insights from magnetic resonance neurography. Ann. Neurol.83, 588–598 (2018). [DOI] [PubMed] [Google Scholar]
  • 95.Yazdanpanah, L., Nasiri, M. & Adarvishi, S. Literature review on the management of diabetic foot ulcer. World J. Diabetes6, 37–53 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Drovandi, A., Seng, L. & Golledge, J. Effectiveness of educational interventions for diabetes-related foot disease: a systematic review and meta-analysis. Diabetes Metab. Res. Rev.40, e3746 (2024). [DOI] [PubMed] [Google Scholar]
  • 97.Abraham, A. et al. Toronto Clinical Neuropathy Score is valid for a wide spectrum of polyneuropathies. Eur. J. Neurol.25, 484–490 (2018). [DOI] [PubMed] [Google Scholar]
  • 98.Fernández-Torres, R., Ruiz-Muñoz, M., Pérez-Panero, A. J., García-Romero, J. C. & Gónzalez-Sánchez, M. Clinician assessment tools for patients with diabetic foot disease: a systematic review. J. Clin. Med.9, 1487 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Herman, W. H. et al. Use of the Michigan Neuropathy Screening Instrument as a measure of distal symmetrical peripheral neuropathy in type 1 diabetes: results from the Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications. Diabet. Med.29, 937–944 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Jensen, T. S. et al. Painful and non-painful diabetic neuropathy, diagnostic challenges and implications for future management. Brain144, 1632–1645 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Selvarajah, D. et al. Diabetic peripheral neuropathy: advances in diagnosis and strategies for screening and early intervention. Lancet Diabetes Endocrinol.7, 938–948 (2019). [DOI] [PubMed] [Google Scholar]
  • 102.Perkins, B. A. & Bril, V. Diabetic neuropathy: a review emphasizing diagnostic methods. Clin. Neurophysiol.114, 1167–1175 (2003). [DOI] [PubMed] [Google Scholar]
  • 103.Peters, E. J. et al. Interventions in the management of infection in the foot in diabetes: a systematic review. Diabetes Metab. Res. Rev.32(Suppl 1), 145–153 (2016). [DOI] [PubMed] [Google Scholar]
  • 104.Polydefkis, M., Hauer, P., Griffin, J. W. & McArthur, J. C. Skin biopsy as a tool to assess distal small fiber innervation in diabetic neuropathy. Diabetes Technol. Ther.3, 23–28 (2001). [DOI] [PubMed] [Google Scholar]
  • 105.England, J. D. et al. Practice parameter: the evaluation of distal symmetric polyneuropathy: the role of autonomic testing, nerve biopsy, and skin biopsy (an evidence-based review). Report of the American Academy of Neurology, the American Association of Neuromuscular and Electrodiagnostic Medicine, and the American Academy of Physical Medicine and Rehabilitation. PM R.1, 14–22 (2009). [DOI] [PubMed] [Google Scholar]
  • 106.Tavakoli, M., Petropoulos, I. N. & Malik, R. A. Corneal confocal microscopy to assess diabetic neuropathy: an eye on the foot. J. Diabetes Sci. Technol.7, 1179–1189 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Ziegler, D. & Fonseca, V. From guideline to patient: a review of recent recommendations for pharmacotherapy of painful diabetic neuropathy. J. Diabetes Complicat.29, 146–156 (2015). [DOI] [PubMed] [Google Scholar]
  • 108.Staff, N. P. et al. Platinum-induced peripheral neurotoxicity: from pathogenesis to treatment. J. Peripher. Nerv. Syst.24, S26–S39 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Papanas, N. & Ziegler, D. Prediabetic neuropathy: does it exist? Curr. Diabetes Rep.12, 376–383 (2012). [DOI] [PubMed] [Google Scholar]
  • 110.Bus, S. A. et al. IWGDF guidance on the prevention of foot ulcers in at-risk patients with diabetes. Diabetes Metab. Res. Rev.32, 16–24 (2016). [DOI] [PubMed] [Google Scholar]
  • 111.Monteiro-Soares, M. et al. Guidelines on the classification of diabetic foot ulcers (IWGDF 2019). Diabetes Metab. Res. Rev.36, e3273 (2020). [DOI] [PubMed] [Google Scholar]
  • 112.Spallone, V. Update on the impact, diagnosis and management of cardiovascular autonomic neuropathy in diabetes: what is defined, what is new, and what is unmet. Diabetes Metab. J.43, 3–30 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Seuring, T., Archangelidi, O. & Suhrcke, M. The economic costs of type 2 diabetes: a global systematic review. Pharmacoeconomics33, 811–831 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Fujita, Y., Murakami, T. & Nakamura, A. Recent advances in biomarkers and regenerative medicine for diabetic neuropathy. Int. J. Mol. Sci.22, 2301 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Tavakoli, M. et al. Diabetic neuropathy: current status and future prospects. J. Diabetes Res.2017, 5825971 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Jermendy, G. Clinical consequences of cardiovascular autonomic neuropathy in diabetic patients. Acta Diabetol.40, S370–S374 (2003). [DOI] [PubMed] [Google Scholar]
  • 117.Benichou, T. et al. Heart rate variability in type 2 diabetes mellitus: a systematic review and meta–analysis. PLoS ONE13, e0195166 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Freeman, R. Assessment of cardiovascular autonomic function. Clin. Neurophysiol.117, 716–730 (2006). [DOI] [PubMed] [Google Scholar]
  • 119.Hermida, R. C., Moyá, A. & Ayala, D. E. Ambulatory blood pressure monitoring in diabetes for the assessment and control of vascular risk. Endocrinol. Nutr.62, 400–410 (2015). [DOI] [PubMed] [Google Scholar]
  • 120.Juraschek, S. P. et al. Abstract MP59: Orthostatic hypotension and risk of falls in the atherosclerosis risk in communities (ARIC) study. Circulation133, AMP59–AMP59 (2016). [Google Scholar]
  • 121.Cheng, Y. F. et al. How inflammation dictates diabetic peripheral neuropathy: an enlightening review. CNS Neurosci. Ther.30, e14477 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Bharucha, A. E., Kudva, Y. C. & Prichard, D. O. Diabetic gastroparesis. Endocr. Rev.40, 1318–1352 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Du, Y. T., Rayner, C. K., Jones, K. L., Talley, N. J. & Horowitz, M. Gastrointestinal symptoms in diabetes: prevalence, assessment, pathogenesis, and management. Diabetes Care41, 627–637 (2018). [DOI] [PubMed] [Google Scholar]
  • 124.Krishnan, B., Babu, S., Walker, J., Walker, A. B. & Pappachan, J. M. Gastrointestinal complications of diabetes mellitus. World J. Diabetes4, 51–63 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Alagiakrishnan, K. & Halverson, T. Holistic perspective of the role of gut microbes in diabetes mellitus and its management. World J. Diabetes12, 1463–1478 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.George, N. S. et al. Distribution of esophageal motor disorders in diabetic patients with dysphagia. J. Clin. Gastroenterol.51, 890–895 (2017). [DOI] [PubMed] [Google Scholar]
  • 127.Saad, R. J. The wireless motility capsule: a one-stop shop for the evaluation of gi motility disorders. Curr. Gastroenterol. Rep.18, 14 (2016). [DOI] [PubMed] [Google Scholar]
  • 128.Maurer, A. H. Gastrointestinal motility, part 1: esophageal transit and gastric emptying. J. Nucl. Med. Technol.44, 1–11 (2016). [DOI] [PubMed] [Google Scholar]
  • 129.Hellström, P. M. et al. The pharmacodynamics, safety and pharmacokinetics of single doses of the motilin agonist, camicinal, in type 1 diabetes mellitus with slow gastric emptying. Br. J. Pharmacol.173, 1768–1777 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Keller, J., Meier, V., Wolfram, K. U., Rosien, U. & Layer, P. Sensitivity and specificity of an abbreviated (13)C-mixed triglyceride breath test for measurement of pancreatic exocrine function. United Eur. Gastroenterol. J.2, 288–294 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Furness, J. B. The enteric nervous system and neurogastroenterology. Nat. Rev. Gastroenterol. Hepatol.9, 286–294 (2012). [DOI] [PubMed] [Google Scholar]
  • 132.Khashab, M. A. et al. Gastric per-oral endoscopic myotomy for refractory gastroparesis: results from the first multicenter study on endoscopic pyloromyotomy (with video). Gastrointest. Endosc.85, 123–128 (2017). [DOI] [PubMed] [Google Scholar]
  • 133.Blair, Y., Wessells, H., Pop-Busui, R., Ang, L. & Sarma, A. V. Urologic complications in diabetes. J. Diabetes Complicat.36, 108288 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Li, C. H. et al. Urinary incontinence and sleep quality in older women with type 2 diabetes: a cross-sectional study. Int. J. Environ. Res. Public. Health19, 15642 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Maiorino, M. I. et al. Erectile dysfunction in young men with type 1 diabetes. Int. J. Impot. Res.29, 17–22 (2017). [DOI] [PubMed] [Google Scholar]
  • 136.Rew, K. T. & Heidelbaugh, J. J. Erectile dysfunction. Am. Fam. Physician94, 820–827 (2016). [PubMed] [Google Scholar]
  • 137.Bąk, E. et al. Sexual function and sexual quality of life in premenopausal women with controlled type 1 and 2 diabetes—preliminary study. Int. J. Environ. Res. Public. Health18, 2536 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Enzlin, P. et al. Sexual dysfunction in women with type 1 diabetes: A controlled study. Diabetes Care25, 672–677 (2002). [DOI] [PubMed] [Google Scholar]
  • 139.Fujita, N. et al. Association between sex hormones and erectile dysfunction in men without hypoandrogenism. Sci. Rep.14, 13433 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Shridharani, A. N. & Brant, W. O. The treatment of erectile dysfunction in patients with neurogenic disease. Transl. Androl. Urol.5, 88–101 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Hotaling, J. M. et al. Cardiovascular autonomic neuropathy, sexual dysfunction, and urinary incontinence in women with type 1 diabetes. Diabetes Care39, 1587–1593 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Kamenov, Z. A. A comprehensive review of erectile dysfunction in men with diabetes. Exp. Clin. Endocrinol. Diabetes123, 141–158 (2015). [DOI] [PubMed] [Google Scholar]
  • 143.Pérez-Aizpurua, X. et al. Stem cell therapy for erectile dysfunction: a step towards a future treatment. Life13, 502 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Zilliox, L. et al. Assessing autonomic dysfunction in early diabetic neuropathy: the Survey of Autonomic Symptoms. Neurology76, 1099–1105 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Abdalrada, A. S., Abawajy, J., Al-Quraishi, T. & Islam, S. M. S. Prediction of cardiac autonomic neuropathy using a machine learning model in patients with diabetes. Ther. Adv. Endocrinol. Metab.13, 20420188221086693 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Casellini, C. M. & Vinik, A. I. Clinical manifestations and current treatment options for diabetic neuropathies. Endocr. Pract.13, 550–566 (2007). [DOI] [PubMed] [Google Scholar]
  • 147.Eleftheriadou, A. et al. The prevalence of cardiac autonomic neuropathy in prediabetes: a systematic review. Diabetologia64, 288–303 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Novak, P. Quantitative autonomic testing. J. Vis. Exp53, 2502 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Lauria, G. & Lombardi, R. Skin biopsy: a new tool for diagnosing peripheral neuropathy. BMJ334, 1159–1162 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Lefaucheur, J. P. The value of electrochemical skin conductance measurement by Sudoscan® for assessing autonomic dysfunction in peripheral neuropathies beyond diabetes. Neurophysiol. Clin.53, 102859 (2023). [DOI] [PubMed] [Google Scholar]
  • 151.Cryer, P. E. Hypoglycemia-associated autonomic failure in diabetes. Handb. Clin. Neurol.117, 295–307 (2013). [DOI] [PubMed] [Google Scholar]
  • 152.Seaquist, E. R. et al. Hypoglycemia and diabetes: a report of a workgroup of the American Diabetes Association and the Endocrine Society. Diabetes Care36, 1384–1395 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Choudhary, P. et al. Evidence-informed clinical practice recommendations for treatment of type 1 diabetes complicated by problematic hypoglycemia. Diabetes Care38, 1016–1029 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Stanley, S., Moheet, A. & Seaquist, E. R. Central mechanisms of glucose sensing and counterregulation in defense of hypoglycemia. Endocr. Rev.40, 768–788 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Forlenza, G. P. et al. Successful at-home use of the tandem control-IQ artificial pancreas system in young children during a randomized controlled trial. Diabetes Technol. Ther.21, 159–169 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Little, S. A. et al. Recovery of hypoglycemia awareness in long-standing type 1 diabetes: a multicenter 2 × 2 factorial randomized controlled trial comparing insulin pump with multiple daily injections and continuous with conventional glucose self-monitoring (HypoCOMPaSS). Diabetes Care37, 2114–2122 (2014). [DOI] [PubMed] [Google Scholar]
  • 157.Ferrari, G. L. et al. Using dynamic pupillometry as a simple screening tool to detect autonomic neuropathy in patients with diabetes: a pilot study. Biomed. Eng. Online9, 26 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Lerner, A. G. et al. Type 2 diabetes and cardiac autonomic neuropathy screening using dynamic pupillometry. Diabet. Med.32, 1470–1478 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Tan, Z., Scheetz, J. & He, M. G. Artificial intelligence in ophthalmology: accuracy, challenges, and clinical application. Asia Pac. J. Ophthalmol.8, 197–199 (2019). [DOI] [PubMed] [Google Scholar]
  • 160.Harati, Y. Diabetic neuropathies: unanswered questions. Neurol. Clin.25, 303–317 (2007). [DOI] [PubMed] [Google Scholar]
  • 161.Lajmi, H. et al. Oculomotor palsy in diabetics. J. Fr. Ophtalmol.41, 45–49 (2018). [DOI] [PubMed] [Google Scholar]
  • 162.Holland, N. J. & Weiner, G. M. Recent developments in Bell’s palsy. BMJ329, 553–557 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Lang, N. W., Ostermann, R. C., Arthold, C., Joestl, J. & Platzer, P. Retrospective case series with one year follow-up after radial nerve palsy associated with humeral fractures. Int. Orthop.41, 191–196 (2016). [DOI] [PubMed] [Google Scholar]
  • 164.Vasavada, K. et al. Predictors using machine learning of complete peroneal nerve palsy recovery after multiligamentous knee injury: a multicenter retrospective cohort study. Orthop. J. Sports Med.10, 23259671221121410 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Zimmerman, M., Gottsäter, A. & Dahlin, L. B. Carpal tunnel syndrome and diabetes-a comprehensive review. J. Clin. Med.11, 1674 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Collins, M. P. & Hadden, R. D. The nonsystemic vasculitic neuropathies. Nat. Rev. Neurol.13, 302–316 (2017). [DOI] [PubMed] [Google Scholar]
  • 167.Latov, N. Diagnosis and treatment of chronic acquired demyelinating polyneuropathies. Nat. Rev. Neurol.10, 435–446 (2014). [DOI] [PubMed] [Google Scholar]
  • 168.Said, G. Diabetic neuropathy–a review. Nat. Clin. Pract. Neurol.3, 331–340 (2007). [DOI] [PubMed] [Google Scholar]
  • 169.David, P. & Barbara, S. Electromyography and neuromuscular disorders: clinical-electrodiagnostic-ultrasound correlations, fourth edition. J. Clin. Neurophysiol.38, e19 (2021). [DOI] [PubMed] [Google Scholar]
  • 170.Barrell, K. & Smith, A. G. Peripheral neuropathy. Med. Clin. North Am.103, 383–397 (2019). [DOI] [PubMed] [Google Scholar]
  • 171.Kieseier, B. C., Mathey, E. K., Sommer, C. & Hartung, H. P. Immune-mediated neuropathies. Nat. Rev. Dis. Prim.4, 31 (2018). [DOI] [PubMed] [Google Scholar]
  • 172.Gwathmey, K. G., Burns, T. M., Collins, M. P. & Dyck, P. J. Vasculitic neuropathies. Lancet Neurol.13, 67–82 (2014). [DOI] [PubMed] [Google Scholar]
  • 173.Callaghan, B. C., Little, A. A., Feldman, E. L. & Hughes, R. A. Enhanced glucose control for preventing and treating diabetic neuropathy. Cochrane Database Syst. Rev.6, Cd007543 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Graham, B. et al. The American Academy of Orthopaedic Surgeons evidence-based clinical practice guideline on: management of carpal tunnel syndrome. J. Bone Jt. Surg. Am.98, 1750–1754 (2016). [DOI] [PubMed] [Google Scholar]
  • 175.Zochodne, D. W. Diabetic polyneuropathy: an update. Curr. Opin. Neurol.21, 527–533 (2008). [DOI] [PubMed] [Google Scholar]
  • 176.Nischal, N., Gupta, S., Lal, K. & Singh, J. P. Performance evaluation of high-resolution ultrasound versus magnetic resonance imaging in diagnosing peripheral nerve pathologies. Indian J. Radiol. Imaging31, 43–48 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Boulton, A. J. et al. Diabetic neuropathies: a statement by the American Diabetes Association. Diabetes Care28, 956–962 (2005). [DOI] [PubMed] [Google Scholar]
  • 178.Massie, R. et al. Diabetic cervical radiculoplexus neuropathy: a distinct syndrome expanding the spectrum of diabetic radiculoplexus neuropathies. Brain135, 3074–3088 (2012). [DOI] [PubMed] [Google Scholar]
  • 179.Ng, P. S. et al. Lumbosacral radiculoplexus neuropathy: incidence and the association with diabetes mellitus. Neurology92, e1188–e1194 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Pinto, M. V. et al. Lumbosacral radiculoplexus neuropathy: neurologic outcomes and survival in a population-based study. Neurology96, e2098–e2108 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Said, G. Diabetic neuropathy. Handb. Clin. Neurol.115, 579–589 (2013). [DOI] [PubMed] [Google Scholar]
  • 182.Albers, J. W., Jacobson, R. D. & Smyth, D. L. Diabetic amyotrophy: from the basics to the bedside. EMJ5, 94–103 (2020). [Google Scholar]
  • 183.Devigili, G. et al. The diagnostic criteria for small fibre neuropathy: from symptoms to neuropathology. Brain131, 1912–1925 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Kelkar, P., Masood, M. & Parry, G. J. Distinctive pathologic findings in proximal diabetic neuropathy (diabetic amyotrophy). Neurology55, 83–88 (2000). [DOI] [PubMed] [Google Scholar]
  • 185.Pascoe, M. K., Low, P. A., Windebank, A. J. & Litchy, W. J. Subacute diabetic proximal neuropathy. Mayo Clin. Proc.72, 1123–1132 (1997). [DOI] [PubMed] [Google Scholar]
  • 186.Younger, D. S. Diabetic lumbosacral radiculoplexus neuropathy: a postmortem studied patient and review of the literature. J. Neurol.258, 1364–1367 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Laughlin, R. S. & Dyck, P. J. Diabetic radiculoplexus neuropathies. Handb. Clin. Neurol.126, 45–52 (2014). [DOI] [PubMed] [Google Scholar]
  • 188.Anandhanarayanan, A., Teh, K., Goonoo, M., Tesfaye, S. & Selvarajah, D. Diabetic neuropathies. In Endotext [Internet] (MDText. com, Inc., 2022).
  • 189.Bhanushali, M. J. & Muley, S. A. Diabetic and non-diabetic lumbosacral radiculoplexus neuropathy. Neurol. India56, 420–425 (2008). [DOI] [PubMed] [Google Scholar]
  • 190.Chan, Y. C., Lo, Y. L. & Chan, E. S. Immunotherapy for diabetic amyotrophy. Cochrane Database Syst. Rev.7, Cd006521 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Tracy, J. A. & Dyck, P. J. The spectrum of diabetic neuropathies. Phys. Med. Rehabil. Clin. N. Am.19, 1–26 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Malone, J. I. Diabetic central neuropathy: CNS damage related to hyperglycemia. Diabetes65, 355–357 (2016). [DOI] [PubMed] [Google Scholar]
  • 193.Kennedy, J. M. & Zochodne, D. W. The regenerative deficit of peripheral nerves in experimental diabetes: its extent, timing and possible mechanisms. Brain123, 2118–2129 (2000). [DOI] [PubMed] [Google Scholar]
  • 194.Grodzinski, B. et al. The effect of ageing on presentation, management and outcomes in degenerative cervical myelopathy: a systematic review. Age Ageing50, 705–715 (2021). [DOI] [PubMed] [Google Scholar]
  • 195.Dyck, P. J. B. & Windebank, A. J. Diabetic and nondiabetic lumbosacral radiculoplexus neuropathies: new insights into pathophysiology and treatment. Muscle Nerve25, 477–491 (2002). [DOI] [PubMed] [Google Scholar]
  • 196.Huang, H. L. & Wu, S. Application of high-resolution ultrasound on diagnosing diabetic peripheral neuropathy. Diabetes Metab. Syndr. Obes.14, 139–152 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Hotta, N. et al. Long-term clinical effects of epalrestat, an aldose reductase inhibitor, on diabetic peripheral neuropathy: the 3-year, multicenter, comparative Aldose Reductase Inhibitor-Diabetes Complications Trial. Diabetes Care29, 1538–1544 (2006). [DOI] [PubMed] [Google Scholar]
  • 198.Matveyenko, A. & Vella, A. Regenerative medicine in diabetes. Mayo Clin. Proc.90, 546–554 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Roy, T. & Lloyd, C. E. Epidemiology of depression and diabetes: a systematic review. J. Affect. Disord.142(Suppl), S8–S21 (2012). [DOI] [PubMed] [Google Scholar]
  • 200.Atti, A. R. et al. Metabolic syndrome, mild cognitive impairment, and dementia: a meta-analysis of longitudinal studies. Am. J. Geriatr. Psychiatry27, 625–637 (2019). [DOI] [PubMed] [Google Scholar]
  • 201.van Elderen, S. G. et al. Progression of brain atrophy and cognitive decline in diabetes mellitus: a 3-year follow-up. Neurology75, 997–1002 (2010). [DOI] [PubMed] [Google Scholar]
  • 202.Williamson, J. D. et al. Intensive vs standard blood pressure control and cardiovascular disease outcomes in adults aged ≥75 years: a randomized clinical trial. JAMA315, 2673–2682 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Biessels, G. J. & Despa, F. Cognitive decline and dementia in diabetes mellitus: mechanisms and clinical implications. Nat. Rev. Endocrinol.14, 591–604 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Omi, M. et al. Transplantation of dental pulp stem cells suppressed inflammation in sciatic nerves by promoting macrophage polarization towards anti-inflammation phenotypes and ameliorated diabetic polyneuropathy. J. Diabetes Investig.7, 485–496 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Chong, Z. Z., Menkes, D. L. & Souayah, N. Targeting neuroinflammation in distal symmetrical polyneuropathy in diabetes. Drug Discov. Today29, 104087 (2024). [DOI] [PubMed] [Google Scholar]
  • 206.Aghamiri, S. H., Komlakh, K. & Ghaffari, M. Toll-like receptors (TLRs) and their potential therapeutic applications in diabetic neuropathy. Int. Immunopharmacol.102, 108398 (2022). [DOI] [PubMed] [Google Scholar]
  • 207.Scholz, J. & Woolf, C. J. The neuropathic pain triad: neurons, immune cells and glia. Nat. Neurosci.10, 1361–1368 (2007). [DOI] [PubMed] [Google Scholar]
  • 208.Jensen, T. S. & Finnerup, N. B. Allodynia and hyperalgesia in neuropathic pain: clinical manifestations and mechanisms. Lancet Neurol.13, 924–935 (2014). [DOI] [PubMed] [Google Scholar]
  • 209.Shakeel, M. Recent advances in understanding the role of oxidative stress in diabetic neuropathy. Diabetes Metab. Syndr.9, 373–378 (2015). [DOI] [PubMed] [Google Scholar]
  • 210.Zhang, W. J. et al. Potential role of Schwann cells in neuropathic pain. Eur. J. Pharmacol.956, 175955 (2023). [DOI] [PubMed] [Google Scholar]
  • 211.Pickering, R. J. et al. Recent novel approaches to limit oxidative stress and inflammation in diabetic complications. Clin. Transl. Immunol.7, e1016 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Wang, Q., Xie, Y. L., Ma, S. C., Luo, H. L. & Qiu, Y. Role of microglia in diabetic neuropathic pain. Front. Cell Dev. Biol.12, 1421191 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Dunnigan, S. K. et al. Conduction slowing in diabetic sensorimotor polyneuropathy. Diabetes Care36, 3684–3690 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Pan, S. & Chan, J. R. Regulation and dysregulation of axon infrastructure by myelinating glia. J. Cell Biol.216, 3903–3916 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Cashman, C. R. & Höke, A. Mechanisms of distal axonal degeneration in peripheral neuropathies. Neurosci. Lett.596, 33–50 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Riccio, A. RNA targeting and translation in axons. Science359, 1331–1332 (2018). [DOI] [PubMed] [Google Scholar]
  • 217.Huang, L. X. et al. Non-coding RNA in schwann cell and peripheral nerve injury: a review. Adv. Biol.26, e2400357 (2024). [DOI] [PubMed] [Google Scholar]
  • 218.Ma, J. C., Pan, P., Anyika, M., Blagg, B. S. & Dobrowsky, R. T. Modulating molecular chaperones improves mitochondrial bioenergetics and decreases the inflammatory transcriptome in diabetic sensory neurons. ACS Chem. Neurosci.6, 1637–1648 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Sanna, M. D., Quattrone, A., Ghelardini, C. & Galeotti, N. PKC-mediated HuD-GAP43 pathway activation in a mouse model of antiretroviral painful neuropathy. Pharmacol. Res.81, 44–53 (2014). [DOI] [PubMed] [Google Scholar]
  • 220.Hur, J. et al. The metabolic syndrome and microvascular complications in a murine model of type 2 diabetes. Diabetes64, 3294–3304 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Kobayashi, M. et al. Diabetic polyneuropathy, sensory neurons, nuclear structure and spliceosome alterations: a role for CWC22. Dis. Model. Mech.10, 215–224 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Fan, B. Y., Chopp, M., Zhang, Z. G. & Liu, X. S. Emerging roles of microRNAs as biomarkers and therapeutic targets for diabetic neuropathy. Front. Neurol.11, 558758 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Han, Y. P. et al. Autophagy and its therapeutic potential in diabetic nephropathy. Front. Endocrinol. (Lausanne)14, 1139444 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Brownlee, M. The pathobiology of diabetic complications: a unifying mechanism. Diabetes54, 1615–1625 (2005). [DOI] [PubMed] [Google Scholar]
  • 225.Giacco, F. & Brownlee, M. Oxidative stress and diabetic complications. Circ. Res.107, 1058–1070 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Li, Q. R. et al. Epalrestat protects against diabetic peripheral neuropathy by alleviating oxidative stress and inhibiting polyol pathway. Neural Regen. Res.11, 345–351 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Vincent, A. M., Calabek, B., Roberts, L. & Feldman, E. L. Biology of diabetic neuropathy. Handb. Clin. Neurol.115, 591–606 (2013). [DOI] [PubMed] [Google Scholar]
  • 228.Satoh, J., Kohara, N., Sekiguchi, K. & Yamaguchi, Y. Effect of ranirestat on sensory and motor nerve function in japanese patients with diabetic polyneuropathy: a randomized double-blind placebo-controlled study. J. Diabetes Res.2016, 5383797 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Hotta, N. et al. Clinical efficacy of fidarestat, a novel aldose reductase inhibitor, for diabetic peripheral neuropathy: a 52-week multicenter placebo-controlled double-blind parallel group study. Diabetes Care24, 1776–1782 (2001). [DOI] [PubMed] [Google Scholar]
  • 230.Abhary, S. et al. Aldose reductase gene polymorphisms and diabetic retinopathy susceptibility. Diabetes Care33, 1834–1836 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Coussement, P., Bauwens, D., Peters, G., Maertens, J. & De Mey, M. Mapping and refactoring pathway control through metabolic and protein engineering: the hexosamine biosynthesis pathway. Biotechnol. Adv.40, 107512 (2020). [DOI] [PubMed] [Google Scholar]
  • 232.Krause, N. & Wegner, A. Fructose metabolism in cancer. Cells9, 2635 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Hammes, H. P. et al. Benfotiamine blocks three major pathways of hyperglycemic damage and prevents experimental diabetic retinopathy. Nat. Med.9, 294–299 (2003). [DOI] [PubMed] [Google Scholar]
  • 234.Fraser, D. A. et al. The effects of long-term oral benfotiamine supplementation on peripheral nerve function and inflammatory markers in patients with type 1 diabetes: a 24-month, double-blind, randomized, placebo-controlled trial. Diabetes Care35, 1095–1097 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Stirban, A. et al. Benfotiamine prevents macro- and microvascular endothelial dysfunction and oxidative stress following a meal rich in advanced glycation end products in individuals with type 2 diabetes. Diabetes Care29, 2064–2071 (2006). [DOI] [PubMed] [Google Scholar]
  • 236.Singh, R., Barden, A., Mori, T. & Beilin, L. Advanced glycation end-products: a review. Diabetologia44, 129–146 (2001). [DOI] [PubMed] [Google Scholar]
  • 237.D’Agati, V. & Schmidt, A. M. RAGE and the pathogenesis of chronic kidney disease. Nat. Rev. Nephrol.6, 352–360 (2010). [DOI] [PubMed] [Google Scholar]
  • 238.Goldin, A., Beckman, J. A., Schmidt, A. M. & Creager, M. A. Advanced glycation end products: Sparking the development of diabetic vascular injury. Circulation114, 597–605 (2006). [DOI] [PubMed] [Google Scholar]
  • 239.Thornalley, P. J. Use of aminoguanidine (Pimagedine) to prevent the formation of advanced glycation endproducts. Arch. Biochem. Biophys.419, 31–40 (2003). [DOI] [PubMed] [Google Scholar]
  • 240.Basta, G., Schmidt, A. M. & De Caterina, R. Advanced glycation end products and vascular inflammation: implications for accelerated atherosclerosis in diabetes. Cardiovasc. Res.63, 582–592 (2004). [DOI] [PubMed] [Google Scholar]
  • 241.Toprak, C. & Yigitaslan, S. Alagebrium and complications of diabetes mellitus. Eurasia. J. Med.51, 285–292 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Zhu, J. X. et al. Diabetic peripheral neuropathy: pathogenetic mechanisms and treatment. Front. Endocrinol. (Lausanne)14, 1265372 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Kamei, J., Mizoguchi, H., Narita, M. & Tseng, L. F. Therapeutic potential of PKC inhibitors in painful diabetic neuropathy. Expert Opin. Investig. Drugs10, 1653–1664 (2001). [DOI] [PubMed] [Google Scholar]
  • 244.Dal-Cim, T. et al. Guanosine prevents oxidative damage and glutamate uptake impairment induced by oxygen/glucose deprivation in cortical astrocyte cultures: involvement of A1 and A2A adenosine receptors and PI3K, MEK, and PKC pathways. Purinergic Signal15, 465–476 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Zan, Y., Kuai, C. X., Qiu, Z. X. & Huang, F. Berberine ameliorates diabetic neuropathy: TRPV1 modulation by PKC pathway. Am. J. Chin. Med.45, 1709–1723 (2017). [DOI] [PubMed] [Google Scholar]
  • 246.Wiggin, T. D. et al. Elevated triglycerides correlate with progression of diabetic neuropathy. Diabetes58, 1634–1640 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Yorek, M. S. et al. Effect of diet-induced obesity or type 1 or type 2 diabetes on corneal nerves and peripheral neuropathy in C57Bl/6J mice. J. Peripher. Nerv. Syst.20, 24–31 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Davidson, E. P., Coppey, L. J., Calcutt, N. A., Oltman, C. L. & Yorek, M. A. Diet-induced obesity in Sprague-Dawley rats causes microvascular and neural dysfunction. Diabetes Metab. Res. Rev.26, 306–318 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Sajic, M. et al. High dietary fat consumption impairs axonal mitochondrial function in vivo. J. Neurosci.41, 4321–4334 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Rumora, A. E. et al. The divergent roles of dietary saturated and monounsaturated fatty acids on nerve function in murine models of obesity. J. Neurosci.39, 3770–3781 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Hinder, L. M. et al. Dietary reversal of neuropathy in a murine model of prediabetes and metabolic syndrome. Dis. Model. Mech.10, 717–725 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Padilla, A., Descorbeth, M., Almeyda, A. L., Payne, K. & De Leon, M. Hyperglycemia magnifies Schwann cell dysfunction and cell death triggered by PA-induced lipotoxicity. Brain Res1370, 64–79 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Vincent, A. M. et al. Dyslipidemia-induced neuropathy in mice: the role of oxLDL/LOX-1. Diabetes58, 2376–2385 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Honjo, T. et al. Essential role of NOXA1 in generation of reactive oxygen species induced by oxidized low-density lipoprotein in human vascular endothelial cells. Endothelium15, 137–141 (2008). [DOI] [PubMed] [Google Scholar]
  • 255.Nowicki, M. et al. Oxidized low-density lipoprotein (oxLDL)-induced cell death in dorsal root ganglion cell cultures depends not on the lectin-like oxLDL receptor-1 but on the toll-like receptor-4. J. Neurosci. Res.88, 403–412 (2010). [DOI] [PubMed] [Google Scholar]
  • 256.Cotter, M. A. & Cameron, N. E. Effect of the NAD(P)H oxidase inhibitor, apocynin, on peripheral nerve perfusion and function in diabetic rats. Life Sci.73, 1813–1824 (2003). [DOI] [PubMed] [Google Scholar]
  • 257.Vincent, A. M. et al. Receptor for advanced glycation end products activation injures primary sensory neurons via oxidative stress. Endocrinology148, 548–558 (2007). [DOI] [PubMed] [Google Scholar]
  • 258.O’Brien, P. D. et al. Integrated lipidomic and transcriptomic analyses identify altered nerve triglycerides in mouse models of prediabetes and type 2 diabetes. Dis. Model. Mech.13, dmm042101 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Hur, J. et al. The identification of gene expression profiles associated with progression of human diabetic neuropathy. Brain134, 3222–3235 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.Kristensen, F. P. et al. Statin therapy and risk of polyneuropathy in type 2 diabetes: a Danish cohort study. Diabetes Care43, 2945–2952 (2020). [DOI] [PubMed] [Google Scholar]
  • 261.Tierney, E. F., Thurman, D. J., Beckles, G. L. & Cadwell, B. L. Association of statin use with peripheral neuropathy in the U.S. population 40 years of age or older. J. Diabetes5, 207–215 (2013). [DOI] [PubMed] [Google Scholar]
  • 262.Rogers, M. A. et al. Dynamin-related protein 1 inhibition reduces hepatic PCSK9 secretion. Cardiovasc. Res.117, 2340–2353 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Cui, N. et al. Protective effect of alirocumab, a PCSK9 inhibitor, on the sciatic nerve of rats with diabetic peripheral neuropathy. Endocr. J.71, 233–244 (2024). [DOI] [PubMed] [Google Scholar]
  • 264.Lin, Q. X. et al. Oxidative stress in diabetic peripheral neuropathy: pathway and mechanism-based treatment. Mol. Neurobiol.60, 4574–4594 (2023). [DOI] [PubMed] [Google Scholar]
  • 265.Wright, E. Jr, Scism-Bacon, J. L. & Glass, L. C. Oxidative stress in type 2 diabetes: the role of fasting and postprandial glycaemia. Int. J. Clin. Pract.60, 308–314 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Rekatsina, M. et al. Pathophysiology and therapeutic perspectives of oxidative stress and neurodegenerative diseases: a narrative review. Adv. Ther.37, 113–139 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Urner, S., Ho, F., Jha, J. C., Ziegler, D. & Jandeleit-Dahm, K. NADPH oxidase inhibition: preclinical and clinical studies in diabetic complications. Antioxid. Redox Signal.33, 415–434 (2020). [DOI] [PubMed] [Google Scholar]
  • 268.Kim, B. & Feldman, E. L. Insulin resistance as a key link for the increased risk of cognitive impairment in the metabolic syndrome. Exp. Mol. Med.47, e149 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269.Cai, C. et al. Mitochondrial quality control in diabetic cardiomyopathy: from molecular mechanisms to therapeutic strategies. Int. J. Biol. Sci.18, 5276–5290 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Tan, Y. et al. Mechanisms of diabetic cardiomyopathy and potential therapeutic strategies: preclinical and clinical evidence. Nat. Rev. Cardiol.17, 585–607 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Chowdhury, S. K., Smith, D. R. & Fernyhough, P. The role of aberrant mitochondrial bioenergetics in diabetic neuropathy. Neurobiol. Dis.51, 56–65 (2013). [DOI] [PubMed] [Google Scholar]
  • 272.Whitley, B. N., Engelhart, E. A. & Hoppins, S. Mitochondrial dynamics and their potential as a therapeutic target. Mitochondrion49, 269–283 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Sims, N. A. & Walsh, N. C. GP130 cytokines and bone remodelling in health and disease. BMB Rep.43, 513–523 (2010). [DOI] [PubMed] [Google Scholar]
  • 274.Zhu, H. et al. PARP inhibitors in pancreatic cancer: molecular mechanisms and clinical applications. Mol. Cancer19, 49 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Patel, S., Pangarkar, A., Mahajan, S. & Majumdar, A. Therapeutic potential of endoplasmic reticulum stress inhibitors in the treatment of diabetic peripheral neuropathy. Metab. Brain Dis.38, 1841–1856 (2023). [DOI] [PubMed] [Google Scholar]
  • 276.Kandhare, A. D., Mukherjee, A. & Bodhankar, S. L. Antioxidant for treatment of diabetic nephropathy: a systematic review and meta-analysis. Chem. Biol. Interact.278, 212–221 (2017). [DOI] [PubMed] [Google Scholar]
  • 277.Hsieh, R. Y., Huang, I. C., Chen, C. & Sung, J. Y. Effects of oral alpha-lipoic acid treatment on diabetic polyneuropathy: a meta-analysis and systematic review. Nutrients15, 3634 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Jiang, Q. et al. Mitochondria-targeted antioxidants: a step towards disease treatment. Oxid. Med. Cell. Longev.2020, 8837893 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279.Rocha, M. et al. Perspectives and potential applications of mitochondria-targeted antioxidants in cardiometabolic diseases and type 2 diabetes. Med. Res. Rev.34, 160–189 (2014). [DOI] [PubMed] [Google Scholar]
  • 280.Sahebkar, A., Serban, M. C., Ursoniu, S. & Banach, M. Effect of curcuminoids on oxidative stress: a systematic review and meta-analysis of randomized controlled trials. J. Funct. Foods18, 898–909 (2015). [Google Scholar]
  • 281.Praet, S. F. & van Loon, L. J. Exercise therapy in type 2 diabetes. Acta Diabetol.46, 263–278 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282.Kahal, H. et al. Effect of induced hypoglycemia on inflammation and oxidative stress in type 2 diabetes and control subjects. Sci. Rep.10, 4750 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283.Ji, R. R., Chamessian, A. & Zhang, Y. Q. Pain regulation by non-neuronal cells and inflammation. Science354, 572–577 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284.Gonçalves, N. P., Vægter, C. B. & Pallesen, L. T. Peripheral glial cells in the development of diabetic neuropathy. Front. Neurol.9, 268 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285.Yorek, M. Treatment for diabetic peripheral neuropathy: what have we learned from animal models? Curr. Diabetes Rev.18, e040521193121 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286.O’Brien, P. D., Sakowski, S. A. & Feldman, E. L. Mouse models of diabetic neuropathy. ILAR J.54, 259–272 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287.Doupis, J. et al. Microvascular reactivity and inflammatory cytokines in painful and painless peripheral diabetic neuropathy. J. Clin. Endocrinol. Metab.94, 2157–2163 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288.Liu, P. et al. Role of macrophages in peripheral nerve injury and repair. Neural Regen. Res.14, 1335–1342 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 289.Dubový, P. Wallerian degeneration and peripheral nerve conditions for both axonal regeneration and neuropathic pain induction. Ann. Anat.193, 267–275 (2011). [DOI] [PubMed] [Google Scholar]
  • 290.Roman-Pintos, L. M., Villegas-Rivera, G., Rodriguez-Carrizalez, A. D., Miranda-Diaz, A. G. & Cardona-Munoz, E. G. Diabetic polyneuropathy in type 2 diabetes mellitus: inflammation, oxidative stress, and mitochondrial function. J. Diabetes Res.2016, 3425617 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291.Sun, Q. et al. Serum adiponectin levels are positively associated with diabetic peripheral neuropathy in Chinese patients with type 2 diabetes. Front. Endocrinol.11, 567959 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292.O’Brien, P. D., Hinder, L. M., Callaghan, B. C. & Feldman, E. L. Neurological consequences of obesity. Lancet Neurol.16, 465–477 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293.Belkouch, M. et al. The chemokine CCL2 increases Nav1.8 sodium channel activity in primary sensory neurons through a Gβγ-dependent mechanism. J. Neurosci.31, 18381–18390 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 294.Ristikj-Stomnaroska, D., Risteska-Nejashmikj, V. & Papazova, M. Role of inflammation in the pathogenesis of diabetic peripheral neuropathy. Open Access Maced. J. Med. Sci.7, 2267–2270 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295.Alexander, J. K. et al. Macrophage migration inhibitory factor (MIF) is essential for inflammatory and neuropathic pain and enhances pain in response to stress. Exp. Neurol.236, 351–362 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296.Vareniuk, I., Pavlov, I. A. & Obrosova, I. G. Inducible nitric oxide synthase gene deficiency counteracts multiple manifestations of peripheral neuropathy in a streptozotocin-induced mouse model of diabetes. Diabetologia51, 2126–2133 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 297.Pop-Busui, R., Ang, L., Holmes, C., Gallagher, K. & Feldman, E. L. Inflammation as a therapeutic target for diabetic neuropathies. Curr. Diab Rep.16, 29 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 298.Herder, C. et al. Proinflammatory cytokines predict the incidence and progression of distal sensorimotor polyneuropathy: KORA F4/FF4 study. Diabetes Care40, 569–576 (2017). [DOI] [PubMed] [Google Scholar]
  • 299.Tesfaye, S. & Selvarajah, D. Advances in the epidemiology, pathogenesis and management of diabetic peripheral neuropathy. Diabetes Metab. Res. Rev.28, 8–14 (2012). [DOI] [PubMed] [Google Scholar]
  • 300.Malik, R. A. et al. Sural nerve pathology in diabetic patients with minimal but progressive neuropathy. Diabetologia48, 578–585 (2005). [DOI] [PubMed] [Google Scholar]
  • 301.Edwards, J. L., Vincent, A. M., Cheng, H. T. & Feldman, E. L. Diabetic neuropathy: mechanisms to management. Pharmacol. Ther.120, 1–34 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302.Hill, R. E. & Williams, P. E. Perineurial cell basement membrane thickening and myelinated nerve fibre loss in diabetic and nondiabetic peripheral nerve. J. Neurol. Sci.217, 157–163 (2004). [DOI] [PubMed] [Google Scholar]
  • 303.Thrainsdottir, S. et al. Endoneurial capillary abnormalities presage deterioration of glucose tolerance and accompany peripheral neuropathy in man. Diabetes52, 2615–2622 (2003). [DOI] [PubMed] [Google Scholar]
  • 304.Malik, R. A. et al. Sural nerve fibre pathology in diabetic patients with mild neuropathy: relationship to pain, quantitative sensory testing and peripheral nerve electrophysiology. Acta Neuropathol.101, 367–374 (2001). [DOI] [PubMed] [Google Scholar]
  • 305.Cameron, N. E., Gibson, T. M., Nangle, M. R. & Cotter, M. A. Inhibitors of advanced glycation end product formation and neurovascular dysfunction in experimental diabetes. Ann. N. Y. Acad. Sci.1043, 784–792 (2005). [DOI] [PubMed] [Google Scholar]
  • 306.Coppey, L. J. et al. ACE inhibitor or angiotensin II receptor antagonist attenuates diabetic neuropathy in streptozotocin-induced diabetic rats. Diabetes55, 341–348 (2006). [DOI] [PubMed] [Google Scholar]
  • 307.Amir, A. A. et al. Management of neuropathic pain in patients with diabetic peripheral neuropathy and low back pain in Saudi Arabia: evidence and gaps. J. Fam. Community Med.28, 155–163 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 308.Schratzberger, P. et al. Reversal of experimental diabetic neuropathy by VEGF gene transfer. J. Clin. Invest.107, 1083–1092 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 309.Javed, S., Petropoulos, I. N., Alam, U. & Malik, R. A. Treatment of painful diabetic neuropathy. Ther. Adv. Chronic Dis.6, 15–28 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 310.Tabit, C. E., Chung, W. B., Hamburg, N. M. & Vita, J. A. Endothelial dysfunction in diabetes mellitus: molecular mechanisms and clinical implications. Rev. Endocr. Metab. Disord.11, 61–74 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 311.Geraldes, P. & King, G. L. Activation of protein kinase C isoforms and its impact on diabetic complications. Circ. Res.106, 1319–1331 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 312.Leinninger, G. M., Vincent, A. M. & Feldman, E. L. The role of growth factors in diabetic peripheral neuropathy. J. Peripher. Nerv. Syst.9, 26–53 (2004). [DOI] [PubMed] [Google Scholar]
  • 313.Østergaard, L. et al. The effects of capillary dysfunction on oxygen and glucose extraction in diabetic neuropathy. Diabetologia58, 666–677 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 314.Vincent, A. M. & Feldman, E. L. New insights into the mechanisms of diabetic neuropathy. Rev. Endocr. Metab. Disord.5, 227–236 (2004). [DOI] [PubMed] [Google Scholar]
  • 315.Chen, J. et al. miRNA-155 silencing reduces sciatic nerve injury in diabetic peripheral neuropathy. J. Mol. Endocrinol.63, 227–238 (2019). [DOI] [PubMed] [Google Scholar]
  • 316.Tesfaye, S. et al. Diagnosis, management and impact of painful diabetic peripheral neuropathy: A patient survey in four European countries. J. Diabetes Complicat.37, 108417 (2023). [DOI] [PubMed] [Google Scholar]
  • 317.Calabek, B., Callaghan, B. & Feldman, E. L. Chapter 22-Therapy for diabetic neuropathy: an overview. in Handbook of Clinical Neurology Vol. 126 (eds Zochodne, D. W. & Malik, R. A.), 317–333 (Elsevier, 2014). [DOI] [PubMed]
  • 318.Huang, T. J., Verkhratsky, A. & Fernyhough, P. Insulin enhances mitochondrial inner membrane potential and increases ATP levels through phosphoinositide 3-kinase in adult sensory neurons. Mol. Cell. Neurosci.28, 42–54 (2005). [DOI] [PubMed] [Google Scholar]
  • 319.Petersen, M. C. & Shulman, G. I. Mechanisms of insulin action and insulin resistance. Physiol. Rev.98, 2133–2223 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 320.Sangwung, P., Petersen, K. F., Shulman, G. I. & Knowles, J. W. Mitochondrial dysfunction, insulin resistance, and potential genetic implications. Endocrinology161, bqaa017 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 321.Grote, C. W. & Wright, D. E. A role for insulin in diabetic neuropathy. Front. Neurosci.10, 581 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 322.Cheng, H. et al. Mitochondrial dysfunction plays a key role in the development of neurodegenerative diseases in diabetes. Am. J. Physiol. Endocrinol. Metab.318, E750–E764 (2020). [DOI] [PubMed] [Google Scholar]
  • 323.Sugimoto, K., Murakawa, Y. & Sima, A. A. Expression and localization of insulin receptor in rat dorsal root ganglion and spinal cord. J. Peripher. Nerv. Syst.7, 44–53 (2002). [DOI] [PubMed] [Google Scholar]
  • 324.Nowicki, M., Kosacka, J., Serke, H., Blüher, M. & Spanel-Borowski, K. Altered sciatic nerve fiber morphology and endoneural microvessels in mouse models relevant for obesity, peripheral diabetic polyneuropathy, and the metabolic syndrome. J. Neurosci. Res.90, 122–131 (2012). [DOI] [PubMed] [Google Scholar]
  • 325.Huang, T. J. et al. Insulin prevents depolarization of the mitochondrial inner membrane in sensory neurons of type 1 diabetic rats in the presence of sustained hyperglycemia. Diabetes52, 2129–2136 (2003). [DOI] [PubMed] [Google Scholar]
  • 326.Coughlan, K. A., Valentine, R. J., Ruderman, N. B. & Saha, A. K. AMPK activation: a therapeutic target for type 2 diabetes? Diabetes Metab. Syndr. Obes.7, 241–253 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 327.Kim, B., McLean, L. L., Philip, S. S. & Feldman, E. L. Hyperinsulinemia induces insulin resistance in dorsal root ganglion neurons. Endocrinology152, 3638–3647 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 328.Roy Chowdhury, S. K. et al. Impaired adenosine monophosphate-activated protein kinase signalling in dorsal root ganglia neurons is linked to mitochondrial dysfunction and peripheral neuropathy in diabetes. Brain135, 1751–1766 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 329.Akhter, F., Chen, D., Akhter, A., Yan, S. F. & Yan, S. S. Age-dependent accumulation of dicarbonyls and advanced glycation endproducts (AGEs) associates with mitochondrial stress. Free Radic. Biol. Med.164, 429–438 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 330.Aziz, N. et al. New horizons in diabetic neuropathies: an updated review on their pathology, diagnosis, mechanism, screening techniques, pharmacological, and future approaches. Curr. Diabetes Rev.20, e201023222416 (2024). [DOI] [PubMed] [Google Scholar]
  • 331.Sullivan, K. A. & Feldman, E. L. New developments in diabetic neuropathy. Curr. Opin. Neurol.18, 586–590 (2005). [DOI] [PubMed] [Google Scholar]
  • 332.Brussee, V., Cunningham, F. A. & Zochodne, D. W. Direct insulin signaling of neurons reverses diabetic neuropathy. Diabetes53, 1824–1830 (2004). [DOI] [PubMed] [Google Scholar]
  • 333.Mohammadi, R., Esmaeil-Sani, Z. & Amini, K. Effect of local administration of insulin-like growth factor I combined with inside-out artery graft on peripheral nerve regeneration. Injury44, 1295–1301 (2013). [DOI] [PubMed] [Google Scholar]
  • 334.Guo, G., Kan, M., Martinez, J. A. & Zochodne, D. W. Local insulin and the rapid regrowth of diabetic epidermal axons. Neurobiol. Dis.43, 414–421 (2011). [DOI] [PubMed] [Google Scholar]
  • 335.Albers, J. W. & Pop-Busui, R. Diabetic neuropathy: mechanisms, emerging treatments, and subtypes. Curr. Neurol. Neurosci. Rep.14, 473 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 336.Wada, R. & Yagihashi, S. Role of advanced glycation end products and their receptors in development of diabetic neuropathy. Ann. N. Y. Acad. Sci.1043, 598–604 (2005). [DOI] [PubMed] [Google Scholar]
  • 337.Grote, C. W., Morris, J. K., Ryals, J. M., Geiger, P. C. & Wright, D. E. Insulin receptor substrate 2 expression and involvement in neuronal insulin resistance in diabetic neuropathy. Exp. Diabetes Res.2011, 212571 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 338.Moran, A. et al. Interleukin-1 antagonism in type 1 diabetes of recent onset: two multicentre, randomised, double-blind, placebo-controlled trials. Lancet381, 1905–1915 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 339.Yorek, M. A. The role of oxidative stress in diabetic vascular and neural disease. Free Radic. Res.37, 471–480 (2003). [DOI] [PubMed] [Google Scholar]
  • 340.Martin, C. L., Albers, J. W., Pop-Busui, R. & Group, D. E. R. Neuropathy and related findings in the diabetes control and complications trial/epidemiology of diabetes interventions and complications study. Diabetes Care37, 31–38 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 341.Li, Y., Wang, S. Y., Xiao, Y. H., Liu, B. & Pang, J. L. Nerve growth factor enhances the therapeutic effect of mesenchymal stem cells on diabetic periodontitis. Exp. Ther. Med.22, 1013 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 342.Lee, H. Y. et al. Human adipose stem cells improve mechanical allodynia and enhance functional recovery in a rat model of neuropathic pain. Tissue Eng. Part A21, 2044–2052 (2015). [DOI] [PubMed] [Google Scholar]
  • 343.Fonseca, V. A. et al. Metanx in type 2 diabetes with peripheral neuropathy: a randomized trial. Am. J. Med.126, 141–149 (2013). [DOI] [PubMed] [Google Scholar]
  • 344.Shevalye, H. et al. Metanx alleviates multiple manifestations of peripheral neuropathy and increases intraepidermal nerve fiber density in Zucker diabetic fatty rats. Diabetes61, 2126–2133 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 345.Yang, H. Nanoparticle-mediated brain-specific drug delivery, imaging, and diagnosis. Pharm. Res.27, 1759–1771 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 346.Duan, L. Y. et al. Nanoparticle-based drug delivery systems: an inspiring therapeutic strategy for neurodegenerative diseases. Polymers15, 2196 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 347.Goncalves, N. P. et al. Schwann cell interactions with axons and microvessels in diabetic neuropathy. Nat. Rev. Neurol.13, 135–147 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 348.Liu, Y. P., Shao, S. J. & Guo, H. D. Schwann cells apoptosis is induced by high glucose in diabetic peripheral neuropathy. Life Sci.248, 117459 (2020). [DOI] [PubMed] [Google Scholar]
  • 349.Zhang, X. et al. TXNIP, a novel key factor to cause Schwann cell dysfunction in diabetic peripheral neuropathy, under the regulation of PI3K/Akt pathway inhibition-induced DNMT1 and DNMT3a overexpression. Cell Death Dis.12, 642 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 350.Ogata, T. et al. Opposing extracellular signal-regulated kinase and Akt pathways control Schwann cell myelination. J. Neurosci.24, 6724–6732 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 351.Viader, A. et al. Aberrant Schwann cell lipid metabolism linked to mitochondrial deficits leads to axon degeneration and neuropathy. Neuron77, 886–898 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 352.Pande, M. et al. Transcriptional profiling of diabetic neuropathy in the BKS db/db mouse: a model of type 2 diabetes. Diabetes60, 1981–1989 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 353.Fideles, S. O. M. et al. Influence of the neuroprotective properties of quercetin on regeneration and functional recovery of the nervous system. Antioxidants12, 149 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 354.Li, J. J., Guan, R. Q. & Pan, L. M. Mechanism of Schwann cells in diabetic peripheral neuropathy: a review. Medicine102, e32653 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 355.Jang, S. Y. et al. Schwann cell dedifferentiation-associated demyelination leads to exocytotic myelin clearance in inflammatory segmental demyelination. Glia65, 1848–1862 (2017). [DOI] [PubMed] [Google Scholar]
  • 356.Babetto, E., Wong, K. M. & Beirowski, B. A glycolytic shift in Schwann cells supports injured axons. Nat. Neurosci.23, 1215–1228 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 357.Beirowski, B. Emerging evidence for compromised axonal bioenergetics and axoglial metabolic coupling as drivers of neurodegeneration. Neurobiol. Dis.170, 105751 (2022). [DOI] [PubMed] [Google Scholar]
  • 358.Selvarajah, D., Wilkinson, I. D., Davies, J., Gandhi, R. & Tesfaye, S. Central nervous system involvement in diabetic neuropathy. Curr. Diab Rep.11, 310–322 (2011). [DOI] [PubMed] [Google Scholar]
  • 359.Tesfaye, S. et al. Diabetic peripheral neuropathy may not be as its name suggests: evidence from magnetic resonance imaging. Pain157(Suppl 1), S72–S80 (2016). [DOI] [PubMed] [Google Scholar]
  • 360.Selvarajah, D. et al. Magnetic resonance neuroimaging study of brain structural differences in diabetic peripheral neuropathy. Diabetes Care37, 1681–1688 (2014). [DOI] [PubMed] [Google Scholar]
  • 361.Gustin, S. M. et al. Thalamic activity and biochemical changes in individuals with neuropathic pain after spinal cord injury. Pain155, 1027–1036 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 362.Cauda, F. et al. Low-frequency BOLD fluctuations demonstrate altered thalamocortical connectivity in diabetic neuropathic pain. BMC Neurosci.10, 138 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 363.Kapur, D. Neuropathic pain and diabetes. Diabetes Metab. Res. Rev.19, S9–S15 (2003). [DOI] [PubMed] [Google Scholar]
  • 364.Hansen, T. M. et al. Brain spectroscopy reveals that N-acetylaspartate is associated to peripheral sensorimotor neuropathy in type 1 diabetes. J. Diabetes Complicat.33, 323–328 (2019). [DOI] [PubMed] [Google Scholar]
  • 365.Selvarajah, D. et al. Thalamic neuronal dysfunction and chronic sensorimotor distal symmetrical polyneuropathy in patients with type 1 diabetes mellitus. Diabetologia51, 2088–2092 (2008). [DOI] [PubMed] [Google Scholar]
  • 366.Selvarajah, D. et al. Structural and functional abnormalities of the primary somatosensory cortex in diabetic peripheral neuropathy: a multimodal MRI study. Diabetes68, 796–806 (2019). [DOI] [PubMed] [Google Scholar]
  • 367.Attal, N. et al. EFNS guidelines on the pharmacological treatment of neuropathic pain: 2010 revision. Eur. J. Neurol.17, 1113–e1188 (2010). [DOI] [PubMed] [Google Scholar]
  • 368.Gonzalez, C. D. et al. The emerging role of autophagy in the pathophysiology of diabetes mellitus. Autophagy7, 2–11 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 369.Zhao, X. et al. The role of autophagy in the treatment of type II diabetes and its complications: a review. Front. Endocrinol.14, 1228045 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 370.Tian, H., Zheng, X. & Wang, H. Isorhapontigenin ameliorates high glucose-induced podocyte and vascular endothelial cell injuries via mitigating oxidative stress and autophagy through the AMPK/Nrf2 pathway. Int. Urol. Nephrol.55, 423–436 (2023). [DOI] [PubMed] [Google Scholar]
  • 371.Liu, F. M., Li, W., Xie, D. Y., Du, X. J. & Chen, X. L. Metformin ameliorates peripheral neuropathy in diabetic rats by downregulating autophagy via the AMPK pathway. Arch. Endocrinol. Metab.68, e240137 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 372.Fleming, A., Noda, T., Yoshimori, T. & Rubinsztein, D. C. Chemical modulators of autophagy as biological probes and potential therapeutics. Nat. Chem. Biol.7, 9–17 (2011). [DOI] [PubMed] [Google Scholar]
  • 373.Pasquier, B. Autophagy inhibitors. Cell. Mol. Life Sci.73, 985–1001 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 374.Yin, Y. D., Qu, H., Yang, Q. N., Fang, Z. H. & Gao, R. Astragaloside IV alleviates Schwann cell injury in diabetic peripheral neuropathy by regulating microRNA-155-mediated autophagy. Phytomedicine92, 153749 (2021). [DOI] [PubMed] [Google Scholar]
  • 375.Jin, Y. et al. Transplantation of endothelial progenitor cells attenuated paraquat-induced acute lung injury via miR-141-3p-Notch-Nrf2 axis. Cell Biosci.8, 21 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 376.Martin, L. J. Neuronal cell death in nervous system development, disease, and injury (Review). Int. J. Mol. Med.7, 455–478 (2001). [PubMed] [Google Scholar]
  • 377.Pradelli, L. A., Beneteau, M. & Ricci, J. E. Mitochondrial control of caspase-dependent and-independent cell death. Cell. Mol. Life Sci.67, 1589–1597 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 378.Erekat, N. S. Programmed cell death in diabetic nephropathy: a review of apoptosis, autophagy, and necroptosis. Med. Sci. Monit.28, e937766 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 379.Zhan, J. F., Huang, H. W., Huang, C., Hu, L. L. & Xu, W. W. Long non-coding RNA NEAT1 regulates pyroptosis in diabetic nephropathy via mediating the miR-34c/NLRP3 axis. Kidney Blood Press. Res.45, 589–602 (2020). [DOI] [PubMed] [Google Scholar]
  • 380.Ramesh, G., MacLean, A. G. & Philipp, M. T. Cytokines and chemokines at the crossroads of neuroinflammation, neurodegeneration, and neuropathic pain. Mediators Inflamm.2013, 480739 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 381.Simon, D. W. et al. The far-reaching scope of neuroinflammation after traumatic brain injury. Nat. Rev. Neurol.13, 171–191 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 382.Zhang, Q. et al. Glucagon-like peptide-1 receptor agonist attenuates diabetic neuropathic pain via inhibition of NOD-like receptor protein 3 inflammasome in brain microglia. Diabetes Res. Clin. Pract.186, 109806 (2022). [DOI] [PubMed] [Google Scholar]
  • 383.Cao, Z. H. et al. Pyroptosis in diabetes and diabetic nephropathy. Clin. Chim. Acta531, 188–196 (2022). [DOI] [PubMed] [Google Scholar]
  • 384.Kang, P. P., Tian, C. W. & Jia, C. Q. Association of RAGE gene polymorphisms with type 2 diabetes mellitus, diabetic retinopathy and diabetic nephropathy. Gene500, 1–9 (2012). [DOI] [PubMed] [Google Scholar]
  • 385.Lango, H. et al. Assessing the combined impact of 18 common genetic variants of modest effect sizes on type 2 diabetes risk. Diabetes57, 3129–3135 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 386.Zhang, Y. F. et al. Genome-wide association studies reveal new genetic targets for five panicle traits of international rice varieties. Rice Sci.22, 217–226 (2015). [Google Scholar]
  • 387.Reddy, M. A. & Natarajan, R. Epigenetic mechanisms in diabetic vascular complications. Cardiovasc. Res.90, 421–429 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 388.Villeneuve, L. M. & Natarajan, R. The role of epigenetics in the pathology of diabetic complications. Am. J. Physiol. Ren. Physiol.299, F14–F25 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 389.Rafehi, H., El-Osta, A. & Karagiannis, T. C. Epigenetic mechanisms in the pathogenesis of diabetic foot ulcers. J. Diabetes Complicat.26, 554–561 (2012). [DOI] [PubMed] [Google Scholar]
  • 390.Gouri, A., Dekaken, A., Chefrour, M. & Guieu, R. Epigenetics and diabetes: current and perspective. Int. J. Diabetes Vasc. Dis. Res3, 94–98 (2015). [Google Scholar]
  • 391.Wu, M., Feng, Y. H. & Shi, X. H. Advances with long non-coding RNAs in diabetic peripheral neuropathy. Diabetes Metab. Syndr. Obes.13, 1429–1434 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 392.Tao, Y. et al. Spinal-specific super enhancer in neuropathic pain. J. Neurosci.43, 8547–8561 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 393.Meydan, C., Üçeyler, N. & Soreq, H. Non-coding RNA regulators of diabetic polyneuropathy. Neurosci. Lett.731, 135058 (2020). [DOI] [PubMed] [Google Scholar]
  • 394.Filardi, T. et al. Non-coding RNA: role in gestational diabetes pathophysiology and complications. Int. J. Mol. Sci.21, 4020 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 395.Gao, X. Y. et al. LncRNA, an emerging approach for neurological diseases treatment by regulating microglia polarization. Front. Neurosci.16, 903472 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 396.Wang, C. et al. The construction and analysis of lncRNA–miRNA–mRNA competing endogenous RNA network of schwann cells in diabetic peripheral neuropathy. Front. Bioeng. Biotechnol.8, 490 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 397.Lu, Y. et al. Ketogenic diet attenuates oxidative stress and inflammation after spinal cord injury by activating Nrf2 and suppressing the NF-kappaB signaling pathways. Neurosci. Lett.683, 13–18 (2018). [DOI] [PubMed] [Google Scholar]
  • 398.Nassirpour, R. et al. Identification of promising urinary microrna biomarkers in two rat models of glomerular injury. Toxicol. Sci.148, 35–47 (2015). [DOI] [PubMed] [Google Scholar]
  • 399.Jiang, P. Y., Zhu, X. J., Zhang, Y. N., Zhou, F. F. & Yang, X. F. Protective effects of apigenin on LPS-induced endometritis via activating Nrf2 signaling pathway. Microb. Pathog.123, 139–143 (2018). [DOI] [PubMed] [Google Scholar]
  • 400.Wang, L. et al. Exosomes derived from Schwann cells ameliorate peripheral neuropathy in type 2 diabetic mice. Diabetes69, 749–759 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 401.Liu, Y. P. et al. Schwann cells-derived exosomal miR-21 participates in high glucose regulation of neurite outgrowth. iScience25, 105141 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 402.Degterev, A. et al. Identification of RIP1 kinase as a specific cellular target of necrostatins. Nat. Chem. Biol.4, 313–321 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 403.Huang, Y. H. et al. Dysbiosis and implication of the gut microbiota in diabetic retinopathy. Front. Cell. Infect. Microbiol.11, 646348 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 404.Zaky, A., Glastras, S. J., Wong, M. Y. W., Pollock, C. A. & Saad, S. The role of the gut microbiome in diabetes and obesity-related kidney disease. Int. J. Mol. Sci.22, 9641 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 405.Snelson, M., de Pasquale, C., Ekinci, E. I. & Coughlan, M. T. Gut microbiome, prebiotics, intestinal permeability and diabetes complications. Best. Pract. Res. Clin. Endocrinol. Metab.35, 101507 (2021). [DOI] [PubMed] [Google Scholar]
  • 406.Haro, C. et al. The gut microbial community in metabolic syndrome patients is modified by diet. J. Nutr. Biochem.27, 27–31 (2016). [DOI] [PubMed] [Google Scholar]
  • 407.Li, J. et al. Gut microbiota dysbiosis contributes to the development of hypertension. Microbiome5, 14 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 408.Yang, Y. et al. Efficacy of fecal microbiota transplantation in type 2 diabetes mellitus: a systematic review and meta-analysis. Endocrine84, 48–62 (2024). [DOI] [PubMed] [Google Scholar]
  • 409.Lahiri, S. et al. The gut microbiota influences skeletal muscle mass and function in mice. Sci. Transl. Med.11, eaan5662 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 410.Aresti Sanz, J. & El Aidy, S. Microbiota and gut neuropeptides: a dual action of antimicrobial activity and neuroimmune response. Psychopharmacology236, 1597–1609 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 411.Burberry, A. et al. C9orf72 suppresses systemic and neural inflammation induced by gut bacteria. Nature582, 89–94 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 412.Nagase, N., Ikeda, Y., Tsuji, A., Kitagishi, Y. & Matsuda, S. Efficacy of probiotics on the modulation of gut microbiota in the treatment of diabetic nephropathy. World J. Diabetes13, 150 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 413.Xie, J. et al. Protective effect of quercetin on streptozotocin-induced diabetic peripheral neuropathy rats through modulating gut microbiota and reactive oxygen species level. Biomed. Pharmacother.127, 110147 (2020). [DOI] [PubMed] [Google Scholar]
  • 414.Rani, M., Akhilesh, Chouhan, D., Uniyal, A. & Tiwari, V. Fecal microbiota transplantation-mediated rebalancing of the gut–brain axis alleviates cisplatin-induced neuropathic pain. ACS Chem. Neurosci.15, 3655–3664 (2024). [DOI] [PubMed] [Google Scholar]
  • 415.Ojo, O., Feng, Q. Q., Ojo, O. O. & Wang, X. H. The role of dietary fibre in modulating gut microbiota dysbiosis in patients with type 2 diabetes: a systematic review and meta-analysis of randomised controlled trials. Nutrients12, 3239 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 416.Zhou, F. et al. Short-chain fatty acids contribute to neuropathic pain via regulating microglia activation and polarization. Mol. Pain17, 1744806921996520 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 417.Młynarska, E. et al. Exploring the significance of gut microbiota in diabetes pathogenesis and management-a narrative review. Nutrients16, 1938 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 418.Whelan, K. Probiotics and prebiotics in the management of irritable bowel syndrome: a review of recent clinical trials and systematic reviews. Curr. Opin. Clin. Nutr. Metab. Care14, 581–587 (2011). [DOI] [PubMed] [Google Scholar]
  • 419.Corriero, A. et al. Gut microbiota modulation and its implications on neuropathic pain: a comprehensive literature review. Pain Ther.13, 33–51 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 420.von Hehn, C. A., Baron, R. & Woolf, C. J. Deconstructing the neuropathic pain phenotype to reveal neural mechanisms. Neuron73, 638–652 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 421.Colloca, L. et al. Neuropathic pain. Nat. Rev. Dis. Prim.3, 17002 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 422.Shillo, P. et al. Painful and painless diabetic neuropathies: what is the difference? Curr. Diab Rep.19, 32 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 423.Spallone, V. & Greco, C. Painful and painless diabetic neuropathy: one disease or two? Curr. Diab Rep.13, 533–549 (2013). [DOI] [PubMed] [Google Scholar]
  • 424.Hoeijmakers, J. G., Faber, C. G., Lauria, G., Merkies, I. S. & Waxman, S. G. Small-fibre neuropathies–advances in diagnosis, pathophysiology and management. Nat. Rev. Neurol.8, 369–379 (2012). [DOI] [PubMed] [Google Scholar]
  • 425.Bonhof, G. J. et al. Patterns of cutaneous nerve fibre loss and regeneration in type 2 diabetes with painful and painless polyneuropathy. Diabetologia60, 2495–2503 (2017). [DOI] [PubMed] [Google Scholar]
  • 426.Cheng, H. T. et al. Increased axonal regeneration and swellings in intraepidermal nerve fibers characterize painful phenotypes of diabetic neuropathy. J. Pain14, 941–947 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 427.Spallone, V. et al. Clinical correlates of painful diabetic neuropathy and relationship of neuropathic pain with sensorimotor and autonomic nerve function. Eur. J. Pain15, 153–160 (2011). [DOI] [PubMed] [Google Scholar]
  • 428.Scheytt, S., Riediger, N., Braunsdorf, S., Sommer, C. & Uceyler, N. Increased gene expression of growth associated protein-43 in skin of patients with early-stage peripheral neuropathies. J. Neurol. Sci.355, 131–137 (2015). [DOI] [PubMed] [Google Scholar]
  • 429.Cheung, A., Podgorny, P., Martinez, J. A., Chan, C. & Toth, C. Epidermal axonal swellings in painful and painless diabetic peripheral neuropathy. Muscle Nerve51, 505–513 (2015). [DOI] [PubMed] [Google Scholar]
  • 430.Alam, U. et al. Vitamin D deficiency is associated with painful diabetic neuropathy. Diabetes Metab. Res. Rev.37, e3361 (2021). [DOI] [PubMed] [Google Scholar]
  • 431.Gandhi, R. A., Marques, J. L., Selvarajah, D., Emery, C. J. & Tesfaye, S. Painful diabetic neuropathy is associated with greater autonomic dysfunction than painless diabetic neuropathy. Diabetes Care33, 1585–1590 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 432.Austin, P. J. & Moalem-Taylor, G. The neuro-immune balance in neuropathic pain: Involvement of inflammatory immune cells, immune-like glial cells and cytokines. J. Neuroimmunol.229, 26–50 (2010). [DOI] [PubMed] [Google Scholar]
  • 433.Sun, J. S. et al. Minocycline attenuates pain by inhibiting spinal microglia activation in diabetic rats. Mol. Med. Rep.12, 2677–2682 (2015). [DOI] [PubMed] [Google Scholar]
  • 434.Bierhaus, A. et al. Methylglyoxal modification of Nav1.8 facilitates nociceptive neuron firing and causes hyperalgesia in diabetic neuropathy. Nat. Med.18, 926–933 (2012). [DOI] [PubMed] [Google Scholar]
  • 435.Purwata, T. E. High TNF-alpha plasma levels and macrophages iNOS and TNF-alpha expression as risk factors for painful diabetic neuropathy. J. Pain Res.4, 169–175 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 436.Yuen, K. C., Baker, N. R. & Rayman, G. Treatment of chronic painful diabetic neuropathy with isosorbide dinitrate spray: a double-blind placebo-controlled cross-over study. Diabetes Care25, 1699–1703 (2002). [DOI] [PubMed] [Google Scholar]
  • 437.Herder, C. et al. Differential association between biomarkers of subclinical inflammation and painful polyneuropathy: results from the KORA F4 study. Diabetes Care38, 91–96 (2015). [DOI] [PubMed] [Google Scholar]
  • 438.Herder, C. et al. Association between pro-and anti-inflammatory cytokines and depressive symptoms in patients with diabetes—potential differences by diabetes type and depression scores. Transl. Psychiatry7, 1 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 439.Matsushita, Y. et al. Inhibition of transient receptor potential vanilloid type 1 through α2 adrenergic receptors at peripheral nerve terminals relieves pain. J. Vet. Med. Sci.83, 1570–1581 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 440.Li, P., Xiong, D. L., Sun, W. P. & Xu, S. Y. Effects of baicalin on diabetic neuropathic pain involving transient receptor potential vanilloid 1 in the dorsal root ganglia of rats. Neuroreport29, 1492–1498 (2018). [DOI] [PubMed] [Google Scholar]
  • 441.Araya, E. I. et al. Role of peripheral and central TRPV1 receptors in facial heat hyperalgesia in streptozotocin-induced diabetic rats. Brain Res.1670, 146–155 (2017). [DOI] [PubMed] [Google Scholar]
  • 442.Andersson, D. A. et al. Methylglyoxal evokes pain by stimulating TRPA1. PLoS ONE8, e77986 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 443.Hiyama, H. et al. TRPA1 sensitization during diabetic vascular impairment contributes to cold hypersensitivity in a mouse model of painful diabetic peripheral neuropathy. Mol. Pain14, 1744806918789812 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 444.Koivisto, A. et al. Inhibiting TRPA1 ion channel reduces loss of cutaneous nerve fiber function in diabetic animals: sustained activation of the TRPA1 channel contributes to the pathogenesis of peripheral diabetic neuropathy. Pharmacol. Res.65, 149–158 (2012). [DOI] [PubMed] [Google Scholar]
  • 445.Xing, H., Chen, M., Ling, J., Tan, W. H. & Gu, J. G. TRPM8 mechanism of cold allodynia after chronic nerve injury. J. Neurosci.27, 13680–13690 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 446.Pabbidi, M. R. & Premkumar, L. S. Role of transient receptor potential channels TRPV1 and TRPM8 in diabetic peripheral neuropathy. J. Diabetes Treat.2017, 029 (2017). [PMC free article] [PubMed] [Google Scholar]
  • 447.Todorovic, S. M. & Jevtovic-Todorovic, V. Targeting of CaV3.2 t-type calcium channels in peripheral sensory neurons for the treatment of painful diabetic neuropathy. Pflug. Arch.466, 701–706 (2014). [DOI] [PubMed] [Google Scholar]
  • 448.Messinger, R. B. et al. In vivo silencing of the Ca(V)3.2 T-type calcium channels in sensory neurons alleviates hyperalgesia in rats with streptozocin-induced diabetic neuropathy. Pain145, 184–195 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 449.Obradovic, A. et al. CaV3.2 T-type calcium channels in peripheral sensory neurons are important for mibefradil-induced reversal of hyperalgesia and allodynia in rats with painful diabetic neuropathy. PLoS ONE9, e91467 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 450.Joksimovic, S. L. et al. Glycosylation of CaV3.2 channels contributes to the hyperalgesia in peripheral neuropathy of type 1 diabetes. Front. Cell. Neurosci.14, 605312 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 451.Franke, H. & Illes, P. Involvement of P2 receptors in the growth and survival of neurons in the CNS. Pharmacol. Ther.109, 297–324 (2006). [DOI] [PubMed] [Google Scholar]
  • 452.Teixeira, J. M. et al. Diabetes-induced neuropathic mechanical hyperalgesia depends on P2X4 receptor activation in dorsal root ganglia. Neuroscience398, 158–170 (2019). [DOI] [PubMed] [Google Scholar]
  • 453.Teh, K. et al. Somatosensory network functional connectivity differentiates clinical pain phenotypes in diabetic neuropathy. Diabetologia64, 1412–1421 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 454.Segerdahl, A. R., Themistocleous, A. C., Fido, D., Bennett, D. L. & Tracey, I. A brain-based pain facilitation mechanism contributes to painful diabetic polyneuropathy. Brain141, 357–364 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 455.Biessels, G. J., Nobili, F., Teunissen, C. E., Simo, R. & Scheltens, P. Understanding multifactorial brain changes in type 2 diabetes: a biomarker perspective. Lancet Neurol.19, 699–710 (2020). [DOI] [PubMed] [Google Scholar]
  • 456.Littlejohns, T. J. et al. The UK Biobank imaging enhancement of 100,000 participants: rationale, data collection, management and future directions. Nat. Commun.11, 2624 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 457.Woolf, C. J. Central sensitization: iImplications for the diagnosis and treatment of pain. Pain152, S2–S15 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 458.Blair, N. T. & Bean, B. P. Roles of tetrodotoxin (TTX)-sensitive Na+ current, TTX-resistant Na+ current, and Ca2+ current in the action potentials of nociceptive sensory neurons. J. Neurosci.22, 10277–10290 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 459.Vileikyte, L. & Gonzalez, J. S. Recognition and management of psychosocial issues in diabetic neuropathy. Handb. Clin. Neurol.126, 195–209 (2014). [DOI] [PubMed] [Google Scholar]
  • 460.Tracey, I. & Mantyh, P. W. The cerebral signature for pain perception and its modulation. Neuron55, 377–391 (2007). [DOI] [PubMed] [Google Scholar]
  • 461.Fischer, T. Z., Tan, A. M. & Waxman, S. G. Thalamic neuron hyperexcitability and enlarged receptive fields in the STZ model of diabetic pain. Brain Res.1268, 154–161 (2009). [DOI] [PubMed] [Google Scholar]
  • 462.Fischer, T. Z. & Waxman, S. G. Neuropathic pain in diabetes–evidence for a central mechanism. Nat. Rev. Neurol.6, 462–466 (2010). [DOI] [PubMed] [Google Scholar]
  • 463.Watanabe, K. et al. Altered cerebral blood flow in the anterior cingulate cortex is associated with neuropathic pain. J. Neurol. Neurosurg. Psychiatry89, 1082–1087 (2018). [DOI] [PubMed] [Google Scholar]
  • 464.Tseng, M. T., Chiang, M. C., Chao, C. C., Tseng, W. Y. & Hsieh, S. T. fMRI evidence of degeneration-induced neuropathic pain in diabetes: enhanced limbic and striatal activations. Hum. Brain Mapp.34, 2733–2746 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 465.Wilkinson, I. D. et al. Determinants of treatment response in painful diabetic peripheral neuropathy: a combined deep sensory phenotyping and multimodal brain MRI study. Diabetes69, 1804–1814 (2020). [DOI] [PubMed] [Google Scholar]
  • 466.Balcıoğlu, A. S. & Müderrisoğlu, H. Diabetes and cardiac autonomic neuropathy: clinical manifestations, cardiovascular consequences, diagnosis and treatment. World J. Diabetes6, 80 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 467.Freeman, R. Autonomic peripheral neuropathy. Lancet365, 1259–1270 (2005). [DOI] [PubMed] [Google Scholar]
  • 468.Hindricks, G. et al. 2020 ESC Guidelines for the diagnosis and management of atrial fibrillation developed in collaboration with the European Association for Cardio-Thoracic Surgery (EACTS) The Task Force for the diagnosis and management of atrial fibrillation of the European Society of Cardiology (ESC) Developed with the special contribution of the European Heart Rhythm Association (EHRA) of the ESC. Eur. Heart J.42, 373–498 (2021). [DOI] [PubMed] [Google Scholar]
  • 469.Ybarra-Muñoz, J. et al. Cardiovascular disease predicts diabetic peripheral polyneuropathy in subjects with type 2 diabetes: a 10-year prospective study. Eur. J. Cardiovasc. Nurs.15, 248–254 (2016). [DOI] [PubMed] [Google Scholar]
  • 470.Camilleri, M., Parkman, H. P., Shafi, M. A., Abell, T. L. & Gerson, L. Clinical guideline: management of gastroparesis. Am. J. Gastroenterol.108, 18–37 (2013). quiz 38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 471.Sellin, J. H. & Chang, E. B. Therapy insight: gastrointestinal complications of diabetes—pathophysiology and management. Nat. Clin. Pract. Gastroenterol. Hepatol.5, 162–171 (2008). [DOI] [PubMed] [Google Scholar]
  • 472.Kaur, N., Kishore, L. & Singh, R. Diabetic autonomic neuropathy: pathogenesis to pharmacological management. J. Diabetes Metab.5, 1–8 (2014). [Google Scholar]
  • 473.Zawada, A. E., Moszak, M., Skrzypczak, D. & Grzymisławski, M. Gastrointestinal complications in patients with diabetes mellitus. Adv. Clin. Exp. Med.27, 567–572 (2018). [DOI] [PubMed] [Google Scholar]
  • 474.Ahmed, M. S. O., Forde, H. & Smith, D. Diabetic gastroparesis: clinical features, diagnosis and management. Ir. J. Med. Sci.192, 1687–1694 (2023). [DOI] [PubMed] [Google Scholar]
  • 475.Brown, J. S. et al. Urologic complications of diabetes. Diabetes Care28, 177–185 (2005). [DOI] [PubMed] [Google Scholar]
  • 476.Koppiker, N., Boolell, M. & Price, D. Recent advances in the treatment of erectile dysfunction in patients with diabetes mellitus. Endocr. Pract.9, 52–63 (2003). [DOI] [PubMed] [Google Scholar]
  • 477.Grundy, L. et al. Activation of MrgprA3 and MrgprC11 on bladder-innervating afferents induces peripheral and central hypersensitivity to bladder distension. J. Neurosci.41, 3900–3916 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 478.Andlib, N., Sajad, M. & Thakur, S. C. Association of diabetes mellitus with risk of reproductive impairment in females: a comprehensive review. Acta Histochem.126, 152173 (2024). [DOI] [PubMed] [Google Scholar]
  • 479.Matz, E. L. & Terlecki, R. P. Stem cell and gene-based therapy for erectile dysfunction: current status and future needs. Urol. Clin. North Am.48, 611–619 (2021). [DOI] [PubMed] [Google Scholar]
  • 480.Thorve, V. S. et al. Diabetes-induced erectile dysfunction: epidemiology, pathophysiology and management. J. Diabetes Complicat.25, 129–136 (2011). [DOI] [PubMed] [Google Scholar]
  • 481.Spallone, V. et al. Diabetic neuropathy: Clinical management—genitourinary dysfunction in diabetes. In Diabetic Neuropathy: Advances in Pathophysiology and Clinical Management 491–529 (Springer International Publishing, 2023).
  • 482.van Heck, J. I. P. et al. The gut microbiome composition is altered in long-standing type 1 diabetes and associates with glycemic control and disease-related complications. Diabetes Care45, 2084–2094 (2022). [DOI] [PubMed] [Google Scholar]
  • 483.Ley, R. E. Gut microbiota in 2015: Prevotella in the gut: choose carefully. Nat. Rev. Gastroenterol. Hepatol.13, 69–70 (2016). [DOI] [PubMed] [Google Scholar]
  • 484.Hinkel, R. et al. Diabetes mellitus-induced microvascular destabilization in the myocardium. J. Am. Coll. Cardiol.69, 131–143 (2017). [DOI] [PubMed] [Google Scholar]
  • 485.Zhou, J. Y. & Zhou, S. W. Inflammation: therapeutic targets for diabetic neuropathy. Mol. Neurobiol.49, 536–546 (2014). [DOI] [PubMed] [Google Scholar]
  • 486.Pitarokoili, K., Gold, R. & Fisse, A. L. Nerve ultrasound for the diagnosis and follow-up of peripheral neuropathies. Curr. Opin. Neurol.36, 373–381 (2023). [DOI] [PubMed] [Google Scholar]
  • 487.Kähler, P. et al. Targeting intensive versus conventional glycaemic control for type 1 diabetes mellitus: a systematic review with meta-analyses and trial sequential analyses of randomised clinical trials. BMJ Open4, e004806 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 488.Bebu, I., Braffett, B. H., Orchard, T. J., Lorenzi, G. M. & Lachin, J. M. Mediation of the effect of glycemia on the risk of CVD outcomes in type 1 diabetes: the DCCT/EDIC study. Diabetes Care42, 1284–1289 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 489.Martin, C. L. et al. Neuropathy among the diabetes control and complications trial cohort 8 years after trial completion. Diabetes Care29, 340–344 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 490.Duckworth, W. et al. Glucose control and vascular complications in veterans with type 2 diabetes. N. Engl. J. Med.360, 129–139 (2009). [DOI] [PubMed] [Google Scholar]
  • 491.Mariam, A. et al. A type 2 diabetes subtype responsive to ACCORD intensive glycemia treatment. Diabetes Care44, 1410–1418 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 492.Nisar, M. U. et al. Association of diabetic neuropathy with duration of type 2 diabetes and glycemic control. Cureus7, e302 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 493.American Diabetes Association. 6. Glycemic targets: Standards of Medical Care in Diabetes-2021. Diabetes Care44, S73–S84 (2021). [DOI] [PubMed] [Google Scholar]
  • 494.Inzucchi, S. E. et al. Management of hyperglycemia in type 2 diabetes, 2015: a patient-centered approach: update to a position statement of the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care38, 140–149 (2015). [DOI] [PubMed] [Google Scholar]
  • 495.Callaghan, B. C. et al. Metabolic syndrome components are associated with symptomatic polyneuropathy independent of glycemic status. Diabetes Care39, 801–807 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 496.Evert, A. B. et al. Nutrition therapy for adults with diabetes or prediabetes: a consensus report. Diabetes Care42, 731 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 497.Esposito, K. et al. Effect of a Mediterranean-style diet on endothelial dysfunction and markers of vascular inflammation in the metabolic syndrome: a randomized trial. JAMA292, 1440–1446 (2004). [DOI] [PubMed] [Google Scholar]
  • 498.Kluding, P. M. et al. Safety of aerobic exercise in people with diabetic peripheral neuropathy: single-group clinical trial. Phys. Ther.95, 223–234 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 499.Mohammad Rahimi, G. R., Niyazi, A. & Alaee, S. The effect of exercise training on osteocalcin, adipocytokines, and insulin resistance: a systematic review and meta-analysis of randomized controlled trials. Osteoporos. Int.32, 213–224 (2021). [DOI] [PubMed] [Google Scholar]
  • 500.Zhu, P., Pan, X. F., Sheng, L., Chen, H. & Pan, A. Cigarette smoking, diabetes, and diabetes complications: call for urgent action. Curr. Diab Rep.17, 78 (2017). [DOI] [PubMed] [Google Scholar]
  • 501.Powers, M. A. et al. Diabetes self-management education and support in adults with type 2 diabetes: a consensus report of the American Diabetes Association, the Association of Diabetes Care & Education Specialists, the Academy of Nutrition and Dietetics, the American Academy of Family Physicians, the American Academy of PAs, the American Association of Nurse Practitioners, and the American Pharmacists Association. Diabetes Care43, 1636–1649 (2020). [DOI] [PubMed] [Google Scholar]
  • 502.Chrvala, C. A., Sherr, D. & Lipman, R. D. Diabetes self-management education for adults with type 2 diabetes mellitus: a systematic review of the effect on glycemic control. Patient Educ. Couns.99, 926–943 (2016). [DOI] [PubMed] [Google Scholar]
  • 503.Sawangjit, R., Thongphui, S., Chaichompu, W. & Phumart, P. Efficacy and safety of mecobalamin on peripheral neuropathy: a systematic review and meta-analysis of randomized controlled trials. J. Altern. Complement. Med.26, 1117–1129 (2020). [DOI] [PubMed] [Google Scholar]
  • 504.Bhrigu, B., Sharma, S., Kumar, N. & Banik, B. K. Assessment for diabetic neuropathy: treatment and neurobiological perspective. Curr. Diabetes Rev.21, e240524230300 (2024). [DOI] [PubMed] [Google Scholar]
  • 505.Ziegler, D. et al. Efficacy and safety of antioxidant treatment with α-lipoic acid over 4 years in diabetic polyneuropathy: the NATHAN 1 trial. Diabetes Care34, 2054–2060 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 506.Mijnhout, G. S., Kollen, B. J., Alkhalaf, A., Kleefstra, N. & Bilo, H. J. Alpha lipoic acid for symptomatic peripheral neuropathy in patients with diabetes: a meta-analysis of randomized controlled trials. Int. J. Endocrinol.2012, 456279 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 507.Han, T. T., Bai, J. F., Liu, W. & Hu, Y. M. A systematic review and meta-analysis of α-lipoic acid in the treatment of diabetic peripheral neuropathy. Eur. J. Endocrinol.167, 465–471 (2012). [DOI] [PubMed] [Google Scholar]
  • 508.Zuo, Q. et al. The dietary flavone luteolin epigenetically activates the Nrf2 pathway and blocks cell transformation in human colorectal cancer HCT116 cells. J. Cell. Biochem.119, 9573–9582 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 509.Deng, H. L. et al. Meta-analysis of methylcobalamin alone and in combination with prostaglandin E1 in the treatment of diabetic peripheral neuropathy. Endocrine46, 445–454 (2014). [DOI] [PubMed] [Google Scholar]
  • 510.Jiang, D. Q. et al. Prostaglandin E1 plus methylcobalamin combination therapy versus prostaglandin E1 monotherapy for patients with diabetic peripheral neuropathy: a meta-analysis of randomized controlled trials. Medicine97, e13020 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 511.Kwai, N. C. et al. Effects of axonal ion channel dysfunction on quality of life in type 2 diabetes. Diabetes Care36, 1272–1277 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 512.Sima, A. A., Calvani, M., Mehra, M. & Amato, A. Acetyl-L-carnitine improves pain, nerve regeneration, and vibratory perception in patients with chronic diabetic neuropathy: an analysis of two randomized placebo-controlled trials. Diabetes Care28, 89–94 (2005). [DOI] [PubMed] [Google Scholar]
  • 513.Veronese, N. et al. Effect of acetyl-l-carnitine in the treatment of diabetic peripheral neuropathy: a systematic review and meta-analysis. Eur. Geriatr. Med.8, 117–122 (2017). [Google Scholar]
  • 514.Zhang, A. X. et al. Efficacy and safety of Mudan granules for painful diabetic peripheral neuropathy: a protocol for a double-blind randomized controlled trial. Medicine101, e28896 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 515.Guo, Y. et al. Salvia miltiorrhiza improves type 2 diabetes: a protocol for systematic review and meta-analysis. Medicine100, e23843 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 516.Cheng, X. D. et al. Compound Danshen dropping pill in treatment of diabetic peripheral neuropathy. Clin. Focus32, 609 (2017). [Google Scholar]
  • 517.Rosenstock, J., Tuchman, M., LaMoreaux, L. & Sharma, U. Pregabalin for the treatment of painful diabetic peripheral neuropathy: a double-blind, placebo-controlled trial. Pain110, 628–638 (2004). [DOI] [PubMed] [Google Scholar]
  • 518.Backonja, M. & Glanzman, R. L. Gabapentin dosing for neuropathic pain: evidence from randomized, placebo-controlled clinical trials. Clin. Ther.25, 81–104 (2003). [DOI] [PubMed] [Google Scholar]
  • 519.Callaghan, B. C. & Feldman, E. L. Painful diabetic neuropathy: many similarly effective therapies with widely dissimilar costs. Ann. Intern. Med.161, 674–675 (2014). [DOI] [PubMed] [Google Scholar]
  • 520.Bril, V. et al. Evidence-based guideline: treatment of painful diabetic neuropathy: report of the American Academy of Neurology, the American Association of Neuromuscular and Electrodiagnostic Medicine, and the American Academy of Physical Medicine and Rehabilitation. Neurology76, 1758–1765 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 521.Goldstein, D. J., Lu, Y. L., Detke, M. J., Lee, T. C. & Iyengar, S. Duloxetine vs. placebo in patients with painful diabetic neuropathy. Pain116, 109–118 (2005). [DOI] [PubMed] [Google Scholar]
  • 522.Price, R. et al. Oral and topical treatment of painful diabetic polyneuropathy: practice guideline update summary: Report of the AAN guideline subcommittee. Neurology98, 31–43 (2022). [DOI] [PubMed] [Google Scholar]
  • 523.Wernicke, J. F. et al. A randomized controlled trial of duloxetine in diabetic peripheral neuropathic pain. Neurology67, 1411–1420 (2006). [DOI] [PubMed] [Google Scholar]
  • 524.Hossain, S. M., Hussain, S. M. & Ekram, A. Duloxetine in painful diabetic neuropathy: a systematic review. Clin. J. Pain32, 1005–1010 (2016). [DOI] [PubMed] [Google Scholar]
  • 525.Rowbotham, M. C., Goli, V., Kunz, N. R. & Lei, D. Venlafaxine extended release in the treatment of painful diabetic neuropathy: a double-blind, placebo-controlled study. Pain110, 697–706 (2004). [DOI] [PubMed] [Google Scholar]
  • 526.Gallagher, H. C., Gallagher, R. M., Butler, M., Buggy, D. J. & Henman, M. C. Venlafaxine for neuropathic pain in adults. Cochrane Database Syst. Rev.2015, Cd011091 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 527.Saarto, T. & Wiffen, P. J. Antidepressants for neuropathic pain. Cochrane Database Syst. Rev.2007, Cd005454 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 528.Boyle, J. et al. Randomized, placebo-controlled comparison of amitriptyline, duloxetine, and pregabalin in patients with chronic diabetic peripheral neuropathic pain: impact on pain, polysomnographic sleep, daytime functioning, and quality of life. Diabetes Care35, 2451–2458 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 529.Schwartz, S. et al. Safety and efficacy of tapentadol ER in patients with painful diabetic peripheral neuropathy: results of a randomized-withdrawal, placebo-controlled trial. Curr. Med. Res. Opin.27, 151–162 (2011). [DOI] [PubMed] [Google Scholar]
  • 530.Duehmke, R. M. et al. Tramadol for neuropathic pain in adults. Cochrane Database Syst. Rev.6, Cd003726 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 531.Dowell, D., Haegerich, T. M. & Chou, R. CDC guideline for prescribing opioids for chronic pain-United States, 2016. MMWR Recomm. Rep.65, 1–49 (2016). [DOI] [PubMed] [Google Scholar]
  • 532.Franklin, G. M. Opioids for chronic noncancer pain: a position paper of the American Academy of Neurology. Neurology83, 1277–1284 (2014). [DOI] [PubMed] [Google Scholar]
  • 533.Balhara, Y. P. S., Singh, S. & Kalra, S. Pragmatic opioid use in painful diabetic neuropathy. Eur. Endocrinol.16, 21–24 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 534.Simpson, D. M. et al. Capsaicin 8% patch in painful diabetic peripheral neuropathy: a randomized, double-blind, placebo-controlled study. J. Pain18, 42–53 (2017). [DOI] [PubMed] [Google Scholar]
  • 535.Ten Hoope, W. et al. Pharmacodynamics and pharmacokinetics of lidocaine in a rodent model of diabetic neuropathy. Anesthesiology128, 609–619 (2018). [DOI] [PubMed] [Google Scholar]
  • 536.Medeiros Dantas, J. et al. Monotherapy versus combination therapy in the treatment of painful diabetic neuropathy: a systematic review and meta-analysis. Clin. Drug Investig.43, 905–914 (2023). [DOI] [PubMed] [Google Scholar]
  • 537.Zhou, L. L., Wu, T., Zhong, Z. S., Yi, L. C. & Li, Y. M. Acupuncture for painful diabetic peripheral neuropathy: a systematic review and meta-analysis. Front. Neurol.14, 1281485 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 538.Zhang, Y. J. & Liu, F. R. Effectiveness of acupuncture for treatment of diabetic peripheral neuropathy: a protocol for systematic review. Medicine98, e17282 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 539.Slangen, R. et al. Spinal cord stimulation and pain relief in painful diabetic peripheral neuropathy: a prospective two-center randomized controlled trial. Diabetes Care37, 3016–3024 (2014). [DOI] [PubMed] [Google Scholar]
  • 540.de Vos, C. C. et al. Spinal cord stimulation in patients with painful diabetic neuropathy: a multicentre randomized clinical trial. Pain155, 2426–2431 (2014). [DOI] [PubMed] [Google Scholar]
  • 541.Tatikola, S. P., Natarajan, V., Desai, V. K., Asirvatham, A. R. & Rajsekhar, H. Effect of various exercise protocols on neuropathic pain in individuals with type 2 diabetes with peripheral neuropathy: a systematic review and meta-analysis. Diabetes Metab. Syndr.16, 102603 (2022). [DOI] [PubMed] [Google Scholar]
  • 542.Dixit, S., Maiya, A. & Shastry, B. Effect of aerobic exercise on quality of life in population with diabetic peripheral neuropathy in type 2 diabetes: a single blind, randomized controlled trial. Qual. Life Res.23, 1629–1640 (2014). [DOI] [PubMed] [Google Scholar]
  • 543.American Diabetes Association. 11. Microvascular complications and foot care: standards of medical care in diabetes-2021. Diabetes Care44, S151–S167 (2021). [DOI] [PubMed] [Google Scholar]
  • 544.Bus, S. A. et al. Footwear and offloading interventions to prevent and heal foot ulcers and reduce plantar pressure in patients with diabetes: a systematic review. Diabetes Metab. Res. Rev.32(Suppl 1), 99–118 (2016). [DOI] [PubMed] [Google Scholar]
  • 545.Bus, S. A. et al. The effectiveness of footwear and offloading interventions to prevent and heal foot ulcers and reduce plantar pressure in diabetes: a systematic review. Diabetes Metab. Res. Rev.24, S162–S180 (2008). [DOI] [PubMed] [Google Scholar]
  • 546.Armstrong, D. G. et al. Five year mortality and direct costs of care for people with diabetic foot complications are comparable to cancer. J. Foot Ankle Res.13, 16 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 547.van Deursen, R. Footwear for the neuropathic patient: offloading and stability. Diabetes Metab. Res. Rev.24(Suppl 1), S96–S100 (2008). [DOI] [PubMed] [Google Scholar]
  • 548.Lacroix, A., Hortobágyi, T., Beurskens, R. & Granacher, U. Effects of supervised vs. unsupervised training programs on balance and muscle strength in older adults: a systematic review and meta-analysis. Sports Med47, 2341–2361 (2017). [DOI] [PubMed] [Google Scholar]
  • 549.Paliwal, Y., Slattum, P. W. & Ratliff, S. M. Chronic health conditions as a risk factor for falls among the community-dwelling US older adults: z zero-inflated regression modeling approach. Biomed. Res. Int.2017, 5146378 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 550.Winkley, K. et al. Psychological interventions to improve glycemic control in adults with type 2 diabetes: a systematic review and meta-analysis. BMJ Open Diabetes Res. Care8, e001150 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 551.Bogusch, L. M. & O’Brien, W. H. The effects of mindfulness-based interventions on diabetes-related distress, quality of life, and metabolic control among persons with diabetes: a meta-analytic review. Behav. Med.45, 19–29 (2019). [DOI] [PubMed] [Google Scholar]
  • 552.Charova, E., Dorstyn, D., Tully, P. & Mittag, O. Web-based interventions for comorbid depression and chronic illness: a systematic review. J. Telemed. Telecare21, 189–201 (2015). [DOI] [PubMed] [Google Scholar]
  • 553.Fanciulli, A. et al. Consensus statement on the definition of neurogenic supine hypertension in cardiovascular autonomic failure by the American Autonomic Society (AAS) and the European Federation of Autonomic Societies (EFAS) : Endorsed by the European Academy of Neurology (EAN) and the European Society of Hypertension (ESH). Clin. Auton. Res.28, 355–362 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 554.Freeman, R. Clinical practice. Neurogenic orthostatic hypotension. N. Engl. J. Med.358, 615–624 (2008). [DOI] [PubMed] [Google Scholar]
  • 555.Gibbons, C. H. & Freeman, R. Delayed orthostatic hypotension: a frequent cause of orthostatic intolerance. Neurology67, 28–32 (2006). [DOI] [PubMed] [Google Scholar]
  • 556.Logan, I. C. & Witham, M. D. Efficacy of treatments for orthostatic hypotension: a systematic review. Age Ageing41, 587–594 (2012). [DOI] [PubMed] [Google Scholar]
  • 557.Jordan, J., Cyndya, S. B. & Biaggioni, I. Multiple system atrophy: using clinical pharmacology to reveal pathophysiology. Clin. Auton. Res.25, 53–59 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 558.Kaufmann, H., Norcliffe-Kaufmann, L. & Palma, J. A. Droxidopa in neurogenic orthostatic hypotension. Expert Rev. Cardiovasc. Ther.13, 875–891 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 559.Liu, A. G. et al. Reducing the glycemic index or carbohydrate content of mixed meals reduces postprandial glycemia and insulinemia over the entire day but does not affect satiety. Diabetes Care35, 1633–1637 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 560.Veazie, S. et al. Fludrocortisone for orthostatic hypotension. Cochrane Database Syst. Rev.5, Cd012868 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 561.Kaufmann, H. et al. Droxidopa for neurogenic orthostatic hypotension: a randomized, placebo-controlled, phase 3 trial. Neurology83, 328–335 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 562.Singer, W., Opfer-Gehrking, T. L., Nickander, K. K., Hines, S. M. & Low, P. A. Acetylcholinesterase inhibition in patients with orthostatic intolerance. J. Clin. Neurophysiol.23, 476–481 (2006). [DOI] [PubMed] [Google Scholar]
  • 563.Bardsley, E. N., Davis, H., Buckler, K. J. & Paterson, D. J. Neurotransmitter switching coupled to β-adrenergic signaling in sympathetic neurons in prehypertensive states. Hypertension71, 1226–1238 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 564.van Vliet, P., Hilt, A. D., Thijs, R. D. & van Dijk, J. G. Effect of orthostatic hypotension on sustained attention in patients with autonomic failure. J. Neurol. Neurosurg. Psychiatry87, 144–148 (2016). [DOI] [PubMed] [Google Scholar]
  • 565.Freeman, R. et al. Orthostatic Hypotension: JACC state-of-the-art review. J. Am. Coll. Cardiol.72, 1294–1309 (2018). [DOI] [PubMed] [Google Scholar]
  • 566.Kaufmann, H., Norcliffe-Kaufmann, L. & Palma, J. A. Baroreflex dysfunction. N. Engl. J. Med.382, 163–178 (2020). [DOI] [PubMed] [Google Scholar]
  • 567.Camilleri, M. et al. Gastroparesis. Nat. Rev. Dis. Prim.4, 41 (2018). [DOI] [PubMed] [Google Scholar]
  • 568.Stanghellini, V. et al. Gastroduodenal disorders. Gastroenterology150, 1380–1392 (2016). [DOI] [PubMed] [Google Scholar]
  • 569.Abell, T. L. et al. Treatment of gastroparesis: a multidisciplinary clinical review. Neurogastroenterol. Motil.18, 263–283 (2006). [DOI] [PubMed] [Google Scholar]
  • 570.Pasricha, P. J., Pehlivanov, N., Sugumar, A. & Jankovic, J. Drug Insight: from disturbed motility to disordered movement–a review of the clinical benefits and medicolegal risks of metoclopramide. Nat. Clin. Pract. Gastroenterol. Hepatol.3, 138–148 (2006). [DOI] [PubMed] [Google Scholar]
  • 571.Mullard, A. FDA approves first drug for Parkinson disease psychosis. Nat. Rev. Drug Discov.15, 375 (2016). [DOI] [PubMed] [Google Scholar]
  • 572.Abell, T. et al. Gastric electrical stimulation for gastroparesis improves nutritional parameters at short, intermediate, and long‐term follow‐up. J. Parenter. Enter. Nutr.27, 277–281 (2003). [DOI] [PubMed] [Google Scholar]
  • 573.Halland, M. & Bharucha, A. E. Relationship between control of glycemia and gastric emptying disturbances in diabetes mellitus. Clin. Gastroenterol. Hepatol.14, 929–936 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 574.Musicki, B. & Burnett, A. L. Endothelial dysfunction in diabetic erectile dysfunction. Int. J. Impot. Res.19, 129–138 (2007). [DOI] [PubMed] [Google Scholar]
  • 575.Yuan, J. et al. Vacuum therapy in erectile dysfunction–science and clinical evidence. Int. J. Impot. Res.22, 211–219 (2010). [DOI] [PubMed] [Google Scholar]
  • 576.Burnett, A. L. Erectile dysfunction. J. Urol.175, S25–S31 (2006). [DOI] [PubMed] [Google Scholar]
  • 577.Hatzimouratidis, K. et al. EAU guidelines on penile curvature. Eur. Urol.62, 543–552 (2012). [DOI] [PubMed] [Google Scholar]
  • 578.Panicker, J. N., de Sèze, M. & Fowler, C. J. Rehabilitation in practice: neurogenic lower urinary tract dysfunction and its management. Clin. Rehabil.24, 579–589 (2010). [DOI] [PubMed] [Google Scholar]
  • 579.Kamble, S. V., Yadav, A. K., Sharma, M. T. & Bhosle, R. R. Bethanechol remains the preferred drug in suspected underactive bladder: findings from nation-wide prescription analysis. Open J. Urol.14, 301–311 (2024). [Google Scholar]
  • 580.Andersson, K. E. & Yoshida, M. Antimuscarinics and the overactive detrusor–which is the main mechanism of action? Eur. Urol.43, 1–5 (2003). [DOI] [PubMed] [Google Scholar]
  • 581.Chapple, C. R. et al. Randomized double-blind, active-controlled phase 3 study to assess 12-month safety and efficacy of mirabegron, a β(3)-adrenoceptor agonist, in overactive bladder. Eur. Urol.63, 296–305 (2013). [DOI] [PubMed] [Google Scholar]
  • 582.Karsenty, G. et al. Botulinum toxin A (Botox) intradetrusor injections in adults with neurogenic detrusor overactivity/neurogenic overactive bladder: a systematic literature review. Eur. Urol.53, 275–287 (2008). [DOI] [PubMed] [Google Scholar]
  • 583.Várkonyi, T. & Kempler, P. Sexual dysfunction in diabetes. Handb. Clin. Neurol.126, 223–232 (2014). [DOI] [PubMed] [Google Scholar]
  • 584.Maiorino, M. I., Bellastella, G. & Esposito, K. Diabetes and sexual dysfunction: current perspectives. Diabetes Metab. Syndr. Obes.7, 95–105 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 585.Simon, J. A., Kokot-Kierepa, M., Goldstein, J. & Nappi, R. E. Vaginal health in the United States: results from the Vaginal Health: Insights, Views & Attitudes survey. Menopause20, 1043–1048 (2013). [DOI] [PubMed] [Google Scholar]
  • 586.Anderson, C. A. et al. Conservative management for postprostatectomy urinary incontinence. Cochrane Database Syst. Rev.1, Cd001843 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 587.Kim, W. O., Kil, H. K., Yoon, D. M. & Cho, M. J. Treatment of compensatory gustatory hyperhidrosis with topical glycopyrrolate. Yonsei Med. J.44, 579–582 (2003). [DOI] [PubMed] [Google Scholar]
  • 588.Restivo, D. A. et al. Improvement of diabetic autonomic gustatory sweating by botulinum toxin type A. Neurology59, 1971–1973 (2002). [DOI] [PubMed] [Google Scholar]
  • 589.Zhang, P. Z. et al. Global epidemiology of diabetic foot ulceration: a systematic review and meta-analysis. Ann. Med.49, 106–116 (2017). [DOI] [PubMed] [Google Scholar]
  • 590.Callaghan, B. & Feldman, E. The metabolic syndrome and neuropathy: therapeutic challenges and opportunities. Ann. Neurol.74, 397–403 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 591.van Dieren, S., Beulens, J. W., van der Schouw, Y. T., Grobbee, D. E. & Neal, B. The global burden of diabetes and its complications: an emerging pandemic. Eur. J. Cardiovasc. Prev. Rehabil.17, S3–S8 (2010). [DOI] [PubMed] [Google Scholar]
  • 592.Sinclair, A. J. et al. An international position statement on the management of frailty in diabetes mellitus: summary of recommendations 2017. J. Frailty Aging7, 10–20 (2018). [DOI] [PubMed] [Google Scholar]
  • 593.Jin, J., Sklar, G. E., Min Sen Oh, V. & Chuen Li, S. Factors affecting therapeutic compliance: a review from the patient’s perspective. Ther. Clin. Risk Manag.4, 269–286 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 594.Tesfaye, S. et al. Duloxetine and pregabalin: high-dose monotherapy or their combination? The “COMBO-DN study”–a multinational, randomized, double-blind, parallel-group study in patients with diabetic peripheral neuropathic pain. Pain154, 2616–2625 (2013). [DOI] [PubMed] [Google Scholar]
  • 595.Tesfaye, S. et al. Comparison of amitriptyline supplemented with pregabalin, pregabalin supplemented with amitriptyline, and duloxetine supplemented with pregabalin for the treatment of diabetic peripheral neuropathic pain (OPTION-DM): a multicentre, double-blind, randomised crossover trial. Lancet400, 680–690 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 596.Oelze, M. et al. The sodium-glucose co-transporter 2 inhibitor empagliflozin improves diabetes-induced vascular dysfunction in the streptozotocin diabetes rat model by interfering with oxidative stress and glucotoxicity. PLoS ONE9, e112394 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 597.Lee, K. A., Jin, H. Y., Lee, N. Y., Kim, Y. J. & Park, T. S. Effect of empagliflozin, a selective sodium-glucose cotransporter 2 inhibitor, on kidney and peripheral nerves in streptozotocin-induced diabetic rats. Diabetes Metab. J.42, 338–342 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 598.El-Haggar, S. M., Hafez, Y. M., El Sharkawy, A. M. & Khalifa, M. Effect of empagliflozin in peripheral diabetic neuropathy of patients with type 2 diabetes mellitus. Med. Clin.163, 53–61 (2024). [DOI] [PubMed] [Google Scholar]
  • 599.Rosenthal, N. et al. Canagliflozin: a sodium glucose co-transporter 2 inhibitor for the treatment of type 2 diabetes mellitus. Ann. N. Y. Acad. Sci.1358, 28–43 (2015). [DOI] [PubMed] [Google Scholar]
  • 600.Naznin, F. et al. Canagliflozin, a sodium glucose cotransporter 2 inhibitor, attenuates obesity-induced inflammation in the nodose ganglion, hypothalamus, and skeletal muscle of mice. Eur. J. Pharmacol.794, 37–44 (2017). [DOI] [PubMed] [Google Scholar]
  • 601.Liao, J. L. et al. The impact of canagliflozin on the risk of neuropathy events: a post-hoc exploratory analysis of the CREDENCE trial. Diabetes Metab.48, 101331 (2022). [DOI] [PubMed] [Google Scholar]
  • 602.Brock, C. et al. Liraglutide treatment reduced interleukin-6 in adults with type 1 diabetes but did not improve established autonomic or polyneuropathy. Br. J. Clin. Pharmacol.85, 2512–2523 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 603.Himeno, T. et al. Beneficial effects of exendin-4 on experimental polyneuropathy in diabetic mice. Diabetes60, 2397–2406 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 604.Jaiswal, M. et al. Effects of exenatide on measures of diabetic neuropathy in subjects with type 2 diabetes: results from an 18-month proof-of-concept open-label randomized study. J. Diabetes Complications29, 1287–1294 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 605.Kimura, T. et al. Comparison of clinical efficacy and safety of weekly glucagon-like peptide-1 receptor agonists dulaglutide and semaglutide in Japanese patients with type 2 diabetes: randomized, parallel-group, multicentre, open-label trial (COMING study). Diabetes Obes. Metab.25, 3632–3647 (2023). [DOI] [PubMed] [Google Scholar]
  • 606.Bril, V., Hirose, T., Tomioka, S. & Buchanan, R. Ranirestat for the management of diabetic sensorimotor polyneuropathy. Diabetes Care32, 1256–1260 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 607.Stracke, H., Gaus, W., Achenbach, U., Federlin, K. & Bretzel, R. G. Benfotiamine in diabetic polyneuropathy (BENDIP): results of a randomised, double blind, placebo-controlled clinical study. Exp. Clin. Endocrinol. Diabetes116, 600–605 (2008). [DOI] [PubMed] [Google Scholar]
  • 608.Bönhof, G. J. et al. BOND study: a randomised double-blind, placebo-controlled trial over 12 months to assess the effects of benfotiamine on morphometric, neurophysiological and clinical measures in patients with type 2 diabetes with symptomatic polyneuropathy. BMJ Open12, e057142 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 609.Casellini, C. M. et al. A 6-month, randomized, double-masked, placebo-controlled study evaluating the effects of the protein kinase C-beta inhibitor ruboxistaurin on skin microvascular blood flow and other measures of diabetic peripheral neuropathy. Diabetes Care30, 896–902 (2007). [DOI] [PubMed] [Google Scholar]
  • 610.Dieckmann, A., Kriebel, M., Andriambeloson, E., Ziegler, D. & Elmlinger, M. Treatment with Actovegin® improves sensory nerve function and pathology in streptozotocin-diabetic rats via mechanisms involving inhibition of PARP activation. Exp. Clin. Endocrinol. Diabetes120, 132–138 (2012). [DOI] [PubMed] [Google Scholar]
  • 611.Ziegler, D. et al. Treatment of symptomatic polyneuropathy with actovegin in type 2 diabetic patients. Diabetes Care32, 1479–1484 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 612.Hijma, H. J., Siebenga, P. S., de Kam, M. L. & Groeneveld, G. J. A phase 1, randomized, double-blind, placebo-controlled, crossover study to evaluate the pharmacodynamic effects of VX-150, a highly selective NaV1.8 inhibitor, in healthy male adults. Pain Med.22, 1814–1826 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 613.Zakrzewska, J. M. et al. Safety and efficacy of a Nav1.7 selective sodium channel blocker in patients with trigeminal neuralgia: a double-blind, placebo-controlled, randomised withdrawal phase 2a trial. Lancet Neurol.16, 291–300 (2017). [DOI] [PubMed] [Google Scholar]
  • 614.Faber, C. G. et al. Efficacy and safety of vixotrigine in idiopathic or diabetes-associated painful small fibre neuropathy (CONVEY): a phase 2 placebo-controlled enriched-enrolment randomised withdrawal study. EClinicalMedicine59, 101971 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 615.Vinik, A. et al. Efficacy and safety of mirogabalin (DS-5565) for the treatment of diabetic peripheral neuropathic pain: a randomized, double-blind, placebo- and active comparator-controlled, adaptive proof-of-concept phase 2 study. Diabetes Care37, 3253–3261 (2014). [DOI] [PubMed] [Google Scholar]
  • 616.Baba, M., Matsui, N., Kuroha, M., Wasaki, Y. & Ohwada, S. Mirogabalin for the treatment of diabetic peripheral neuropathic pain: a randomized, double-blind, placebo-controlled phase III study in Asian patients. J. Diabetes Investig.10, 1299–1306 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 617.Ziegler, D., Duan, W. R., An, G. H., Thomas, J. W. & Nothaft, W. A randomized double-blind, placebo-, and active-controlled study of T-type calcium channel blocker ABT-639 in patients with diabetic peripheral neuropathic pain. Pain156, 2013–2020 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 618.Guo, X. H. et al. GABA analogue HSK16149 in Chinese patients with diabetic peripheral neuropathic pain: a phase 3 randomized clinical trial. JAMA Netw. Open7, e2425614 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 619.Davenport, A. J. et al. Eliapixant is a selective P2X3 receptor antagonist for the treatment of disorders associated with hypersensitive nerve fibers. Sci. Rep.11, 19877 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 620.Bouhassira, D. et al. Efficacy and safety of eliapixant in diabetic neuropathic pain and prediction of placebo responders with an exploratory novel algorithm: results from the randomized controlled phase 2a PUCCINI study. Pain165, 785–795 (2024). [DOI] [PubMed] [Google Scholar]
  • 621.Kharatmal, S. B., Singh, J. N. & Sharma, S. S. Calpain inhibitor, MDL 28170 confer electrophysiological, nociceptive and biochemical improvement in diabetic neuropathy. Neuropharmacology97, 113–121 (2015). [DOI] [PubMed] [Google Scholar]
  • 622.Jain, S. M. et al. Randomized, double-blind, placebo-controlled trial of ISC 17536, an oral inhibitor of transient receptor potential ankyrin 1, in patients with painful diabetic peripheral neuropathy: Impact of preserved small nerve fiber function. Pain163, e738–e747 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 623.Sang, C. N., Booher, S., Gilron, I., Parada, S. & Max, M. B. Dextromethorphan and memantine in painful diabetic neuropathy and postherpetic neuralgia: efficacy and dose-response trials. Anesthesiology96, 1053–1061 (2002). [DOI] [PubMed] [Google Scholar]
  • 624.Houck, D. R. et al. NYX-2925, A novel N-methyl-D-aspartate receptor modulator: a first-in-human, randomized, double-blind study of safety and pharmacokinetics in adults. Clin. Transl. Sci.12, 164–171 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 625.Tsuda, M. et al. Lyn tyrosine kinase is required for P2X4 receptor upregulation and neuropathic pain after peripheral nerve injury. Glia56, 50–58 (2008). [DOI] [PubMed] [Google Scholar]
  • 626.Tiecke, E. et al. NRD.E1, an innovative non-opioid therapy for painful diabetic peripheral neuropathy-a randomized proof of concept study. Eur. J. Pain26, 1665–1678 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 627.Rice, A. S. C. et al. Efficacy and safety of EMA401 in peripheral neuropathic pain: results of 2 randomised, double-blind, phase 2 studies in patients with postherpetic neuralgia and painful diabetic neuropathy. Pain162, 2578–2589 (2021). [DOI] [PubMed] [Google Scholar]
  • 628.Brines, M. et al. ARA 290, a nonerythropoietic peptide engineered from erythropoietin, improves metabolic control and neuropathic symptoms in patients with type 2 diabetes. Mol. Med.20, 658–666 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 629.Simpson, R. W. & Wlodarczyk, J. H. Transdermal buprenorphine relieves neuropathic pain: a randomized, double-blind, parallel-group, placebo-controlled trial in diabetic peripheral neuropathic pain. Diabetes Care39, 1493–1500 (2016). [DOI] [PubMed] [Google Scholar]
  • 630.Tesfaye, S. PL37: A new hope in the treatment of painful diabetic neuropathy? Pain Manag.6, 129–132 (2016). [DOI] [PubMed] [Google Scholar]
  • 631.Thibault, K. et al. Antinociceptive and anti-allodynic effects of oral PL37, a complete inhibitor of enkephalin-catabolizing enzymes, in a rat model of peripheral neuropathic pain induced by vincristine. Eur. J. Pharmacol.600, 71–77 (2008). [DOI] [PubMed] [Google Scholar]
  • 632.Gupta, S. & Wright, H. M. Nebivolol: a highly selective beta1-adrenergic receptor blocker that causes vasodilation by increasing nitric oxide. Cardiovasc. Ther.26, 189–202 (2008). [DOI] [PubMed] [Google Scholar]
  • 633.Bhadri, N., Razdan, R. & Goswami, S. K. Nebivolol, a β-blocker abrogates streptozotocin-induced behavioral, biochemical, and neurophysiological deficit by attenuating oxidative-nitrosative stress: a possible target for the prevention of diabetic neuropathy. Naunyn. Schmiedebergs Arch. Pharmacol.391, 207–217 (2018). [DOI] [PubMed] [Google Scholar]
  • 634.Campbell, C. M. et al. Randomized control trial of topical clonidine for treatment of painful diabetic neuropathy. Pain153, 1815–1823 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 635.Kostich, W. et al. Inhibition of aak1 kinase as a novel therapeutic approach to treat neuropathic pain. J. Pharmacol. Exp. Ther.358, 371–386 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 636.Pop-Busui, R. et al. Efficacy and safety of LX9211 for relief of diabetic peripheral neuropathic pain (RELIEF-DPN 1): results of a double-blind, randomized, placebo-controlled, proof-of-concept study. Diabetes Care47, 1325–1332 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 637.Peng, Y. Y., Zhang, A. H., Wei, L. P. & Welsh, W. J. Preclinical evaluation of sigma 1 receptor antagonists as a novel treatment for painful diabetic neuropathy. ACS Pharmacol. Transl. Sci.7, 2358–2368 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 638.Paniagua, N. et al. Blockade of sigma 1 receptors alleviates sensory signs of diabetic neuropathy in rats. Eur. J. Pain21, 61–72 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 639.Wang, H. et al. Fulranumab for treatment of diabetic peripheral neuropathic pain: a randomized controlled trial. Neurology83, 628–637 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 640.Bramson, C. et al. Exploring the role of tanezumab as a novel treatment for the relief of neuropathic pain. Pain Med.16, 1163–1176 (2015). [DOI] [PubMed] [Google Scholar]
  • 641.Hasan, I., Rainsford, K. D. & Ross, J. S. Salsalate: a pleotropic anti-inflammatory drug in the treatment of diabetes, obesity, and metabolic diseases. Inflammopharmacology31, 2781–2797 (2023). [DOI] [PubMed] [Google Scholar]
  • 642.Rolim, L. C., da Silva, E. M., Flumignan, R. L., Abreu, M. M. & Dib, S. A. Acetyl-L-carnitine for the treatment of diabetic peripheral neuropathy. Cochrane Database Syst. Rev.6, Cd011265 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 643.Guo, L. X. et al. Acetyllevocarnitine hydrochloride for the treatment of diabetic peripheral neuropathy: a phase 3, randomized clinical trial in China. Diabetes73, 797–805 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 644.Wahren, J. & Larsson, C. C-peptide: new findings and therapeutic possibilities. Diabetes Res. Clin. Pract.107, 309–319 (2015). [DOI] [PubMed] [Google Scholar]
  • 645.Wahren, J., Foyt, H., Daniels, M. & Arezzo, J. C. Long-acting C-peptide and neuropathy in type 1 diabetes: a 12-month clinical trial. Diabetes Care39, 596–602 (2016). [DOI] [PubMed] [Google Scholar]
  • 646.Pulya, S. et al. HDAC6 as privileged target in drug discovery: a perspective. Pharmacol. Res.163, 105274 (2021). [DOI] [PubMed] [Google Scholar]
  • 647.Michelson, D. et al. A randomized, double-blind, placebo-controlled study of histone deacetylase type 6 inhibition for the treatment of painful diabetic peripheral neuropathy. Pain Rep.8, e1114 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 648.Crigler, L., Robey, R. C., Asawachaicharn, A., Gaupp, D. & Phinney, D. G. Human mesenchymal stem cell subpopulations express a variety of neuro-regulatory molecules and promote neuronal cell survival and neuritogenesis. Exp. Neurol.198, 54–64 (2006). [DOI] [PubMed] [Google Scholar]
  • 649.Monfrini, M. et al. Therapeutic potential of mesenchymal stem cells for the treatment of diabetic peripheral neuropathy. Exp. Neurol.288, 75–84 (2017). [DOI] [PubMed] [Google Scholar]
  • 650.Kawai, M. et al. Long-term selective stimulation of transplanted neural stem/progenitor cells for spinal cord injury improves locomotor function. Cell Rep.37, 110019 (2021). [DOI] [PubMed] [Google Scholar]
  • 651.Kessler, J. A. et al. Gene therapy for diabetic peripheral neuropathy: a randomized, placebo-controlled phase III study of VM202, a plasmid DNA encoding human hepatocyte growth factor. Clin. Transl. Sci.14, 1176–1184 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 652.Ajroud-Driss, S., Christiansen, M., Allen, J. A. & Kessler, J. A. Phase 1/2 open-label dose-escalation study of plasmid DNA expressing two isoforms of hepatocyte growth factor in patients with painful diabetic peripheral neuropathy. Mol. Ther.21, 1279–1286 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 653.Simovic, D., Isner, J. M., Ropper, A. H., Pieczek, A. & Weinberg, D. H. Improvement in chronic ischemic neuropathy after intramuscular phVEGF165 gene transfer in patients with critical limb ischemia. Arch. Neurol.58, 761–768 (2001). [DOI] [PubMed] [Google Scholar]
  • 654.Yang, J. P. et al. Gut microbiota modulate distal symmetric polyneuropathy in patients with diabetes. Cell Metab.35, 1548–1562.e1547 (2023). [DOI] [PubMed] [Google Scholar]
  • 655.Zheng, H. J. et al. The effect of probiotic and synbiotic supplementation on biomarkers of inflammation and oxidative stress in diabetic patients: a systematic review and meta-analysis of randomized controlled trials. Pharmacol. Res.142, 303–313 (2019). [DOI] [PubMed] [Google Scholar]
  • 656.Lewis, E. J. H. et al. Effect of omega-3 supplementation on neuropathy in type 1 diabetes: a 12-month pilot trial. Neurology88, 2294–2301 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 657.Zhao, L. P. et al. Gut bacteria selectively promoted by dietary fibers alleviate type 2 diabetes. Science359, 1151–1156 (2018). [DOI] [PubMed] [Google Scholar]
  • 658.Cooper, M. A. et al. A ketogenic diet reduces metabolic syndrome-induced allodynia and promotes peripheral nerve growth in mice. Exp. Neurol.306, 149–157 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 659.Karwasra, R., Sharma, S., Sharma, I., Shahid, N. & Umar, T. Diabetology and nanotechnology: a compelling combination. Recent Pat. Nanotechnol. 10.2174/0118722105253055231016155618 (2023). [DOI] [PubMed]
  • 660.Bhandari, R., Sharma, A. & Kuhad, A. Novel nanotechnological approaches for targeting dorsal root ganglion (DRG) in mitigating diabetic neuropathic pain (DNP). Front. Endocrinol. (Lausanne)12, 790747 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 661.Latorre, R. et al. Sustained endosomal release of a neurokinin-1 receptor antagonist from nanostars provides long-lasting relief of chronic pain. Biomaterials285, 121536 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 662.Imtiaz, F. et al. Neutrophil lymphocyte ratio as a measure of systemic inflammation in prevalent chronic diseases in Asian population. Int. Arch. Med.5, 2 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 663.Feng, Y. H. et al. Involvement of microRNA-146a in diabetic peripheral neuropathy through the regulation of inflammation. Drug Des. Dev. Ther.12, 171–177 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 664.Peng, H. Y. et al. lncRNA NONRATT021972 siRNA decreases diabetic neuropathic pain mediated by the P2X3 receptor in dorsal root ganglia. Mol. Neurobiol.54, 511–523 (2017). [DOI] [PubMed] [Google Scholar]
  • 665.Yu, W., Zhao, G. Q., Cao, R. J., Zhu, Z. H. & Li, K. LncRNA NONRATT021972 was associated with neuropathic pain scoring in patients with type 2 diabetes. Behav. Neurol.2017, 2941297 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 666.Li, J. B. et al. NSE, a potential biomarker, is closely connected to diabetic peripheral neuropathy. Diabetes Care36, 3405–3410 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 667.Li, Y. P. et al. The Association between phosphorylated neurofilament heavy chain (pNF-H) and small fiber neuropathy (SFN) in patients with impaired glucose tolerance. Diabetes Ther.11, 71–81 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 668.Bugada, D., Lorini, L. F., Fumagalli, R. & Allegri, M. Genetics and opioids: towards more appropriate prescription in cancer pain. Cancers12, 1951 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 669.Kim, O. Y. & Song, J. The importance of BDNF and RAGE in diabetes-induced dementia. Pharmacol. Res.160, 105083 (2020). [DOI] [PubMed] [Google Scholar]
  • 670.Farhadi, A., Vosough, M., Zhang, J. S., Tahamtani, Y. & Shahpasand, K. A possible neurodegeneration mechanism triggered by diabetes. Trends Endocrinol. Metab.30, 692–700 (2019). [DOI] [PubMed] [Google Scholar]
  • 671.Zhao, Y. Y. Metabolomics in chronic kidney disease. Clin. Chim. Acta422, 59–69 (2013). [DOI] [PubMed] [Google Scholar]
  • 672.Slobodyanyuk, M. et al. Directional integration and pathway enrichment analysis for multi-omics data. Nat. Commun.15, 5690 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 673.Agamah, F. E. et al. Computational/in silico methods in drug target and lead prediction. Brief. Bioinform.21, 1663–1675 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 674.Guo, G. J., Chen, J., Shen, Q. X. & Chen, Z. B. Single-cell transcriptome analysis reveals distinct cell populations in dorsal root ganglia and their potential roles in diabetic peripheral neuropathy. PLoS One19, e0306424 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 675.Yu, Z. Y. et al. Metabolomic and lipidomic profiling of the spinal cord in type 2 diabetes mellitus rats with painful neuropathy. Metab. Brain Dis.39, 1117–1130 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 676.Fan, Y. H. et al. Expansion spatial transcriptomics. Nat. Methods20, 1179–1182 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 677.Atmaca, A. et al. Expert opinion on screening, diagnosis and management of diabetic peripheral neuropathy: a multidisciplinary approach. Front. Endocrinol.15, 1380929 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 678.Lavery, L. A. et al. Comprehensive foot examination and risk assessment. Diabetes Care31, 1679 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 679.de Vries, N. M. et al. Effects of physical exercise therapy on mobility, physical functioning, physical activity and quality of life in community-dwelling older adults with impaired mobility, physical disability and/or multi-morbidity: a meta-analysis. Ageing Res. Rev.11, 136–149 (2012). [DOI] [PubMed] [Google Scholar]
  • 680.Yao, Y. et al. Integration of interstitial fluid extraction and glucose detection in one device for wearable non-invasive blood glucose sensors. Biosens. Bioelectron.179, 113078 (2021). [DOI] [PubMed] [Google Scholar]
  • 681.Wu, S. B. et al. MhNet: multi-scale spatio-temporal hierarchical network for real-time wearable fall risk assessment of the elderly. Comput. Biol. Med.144, 105355 (2022). [DOI] [PubMed] [Google Scholar]
  • 682.Tchero, H. et al. Telemedicine in diabetic foot care: a systematic literature review of interventions and meta-analysis of controlled trials. Int. J. Lower Extrem. Wounds16, 274–283 (2017). [DOI] [PubMed] [Google Scholar]
  • 683.Song, T. et al. Measuring success of patients’ continuous use of mobile health services for self-management of chronic conditions: model development and validation. J. Med. Internet Res.23, e26670 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 684.Lopes, R. O. P. et al. Benchmarking mobile applications for the health of people with diabetes mellitus. Rev. Lat. Am. Enferm.32, e4221 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 685.McDowell, A. R. et al. Initial findings using high-resolution magnetic resonance imaging for visualisation of the sural nerve and surrounding anatomy in healthy volunteers at 7 Tesla. J. Peripher. Nerv. Syst.29, 368–375 (2024). [DOI] [PubMed] [Google Scholar]
  • 686.Singh, K. P., Gupta, K., Kataria, N., Arora, V. & Nagpal, N. High-resolution ultrasonography of the sural nerve in diabetic peripheral neuropathy. J. Ultrason.20, e83–e89 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 687.Zhang, X. Y. et al. Machine learning models for diabetic neuropathy diagnosis using microcirculatory parameters in type 2 diabetes patients. Int. Angiol.42, 191–200 (2023). [DOI] [PubMed] [Google Scholar]
  • 688.Lai, Y. R. et al. Longitudinal artificial intelligence-based deep learning models for diagnosis and prediction of the future occurrence of polyneuropathy in diabetes and prediabetes. Neurophysiol. Clin.54, 102982 (2024). [DOI] [PubMed] [Google Scholar]
  • 689.Sheng, B. et al. Artificial intelligence for diabetes care: current and future prospects. Lancet Diabetes Endocrinol.12, 569–595 (2024). [DOI] [PubMed] [Google Scholar]
  • 690.Bini, S. A. Artificial intelligence, machine learning, deep learning, and cognitive computing: what do these terms mean and how will they impact health care? J. Arthroplast.33, 2358–2361 (2018). [DOI] [PubMed] [Google Scholar]
  • 691.Cho, H. et al. Privacy-enhancing technologies in biomedical data science. Annu. Rev. Biomed. Data Sci.7, 317–343 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 692.Zuchowski, L. C., Zuchowski, M. L. & Nagel, E. A trust based framework for the envelopment of medical AI. NPJ Digit. Med.7, 230 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Signal Transduction and Targeted Therapy are provided here courtesy of Nature Publishing Group

RESOURCES