Abstract
Diabetes mellitus (DM) and Parkinson’s disease (PD) are both age-related diseases of global concern being among the most common chronic metabolic and neurodegenerative diseases, respectively. While both diseases can be genetically inherited, environmental factors play a vital role in their pathogenesis. Moreover, DM and PD have common underlying molecular mechanisms, such as misfolded protein aggregation, mitochondrial dysfunction, oxidative stress, chronic inflammation, and microbial dysbiosis. Recently, epidemiological and experimental studies have reported that DM affects the incidence and progression of PD. Moreover, certain antidiabetic drugs have been proven to decrease the risk of PD and delay its progression. In this review, we elucidate the epidemiological and pathophysiological association between DM and PD and summarize the antidiabetic drugs used in animal models and clinical trials of PD, which may provide reference for the clinical translation of antidiabetic drugs in PD treatment.
Keywords: diabetes mellitus, Parkinson’s disease;, epidemiology, pathophysiology, antidiabetic drugs, treatment
1.Introduction
Diabetes mellitus (DM), the most common chronic metabolic disease globally, has become a heavy public health burden because of its high prevalence and associated disability and mortality [1]. DM is a heterogeneous group of disorders characterized by hyperglycemia and glucose intolerance. It mainly comprises two subclasses: type 1 diabetes mellitus (T1DM) (insulin-dependent diabetes) and type 2 diabetes mellitus (T2DM) (noninsulin-dependent diabetes). Its progression is accompanied by several complications including vision loss, stroke, kidney failure, heart attack, leg amputation, and nerve damage [2]. Parkinson’s disease (PD) is the second most prevalent neurodegenerative disease following Alzheimer’s disease (AD). PD is characterized by typical motor symptoms, including resting tremor, bradykinesia, rigidity, and postural disability. Other nonmotor symptoms such as constipation, anxiety, depression, and rapid eye movement sleep behavior disorder (RBD), can also be present before or during the occurrence of motor symptoms [3, 4]. Recently, the association between DM and AD has been intensively studied. Researchers have recently proposed AD as ‘type 3 diabetes mellitus (T3DM)’ because of the overlap of risk factors and pathophysiological mechanisms [5]. Considerable evidence has confirmed the association of T2DM with memory decline and cognitive impairment. Insulin resistance is the core characteristic of T2DM that contributes to AD and related dementias (ADRDs) [6]. However, the association between DM and PD remains to be elucidated.
2.Epidemiological association between DM and PD
Both DM, particularly adult-onset DM (T2DM), and PD affect the aging population, with rising prevalence due to increasing longevity. In the past three decades, the prevalence of DM has increased substantially with approximately 422 million people affected worldwide. A recent study showed the prevalence of DM in China and the US to be 11.2% and 14.3%, respectively [7, 8]. PD prevalence is generally estimated at 0.3% of the entire population and approximately 1% among people over 60 years old, increasing the burden on patients’ families and society [9-11].
Recently, some epidemiological studies have shown an association between DM and PD. Many cohort and case-control studies have investigated the association between DM and the risk of PD, and several meta-analyses have been performed. However, the results remain conflicting (Table 1) [12-16]. Prospective cohort studies have suggested pre-existing DM as a risk factor for the onset of PD [12, 13, 15]. However, retrospective case-control studies indicated that DM could decrease the incidence of PD, which is contrary to the findings of cohort studies [12, 14]. This may be due to heterogeneity among studies, confounding factors, and bias such as inclusion and recall biases [12]. In the most recent meta-analysis, Chohan et al investigated the association between DM (particularly T2DM) and PD risk and progression. They found that any type of DM slightly increased the risk of PD, but T2DM was associated with an increased risk of PD (odds ratio (OR) 1.21; 95% confidence interval (CI) 1.07-1.36). Moreover, they showed that T2DM was associated with faster progression of motor symptoms, with higher Unified Parkinson's Disease Rating Scale (UPDRS) Part III score (standard mean difference (SMD) 0.55; 95% CI 0.39-0.72), and faster cognitive decline, with decreased Montreal cognitive assessment score (SMD -0.92; 95% CI -1.50 to -0.34) [17]. Furthermore, Sánchez-Gómez et al first reported that prediabetes could increase the risk of subsequent PD (hazard ratio (HR) 1.07, 95% CI 1.00-1.14) [18]. Meanwhile, Rhee et al found that the risk of PD increased with the increasing duration of diabetes. Compared with the non-DM group, the adjusted HR of PD was 1.185 (95% CI 1.143-1.229) in the DM duration <5 years group, and 1.618 (95% CI 1.566-1.672) in the DM duration >5 years group (Table 1) [19].
Table 1.
Association between DM and PD in some epidemiological studies and meta-analyses.
| Authors, pulication year and references | Study types | Main results |
|---|---|---|
| Cerada et al., 2011 [12] | meta-analysis | DM is a risk factor for future PD (RR 1.34; 95% CI 1.14-1.58) in cohort studies (n=4). |
| in case control studies (n=5), lack of a relationship between PD and DM (OR 0.56; 95% CI 0.28-1.15) | ||
| Cerada et al., 2013 [13] | meta-analysis | DM is a risk factor for future PD (RR 1.26; 95% CI 1.00-1.58) in cohort studies (n=5) |
| Lu et al., 2014 [14] | meta-analysis | in case control studies (n=14), DM have a negative association with future PD (OR 0.75; 95% CI 0.58-0.98) |
| Yue et al., 2016 [15] | meta-analysis | DM is a risk factor for future PD (RR 1.38; 95% CI 1.18-1.62) in cohort studies (n=7) |
| Chohan et al., 2021 [17] | meta-analysis | T2DM was associated with an increased risk of PD (OR 1.21; 95% CI 1.07-1.36); T2DM was associated with faster progression of motor symptoms (SMD 0.55; 95% CI 0.39-0.72) and cognitive decline (SMD -0.92; 95% CI -1.50 to -0.34) |
| mendelian randomization | a causal effect of T2DM on PD risk (IVW OR 1.08; 95% CI 1.02-1.14); an effect of T2DM on motor progression (IVW OR 1.10; 95% CI 1.01-1.20) but not on cognitive progression | |
| Komici et al., 2021 [16] | meta-analysis | the prevalence of DM in PD patients was 10.02%, (95% CI 7.88-12.16) in observational studies (n=21); DM patients showed a higher risk of developing PD (OR 1.34; 95% CI 1.26-1.43) in observational studies (n=12) |
| Rhee et al., 2020 [19] | cohort study | Compared with the non-DM group, the adjusted HR of PD was 1.038 (95% CI 1.009-1.067) in the IFG group, 1.185 (95% CI 1.143-1.229) in the DM duration <5 years group, and 1.618 (95% CI 1.566-1.672) in the DM duration >5 years group |
| S´anchez-G´omez et al., 2021 [18] | cohort study | T2DM and prediabetes were associated with higher risk of PD (HR 1.19; 95% CI 1.13-1.25, and HR 1.07; 95% CI 1.00-1.14, respectively) |
PD: Parkinson’s Disease; DM: diabetes mellitus; T2DM: type 2 diabetes mellitus; OR: odds ratio; RR: risk ratio; HR: hazard ratio; CI: confidence interval; SMD: standard mean derivation; IFG: impaired fasting glucose; IVW: inverse-variance weighted method
In addition to meta-analysis of observational studies, Mendelian randomization (MR) have been used to investigate the causal effect of T2DM on the risk and progression of PD using data from genome-wide association studies (GWAS) [20-22]. MR results suggest a causal effect of T2DM on PD risk and faster progression of motor dysfunction but not on cognitive decline [17]. In conclusion, DM, particularly T2DM as well as prediabetes, play an important role in the risk and progression of PD.
3.Pathophysiological crosstalk between DM and PD
Both genetic variants and environmental factors play roles in the pathophysiology of DM and PD. Some DM animal models showed similar pathological characteristics to PD animal models [23]. There are potential mechanisms common to the pathophysiology of both diseases, including misfolded protein aggregation, cell death, mitochondrial dysfunction, oxidative stress, chronic inflammation and microbial dysbiosis [24].
3.1Misfolded protein aggregation and cell death in DM and PD
Misfolded amyloid protein aggregation and specific type of cell death are involved in both DM and PD. In T2DM, islet amyloid polypeptide (IAPP) or amylin is produced by islet β cells to regulate insulin secretion. Fibrillar IAPP aggregates are the main constituents of protein deposits in pancreatic islets, which cause dysfunction in islet β cells and their death in most patients with T2DM [25]. In PD, misfolded α-synuclein (α-Syn) is aggregated primarily in the cytoplasm of dopaminergic neurons to form Lewy bodies (LBs) or Lewy neurites (LNs), which can cause progressive death of neurons [26] (Fig. 1).
Figure 1.
Pathophysiological crosstalk between DM and PD. Various genetic variants and environmental toxins can damage the mitochondria, causing excess ROS production and thereby resulting in insulin resistance. Subsequently, peripheral insulin resistance can cause islet amyloid polypeptide aggregation and pancreatic β-cell apoptosis, and central insulin resistance can cause α-Syn aggregation and dopaminergic degeneration.
Crosstalk between IAPP and α-Syn has been described previously [27]. Using in vitro cross-seeding experiments, Horvath et al found that IAPP amyloids promote α-Syn aggregation and that mixing IAPP and α-Syn monomers results in faster co-aggregation than either alone [28]. In cynomolgus monkeys with T2DM, Sun et al found increased aggregation and phosphorylation of α-Syn and partial co-localization with IAPP in the pancreatic islets and brain without obvious dopaminergic neuron death in the substantia nigra (SN), which suggests that T2DM may develop prodromal alterations in PD [29]. In streptozotocin (STZ)-induced T1DM mice models, the proportion of insulin-positive areas to the islet areas in the pancreases of α-Syn-overexpressing mice was significantly lower than that of the wild-type mice, indicating a more severe pancreatic β cell loss in the α-Syn-overexpressing mice [30]. Furthermore, Martinez-Valbuena et al found phosphorylated α-Syn deposited in pancreatic β cells in 93% of patients with PD and 68% of patients with T2DM who had no clear neuropathological alterations, and IAPP/α-Syn interactions occurred in patients with pancreatic inclusions of phosphorylated α-Syn [31]. Therefore, co-aggregation of IAPP and α-Syn may be the underlying pathological mechanisms of increased incidence of PD in patients with DM.
3.2Insulin resistance and synaptic plasticity in DM and PD
Insulin is a vital hormone secreted by pancreatic β cells to maintain peripheral glucose homeostasis. Insulin functions via insulin receptor (IR), which activates insulin receptor substrates (IRS). Insulin binds to IR/IRS to stimulate various downstream signaling pathways [32]. In addition to its functions in the periphery, it also plays a role in the central nervous system (CNS) [33, 34]. IRs are highly expressed in several brain regions, including the olfactory bulb, hypothalamus, hippocampus, and midbrain, in rats [35, 36]. Insulin resistance is a pathological condition in which cells fail to sense and respond normally to insulin, with decreased insulin/insulin-like growth factor 1 (IGF-1) signaling (IIS). There may be an association between peripheral insulin resistance in T2DM and central insulin resistance in PD [37] (Fig. 1).
Decreased IIS may play a role in the pathophysiology of PD [38]. A cross-sectional study suggested that insulin resistance is higher in the brains of patients with PD [39]. Additionally, IR mRNA levels decreased in the SN from the brains of patients with PD [40]. In the SNpc and putamen of postmortem brain tissue of patients with PD, Bassil et al found higher IRS-1-pS312 levels in nigral dopaminergic neurons and a trend toward higher IRS-1-pS312 levels in putaminal neurons but no differences in IRS-1-pS312 staining intensity in glial cells. Moreover, IRS-1-pS312 showed high co-localization with α-Syn within the core of nigral LBs and putamen of PD patients [38]. In PD rat models, IRS-2 phosphorylation at serine 731 increased in the 6-hydroxydopamine (6-OHDA)-induced dopamine-depleted striatum [41]. In α-Syn-overexpressing cells and a mouse model, Gao et al found increased IRS-1 phosphorylation at serine 636 and decreased tyrosine phosphorylation, which accelerated IRS-1 turnover and reduced insulin signaling [42]. These results suggest that insulin resistance indeed occurs in PD. In addition, faster disease progression in PD patients with T2DM may be attributed to brain insulin resistance [43]. In ob/ob and db/db T2DM mice, insulin signaling impairment and α-Syn aggregation occurred in both pancreas and midbrain, and T2DM mice were more susceptible to neurotoxicity induced by acute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) admini-stration [44]. Similarly, high-fat-diet (HFD)-induced insulin-resistant mice may be more vulnerable to 6-OHDA, which exacerbates nigrostriatal dopamine depletion, vascular alterations, and motor symptoms [45-47].
Moreover, insulin plays a role in regulating brain dopamine concentrations. Insulin can increase dopamine transporter (DAT) mRNA expression in the SN [48, 49]. Moreover, insulin enhances dopamine uptake by activating the phosphatidylinositol-3-kinase (PI3K) pathway and increases dopamine release by activating cholinergic interneurons [50]. Abnormal synaptic plasticity is associated with the motor symptoms of PD. Simultaneously, insulin can regulate synaptic plasticity by controlling synapse density and affecting the formation of synaptic junction and dopamine-mediated synaptic and behavioral plasticity via mammalian target of rapamycin C1 (mTORC1) [51]. These studies provide evidence for brain insulin resistance in PD and support the rationale for repurposing antidiabetic drugs for PD treatment.
3.3 Mitochondrial dysfunction and oxidative stress in DM and PD
Different genetic factors and environmental toxins can impair mitochondrial function and subsequently induce oxidative stress, defined as an imbalance between reactive oxygen species (ROS) generation and antioxidant cellular defenses [52]. Although most PD cases are of sporadic type, familial PD accounts for 5%-10% of all patients worldwide [53]. Many familial PD-related genes are involved in mitochondrial function [54]. Autosomal dominant forms of PD are associated with mutations in the SNCA (PARK1/4) gene, encoding α-Syn, and LRRK2 (PARK8), encoding leucine-rich repeat kinase 2. Furthermore, mutations in the Parkin (PARK2) gene, encoding E3 ubiquitin protein ligase parkin, PINK1 (PARK6), encoding PTEN-induced putative kinase 1, and DJ-1 (PARK7), encoding the deglycase protein DJ-1, are involved in the autosomal recessive forms of PD [55]. All these, PD-related genes are related to mitochondrial function. α-Syn can interact with the mitochondrial membrane, causing direct mitochondrial toxicity [56]. LRRK2 mutation inhibits mitochondrial fission and increases ROS production [57]. PINK1 and Parkin participate in mitophagy, and mutations in these genes result in the accumulation of defective mitochondria [58, 59]. DJ-1 is primarily located on the mitochondrial inner membrane and matrix and is translocated to the mitochondrial outer membrane in response to oxidative stress [60]. Inhibition of DJ-1 influences dopaminergic neuron activity in the SN [61]. In addition to genetic mutations, some environmental toxins can cause mitochondrial damage and thereby lead to parkinsonism. Dopaminergic toxins including 6-OHDA, MPTP, rotenone, and paraquat mainly impair the mitochondrial respiratory chain, triggering ROS-induced dopaminergic neuronal apoptosis [62].
In addition, mitochondrial dysfunction plays a central role in insulin resistance [63]. Mitochondria are the primary site of ROS and are critical to energy production, metabolism, redox homeostasis, and multiple cell biological processes [64]. ROS production can activate the phosphorylation of IRS proteins and impair insulin signaling, directly related to mitochondrial dysfunction and insulin resistance [65, 66]. Moreover, insulin resistance can cause mitochondrial biogenesis impairment, oxidative stress, membrane depolarization, and increased dopaminergic neuronal degeneration in the SN [67]. Additionally, in db/db or HFD diabetic mice, the expression of Parkin reduced with the accumulation of the zinc finger protein Parkin interacting substrate (PARIS) and the inhibition of peroxisome proliferator-activated receptor gamma coactivator 1α (PGC1α). Thus, disruption of the Parkin-PARIS-PGC1α pathway may explain the association among mitochondrial dysfunction, insulin resistance, and dopaminergic neuronal degeneration [68]. PGC1α, a key transcriptional regulator of enzymes in mitochondrial respiration and insulin resistance, is potentially pivotal in the pathogenesis of DM and PD [69, 70] (Figure 1). Furthermore, dysfunction of PINK1-Parkin-mediated mitophagy in PD has been associated with DM [71]. Impairment of mitophagy reduces insulin secretion by β cells [72]. Therefore, there is a potential for improving mitophagy to preserve β cell function and delay DM progression [73].
3.4 Chronic inflammation in DM and PD
DM and PD are age-related diseases in which chronic inflammation is a factor, and a new concept “senoinflammation,” was proposed by Chung et al. [74]. Metabolically active tissues such as adipose tissue, muscle, the pancreas, and the liver commonly induce inflammation in aging, which may cause metabolic disorders, including obesity, insulin resistance, and DM [75, 76]. In the CNS, microglia are primary immunological cells important in regulating brain immunity and inflammation [77, 78]. In PD postmortem brains, reactive microglia are involved in the degeneration of dopaminergic neurons [79]. Under various neuroinflammatory stimuli, resting microglia can be activated and polarized into two phenotypes: classical activation of the proinflammatory M1 phenotype and alternative activation of the anti-inflammatory M2 phenotype [80]. There are elevated proinflammatory cytokines, including IL-6, TNF-α and IL-1β, in the cerebral spinal fluid (CSF), serum, and brain tissue of patients with PD. Some PD animal models have demonstrated that both M1 proinflammatory and M2 anti-inflammatory phenotypes of microglia coexist in the early stage of PD, converting to prevalence of M1 proinflammatory microglia in later stages [81]. Moreover, the blockade of neurotoxic A1 astrocytes induced by M1 microglia is neuroprotective in PD animal models [82, 83]. Intriguingly, metabolic disorders including obesity, insulin resistance, and T2DM can regulate the transition of microglia from M2 neuroprotective phenotype to M1 neurotoxic phenotype [84, 85].
The inflammasome pathway is pivotal in chronic systemic inflammation and brain inflammation, linking DM and PD [71, 86]. After sensing pathogen-associated molecular patterns (PAMPs) or danger-associated molecular patterns (DAMPs), canonical nucleotide-binding oligomerization domain- and leucine-rich-repeat- and pyrin-domain-containing 3 (NLRP3) inflammasomes assemble in the cytosol to recruit inactive pro-caspase-1 and cleave into active caspase-1, which cleaves the precursor cytokines pro-IL-1β and pro-IL-18 to generate the active cytokines IL-1β and IL-18, respectively [87]. IL-1β and IL-18 can cause systematic inflammation, impair pancreatic β-cell function, induce cell apoptosis, and exacerbate insulin resistance [88]. Then, peripheral proinflammatory stimuli activate microglial metabolic reprogramming and increase glycolysis and glutaminolysis in microglia to cause neuroinflammation, which excretes more proinflammatory cytokines initiating a vicious cycle [89, 90] (Fig. 2). Notably, mitochondrial damage plays a critical role in activating NLRP3 inflammasome. The accumulation of damaged mitochondria and an increase in mitochondrial ROS (mtROS) due to mitophagy inhibition are associated with NLRP3 inflammasome activation [91].
Figure 2.

Roles of gut-brain axis in DM and PD. Gut microbiota dysbiosis causes beneficial bacterial metabolites to decrease and proinflammatory cytokine levels to increase, leading to systemic inflammation and pancreatic β-cell apoptosis. Proinflammatory cytokines then propagate from the gut-brain axis, resulting in microglial activation and NLRP3 pathway activation and further causing neuroinflammation.
3.5 Microbiota dysbiosis of gut-brain axis in DM and PD
Currently, gut microbiota dysbiosis plays an important role in intestinal disease and in many extraintestinal diseases, including metabolic and neurodegenerative diseases [92, 93]. The gut microbiota mainly comprises six phyla, including Firmicutes, Proteobacteria, Verrucomicrobia, Fusobacteria, Actinobacteria, and Bacteroidetes [94]. Microbiota dysbiosis results in a deficiency of short-chain fatty acids (SCFAs) such as acetate, propionate, and butyrate, which are metabolites of healthy gut microbiota, and has been demonstrated in obesity, T1DM, and T2DM [95]. Some studies demonstrated that the ratio of Firmicutes to Bacteroidetes decreased in T1DM and T2DM [96, 97]. At the genus level, patients with T2DM have reduced levels of butyrate-producing bacteria, such as Bifidobacterium, Akkermansia, and Faecalibacterium, and increased levels of Dorea, which are involved in chronic inflammation [98, 99]. Similarly, the abundances of Bacteroidetes, Verrucomicrobia, and Proteobacteria were higher, whereas that of Firmicutes was lower in the feces of patients with PD. Additionally, at the genus level, putative proinflammatory bacteria, Oscillospira spp., Akkermansia spp., and Bacteroides spp., were significantly increased in patients with PD. In contrast, the abundance of putative anti-inflammatory butyrate-producing bacteria Blautias spp., Coprococcus spp., and Roseburia spp. significantly decreased in PD [100, 101]. Yan et al. reported that the composition of gut microbiota and metabolites in A53T monkeys showed high similarity to those in patients with PD. Therefore, metabolites mainly associated with mitochondrial dysfunction may be related to the pathogenesis of PD [102].
The mechanism linking DM and PD by microbiota dysbiosis is mainly by the gut-brain axis. In the presence of gut microbiota-caused dysbiosis, harmful metabolites and proinflammatory cytokines can damage the intestinal barrier, causing systematic inflammation and pancreatic β-cell dysfunction and further damaging the brain-blood barrier to cause neuroinflammation, immune activation, and oxidative stress [103, 104]. The gut microbiota mediates the inflammatory response based on interactions between lipopolysaccharides (LPS) produced by bacteria with toll-like receptors (TLRs) [96]. Moreover, the level of LPS in the blood was correlated with cell death, DM, and other diseases such as sepsis, inflammatory bowel disease, and neurodegenerative diseases including PD [105, 106]. Microbiota plays a significant role in the pathogenesis and progression of PD [107]. Patients with PD can exhibit microbiota-related dysbiosis and gastrointestinal complications even before the clinical onset of motor symptoms, such as hypersalivation, dysphagia, and constipation [108]. Therefore, targeting gut microbiota homeostasis with probiotic, prebiotic, synbiotic, and postbiotic (PPSP) supplementation as well as fecal microbiota transplantation (FMT) is a promising therapy for both DM and PD [109-111] (Fig. 2).
4. Repurposing antidiabetic drugs for the prevention and treatment of PD
Currently, medication only improves the clinical symptoms of PD. There is an urgent need to discover effective drugs to delay its progression, if not cure PD. Common pathophysiological processes linking DM and PD offer a new prospective for repurposing antidiabetic drugs for the treatment of PD. Some antidiabetic drugs have antioxidative and anti-inflammatory roles, which may be neuroprotective in the prevention and treatment of PD [112] (Fig. 3).
Figure 3.

Roles of antidiabetic drugs in PD treatment. Antidiabetic drugs can be neuroprotective by improving insulin resistance. Moreover, they can inhibit α-Syn aggregation and decrease dopaminergic neuron degeneration by antioxidative stress and anti-inflammatory effects, thus ameliorating motor symptoms in animal models and patients with PD.
4.1 Antidiabetic drugs for the prevention of PD
In patients with diabetes, some antidiabetic drugs can decrease the risk of PD. A cohort study of patients with diabetes by Brauer et al. showed that glucagon-like peptide-1 (GLP-1) receptor agonists or dipeptidyl peptidase 4 (DPP4) inhibitors are associated with a lower incidence rate ratio (IRR) of PD compared with other oral antidiabetic drugs (IRR 0.38; 95% CI 0.17-0.60, and IRR 0.64; 95% CI 0.43-0.88, respectively), but thiazolidinediones (TZDs) or glitazones (GTZs) do not decrease the risk of PD [113]. However, a systematic review and meta-analysis by Qin et al. suggested that GLP-1 receptor agonists decrease the risk of PD (HR 0.41; 95% CI, 0.19-0.87), while DPP4 inhibitors, GTZ, metformin, and sulfonylurea do not [114].
4.2 Antidiabetic drugs for the treatment of PD
Wang et al. found that the GLP-1 receptor agonist exenatide, but not pioglitazone, can ameliorate cognitive, motor, and nonmotor symptoms in patients with PD [115]. Furthermore, Jeong et al. suggested that DPP4 inhibitors showed beneficial effects on baseline dopamine degeneration and long-term motor symptoms in diabetic patients with PD. However, studies with large sample sizes should be conducted to prove these conclusions, which may extend the effect of antidiabetic drugs to nondiabetic patients with PD [116]. In this section, we will discuss several antidiabetic drugs used in cellular and animal models of PD as well as some current clinical trials (Table 2).
Table 2.
Roles of antidiabetic drugs on PD treatment.
| Antidiabeticdrugs | Animal models or clinical trials | Roles in PD | Refs. |
|---|---|---|---|
| Insulin | 6-OHDA-induced PD animal model | Alleviate motor deficits Protect dopaminergic neuron | [118] |
| PD and MSA patients (NCT02064166) | Improve UPDRS score and H-Y scale | [119] | |
| Metformin | MPTP-induced or rotenone-induced PD animal model | Improve motor symptoms Protect dopaminergic neuron Inhibit α-Syn phosphorylation Antioxidative stress Rescue mitochondrial function | [126-131] |
| LPS-induced PD animal model | Fail to protect dopaminergic neuron | [138] | |
| TZDs or GTZs | 6-OHDA-induced or MPTP-induced or LPS-induced PD animal model | Anti-inflammation Antioxidative stress Improve mitochondrial function | [143-148] |
| PD patients (NCT01280123) | Fail to improve UPDRS score | [150] | |
| GLP-1 receptor agonists | 6-OHDA-induced or MPTP-induced or LPS-induced PD animal model | Improve motor function Protect dopaminergic neuron Anti-inflammation Antioxidative stress | [154-156] |
| PD patients (NCT01174810) | Improve UPDRS score Improve cognitive function | [160, 161] | |
| PD patients (NCT01971242) | Improve UPDRS score | [162, 163] | |
| DPP4 inhibitors | Rotenone-induced PD animal model | Anti-inflammation Antioxidative stress | [168-172] |
PD: Parkinson’s Disease; UPDRS: Unified Parkinson’s Disease Rating Scale; H-Y scale: Hoehn and Yahr scale.
4.2.1 Insulin and PD
Due to the potential role of insulin resistance in PD pathogenesis, targeting insulin resistance may have therapeutic implications for PD. To study the role of insulin in the treatment of PD, researchers have focused on intranasal insulin administration to avoid hypoglycemia resulting from peripheral administration [117]. Pang et al. showed that intranasal insulin provided strong neuroprotection in a 6-OHDA-induced PD rat model without affecting body weight or blood glucose levels, which proved that insulin signaling may be a novel therapeutic target in PD [118]. A double-blind placebo-controlled pilot study showed that 40 international units (IUs) of intranasal insulin administered once daily for 4 weeks improved verbal fluency and motor symptoms in patients with PD or multiple system atrophy (MSA) [119]. However, a larger study with long-term observation is required to verify the role of IIS in cognitive function and motor symptoms in patients with PD [120-122].
4.2.2 Metformin and PD
Metformin is the most commonly used first-line oral antidiabetic drug belonging to the biguanide family [123]. Metformin can cross the BBB and act via AMP-activated protein kinase (AMPK)-dependent or AMPK-independent mechanisms in the CNS [124, 125]. Metformin is neuroprotective and can improve motor impairment in MPTP- or rotenone-induced PD models [126-131]. The neuroprotective role of metformin may be mediated by the inhibition of α-Syn phosphorylation and improvement of mitochondrial function [132, 133]. In SH-SY5Y and HeLa cells, as well as in wild-type C57BL/6 mice, metformin reduced the phosphorylation of Ser129 α-Syn, which was associated with the inhibition of mTOR and the activation of protein phosphatase 2A (PP2A) [134]. Moreover, metformin rescues PD phenotypes caused by hyperactive mitochondria via its role in the inhibition of the mitochondrial respiratory chain [135-137]. However, in LPS-induced PD models, metformin fails to protect dopaminergic neurons in the SN [138]. Furthermore, a meta-analysis of observational studies showed that the use of metformin in patients with DM may lead to the development of PD [139]. Thus, metformin has both beneficial and harmful effects in various PD models, particularly as a weak inhibitor of complex I of the mitochondrial respiratory chain, limiting the use of metformin for the prevention and treatment of PD.
4.2.3 TZDs (or GTZs) and PD
TZDs, or GTZs, are another class of oral antidiabetic drugs. TZDs are selective agonists of peroxisome proliferator-activated receptor gamma (PPARγ) that regulate glycemia and lipid homeostasis [140]. PPARγ is expressed not only in insulin-sensitive tissues but also in the SN and putamen, which expands its function in other diseases, such as PD [141]. PPARγ can activate central PGC1α signaling to exhibit promising protective effects in anti-parkinsonism therapeutics [142]. Moreover, pioglitazone and rosiglitazone have exhibited neuroprotective effects on behavioral and motor symptoms in some toxin-induced PD models, including 6-OHDA [143, 144], MPTP [145, 146], and LPS [147, 148]. Its protective roles are associated with anti-inflammation, antioxidative stress, and improvement of mitochondrial function [149]. In a phase 2, multicenter, double-blind, randomized trial of pioglitazone in early PD, 15 or 45 mg of pioglitazone per day for 1 year failed to demonstrate obvious protective effects on the UPDRS score [150]. Moreover, the translation of TZDs for PD treatment could be limited by the side effects of cardiovascular risk and its poor capacity to cross the BBB [140, 151].
4.2.4 GLP-1 receptor agonists and PD
Incretins, including glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), can stimulate insulin secretion by pancreatic β cells in a glucose-dependent manner, which plays an important role in insulin and glucose homeostasis [152]. GLP-1 receptor agonists, or dual GLP-1/GIP receptor agonists, have promising therapeutic potential for PD [153]. The natural GLP-1 mimetic exendin-4 has neuroprotective roles in several PD animal models [154, 155]. Moreover, Liu et al. found that other long-acting GLP-1 receptor agonists, liraglutide and lixisenatide, are superior to exendin-4 in improving motor function and protecting dopaminergic neurons in MPTP models of PD [156]. The protective mechanisms of GLP-1 receptor agonists are because of the preservation of mitochondrial function, inhibition of dopaminergic neurons apoptosis, and microglial activation [157, 158]. Several clinical trials have proven the beneficial effects of GLP-1 receptor agonists in patients with PD [159]. Aviles-Olmos et al. conducted the first clinical trial of exenatide for patients with PD. They found that exenatide can improve motor and cognitive functions, which persists even 12 months after its discontinuation [160, 161]. Moreover, Athauda et al. found that exenatide once weekly versus placebo for 48 weeks followed by a 12-week washout period can improve the UPDRS Part III score at 60 weeks. Exenatide-treated patients had augmented tyrosine phosphorylation of IRS1 and elevated expression of downstream substrates, including total Akt and phosphorylated mTOR, in serum exosomes [162, 163]. Normalizing insulin signaling and delaying disease progression make GLP-1 receptor agonists potential drugs for disease-modifying treatment [164].
4.2.5 DPP4 inhibitors and PD
Natural GLP-1 has a half-life of only 2-3 min and can undergo immediate degradation by DPP4. Therefore, DPP4 inhibitors can enhance the function of GLP-1 [165]. Oral DPP4 inhibitors can increase insulin secretion and decrease glucagon secretion without affecting body weight. In addition to their effect in the endocrinological system, DPP4 inhibitors have a protective role in other systems, including the cardiovascular system and CNS, due to their immunosuppressive functions [166, 167]. Abdelsalam et al. showed that vildagliptin reduced dopaminergic neuron degeneration and improved motor performance by targeting the receptor for advanced glycated end product (RAGE)/nuclear factor kappa B (NF-κB) anti-inflammatory pathway and Nrf-2 antioxidative pathway in rotenone-induced PD models [168]. Moreover, saxagliptin, sitagliptin, and alogliptin showed neuroprotective functions in rotenone-induced PD models [169-172]. However, gliptins cannot penetrate the BBB, whereas omarigliptin was the first gliptin able to cross it and increase GLP-1 concentration in the brain, which makes omarigliptin a potent candidate for PD treatment [173].
5. Conclusions
In this review, we discussed the association between PD and DM. First, epidemiological studies demonstrated that DM, or prediabetic conditions, can increase the risk of PD development, and DM can accelerate the progression of PD, including motor dysfunction and cognitive impairment. Second, we discussed the underlying pathophysiological links between PD and DM, including amyloid protein aggregation, insulin resistance, mitochondrial dysfunction, chronic inflammation, and gut microbiota dysbiosis, which may explain why patients with DM are prone to develop PD. Third, we summarized some publicly available antidiabetic drugs that show potential for repurposing in PD treatment. While the clinical safety of antidiabetic drugs has been verified, large-scale clinical trials are required to prove their effectiveness in preventing and treating PD.
Acknowledgments
We gratefully thank members of Feng’s lab and Li’s lab for their intellectual input. This review was supported by the Outstanding Scientific Fund of Shengjing Hospital. This review was also generously supported by the National Natural Science Foundation of China (817712 71, 31800898, U1801681), the Key Field Research Development Program of Guangdong Province (2018B030337001), and the Swedish Research Council (2019-01551), ParkinsonFonden, the Strategic Research Area Multipark (Multidisciplinary research in Parkinson’s disease at Lund University).
Footnotes
Conflicts of interest
The authors declare no conflicts of interest.
References
- [1].Collaborators GRF (2018). Global, regional, and national comparative risk assessment of 84 behavioural, environmental and occupational, and metabolic risks or clusters of risks for 195 countries and territories, 1990-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet, 392:1923-94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [2].Association AD (2020). 2. Classification and Diagnosis of Diabetes. Diabetes Care, 43:S14-S31. [DOI] [PubMed] [Google Scholar]
- [3].Schapira AHV, Chaudhuri KR, Jenner P (2017). Non-motor features of Parkinson disease. Nat Rev Neurosci, 18:435-50. [DOI] [PubMed] [Google Scholar]
- [4].Armstrong MJ, Okun MS (2020). Diagnosis and Treatment of Parkinson Disease: A Review. JAMA, 323:548-60. [DOI] [PubMed] [Google Scholar]
- [5].Kandimalla R, Thirumala V, Reddy PH (2017). Is Alzheimer's disease a Type 3 Diabetes? A critical appraisal. Biochim Biophys Acta Mol Basis Dis, 1863:1078-89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Arnold SE, Arvanitakis Z, Macauley-Rambach SL, Koenig AM, Wang H-Y, Ahima RS, et al. (2018). Brain insulin resistance in type 2 diabetes and Alzheimer disease: concepts and conundrums. Nat Rev Neurol, 14:168-81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Li Y, Teng D, Shi X, Qin G, Qin Y, Quan H, et al. (2020). Prevalence of diabetes recorded in mainland China using 2018 diagnostic criteria from the American Diabetes Association: national cross sectional study. BMJ, 369:m997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Wang L, Li X, Wang Z, Bancks MP, Carnethon MR, Greenland P, et al. (2021). Trends in Prevalence of Diabetes and Control of Risk Factors in Diabetes Among US Adults, 1999-2018. JAMA. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].de Lau LML, Breteler MMB (2006). Epidemiology of Parkinson's disease. Lancet Neurol, 5:525-35. [DOI] [PubMed] [Google Scholar]
- [10].Ascherio A, Schwarzschild MA (2016). The epidemiology of Parkinson's disease: risk factors and prevention. Lancet Neurol, 15:1257-72. [DOI] [PubMed] [Google Scholar]
- [11].Feigin VL, Vos T, Alahdab F, Amit AML, Bärnighausen TW, Beghi E, et al. (2021). Burden of Neurological Disorders Across the US From 1990-2017: A Global Burden of Disease Study. JAMA Neurol, 78:165-76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Cereda E, Barichella M, Pedrolli C, Klersy C, Cassani E, Caccialanza R, et al. (2011). Diabetes and risk of Parkinson's disease: a systematic review and meta-analysis. Diabetes Care, 34:2614-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Cereda E, Barichella M, Pedrolli C, Klersy C, Cassani E, Caccialanza R, et al. (2013). Diabetes and risk of Parkinson's disease. Mov Disord, 28:257. [DOI] [PubMed] [Google Scholar]
- [14].Lu L, Fu D-L, Li H-Q, Liu A-J, Li J-H, Zheng G-Q (2014). Diabetes and risk of Parkinson's disease: an updated meta-analysis of case-control studies. PLoS One, 9:e85781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Yue X, Li H, Yan H, Zhang P, Chang L, Li T (2016). Risk of Parkinson Disease in Diabetes Mellitus: An Updated Meta-Analysis of Population-Based Cohort Studies. Medicine (Baltimore), 95:e3549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Komici K, Femminella GD, Bencivenga L, Rengo G, Pagano G (2021). Diabetes Mellitus and Parkinson's Disease: A Systematic Review and Meta-Analyses. J Parkinsons Dis, 11:1585-96. [DOI] [PubMed] [Google Scholar]
- [17].Chohan H, Senkevich K, Patel RK, Bestwick JP, Jacobs BM, Bandres Ciga S, et al. (2021). Type 2 Diabetes as a Determinant of Parkinson's Disease Risk and Progression. Mov Disord, 36:1420-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Sánchez-Gómez A, Díaz Y, Duarte-Salles T, Compta Y, Martí MJ (2021). Prediabetes, type 2 diabetes mellitus and risk of Parkinson's disease: A population-based cohort study. Parkinsonism Relat Disord, 89:22-7. [DOI] [PubMed] [Google Scholar]
- [19].Rhee SY, Han K-D, Kwon H, Park S-E, Park Y-G, Kim Y-H, et al. (2020). Association Between Glycemic Status and the Risk of Parkinson Disease: A Nationwide Population-Based Study. Diabetes Care, 43:2169-75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Vujkovic M, Keaton JM, Lynch JA, Miller DR, Zhou J, Tcheandjieu C, et al. (2020). Discovery of 318 new risk loci for type 2 diabetes and related vascular outcomes among 1.4 million participants in a multi-ancestry meta-analysis. Nat Genet, 52:680-91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Nalls MA, Blauwendraat C, Vallerga CL, Heilbron K, Bandres-Ciga S, Chang D, et al. (2019). Identification of novel risk loci, causal insights, and heritable risk for Parkinson's disease: a meta-analysis of genome-wide association studies. Lancet Neurol, 18:1091-102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Iwaki H, Blauwendraat C, Leonard HL, Kim JJ, Liu G, Maple-Grødem J, et al. (2019). Genomewide association study of Parkinson's disease clinical biomarkers in 12 longitudinal patients' cohorts. Mov Disord, 34:1839-50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].de Bem AF, Krolow R, Farias HR, de Rezende VL, Gelain DP, Moreira JCF, et al. (2020). Animal Models of Metabolic Disorders in the Study of Neurodegenerative Diseases: An Overview. Front Neurosci, 14:604150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Camargo Maluf F, Feder D, Alves de Siqueira Carvalho A (2019). Analysis of the Relationship between Type II Diabetes Mellitus and Parkinson's Disease: A Systematic Review. Parkinsons Dis, 2019:4951379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Höppener JW, Ahrén B, Lips CJ (2000). Islet amyloid and type 2 diabetes mellitus. N Engl J Med, 343:411-9. [DOI] [PubMed] [Google Scholar]
- [26].Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M (1997). Alpha-synuclein in Lewy bodies. Nature, 388:839-40. [DOI] [PubMed] [Google Scholar]
- [27].Nguyen PH, Ramamoorthy A, Sahoo BR, Zheng J, Faller P, Straub JE, et al. (2021). Amyloid Oligomers: A Joint Experimental/Computational Perspective on Alzheimer's Disease, Parkinson's Disease, Type II Diabetes, and Amyotrophic Lateral Sclerosis. Chem Rev, 121:2545-647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Horvath I, Wittung-Stafshede P (2016). Cross-talk between amyloidogenic proteins in type-2 diabetes and Parkinson's disease. Proc Natl Acad Sci U S A, 113:12473-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Sun Y, Guo C, Yuan L, Li W, Wang Z-Y, Yue F, et al. (2020). Cynomolgus Monkeys With Spontaneous Type-2-Diabetes-Mellitus-Like Pathology Develop Alpha-Synuclein Alterations Reminiscent of Prodromal Parkinson's Disease and Related Diseases. Front Neurosci, 14:63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Lv Y-Q, Yuan L, Sun Y, Dou H-W, Su J-H, Hou Z-P, et al. (2022). Long-term hyperglycemia aggravates α-synuclein aggregation and dopaminergic neuronal loss in a Parkinson's disease mouse model. Transl Neurodegener, 11:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Martinez-Valbuena I, Amat-Villegas I, Valenti-Azcarate R, Carmona-Abellan MDM, Marcilla I, Tuñon M-T, et al. (2018). Interaction of amyloidogenic proteins in pancreatic β cells from subjects with synucleinopathies. Acta Neuropathol, 135:877-86. [DOI] [PubMed] [Google Scholar]
- [32].Haeusler RA, McGraw TE, Accili D (2018). Biochemical and cellular properties of insulin receptor signalling. Nat Rev Mol Cell Biol, 19:31-44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Unger JW, Livingston JN, Moss AM (1991). Insulin receptors in the central nervous system: localization, signalling mechanisms and functional aspects. Prog Neurobiol, 36:343-62. [DOI] [PubMed] [Google Scholar]
- [34].Kim B, Feldman EL (2012). Insulin resistance in the nervous system. Trends Endocrinol Metab, 23:133-41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Werther GA, Hogg A, Oldfield BJ, McKinley MJ, Figdor R, Allen AM, et al. (1987). Localization and characterization of insulin receptors in rat brain and pituitary gland using in vitro autoradiography and computerized densitometry. Endocrinology, 121:1562-70. [DOI] [PubMed] [Google Scholar]
- [36].Baskin DG, Porte D, Guest K, Dorsa DM (1983). Regional concentrations of insulin in the rat brain. Endocrinology, 112:898-903. [DOI] [PubMed] [Google Scholar]
- [37].Aviles-Olmos I, Limousin P, Lees A, Foltynie T (2013). Parkinson's disease, insulin resistance and novel agents of neuroprotection. Brain, 136:374-84. [DOI] [PubMed] [Google Scholar]
- [38].Bassil F, Delamarre A, Canron MH, Dutheil N, Vital A, Négrier-Leibreich ML, et al. (2021). Impaired brain insulin signalling in Parkinson's disease. Neuropathol Appl Neurobiol. [DOI] [PubMed] [Google Scholar]
- [39].Morris JK, Vidoni ED, Perea RD, Rada R, Johnson DK, Lyons K, et al. (2014). Insulin resistance and gray matter volume in neurodegenerative disease. Neuroscience, 270:139-47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Takahashi M, Yamada T, Tooyama I, Moroo I, Kimura H, Yamamoto T, et al. (1996). Insulin receptor mRNA in the substantia nigra in Parkinson's disease. Neurosci Lett, 204:201-4. [DOI] [PubMed] [Google Scholar]
- [41].Morris JK, Seim NB, Bomhoff GL, Geiger PC, Stanford JA (2011). Effects of unilateral nigrostriatal dopamine depletion on peripheral glucose tolerance and insulin signaling in middle aged rats. Neurosci Lett, 504:219-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Gao S, Duan C, Gao G, Wang X, Yang H (2015). Alpha-synuclein overexpression negatively regulates insulin receptor substrate 1 by activating mTORC1/S6K1 signaling. Int J Biochem Cell Biol, 64:25-33. [DOI] [PubMed] [Google Scholar]
- [43].de Pablo-Fernández E, Courtney R, Rockliffe A, Gentleman S, Holton JL, Warner TT (2021). Faster disease progression in Parkinson's disease with type 2 diabetes is not associated with increased α-synuclein, tau, amyloid-β or vascular pathology. Neuropathol Appl Neurobiol. [DOI] [PubMed] [Google Scholar]
- [44].Wang L, Zhai Y-Q, Xu L-L, Qiao C, Sun X-L, Ding J-H, et al. (2014). Metabolic inflammation exacerbates dopaminergic neuronal degeneration in response to acute MPTP challenge in type 2 diabetes mice. Exp Neurol, 251:22-9. [DOI] [PubMed] [Google Scholar]
- [45].Morris JK, Bomhoff GL, Stanford JA, Geiger PC (2010). Neurodegeneration in an animal model of Parkinson's disease is exacerbated by a high-fat diet. Am J Physiol Regul Integr Comp Physiol, 299:R1082-R90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Sharma S, Taliyan R (2018). High fat diet feeding induced insulin resistance exacerbates 6-OHDA mediated neurotoxicity and behavioral abnormalities in rats. Behav Brain Res, 351:17-23. [DOI] [PubMed] [Google Scholar]
- [47].Elabi OF, Cunha JPMCM, Gaceb A, Fex M, Paul G (2021). High-fat diet-induced diabetes leads to vascular alterations, pericyte reduction, and perivascular depletion of microglia in a 6-OHDA toxin model of Parkinson disease. J Neuroinflammation, 18:175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Craft S, Watson GS (2004). Insulin and neurodegenerative disease: shared and specific mechanisms. Lancet Neurol, 3:169-78. [DOI] [PubMed] [Google Scholar]
- [49].Cardoso S, Correia S, Santos RX, Carvalho C, Santos MS, Oliveira CR, et al. (2009). Insulin is a two-edged knife on the brain. J Alzheimers Dis, 18:483-507. [DOI] [PubMed] [Google Scholar]
- [50].Shaughness M, Acs D, Brabazon F, Hockenbury N, Byrnes KR (2020). Role of Insulin in Neurotrauma and Neurodegeneration: A Review. Front Neurosci, 14:547175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Athauda D, Foltynie T (2016). Insulin resistance and Parkinson's disease: A new target for disease modification? Prog Neurobiol, 145-146. [DOI] [PubMed] [Google Scholar]
- [52].Schapira AHV (2008). Mitochondria in the aetiology and pathogenesis of Parkinson's disease. Lancet Neurol, 7. [DOI] [PubMed] [Google Scholar]
- [53].Tysnes O-B, Storstein A (2017). Epidemiology of Parkinson's disease. J Neural Transm (Vienna), 124:901-5. [DOI] [PubMed] [Google Scholar]
- [54].Larsen SB, Hanss Z, Krüger R (2018). The genetic architecture of mitochondrial dysfunction in Parkinson's disease. Cell Tissue Res, 373:21-37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Dzamko N, Geczy CL, Halliday GM (2015). Inflammation is genetically implicated in Parkinson's disease. Neuroscience, 302. [DOI] [PubMed] [Google Scholar]
- [56].Mullin S, Schapira A (2013). α-Synuclein and mitochondrial dysfunction in Parkinson's disease. Mol Neurobiol, 47:587-97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Saez-Atienzar S, Bonet-Ponce L, Blesa JR, Romero FJ, Murphy MP, Jordan J, et al. (2014). The LRRK2 inhibitor GSK2578215A induces protective autophagy in SH-SY5Y cells: involvement of Drp-1-mediated mitochondrial fission and mitochondrial-derived ROS signaling. Cell Death Dis, 5:e1368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Clark IE, Dodson MW, Jiang C, Cao JH, Huh JR, Seol JH, et al. (2006). Drosophila pink1 is required for mitochondrial function and interacts genetically with parkin. Nature, 441:1162-6. [DOI] [PubMed] [Google Scholar]
- [59].Gegg ME, Schapira AHV (2011). PINK1-parkin-dependent mitophagy involves ubiquitination of mitofusins 1 and 2: Implications for Parkinson disease pathogenesis. Autophagy, 7:243-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Bonifati V, Rizzu P, van Baren MJ, Schaap O, Breedveld GJ, Krieger E, et al. (2003). Mutations in the DJ-1 gene associated with autosomal recessive early-onset parkinsonism. Science, 299:256-9. [DOI] [PubMed] [Google Scholar]
- [61].Guzman JN, Sanchez-Padilla J, Wokosin D, Kondapalli J, Ilijic E, Schumacker PT, et al. (2010). Oxidant stress evoked by pacemaking in dopaminergic neurons is attenuated by DJ-1. Nature, 468:696-700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Prasad EM, Hung S-Y (2020). Behavioral Tests in Neurotoxin-Induced Animal Models of Parkinson's Disease. Antioxidants(Basel), 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Petersen KF, Dufour S, Befroy D, Garcia R, Shulman GI (2004). Impaired mitochondrial activity in the insulin-resistant offspring of patients with type 2 diabetes. N Engl J Med, 350:664-71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Wallace DC (2007). Why do we still have a maternally inherited mitochondrial DNA? Insights from evolutionary medicine. Annu Rev Biochem, 76:781-821. [DOI] [PubMed] [Google Scholar]
- [65].Rovira-Llopis S, Apostolova N, Bañuls C, Muntané J, Rocha M, Victor VM (2018). Mitochondria, the NLRP3 Inflammasome, and Sirtuins in Type 2 Diabetes: New Therapeutic Targets. Antioxid Redox Signal, 29:749-91. [DOI] [PubMed] [Google Scholar]
- [66].Morino K, Petersen KF, Dufour S, Befroy D, Frattini J, Shatzkes N, et al. (2005). Reduced mitochondrial density and increased IRS-1 serine phosphorylation in muscle of insulin-resistant offspring of type 2 diabetic parents. J Clin Invest, 115:3587-93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Kleinridders A, Cai W, Cappellucci L, Ghazarian A, Collins WR, Vienberg SG, et al. (2015). Insulin resistance in brain alters dopamine turnover and causes behavioral disorders. Proc Natl Acad Sci U S A, 112:3463-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].Khang R, Park C, Shin JH (2015). Dysregulation of parkin in the substantia nigra of db/db and high-fat diet mice. Neuroscience, 294:182-92. [DOI] [PubMed] [Google Scholar]
- [69].Stark R, Roden M (2007). ESCI Award 2006. Mitochondrial function and endocrine diseases. Eur J Clin Invest, 37:236-48. [DOI] [PubMed] [Google Scholar]
- [70].Zheng B, Liao Z, Locascio JJ, Lesniak KA, Roderick SS, Watt ML, et al. (2010). PGC-1α, a potential therapeutic target for early intervention in Parkinson's disease. Sci Transl Med, 2:52ra73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Rocha M, Apostolova N, Diaz-Rua R, Muntane J, Victor VM (2020). Mitochondria and T2D: Role of Autophagy, ER Stress, and Inflammasome. Trends Endocrinol Metab, 31:725-41. [DOI] [PubMed] [Google Scholar]
- [72].Chen L, Liu C, Gao J, Xie Z, Chan LWC, Keating DJ, et al. (2017). Inhibition of Miro1 disturbs mitophagy and pancreatic β-cell function interfering insulin release via IRS-Akt-Foxo1 in diabetes. Oncotarget, 8:90693-705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Bhansali S, Bhansali A, Walia R, Saikia UN, Dhawan V (2017). Alterations in Mitochondrial Oxidative Stress and Mitophagy in Subjects with Prediabetes and Type 2 Diabetes Mellitus. Frontiers in endocrinology, 8:347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [74].Chung HY, Kim DH, Lee EK, Chung KW, Chung S, Lee B, et al. (2019). Redefining Chronic Inflammation in Aging and Age-Related Diseases: Proposal of the Senoinflammation Concept. Aging Dis, 10:367-82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [75].Hotamisligil GS (2006). Inflammation and metabolic disorders. Nature, 444:860-7. [DOI] [PubMed] [Google Scholar]
- [76].Esser N, Legrand-Poels S, Piette J, Scheen AJ, Paquot N (2014). Inflammation as a link between obesity, metabolic syndrome and type 2 diabetes. Diabetes Res Clin Pract, 105:141-50. [DOI] [PubMed] [Google Scholar]
- [77].Ransohoff RM, Cardona AE (2010). The myeloid cells of the central nervous system parenchyma. Nature, 468:253-62. [DOI] [PubMed] [Google Scholar]
- [78].Calabrese V, Santoro A, Monti D, Crupi R, Di Paola R, Latteri S, et al. (2018). Aging and Parkinson's Disease: Inflammaging, neuroinflammation and biological remodeling as key factors in pathogenesis. Free Radic Biol Med, 115:80-91. [DOI] [PubMed] [Google Scholar]
- [79].Imamura K, Hishikawa N, Sawada M, Nagatsu T, Yoshida M, Hashizume Y (2003). Distribution of major histocompatibility complex class II-positive microglia and cytokine profile of Parkinson's disease brains. Acta Neuropathol, 106:518-26. [DOI] [PubMed] [Google Scholar]
- [80].Martinez FO, Gordon S (2014). The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep, 6:13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Joers V, Tansey MG, Mulas G, Carta AR (2017). Microglial phenotypes in Parkinson's disease and animal models of the disease. Prog Neurobiol, 155:57-75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, et al. (2017). Neurotoxic reactive astrocytes are induced by activated microglia. Nature, 541:481-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Yun SP, Kam T-I, Panicker N, Kim S, Oh Y, Park J-S, et al. (2018). Block of A1 astrocyte conversion by microglia is neuroprotective in models of Parkinson's disease. Nat Med, 24:931-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [84].Kwon HS, Koh S-H (2020). Neuroinflammation in neurodegenerative disorders: the roles of microglia and astrocytes. Transl Neurodegener, 9:42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Tang Y, Le W (2016). Differential Roles of M1 and M2 Microglia in Neurodegenerative Diseases. Mol Neurobiol, 53:1181-94. [DOI] [PubMed] [Google Scholar]
- [86].Haque ME, Akther M, Jakaria M, Kim I-S, Azam S, Choi D-K (2020). Targeting the microglial NLRP3 inflammasome and its role in Parkinson's disease. Mov Disord, 35:20-33. [DOI] [PubMed] [Google Scholar]
- [87].Guo H, Callaway JB, Ting JPY (2015). Inflammasomes: mechanism of action, role in disease, and therapeutics. Nat Med, 21:677-87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].O'Neill CM, Lu C, Corbin KL, Sharma PR, Dula SB, Carter JD, et al. (2013). Circulating levels of IL-1B+IL-6 cause ER stress and dysfunction in islets from prediabetic male mice. Endocrinology, 154:3077-88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Lynch MA (2020). Can the emerging field of immunometabolism provide insights into neuroinflammation? Prog Neurobiol, 184:101719. [DOI] [PubMed] [Google Scholar]
- [90].Bernier L-P, York EM, MacVicar BA (2020). Immunometabolism in the Brain: How Metabolism Shapes Microglial Function. Trends Neurosci, 43:854-69. [DOI] [PubMed] [Google Scholar]
- [91].Yu J-W, Lee M-S (2016). Mitochondria and the NLRP3 inflammasome: physiological and pathological relevance. Arch Pharm Res, 39:1503-18. [DOI] [PubMed] [Google Scholar]
- [92].Castaner O, Goday A, Park Y-M, Lee S-H, Magkos F, Shiow S-ATE, et al. (2018). The Gut Microbiome Profile in Obesity: A Systematic Review. Int J Endocrinol, 2018:4095789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [93].Haikal C, Chen Q-Q, Li J-Y (2019). Microbiome changes: an indicator of Parkinson's disease? Transl Neurodegener, 8:38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Rinninella E, Raoul P, Cintoni M, Franceschi F, Miggiano GAD, Gasbarrini A, et al. (2019). What is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Zaky A, Glastras SJ, Wong MYW, Pollock CA, Saad S (2021). The Role of the Gut Microbiome in Diabetes and Obesity-Related Kidney Disease. Int J Mol Sci, 22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Demirci M, Bahar Tokman H, Taner Z, Keskin FE, Çağatay P, Ozturk Bakar Y, et al. (2020). Bacteroidetes and Firmicutes levels in gut microbiota and effects of hosts TLR2/TLR4 gene expression levels in adult type 1 diabetes patients in Istanbul, Turkey. J Diabetes Complications, 34:107449. [DOI] [PubMed] [Google Scholar]
- [97].Doumatey AP, Adeyemo A, Zhou J, Lei L, Adebamowo SN, Adebamowo C, et al. (2020). Gut Microbiome Profiles Are Associated With Type 2 Diabetes in Urban Africans. Front Cell Infect Microbiol, 10:63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Li Q, Chang Y, Zhang K, Chen H, Tao S, Zhang Z (2020). Implication of the gut microbiome composition of type 2 diabetic patients from northern China. Sci Rep, 10:5450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Balvers M, Deschasaux M, van den Born B-J, Zwinderman K, Nieuwdorp M, Levin E (2021). Analyzing Type 2 Diabetes Associations with the Gut Microbiome in Individuals from Two Ethnic Backgrounds Living in the Same Geographic Area. Nutrients, 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [100].Keshavarzian A, Green SJ, Engen PA, Voigt RM, Naqib A, Forsyth CB, et al. (2015). Colonic bacterial composition in Parkinson's disease. Mov Disord, 30:1351-60. [DOI] [PubMed] [Google Scholar]
- [101].Cirstea MS, Yu AC, Golz E, Sundvick K, Kliger D, Radisavljevic N, et al. (2020). Microbiota Composition and Metabolism Are Associated With Gut Function in Parkinson's Disease. Mov Disord, 35:1208-17. [DOI] [PubMed] [Google Scholar]
- [102].Yan Y, Ren S, Duan Y, Lu C, Niu Y, Wang Z, et al. (2021). Gut microbiota and metabolites of α-synuclein transgenic monkey models with early stage of Parkinson's disease. NPJ Biofilms Microbiomes, 7:69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [103].Rowland I, Gibson G, Heinken A, Scott K, Swann J, Thiele I, et al. (2018). Gut microbiota functions: metabolism of nutrients and other food components. Eur J Nutr, 57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [104].Vamanu E, Rai SN (2021). The Link between Obesity, Microbiota Dysbiosis, and Neurodegenerative Pathogenesis. Diseases, 9:45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [105].Kell DB, Pretorius E (2015). On the translocation of bacteria and their lipopolysaccharides between blood and peripheral locations in chronic, inflammatory diseases: the central roles of LPS and LPS-induced cell death. Integr Biol (Camb), 7:1339-77. [DOI] [PubMed] [Google Scholar]
- [106].Loffredo L, Ettorre E, Zicari AM, Inghilleri M, Nocella C, Perri L, et al. (2020). Oxidative Stress and Gut-Derived Lipopolysaccharides in Neurodegenerative Disease: Role of NOX2. Oxid Med Cell Longev, 2020:8630275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [107].Yang D, Zhao D, Ali Shah SZ, Wu W, Lai M, Zhang X, et al. (2019). Corrigendum: The Role of the Gut Microbiota in the Pathogenesis of Parkinson's Disease. Front Neurol, 10:1412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [108].Dutta SK, Verma S, Jain V, Surapaneni BK, Vinayek R, Phillips L, et al. (2019). Parkinson's Disease: The Emerging Role of Gut Dysbiosis, Antibiotics, Probiotics, and Fecal Microbiota Transplantation. J Neurogastroenterol Motil, 25:363-76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [109].Li H-Y, Zhou D-D, Gan R-Y, Huang S-Y, Zhao C-N, Shang A, et al. (2021). Effects and Mechanisms of Probiotics, Prebiotics, Synbiotics, and Postbiotics on Metabolic Diseases Targeting Gut Microbiota: A Narrative Review. Nutrients, 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [110].Snigdha S, Ha K, Tsai P, Dinan TG, Bartos JD, Shahid M (2021). Probiotics: Potential novel therapeutics for microbiota-gut-brain axis dysfunction across gender and lifespan. Pharmacol Ther: 107978. [DOI] [PubMed] [Google Scholar]
- [111].Liptak R, Gromova B, Gardlik R (2021). Fecal Microbiota Transplantation as a Tool for Therapeutic Modulation of Non-gastrointestinal Disorders. Front Med (Lausanne), 8:665520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [112].Fiory F, Perruolo G, Cimmino I, Cabaro S, Pignalosa FC, Miele C, et al. (2019). The Relevance of Insulin Action in the Dopaminergic System. Front Neurosci, 13:868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [113].Brauer R, Wei L, Ma T, Athauda D, Girges C, Vijiaratnam N, et al. (2020). Diabetes medications and risk of Parkinson's disease: a cohort study of patients with diabetes. Brain, 143:3067-76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [114].Qin X, Zhang X, Li P, Wang M, Yan L, Bao Z, et al. (2021). Association Between Diabetes Medications and the Risk of Parkinson's Disease: A Systematic Review and Meta-Analysis. Front Neurol, 12:678649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [115].Wang S-Y, Wu S-L, Chen T-C, Chuang C-S (2020). Antidiabetic Agents for Treatment of Parkinson's Disease: A Meta-Analysis. Int J Environ Res Public Health, 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [116].Jeong SH, Chung SJ, Yoo HS, Hong N, Jung JH, Baik K, et al. (2021). Beneficial effects of dipeptidyl peptidase-4 inhibitors in diabetic Parkinson's disease. Brain, 144:1127-37. [DOI] [PubMed] [Google Scholar]
- [117].Hölscher C (2014). First clinical data of the neuroprotective effects of nasal insulin application in patients with Alzheimer's disease. Alzheimers Dement, 10:S33-S7. [DOI] [PubMed] [Google Scholar]
- [118].Pang Y, Lin S, Wright C, Shen J, Carter K, Bhatt A, et al. (2016). Intranasal insulin protects against substantia nigra dopaminergic neuronal loss and alleviates motor deficits induced by 6-OHDA in rats. Neuroscience, 318:157-65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [119].Novak P, Pimentel Maldonado DA, Novak V (2019). Safety and preliminary efficacy of intranasal insulin for cognitive impairment in Parkinson disease and multiple system atrophy: A double-blinded placebo-controlled pilot study. PLoS One, 14:e0214364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [120].Cohen E, Dillin A (2008). The insulin paradox: aging, proteotoxicity and neurodegeneration. Nat Rev Neurosci, 9:759-67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [121].Akintola AA, van Heemst D (2015). Insulin, aging, and the brain: mechanisms and implications. Frontiers in endocrinology, 6:13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [122].Castilla-Cortázar I, Aguirre GA, Femat-Roldán G, Martín-Estal I, Espinosa L (2020). Is insulin-like growth factor-1 involved in Parkinson's disease development? Journal of translational medicine, 18:70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [123].Viollet B, Guigas B, Sanz Garcia N, Leclerc J, Foretz M, Andreelli F (2012). Cellular and molecular mechanisms of metformin: an overview. Clin Sci (Lond), 122:253-70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [124].Rena G, Hardie DG, Pearson ER (2017). The mechanisms of action of metformin. Diabetologia, 60:1577-85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [125].Nath N, Khan M, Paintlia MK, Singh I, Hoda MN, Giri S (2009). Metformin attenuated the autoimmune disease of the central nervous system in animal models of multiple sclerosis. J Immunol, 182:8005-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [126].Katila N, Bhurtel S, Shadfar S, Srivastav S, Neupane S, Ojha U, et al. (2017). Metformin lowers α-synuclein phosphorylation and upregulates neurotrophic factor in the MPTP mouse model of Parkinson's disease. Neuropharmacology, 125:396-407. [DOI] [PubMed] [Google Scholar]
- [127].Lu M, Su C, Qiao C, Bian Y, Ding J, Hu G (2016). Metformin Prevents Dopaminergic Neuron Death in MPTP/P-Induced Mouse Model of Parkinson's Disease via Autophagy and Mitochondrial ROS Clearance. Int J Neuropsychopharmacol, 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [128].Patil SP, Jain PD, Ghumatkar PJ, Tambe R, Sathaye S (2014). Neuroprotective effect of metformin in MPTP-induced Parkinson's disease in mice. Neuroscience, 277:747-54. [DOI] [PubMed] [Google Scholar]
- [129].Ozbey G, Nemutlu-Samur D, Parlak H, Yildirim S, Aslan M, Tanriover G, et al. (2020). Metformin protects rotenone-induced dopaminergic neurodegeneration by reducing lipid peroxidation. Pharmacol Rep, 72:1397-406. [DOI] [PubMed] [Google Scholar]
- [130].Katila N, Bhurtel S, Park P-H, Choi D-Y (2021). Metformin attenuates rotenone-induced oxidative stress and mitochondrial damage via the AKT/Nrf2 pathway. Neurochem Int, 148:105120. [DOI] [PubMed] [Google Scholar]
- [131].El-Ghaiesh SH, Bahr HI, Ibrahiem AT, Ghorab D, Alomar SY, Farag NE, et al. (2020). Metformin Protects From Rotenone-Induced Nigrostriatal Neuronal Death in Adult Mice by Activating AMPK-FOXO3 Signaling and Mitigation of Angiogenesis. Front Mol Neurosci, 13:84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [132].Sportelli C, Urso D, Jenner P, Chaudhuri KR (2020). Metformin as a Potential Neuroprotective Agent in Prodromal Parkinson's Disease-Viewpoint. Front Neurol, 11:556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [133].Paudel YN, Angelopoulou E, Piperi C, Shaikh MF, Othman I (2020). Emerging neuroprotective effect of metformin in Parkinson's disease: A molecular crosstalk. Pharmacol Res, 152:104593. [DOI] [PubMed] [Google Scholar]
- [134].Pérez-Revuelta BI, Hettich MM, Ciociaro A, Rotermund C, Kahle PJ, Krauss S, et al. (2014). Metformin lowers Ser-129 phosphorylated α-synuclein levels via mTOR-dependent protein phosphatase 2A activation. Cell Death Dis, 5:e1209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [135].Mor DE, Sohrabi S, Kaletsky R, Keyes W, Tartici A, Kalia V, et al. (2020). Metformin rescues Parkinson's disease phenotypes caused by hyperactive mitochondria. Proc Natl Acad Sci U S A, 117:26438-47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [136].Fitzgerald JC, Zimprich A, Carvajal Berrio DA, Schindler KM, Maurer B, Schulte C, et al. (2017). Metformin reverses TRAP1 mutation-associated alterations in mitochondrial function in Parkinson's disease. Brain, 140:2444-59. [DOI] [PubMed] [Google Scholar]
- [137].Bharath LP, Agrawal M, McCambridge G, Nicholas DA, Hasturk H, Liu J, et al. (2020). Metformin Enhances Autophagy and Normalizes Mitochondrial Function to Alleviate Aging-Associated Inflammation. Cell Metab, 32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [138].Tayara K, Espinosa-Oliva AM, García-Domínguez I, Ismaiel AA, Boza-Serrano A, Deierborg T, et al. (2018). Divergent Effects of Metformin on an Inflammatory Model of Parkinson's Disease. Front Cell Neurosci, 12:440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [139].Ping F, Jiang N, Li Y (2020). Association between metformin and neurodegenerative diseases of observational studies: systematic review and meta-analysis. BMJ Open Diabetes Res Care, 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [140].Davidson MA, Mattison DR, Azoulay L, Krewski D (2018). Thiazolidinedione drugs in the treatment of type 2 diabetes mellitus: past, present and future. Crit Rev Toxicol, 48. [DOI] [PubMed] [Google Scholar]
- [141].Swanson C, Emborg M (2014). Expression of peroxisome proliferator-activated receptor-gamma in the substantia nigra of hemiparkinsonian nonhuman primates. Neurol Res, 36:634-46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [142].P P, Justin A, Ananda Kumar TD, Chinaswamy M, Kumar BRP (2021). Glitazones Activate PGC-1α Signaling via PPAR-γ: A Promising Strategy for Antiparkinsonism Therapeutics. ACS Chem Neurosci, 12:2261-72. [DOI] [PubMed] [Google Scholar]
- [143].Lee EY, Lee JE, Park JH, Shin IC, Koh HC (2012). Rosiglitazone, a PPAR-γ agonist, protects against striatal dopaminergic neurodegeneration induced by 6-OHDA lesions in the substantia nigra of rats. Toxicol Lett, 213:332-44. [DOI] [PubMed] [Google Scholar]
- [144].Bonato JM, Bassani TB, Milani H, Vital MABF, de Oliveira RMW (2018). Pioglitazone reduces mortality, prevents depressive-like behavior, and impacts hippocampal neurogenesis in the 6-OHDA model of Parkinson's disease in rats. Exp Neurol, 300:188-200. [DOI] [PubMed] [Google Scholar]
- [145].Swanson CR, Joers V, Bondarenko V, Brunner K, Simmons HA, Ziegler TE, et al. (2011). The PPAR-γ agonist pioglitazone modulates inflammation and induces neuroprotection in parkinsonian monkeys. J Neuroinflammation, 8:91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [146].Pisanu A, Lecca D, Mulas G, Wardas J, Simbula G, Spiga S, et al. (2014). Dynamic changes in pro- and anti-inflammatory cytokines in microglia after PPAR-γ agonist neuroprotective treatment in the MPTPp mouse model of progressive Parkinson's disease. Neurobiol Dis, 71:280-91. [DOI] [PubMed] [Google Scholar]
- [147].Hunter RL, Choi D-Y, Ross SA, Bing G (2008). Protective properties afforded by pioglitazone against intrastriatal LPS in Sprague-Dawley rats. Neurosci Lett, 432:198-201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [148].He X, Feng L, Meng H, Wang X, Liu S (2012). Rosiglitazone protects dopaminergic neurons against lipopolysaccharide-induced neurotoxicity through inhibition of microglia activation. Int J Neurosci, 122:532-40. [DOI] [PubMed] [Google Scholar]
- [149].Pinto M, Nissanka N, Peralta S, Brambilla R, Diaz F, Moraes CT (2016). Pioglitazone ameliorates the phenotype of a novel Parkinson's disease mouse model by reducing neuroinflammation. Mol Neurodegener, 11:25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [150].Investigators NETiPDN-PF-Z (2015). Pioglitazone in early Parkinson's disease: a phase 2, multicentre, double-blind, randomised trial. Lancet Neurol, 14:795-803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [151].Chang KL, Pee HN, Yang S, Ho PC (2015). Influence of drug transporters and stereoselectivity on the brain penetration of pioglitazone as a potential medicine against Alzheimer's disease. Sci Rep, 5:9000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [152].Nauck MA, Meier JJ (2016). The incretin effect in healthy individuals and those with type 2 diabetes: physiology, pathophysiology, and response to therapeutic interventions. Lancet Diabetes Endocrinol, 4:525-36. [DOI] [PubMed] [Google Scholar]
- [153].Hölscher C (2020). Brain insulin resistance: role in neurodegenerative disease and potential for targeting. Expert Opin Investig Drugs, 29:333-48. [DOI] [PubMed] [Google Scholar]
- [154].Harkavyi A, Abuirmeileh A, Lever R, Kingsbury AE, Biggs CS, Whitton PS (2008). Glucagon-like peptide 1 receptor stimulation reverses key deficits in distinct rodent models of Parkinson's disease. J Neuroinflammation, 5:19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [155].Li Y, Perry T, Kindy MS, Harvey BK, Tweedie D, Holloway HW, et al. (2009). GLP-1 receptor stimulation preserves primary cortical and dopaminergic neurons in cellular and rodent models of stroke and Parkinsonism. Proc Natl Acad Sci U S A, 106:1285-90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [156].Liu W, Jalewa J, Sharma M, Li G, Li L, Hölscher C (2015). Neuroprotective effects of lixisenatide and liraglutide in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson's disease. Neuroscience, 303:42-50. [DOI] [PubMed] [Google Scholar]
- [157].Muscogiuri G, DeFronzo RA, Gastaldelli A, Holst JJ (2017). Glucagon-like Peptide-1 and the Central/Peripheral Nervous System: Crosstalk in Diabetes. Trends Endocrinol Metab, 28. [DOI] [PubMed] [Google Scholar]
- [158].Erbil D, Eren CY, Demirel C, Küçüker MU, Solaroğlu I, Eser HY (2019). GLP-1's role in neuroprotection: a systematic review. Brain Inj, 33:734-819. [DOI] [PubMed] [Google Scholar]
- [159].Mulvaney CA, Duarte GS, Handley J, Evans DJ, Menon S, Wyse R, et al. (2020). GLP-1 receptor agonists for Parkinson's disease. Cochrane Database Syst Rev, 7:CD012990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [160].Aviles-Olmos I, Dickson J, Kefalopoulou Z, Djamshidian A, Ell P, Soderlund T, et al. (2013). Exenatide and the treatment of patients with Parkinson's disease. J Clin Invest, 123:2730-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [161].Aviles-Olmos I, Dickson J, Kefalopoulou Z, Djamshidian A, Kahan J, Ell P, et al. (2014). Motor and cognitive advantages persist 12 months after exenatide exposure in Parkinson's disease. J Parkinsons Dis, 4:337-44. [DOI] [PubMed] [Google Scholar]
- [162].Athauda D, Maclagan K, Skene SS, Bajwa-Joseph M, Letchford D, Chowdhury K, et al. (2017). Exenatide once weekly versus placebo in Parkinson's disease: a randomised, double-blind, placebo-controlled trial. Lancet, 390:1664-75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [163].Athauda D, Gulyani S, Karnati HK, Li Y, Tweedie D, Mustapic M, et al. (2019). Utility of Neuronal-Derived Exosomes to Examine Molecular Mechanisms That Affect Motor Function in Patients With Parkinson Disease: A Secondary Analysis of the Exenatide-PD Trial. JAMA Neurol, 76:420-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [164].Kim DS, Choi HI, Wang Y, Luo Y, Hoffer BJ, Greig NH (2017). A New Treatment Strategy for Parkinson's Disease through the Gut-Brain Axis: The Glucagon-Like Peptide-1 Receptor Pathway. Cell transplantation, 26:1560-71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [165].Meier JJ (2012). GLP-1 receptor agonists for individualized treatment of type 2 diabetes mellitus. Nat Rev Endocrinol, 8:728-42. [DOI] [PubMed] [Google Scholar]
- [166].Jose T, Inzucchi SE (2012). Cardiovascular effects of the DPP-4 inhibitors. Diab Vasc Dis Res, 9:109-16. [DOI] [PubMed] [Google Scholar]
- [167].Mousa SA, Ayoub BM (2019). Repositioning of dipeptidyl peptidase-4 inhibitors and glucagon like peptide-1 agonists as potential neuroprotective agents. Neural Regen Res, 14:745-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [168].Abdelsalam RM, Safar MM (2015). Neuroprotective effects of vildagliptin in rat rotenone Parkinson's disease model: role of RAGE-NFκB and Nrf2-antioxidant signaling pathways. J Neurochem, 133:700-7. [DOI] [PubMed] [Google Scholar]
- [169].Nassar NN, Al-Shorbagy MY, Arab HH, Abdallah DM (2015). Saxagliptin: a novel antiparkinsonian approach. Neuropharmacology, 89:308-17. [DOI] [PubMed] [Google Scholar]
- [170].Badawi GA, Abd El Fattah MA, Zaki HF, El Sayed MI (2017). Sitagliptin and liraglutide reversed nigrostriatal degeneration of rodent brain in rotenone-induced Parkinson's disease. Inflammopharmacology, 25:369-82. [DOI] [PubMed] [Google Scholar]
- [171].Badawi GA, Abd El Fattah MA, Zaki HF, El Sayed MI (2019). Sitagliptin and Liraglutide Modulate L-dopa Effect and Attenuate Dyskinetic Movements in Rotenone-Lesioned Rats. Neurotox Res, 35:635-53. [DOI] [PubMed] [Google Scholar]
- [172].Safar MM, Abdelkader NF, Ramadan E, Kortam MA, Mohamed AF (2021). Novel mechanistic insights towards the repositioning of alogliptin in Parkinson's disease. Life Sci, 287:120132. [DOI] [PubMed] [Google Scholar]
- [173].Ayoub BM, Mowaka S, Safar MM, Ashoush N, Arafa MG, Michel HE, et al. (2018). Repositioning of Omarigliptin as a once-weekly intranasal Anti-parkinsonian Agent. Sci Rep, 8:8959. [DOI] [PMC free article] [PubMed] [Google Scholar]

