Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Dec 1.
Published in final edited form as: Curr Opin Pharmacol. 2020 Nov 1;55:60–72. doi: 10.1016/j.coph.2020.09.004

Lipid Deposition and Metaflammation in Diabetic Kidney Disease

Alla Mitrofanova 1,2,3, Antonio M Fontanella 1,2, Sandra Merscher 1,2, Alessia Fornoni 1,2,*
PMCID: PMC7769904  NIHMSID: NIHMS1644768  PMID: 33137677

Abstract

A critical link between metabolic disorders and a form of low-grade systemic and chronic inflammation has been recently established and named “Metaflammation”. Metaflammation has been recognized as a key mediator of both micro- and macrovascular complications of diabetes and as a significant contributor to the development of diabetic kidney disease (DKD). The goal of this review is to summarize the contribution of diabetes-induced inflammation and the related signaling pathways to diabetic complications, with a particular focus on how innate immunity and lipid metabolism influence each other.

Introduction

Obesity and diabetes mellitus (DM) have become a worldwide pandemic, and diabetes-related complications represent a major health problem, with diabetic kidney disease (DKD) remaining the leading cause of kidney disease worldwide. Among several contributors to DKD, inflammation is currently recognized to contribute to the progression of chronic kidney disease (CKD). Moreover, several inflammatory biomarkers, such as adhesion molecules (ICAM-1, VCAM-1), cytokines (IL-6, TNF), c-reactive protein, fibrinogen, serum albumin, white blood cell counts, have been reported to be prognostic to risk-stratify patients for disease progression and all-cause mortality [1].

In obesity and type 2 diabetes, inflammatory biomarkers are traditionally thought to be derived from adipose tissue. As an example, secretion of inflammatory cytokines such as interleukin-1β (IL-1β) [2] and tumor necrosis factor (TNF) [3] has been shown to contribute to the development of insulin resistance and type 2 diabetes (T2D). Secretion of inflammation cytokines may be the consequence of inflammasome activation via caspase-1. Besides the inflammatory cytokine secretion from adipose tissue, de novo recruitment of macrophages into adipose tissue has been described (reviewed in Ref. [4] Once recruited, they become activated by the uptake of saturated fatty acids via Toll-like receptors 2 and 4, which triggers the activation of inflammatory signaling pathways via interferon regulatory factor 3 (IRF3), activator protein 1 (AP1) and nuclear factor-κB (NFκB). Inflammatory mediators can perpetuate lipid dysmetabolism locally in adipose tissue, or affect the concentration of other circulating inflammatory cytokines that in turn will influence the function of peripheral organs including the kidneys. Finally, inflammatory cytokines produced locally in peripheral organs can also influence disease progression, as clearly shown in the pathogenesis of DKD.

In CKD, the concentration of several circulating cytokines correlates with albuminuria. In particular, increased levels of circulating tumor necrosis factor (TNF) and TNF-receptors 1 and 2 are independent predictors of decline in renal function [5]. Several circulating inflammatory cytokines have been linked to progression to end stage renal disease (ESRD) in diabetes and referred to as the Kidney Risk Inflammatory Signature (KRIS). Seventeen of KRIS proteins in the T2D Validation Joslin Cohort and 16 of KRIS proteins in the T2D Replication Pima Cohort were significantly upregulated in patients who developed ESRD compared to those who did not develop ESRD, as discussed in the following section [6]. However, whether systemic or local inflammation contributes to disease progression is still being debated. In fact, increased local production of TNF, independently of circulating TNF levels, has shown to correlate with the loss of eGFR and to contribute to free cholesterol-mediated podocyte apoptosis via nuclear factor of activated T cells (NFAT)-mediated ATP-binding cassette transporter A1 (ABCA1) suppression [7]. In addition, a role of locally produced sphingolipids in kidney diseases has been recently established, and resulted in the discovery of novel key mediators of cell injury such as sphingosine-1-phosphate [8], ceramide-1-phosphate [9] and C16 ceramide [10].

This review will summarize the recent studies on the role of inflammatory mediators in diabetes with focus on the link between innate immunity and lipid metabolism patients, a small number of cross-sectional studies [11,12] and more longitudinal studies [1317] demonstrated that C-reactive protein (CRP), IL-6 and TNF are associated with an increased risk of developing T2D.

Circulating Inflammatory Mediators and Metaflammation in DKD

Under physiological conditions, inflammation is a process mediated by cytokines, chemokines, and lipid mediators, including prostaglandins and leukotrienes. In healthy patients, a small number of cross-sectional studies [11,12] and more longitudinal studies [1317] demonstrated that C-reactive protein (CRP), IL-6 and TNF are associated with an increased risk of developing T2D.

Inflammation in diabetes.

T2D is characterized by peripheral insulin resistance, which is accompanied by chronic low-grade inflammation in peripheral tissues such as muscles, liver and adipose tissue, redefining diabetes as an immune disorder. Classic inflammatory markers in diabetes include lectin, TNF and its receptors 1 and 2, interleukins (IL-6, IL-1β, IL-17, IL-18), monocyte chemoattractant protein 1 (MCP1), adhesion molecules VCAM1 and ICAM1, c-reactive protein (CRP) and nuclear factor-κB (NFκB). Increased production of interleukins has been shown in many animal models of diabetes [1821] and these findings were confirmed in a cohort of T2D patients, where elevated levels of IL-1α, IL-8 and IL-18 were associated with early stages of DKD and correlated with podocyte damage and proximal tubular dysfunction [22]. Interestingly, IL-37 treatment of C57BL/6 mice exposed to high fat diet was associated with decreased secretion of pro-inflammatory cytokines such as IL-1β, TNF and IL-6 and improved established insulin resistance and glucose intolerance in the liver and the adipose tissue [23], suggesting that IL-37 may be a potential treatment strategy in metabolic diseases. Similar findings were demonstrated in humans as mentioned earlier, in the T2D Validation Joslin Cohort and in the T2D Replication Pima Cohort, where besides the TNFR superfamily proteins, other cytokines including IL15RA, IL17F, CD55, CD300C, CCL14, CCL15, CSF1, HAVCR2, IL1R1 and IL18R1 have been reported to be associated with increased risk of progression to ESRD [6]. In the Tatar ethnic group, Russia (n=440), CCL20-rs6749704 and CCL5-rs2107538 were shown to be associated with T2D [24]. Fractalkine (CX3CL1) was also found to be associated with development of inflammatory conditions in metabolic diseases. In T2D patients (n=47), plasma levels of CX3CL1 has been shown to correlate with inflammatory chemokines/cytokines, such as CCL3, CCL4, CCL11, CXCL1, INF-α2, IL-17A, IL-1β, IL-6 and TNF [25]. Similarly, another study of patients with advanced DKD (n=70) demonstrated strong correlation of C-X-C motif ligand 16 (CXCL16) and endostatin with interstitial fibrosis and tubular atrophy [26].

MCP1 signaling was found significantly increased in patients with T2D [2729]. A common A/G polymorphism located at the position −2518 in the MCP1 distal regulatory region has been shown to be associated with diabetes. For example, in Turkish T2D patients [30] and Chinese T2D population [31], prevalence of T2D was reported in individuals with the MCP1 G-2518 genotype. A study on Japanese T2D patients revealed that −2518AA carriers have higher serum MCP1 concentration and increased insulin resistance compared to −2518G carriers [32]. The methylation status of CpG sites in the MCP1 promotor was also reported to be associated with diabetes, suggesting that hypomethylation of CpG motifs in T2D patients results in increased serum MCP1 levels [33]. Interestingly, blockade or lack of MCP1 in animal models resulted in reduced renal macrophage/monocyte infiltration and reduction of interstitial fibrosis [34], which will be discussed in details below.

Several adhesion molecules, such as vascular cell adhesion molecule 1 (VCAM1), also known as CD106, and intercellular adhesion molecule 1 (ICAM1), also known as CD54, were also shown to contribute to both types of diabetes. Subsequent studies have demonstrated that elevated levels of plasma ICAM1 in T1D patients (n=1,441) are independently associated with the onset of sustained microalbuminuria [35,36]. Similarly, levels of VCAM1 and ICAM1 were significantly higher in plasma of T2D patients who were enrolled in a cross-sectional study in Saudi Arabia regardless of associated cardiovascular complications [37], as well as in T2D patients with hypertension [38]. In a study on Mexican Americans, a prospective association of serum ICAM1 and VCAM1 with incidence of T2D was determined [39].

Inflammatory mediators and biomarkers of DKD.

Low-grade inflammation is also a common feature of CKD, and evidence suggests that persistent inflammation may be a risk factor for the progression of CKD per se and for the cardiovascular morbidity and mortality associated with CKD. Interestingly, other studies demonstrated that the number of white blood cells (neutrophils, eosinophils, basophils, lymphocytes, and monocytes) is increased in the transition from normalbuminuria to microalbuminuria [40,41], suggesting that white blood cells are also implicated in the development of DKD.

While the molecular mechanisms that attract monocytes to renal sites are not fully understood, endothelial leukocyte adhesion molecules, including VCAM1and ICAM1, have been shown to play an important role in the initiation of renal inflammation. These molecules are abundantly expressed in kidneys of patients with DKD and VCAM1/ICAM1 circulating levels are independently associated with DKD progression [42]. In a case-control study of a Diabetic Kidney Disease cohort of patients from Saudi Arabia, T2D patients with DKD had higher levels of VCAM1 and ICAM1 compared to those without DKD [43]. Another study, based on a meta-analysis, revealed that ICAM1-rs5498 might be a risk factor of DKD in T1D patients [44]. In T1D patients, ICAM1 K469E polymorphism was also shown to be associated with DKD [45]. In Turkish T2D patients with DKD, elevated levels of plasma VCAM1 and ICAM1 were also demonstrated and what is of particular interest, haemodialysis did not reduce those values [46]. While we do not know the exact role of adhesion molecules in the development of DKD, a recent study using the STZ mouse model of diabetes showed that micro RNA miR-146a expression may regulate levels of VCAM1 and ICAM1 (more miR-146a, less VCAM1 and ICAM1) [47], but no evidence was provided linking the miR-146a/VCAM/ICAM axis to the functional or structural alterations in DKD. Interestingly, ICAM1-deficient mice failed to develop diabetes in a STZ model [48], while the deletion of ICAM1 in db/db mice resulted in decreased renal inflammation [49], suggesting that ICAM1 is critically involved in the pathogenesis of DKD.

Circulating levels of TNF, TNF receptor 1 (TNFR1) and 2 (TNFR2) predict progression to ESRD in patients with type 1 diabetes (T1D) and T2D [5]. Our studies on a small number of patients with T1D enrolled in the FinnDiane Study cohort demonstrated that elevated levels of TNF, TNFR1 and TNFR2 in serum are associated with presence of DKD [7]. In Joslin and Pima cohorts of patients with T2D, elevated plasma concentrations of TNFR1 and TNFR2 were strongly associated with a risk of early renal function decline [6]. Additionally, patients with higher renal TNFR2 values were threefold more likely to experience renal function decline compared to controls [50]. In the Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications cohort, increased levels of soluble TNFR1 and TNFR2 was associated with an increase in the odds to develop proteinuria [51]. Table 1 summarizes the clinical studies linking TNF, TNFR1 and TNFR2 to DKD.

Table 1.

Summary of clinical studies linking circulating TNF and TNF receptors and DKD.

Population Key Findings Ref
TNF T1D Joslin Cohort (n=219) No association with ESRD risk was found. [6]
T2D Joslin Cohort (n=144) No association with ESRD risk was found. [6]
T2D normalbuminuric patients Association with reduced renal function (eGFR<60mL/min/1.73m2), which was lost after adjusting for TNFR covariates. [132]
TNFR1/TNFR2 T1D Joslin Cohort (n=275) Elevated serum levels predict early renal function loss in T1D patients without proteinuria. [50]
T1D Joslin Cohort (n=219) and T2D Joslin Cohort (n=144) Elevated serum levels predict the highest adjusted hazard ratios (HR) for ESRD. [6]
Type 1 Diabetes Control and Complications Trial (DCCT)/Epidemiology of Diabetes Interventions and Complications Cohort (n=1,237) Elevated serum levels predict macroalbuminuria in patients [51]
T2D Joslin Cohort (n=267) Stronger association of elevated serum levels with ESRD in patients without proteinuria. [133]
T2D Pima Indians Cohort (n=193) Elevated serum concentrations are associated with increased risk of ESRD in American Indians with T2D. [134]
Indigenous Australians with Diabetes Association of TNFR1 with ESRD progression independent of albuminuria and eGFR. [135]
T2D normalbuminuric patients Association with reduced renal function (eGFR<60mL/min/1.73m2). [132]

Higher levels of MCP1 were also found in kidneys of the STZ-induced experimental mouse model of diabetes [52], which were also associated with overexpression of alpha-kinase 1 (ALPK1), an enzyme involved in TNF secretion, NFκB activation and production of pro-inflammatory cytokines (including IL-1β, IL-8 and TGF-β1). Additionally, MCP1 has been shown to be strongly associated with a decline in renal function in patients with diabetes [53,54]. In Malaysian T2D patients, a polymorphism in MCP1 (MCP1-rs3917887) was associated with DKD [55]. The MCP1-rs1024611 polymorphism was also found to be associated with foot ulcers in a Chinese Han population with T2D (n=400) [56]. The role of the MCP1 −2518 polymorphism in DKD remains unclear. While in an Indian population the presence of −2518GG [57] and in Korean patients, the −2518A allele of the MCP1 gene [58] are associated with increased risk of DKD, another study of Korean patients with T2D did not reveal an association of the MCP1 −2518 A/G polymorphism with DKD. However, these differences might be explained by the variations in the experiment design and need further investigation.

In contrast to MCP1, epidermal growth factor (EGF) has a protective role in kidney injury. Recent studies have suggested that urinary EGF/MCP1 ratio might be another reliable biomarker of progression to DKD. Thus, in a prospective T2D cohort from Thailand (n=83) a significantly urinary EGF/MCP1 ratio was associated with rapid renal decline, showing significantly lower urine EGF/MCP1 ratio in patients with a glomerular filtration rate (GFR) decline more than 25%/year [54]. The cross-sectional study on Chinese patients (n=1811) showed that urinary EGF- or MCP1-to-creatinine ratios negatively correlated with the occurrence of DKD, while in the longitudinal cohort (n=208) from the same study urinary EFG/MCP1 ratio also correlated with the percentage change of estimated GFR slope [59].

Nuclear factor-κB (NFκB) is another important transcription factor involved in the pathophysiology of diabetes and DKD. Glucose can activate NFκB, resulting in increased inflammatory genes expression. Activation of NFκB leads to activation of MCP1 and macrophage infiltration [60], activation of transforming growth factor β (TGFβ) with extracellular matrix accumulation and fibrosis [61]. A recent study demonstrated that induction of diabetes mellitus in Psammomys obesus using high fat diet results in increased expression of NFκB after three months suggesting its involvement in DKD development [62]. Mesangial cells cultured in high glucose (30 mmol/L), demonstrated activation of TLR4/NFκB p65/NGAL signaling pathway and promoted inflammatory reaction via suppression of Klotho [63].

Taken together, several circulating cytokines have been proposed as biomarkers for risk stratification and may be involved in the pathogenesis of diabetes and DKD (Figure 1), making them a potential drug target for the treatment of patients with DKD. However, as a cluster of cytokines rather than a single cytokine is often involved, targeting of common pathways activated by multiple cytokines rather than targeting single cytokines may prove more efficacious in the treatment of patients with DKD.

Figure 1. The role of lipid deposition and Metaflammation in the pathogenesis of diabetes and its complications.

Figure 1.

Hypercaloric diet and genetics affect the function of pancreas, liver, muscles, and adipose tissue, leading to the formation of insulin resistance, hyperglycemia and hyperlipidemia via activation of the innate immune system, the production of pro-inflammatory cytokines and the deposition of lipids. Consequently, this results in diabetic micro- and macro-complications. This image was created using BioRender software (biorender.com).

Abbreviations: TLRs – Toll-like receptors; CLRs – C-type lectin receptors; NLRs – nucleotide-binding domain and leucine-rich repeat-containing receptors; RLRs – retinoic acid-inducible gene I-like receptors; cGAS – cyclic GMP-AMP synthase; STING – stimulator of interferon genes; JNK – c-Jun N-terminal kinase; NF-κB – nuclear factor kappa-light-chain-enchancer of activated B cells; IL – interleukin; TNFα – tumor necrosis factor alpha; ICAM1 – intercellular adhesion molecule 1; VCAM1 – vascular cell adhesion molecule 1; CX3CL1 – C-X3-C motif chemokine ligand 1; CCL5 – C-C motif ligand 5; MCP-1 – monocyte chemoattractant protein-1; TAG – triacylglycerol; FFA – free fatty acids; CD36 – scavenger receptor class B member 3; ABCA1 – ATP-binding cassette subfamily A member 1; ABCG1 – ATP-binding cassette subfamily G member 1; DAG – diacylglycerol; Cer – ceramide; Sph – sphingosine; SphK2 – sphingosine kinase 2; S1P – sphingosine-1-phospahte; C1P – ceramide-1-phosphate; SMPDL3b – sphingomyelin phosphodiesterase acid-like 3b.

Targeting the inflammatory pathways in DKD.

Several approaches have been proposed to treat inflammation in DKD. Besides lifestyle modifications (diet and exercise), some drugs have been reported to be promising in reduction of inflammation in DKD patients.

Bardoxolone is known to target oxidative stress and reduce inflammation via decreasing TGFβ. Clinical trials of Bardoxolone Methyl (BEACON) showed preliminary significant improvement in renal function in CKD patients with T2D, but this trial was stopped due to an increase in heart failure events [64]. The BEACON trial post-hoc analysis (n=2185), however, demonstrated that Bardoxolone Methyl preserves kidney function by preventing decline of estimated GFR, which was sustained 4 weeks post-cessation of treatment [65]. Currently, another trial of Bardoxolone in Japanese T2D patients (AYAME study, NCT03550443) is ongoing with an estimated completion date of March 2022.

Post-hoc analysis of using of Selonsertib, a highly selective small-molecular inhibitor of apoptosis-regulating kinase 1 (which in turn regulates activity of p38 MAPK and c-JNK), suggest that it may slow DKD progression [66]. Currently, Selonsertib is in a clinical trial Phase 3 (MOSAIC; NCT04026165).

Another potential medication to treat inflammation in DKD is Pentoxifylline, a methylxantine derivative with nonspecific inhibition of phosphodiesterases. In animal models, Pentoxifylline was shown to have a marked antiproteinuric effect with attenuating interstitial inflammation and renal injury progression [67,68] and decreasing levels of TNF and IL-6 [69]. In the PREDIAN trial, using of 2 years of Pentoxifylline therapy in combination with RAAS blockade resulted in decreased proteinuria and urinary concentration of TNF and slowed the decline in estimated GFR [70].

Metaflammation Activates Pathways of the Innate Immune System Involved in Insulin Resistance and DKD

Even if DKD is not considered as a primary “immune-mediated” renal disease, more and more evidence supports the involvement of many components of the immune system in DKD initiation and progression. As an evolutionarily conserved system, the innate immune system is the first line of host defense against pathogens and internal danger signals, which relies on pattern recognition receptors, such as toll-like receptors (TLRs), C-type lectin receptors (CLRs), RIG-I-like receptors (RLRs), intracellular Nod-like receptors (NLRs), HIN-200 receptors and the recently discovered cyclic GMP-AMP synthase-stimulator of interferon genes pathway (cGAS-STING).

More than 90% of scientific reports investigating role of TLRs in diabetes revealed that TLR2 is positively associated with T2D and its complications, while T1D is also associated with TLR1, TLR3, TLR4, TLR7 and TLR9 [7175]. In NOD mice, a model of T1D, TLR4 mediates cardiac lipid accumulation and diabetic heart disease [76], while deletion of TLR4 results in acceleration of diabetes onset [77]. In human monocytes, it has been demonstrated that high glucose induces increased expression of TLR2 and TLR4 via protein kinase C (PKC-α and PKC-δ, respectively) [78].

NLRs signal via the inflammasome, a cytosolic multiprotein complex of the innate immune system that detects pathogens and activates pro-inflammatory response via IL-1β and IL-18. Within the NLR family of proteins, pyrin domain containing 1 (NLRP1) and 3 (NLRP3) have attracted much attention in recent studies as potential targets to modulate inflammation in T1D and T2D. NLRP1 was the first protein shown to form inflammasome [79]. A recent study demonstrated that alleles of the Nlrp1b gene in C57BL/6 mice play an important role in the glucose metabolism when fed with a high fat diet, where the Nlrp1b1 allele downregulates inflammation and attenuates glucose intolerance and insulin resistance in adipose tissue in obese mice [80]. Polymorphisms of NLRP1 were found to be associated with lupus erythematosus and T1D [81,82]. Interestingly, in the blood mononuclear cells and granulocytes of patients with T1D expression of NLRP1 and NLRP3 was markedly decreased, suggesting that NLRP1/NLRP3 inflammasomes signaling pathways may be a protective factor in the early stage of T1D [83]. In a recent study the functional specificity of the NLRP1 inflammasome for the activation of IL-18 in adipose tissue of mice on a high fat diet was demonstrated [84], which opens novel avenues for the targeting therapy.

Activation of NLRP3 is promoted by TLRs activation, which senses dangerous signals such as lipopolysaccharides and high glucose levels, thereby triggering NFκB pathway and production of IL-1β, IL-18 and IL-33 proinflammatory cytokines [85,86]. Another study also demonstrated that high glucose promotes expression of NLRP3, IL-1β, IL-18 in rat glomerular mesangial cells [87]. A decline in renal function is associated with accumulation of inflammatory cells in the kidney, which suggests the NLRP3 inflammasome as a potential therapeutic target in patients with CKD [88]. In glomerular mesangial cells of the STZ-treated rats, high glucose and lipopolysaccharides activate reactive oxygen species (ROS) and the NLRP3 inflammasome, contributing to the development of DKD [89]. In addition, in rats with STZ-induced DKD renal NLRP3 inflammasome activation is shown to be associated with lipid accumulation (based on the Oil Red O staining), which was ameliorated using quercertin (100 mg/kg) and allopurinol (10 mg/kg) [90]. In T2D patients with DKD (n=45), increased mRNA expression of NLRP3 and caspase-1 compared to T2D patients without DKD (n=45) was also reported, while mRNA expression of caspase-1 only was significantly associated with decreased estimated glomerular filtration rate, representing the severity of DKD [91]. Thus, the NLRP3-IL-1β pathway seems to be activated by food intake, obesity, and insulin resistance, while the NLRP1-IL-18 pathway reduces energy intake, lipid accumulation and improves insulin sensitivity.

The recently discovered cGAS-STING pathway was originally described as a signaling cascade that can be activated by double-stranded DNA during a pathogen infection. Studies suggest that host DNA damage induces cytokines through the cGAS-STING signaling pathway thus contributing to increased sterile inflammation. More evidences appeared connecting the cGAS-STING signaling pathway with a state of chronic inflammation in kidneys. A recent study demonstrated that STING activation in tubular epithelial cells contributes to renal inflammation and fibrosis and that pharmacological inhibition of STING has a beneficial effect in a mouse model of chronic kidney disease [92], thus providing further evidence that connects the cGAS-STING signaling pathway with a state of chronic inflammation in kidneys. However, less is known about the role of the cGAS-STING pathway in DKD. A recent study demonstrated that in eNOSdb/db mice and T2D rats the cGAS-STING pathway contributes to the development of DKD particularly in [93]. Thus, targeting of the cGAS-STING signaling pathway may present another novel treatment approach in DKD. However, further investigations are needed.

Overall, there is an increasing number of studies linking Metaflammation to diabetic complications, including DKD (Figure 1). While it is important to understand the roles of innate immunity and adaptive immunity in obesity/diabetes, which could reveal novel immunotherapeutic approaches to modulate metabolic inflammation and insulin resistance, the contribution of lipid metabolism to inflammation remains largely unexplored.

Role of Metaflammation and Lipid/Sphingolipid Dysmetabolism in Kidney Injury

Basal fat cell lipolysis, a process of triacylglycerol breakdown into fatty acids and glycerol, is elevated in obesity conditions and is closely associated with insulin resistance. Both circulating lipids and lipid tissue deposition have been shown to contribute to cellular dysfunction and injury. Lipotoxicity is a process of lipid accumulation in non-adipose tissue such as kidney, liver, heart and muscles. Lipid droplets, which are specialized cellular organelles that play a dynamic role in energy homeostasis, were shown to accumulate in islets as well as in podocytes in diabetes and DKD [9496]. While accumulation of lipid droplets was initially considered a defense mechanism, it is now well recognized that intracellular lipid deposition may play an essential causative role in the progression of diabetic complications.

Both fatty acid and cholesterol metabolism have been shown to contribute to lipotoxic damage in metabolic tissues in patients with diabetes. High glucose has been shown to exacerbate fatty acid uptake and deposition by increasing the class B scavenger receptor CD36 expression via AKT-PPARγ signaling pathway. Plasma CD36 is associated with microparticles in patients with T2D and suggested to be a biomarker of T2D [97]. Interestingly, fatty acids, which are often increased in conditions of insulin resistance, stimulate the production of pro-inflammatory cytokines such as IL-1β directly via activation of TLR2 and TLR4 or indirectly via ceramide production [98]. Another study demonstrated an important role of fatty acid synthase (FAS) in membrane lipid composition and inflammation in diabetes [99], where deletion of Fasn in macrophages of HFD-fed mice prevented insulin resistance, recruitment of macrophages into adipose tissue and chronic inflammation by impairing the retention of plasma membrane cholesterol and disturbing Rho GTPase trafficking. However, lipid deposition may also be glucose-independent and driven by altered cholesterol metabolism rather than altered fatty acid metabolism. In fact, we have demonstrated that human podocytes exposed to the sera of patients with DKD accumulate lipid droplets when compared to the euglycemic sera of patients with diabetes without DKD [100]. Studies investigating the role of ABCA1, a protein involved in cholesterol and phospholipid efflux to apolipoproteins, demonstrated reduced expression of ABCA1 in podocytes treated with sera obtained from patients with both T1D and T2D [7,95] as well as in the kidney cortexes of db/db and ob/ob DKD mouse models. While reduced ABCA1 expression is not sufficient to cause DKD, genetic or pharmacological ABCA1 overexpression were sufficient to completely rescue mice from DKD or DKD-like glomerulosclerosis [95], suggesting that ABCA1 may be a potential therapeutic target. Moreover, glomerular ABCA1 expression in early disease stages inversely correlates with markers of DKD progression both clinically and experimentally [7], suggesting that ABCA1 may be a susceptibility factor contributing to podocyte injury in DKD.

A tight connection between metabolic and inflammatory pathways exists. Persistent inflammation or the failure of tissue to return to the normal physiological state favors tissue destruction and resolution of inflammation is a critical step in the process of regaining tissue homeostasis. Over recent years, pro-resolution lipid mediators were brought into focus in the pathogenesis of diabetes.

Prostaglandins are important lipid mediators of arachidonic acid produced via the sequential catalyzation of cyclooxygenases (COXs) and downstream prostaglandin synthases. One of the best studied lipid mediators is prostaglandin E2 (PGE2), which has been shown to be associated with hypertension and diabetes [101,102]. Urinary PGE2 production is elevated in parallel with activation of COX2 in mice with STZ-induced diabetes [103] and B6-Ins(Akita) mice [104], while using of PGE2 receptor inhibitors [105] and COX2 inhibition [106] may protect against DKD progression. Deletion of PTGER1, a G-protein coupled receptor of PGE2 action, in diabetic OVE26 mice reduced diabetes-induced expression of fibrotic markers, prevented downregulation of nephrin mRNA expression down-regulation and ameliorated glomerular basement membrane thickening and foot processes effacement [107]. Interestingly, in humans with uncomplicated T1D, inhibition of COX2 using 200 mg celecoxib resulted in a significant decrease of glomerular filtration rate (GFR) in the hyperfiltration group and increased in GFR in the normofiltration group [108].

Leukotrienes are a family of eicosanoid derived from arachidonic acid and the essential fatty acid eicosapentaenoic acid by the enzyme arachidonate-5-lipoxygenase (5-LO). Increased levels of leukotrienes synthesis was also reported in diabetic retinopathy (reviewed in Ref. [109]). Like PGE2, urinary excretion levels of leukotriene E4 (LTE4) was increased in T1D patients [110], suggesting a distinguished role of LTE4 in renal inflammation. In addition, in patients with T2D (n=30) lower urinary LTE4 levels were associated with decreased renal function and correlated with serum creatinine and estimated glomerular filtration rate [111].

A Role for eicosanoids, including protectins, resolvins and lipoxins was reported in T1D and T2D. Many studies report low concentrations of eicosanoids in diabetes milieus [112117], suggesting affected resolution of inflammation. However, not too many data exist on the role of eicosanoids in DKD. Thus, in human renal mesangial cells in DKD, LXA4 is a potent modulator of matrix accumulation, where it affects the expression of many genes, including transforming growth factor beta 1, fibronectin, matrix metalloproteinase 1, and several collagens [118]. The role of prostaglandins, leukotrienes and eicosanoids in diabetes and its complications is also summarized in Figure 2.

Figure 2. Prostaglandins, leukotrienes and eicosanoids in the pathogenesis of diabetes and its complications.

Figure 2.

Hyperglycemia and release of cytokines lead to the malfunction in the polyunsaturated fatty acids metabolism. In turn it causes overexpression of prostaglandins (mainly PGE2) and leukotrienes (mainly LTB4 and LTE4) and inhibition of eicosanoids (mainly RvE1, LXA4, LXB4 and RvD1), resulting in inability to inhibit inflammation and promotion of diabetic complications.

The image is created using BioRender software (biorender.com).

Abbreviations: ω3-PUFA – omega 3 polyunsaturated fatty acids; ω6-PUFA – omega 6 polyunsaturated fatty acids; EPA – eicosapentaenoic acid; DHA – docosahexaenoic acid; COX2 – cyclooxygenase 2; HPETE – hydroperoxyeicosatetraenoic acid; HPDHA – hydroperoxydocahexaenoic acid; LOX – lipoxygenase; HDHA – hydroxy-docosahexaenoic acid; RvD1 – resolvin D 1; RvE1, RvE2, RvE3 – resolvin E 1, 2 and 3, respectively; LXA4 – lipoxin A4; LXB4 – lipoxin B4; AA – arachidonic acid; PGG2 – prostaglandin G2; PGH2 – prostaglandin H2; PGE2 – prostaglandin E2; mPGES-1 – microsomal prostaglandin E synthase-1; 5-LO – 5-lipoxygenase; LTA4 – leukotriene A4; LTB4 – leukotriene B4; LTC4 – leukotriene C4; LTD4 – leukotriene D4; LTE4 – leukotriene E4; MyD88 – innate immune signal transduction adaptor.

Involvement of the sphingolipid metabolic pathway in renal injury in diabetes [119] and in inflammation [120] has been also reported. Major bioactive sphingolipids include ceramide, sphingosine, sphingosine-1-phosphate (S1P), and ceramide-1-phosphate (C1P). A role of ceramides has been demonstrated in inflammation of adipose tissue as well, suggesting that regulation of C16-ceramide synthesis may be beneficial in the treatment and even prevention of diabetes [10]. Elevation of muscle C20- and C22-ceramide was shown in patients with general or abdominal obesity in association with muscle insulin resistance, suggesting that tissue ceramide levels may predict insulin resistance [121].

S1P is a member of the sphingolipid family, which is involved in lymphocyte development, B- and T-cell recirculation and mediations of the effects of numerous of biological stimuli, including cytokines. S1P is positively associated with obesity. Circulating levels of S1P have been shown to be significantly increased in diabetic db/db mice and obese humans where a positive correlation of human plasma S1P with homeostatic model assessment of insulin resistance (HOMA-IR), hemoglobin A1C, low density lipoproteins and total cholesterol was detected [122]. A cross-sectional study demonstrated a significant association between serum S1P and TNF in healthy overweight adolescents [123], suggesting increased levels of S1P as a risk factor for metabolic syndrome. Additionally, a previous study demonstrated elevated levels of S1P in obese diabetic BTBR ob/ob mice [124]. The same study showed that S1P and ceramide, another important centerpiece of sphingolipid metabolism, induced expression of IL-6, TNF, and MCP1. S1P can also induce adipocyte dysfunction by promoting chronic inflammation and blocking of insulin signaling [125]. Finally, studies in MIN6 pancreatic β-cells showed that decreased levels of S1P via treatment with sphingosine kinase inhibitor results in reduced glucose-stimulated insulin secretion [126]. However, the role of S1P in DKD remains largely unknown. Some studies demonstrated that excessive S1P/S1P2 receptor pathway activation in tubular cells in DKD may induce activation of Rho kinase and facilitate renal fibrosis in DKD [127], while the activation of S1P/S1P1 receptor pathway may attenuate early-stage DKD [128].

Sphingomyelin phosphodiesterase acid-like 3b (SMPDL3b), an enzyme localized in lipid raft domains and upregulated in DKD [129], was previously shown to act as a major regulator of TLR4 signaling in macrophages [130]. We demonstrated that overexpression of SMPDL3b in human podocytes results in the activation of multiple inflammatory pathways [9], suggesting an important role of sphingolipids in the immune system homeostasis. In the same study, we showed that in vitro treatment of SMPDL3b overexpression human podocytes with C1P or exogenous administration of C1P in db/db mice results in restored AKT phosphorylation and improved renal outcome [9], suggesting that SMPDL3b excess interferes with ceramide kinase function in podocytes [131].

Concluding Remarks

The existence of ample cumulative data that support a role of inflammation in the pathogenesis of diabetes and its complications such as DKD thus warranting their consideration as therapeutic strategies. Many clinical trials have been initiated to study anti-inflammatory approaches to treat T2D or pre-diabetes. Based on the reviewed literature, we can conclude that abnormalities in lipid metabolism per se are not sufficient to induce renal injury, but that lipotoxicity represents an important component of the disease mechanism that may involve multiple hits. Patterns of cytokine production differ due to various inflammatory conditions in patients with diabetes, making a personalized approach urgently important when planning a treatment for a patient. Many of the inflammatory and lipid mediators are measurable in blood and urine and may therefore serve as candidate DKD biomarkers, however, therapeutic developments specifically targeting these mediators for the treatment of patients with DKD still need to be established. Moreover, the crosstalk between lipids and the immune system is still elusive. Further studies are needed to investigate whether anti-inflammatory treatment alone or together with drugs preventing peripheral lipid dysmetabolism can prevent the progression of diabetes and its complications.

Highlights.

  • Lipotoxicity and glucotoxicity are linked to the activation of pro-inflammatory cytokines in diabetic kidney disease.

  • The innate immune system, including the cGAS-STING signaling pathway, is highly involved in the pathogenesis of diabetic complications.

  • Impaired cholesterol efflux (primarily via ATP-binding cassette transporter 1) and an imbalance in biologically active sphingolipids (such as ceramide-1-phosphate and sphingosine-1-phosphate) contribute to the development of diabetic kidney disease.

Acknowledgements

Research in Dr. Alessia Fornoni’s laboratory is supported by the National Institute of Health [grant numbers R01DK117599, R01DK104753, R01CA227493, U54DK083912, UM1DK100846, U01DK116101] and by the Miami Clinical Translational Science Institute [grant number UL1TR000460]. We give a special thanks to the Katz family for continuous support.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflict of Interest

AF and SM are an investors on pending or issued patents (US 10,183,038 and US 10,052,345) aimed at diagnosing or treating proteinuric kidney diseases. They stand to gain royalties from the future commercialization of these patents. A.F. is Vice-President of L&F Health LLC and is consultant for ZyVersa Therapeutics, Inc. ZyVersa Therapeutics, Inc has licensed worldwide rights to develop and commercialize hydroxypropyl-beta-cyclodextrin from L&F Research for the treatment of kidney disease. A.F. is founder of LipoNexT LLC. S.M. is a consultant for Kintai Therapeutics, Inc and holds equity interest in L&F Research. AF and SM are supported by Hoffman-La Roche and by Boehringer Ingelheim.

AM and AMF declare that they have no conflict of interest.

References

Papers of particular interest, published recently, have been highlighted as:

* Of special interest

** Of outstanding interest

  • 1.Proctor MJ, McMillan DC, Horgan PG, Fletcher CD, Talwar D, Morrison DS: Systemic inflammation predicts all-cause mortality: a glasgow inflammation outcome study. PLoS One 2015, 10:e0116206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Larsen CM, Faulenbach M, Vaag A, Volund A, Ehses JA, Seifert B, Mandrup-Poulsen T, Donath MY: Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N Engl J Med 2007, 356:1517–1526. [DOI] [PubMed] [Google Scholar]
  • 3.Akash MSH, Rehman K, Liaqat A: Tumor Necrosis Factor-Alpha: Role in Development of Insulin Resistance and Pathogenesis of Type 2 Diabetes Mellitus. Journal of Cellular Biochemistry 2018, 119:105–110. [DOI] [PubMed] [Google Scholar]
  • 4.Chawla A, Nguyen KD, Goh YP: Macrophage-mediated inflammation in metabolic disease. Nat Rev Immunol 2011, 11:738–749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Al-Lamki RS, Mayadas TN: TNF receptors: signaling pathways and contribution to renal dysfunction. Kidney Int 2015, 87:281–296. [DOI] [PubMed] [Google Scholar]
  • 6. **.Niewczas MA, Pavkov ME, Skupien J, Smiles A, Md Dom ZI, Wilson JM, Park J, Nair V, Schlafly A, Saulnier PJ, et al. : A signature of circulating inflammatory proteins and development of end-stage renal disease in diabetes. Nat Med 2019, 25:805–813. Identified kidney risk inflammatory signature (KRIS) proteins in T1D and T2D Joslin Cohorts and T2D Pima Indian Cohort, where 17 proteins enriched in tumor necrosis factor-receptor superfamily members are associated with ESRD progression.
  • 7.Pedigo CE, Ducasa GM, Leclercq F, Sloan A, Mitrofanova A, Hashmi T, Molina-David J, Ge M, Lassenius MI, Forsblom C, et al. : Local TNF causes NFATc1-dependent cholesterol-mediated podocyte injury. J Clin Invest 2016, 126:3336–3350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. **.Lovric S, Goncalves S, Gee HY, Oskouian B, Srinivas H, Choi WI, Shril S, Ashraf S, Tan W, Rao J, et al. : Mutations in sphingosine-1-phosphate lyase cause nephrosis with ichthyosis and adrenal insufficiency. J Clin Invest 2017, 127:912–928. Identification of different mutations in SGPL1, which encodes sphingosine-1-phosphate lyase, are associated with nephrotic syndrome. This is the first report linking abnormalities in sphingolipid metabolism and kidney inury.
  • 9. *.Mitrofanova A, Mallela SK, Ducasa GM, Yoo TH, Rosenfeld-Gur E, Zelnik ID, Molina J, Varona Santos J, Ge M, Sloan A, et al. : SMPDL3b modulates insulin receptor signaling in diabetic kidney disease. Nat Commun 2019, 10:2692. This study demonstrated that SMPDL3b, an enzyme of sphingolipid pathway with poorly understood function, plays a pivotal role in regulation of C1P availability in diabetic kidneys and modulates insulin signaling. Regulation of SMPDL3b expresion may represent a therapeutic target in DKD tereatment.
  • 10. **.Hammad SM, Twal WO, Arif E, Semler AJ, Klein RL, Nihalani D: Transcriptomics Reveal Altered Metabolic and Signaling Pathways in Podocytes Exposed to C16 Ceramide-Enriched Lipoproteins. Genes (Basel) 2020, 11. Using cultured human podocyte exposed to lipoproteins (HDL2 and HDL3) and comprehensive transcriptomic analysis, authors uncovered that C16 ceramide-enriched HDL2 downregulates phosphorylatin of mTOR intermediates, while C16 ceramide-enriched HDL3 upregulates phosphorylation of mTOR intermediates. This report highlights an important role for lipoprotein-associated sphingolipids in regulating metabolic pathways.
  • 11.Muller S, Martin S, Koenig W, Hanifi-Moghaddam P, Rathmann W, Haastert B, Giani G, Illig T, Thorand B, Kolb H: Impaired glucose tolerance is associated with increased serum concentrations of interleukin 6 and co-regulated acute-phase proteins but not TNF-alpha or its receptors. Diabetologia 2002, 45:805–812. [DOI] [PubMed] [Google Scholar]
  • 12.Pitsavos C, Tampourlou M, Panagiotakos DB, Skoumas Y, Chrysohoou C, Nomikos T, Stefanadis C: Association Between Low-Grade Systemic Inflammation and Type 2 Diabetes Mellitus Among Men and Women from the ATTICA Study. Rev Diabet Stud 2007, 4:98–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Pradhan AD, Manson JE, Rifai N, Buring JE, Ridker PM: C-reactive protein, interleukin 6, and risk of developing type 2 diabetes mellitus. Jama 2001, 286:327–334. [DOI] [PubMed] [Google Scholar]
  • 14.Hu FB, Meigs JB, Li TY, Rifai N, Manson JE: Inflammatory markers and risk of developing type 2 diabetes in women. Diabetes 2004, 53:693–700. [DOI] [PubMed] [Google Scholar]
  • 15.Freeman DJ, Norrie J, Caslake MJ, Gaw A, Ford I, Lowe GD, O’Reilly DS, Packard CJ, Sattar N: C-reactive protein is an independent predictor of risk for the development of diabetes in the West of Scotland Coronary Prevention Study. Diabetes 2002, 51:1596–1600. [DOI] [PubMed] [Google Scholar]
  • 16.Laaksonen DE, Niskanen L, Nyyssonen K, Punnonen K, Tuomainen TP, Valkonen VP, Salonen R, Salonen JT: C-reactive protein and the development of the metabolic syndrome and diabetes in middle-aged men. Diabetologia 2004, 47:1403–1410. [DOI] [PubMed] [Google Scholar]
  • 17.Lee CC, Adler AI, Sandhu MS, Sharp SJ, Forouhi NG, Erqou S, Luben R, Bingham S, Khaw KT, Wareham NJ: Association of C-reactive protein with type 2 diabetes: prospective analysis and meta-analysis. Diabetologia 2009, 52:1040–1047. [DOI] [PubMed] [Google Scholar]
  • 18.Hasegawa G, Nakano K, Sawada M, Uno K, Shibayama Y, Ienaga K, Kondo M: Possible role of tumor necrosis factor and interleukin-1 in the development of diabetic nephropathy. Kidney Int 1991, 40:1007–1012. [DOI] [PubMed] [Google Scholar]
  • 19.Li CR, Mueller EE, Bradley LM: Islet antigen-specific Th17 cells can induce TNF-alpha-dependent autoimmune diabetes. J Immunol 2014, 192:1425–1432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Kuriya G, Uchida T, Akazawa S, Kobayashi M, Nakamura K, Satoh T, Horie I, Kawasaki E, Yamasaki H, Yu L, et al. : Double deficiency in IL-17 and IFN-gamma signalling significantly suppresses the development of diabetes in the NOD mouse. Diabetologia 2013, 56:1773–1780. [DOI] [PubMed] [Google Scholar]
  • 21.Ablamunits V, Henegariu O, Hansen JB, Opare-Addo L, Preston-Hurlburt P, Santamaria P, Mandrup-Poulsen T, Herold KC: Synergistic Reversal of Type 1 Diabetes in NOD Mice With Anti-CD3 and Interleukin-1 Blockade. Evidence of Improved Immune Regulation 2012, 61:145–154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. *.Milas O, Gadalean F, Vlad A, Dumitrascu V, Velciov S, Gluhovschi C, Bob F, Popescu R, Ursoniu S, Jianu DC, et al. : Pro-inflammatory cytokines are associated with podocyte damage and proximal tubular dysfunction in the early stage of diabetic kidney disease in type 2 diabetes mellitus patients. J Diabetes Complications 2019:107479. An association between podocyte dysfunction and pro-inflammatory cytokines has been demosntrated, which may be used an early diagnostic parameter to predict DKD development in patients with T2D, before albuminuria onset.
  • 23.Ballak DB, Li S, Cavalli G, Stahl JL, Tengesdal IW, van Diepen JA, Kluck V, Swartzwelter B, Azam T, Tack CJ, et al. : Interleukin-37 treatment of mice with metabolic syndrome improves insulin sensitivity and reduces pro-inflammatory cytokine production in adipose tissue. J Biol Chem 2018, 293:14224–14236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Kochetova OV, Avzaletdinova DS: Chemokine gene polymorphisms association with increased risk of type 2 diabetes mellitus in Tatar ethnic group, Russia. 2019, 46:887–896. [DOI] [PubMed] [Google Scholar]
  • 25.Sindhu S, Akhter N, Arefanian H, Al-Roub AA, Ali S, Wilson A, Al-Hubail A, Al-Beloushi S, Al-Zanki S, Ahmad R: Increased circulatory levels of fractalkine (CX3CL1) are associated with inflammatory chemokines and cytokines in individuals with type-2 diabetes. J Diabetes Metab Disord 2017, 16:15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Lee YH, Kim KP, Park SH, Kim DJ, Kim YG, Moon JY, Jung SW, Kim JS, Jeong KH, Lee SY, et al. : Urinary chemokine C-X-C motif ligand 16 and endostatin as predictors of tubulointerstitial fibrosis in patients with advanced diabetic kidney disease. Nephrol Dial Transplant 2019. [DOI] [PubMed] [Google Scholar]
  • 27.Bláha V, Andrýs C, Smahelová A, Knízek J, Hyspler R, Solichová D, Bláha M, Zadák Z: Effect of atorvastatin on soluble CD14, CD40 Ligand, sE- and sP-selectins and MCP-1 in patients with type 2 diabetes mellitus: relationship to cholesterol turnover. Pharmacol Res 2006, 54:421–428. [DOI] [PubMed] [Google Scholar]
  • 28.Piemonti L, Calori G, Lattuada G, Mercalli A, Ragogna F, Garancini MP, Ruotolo G, Luzi L, Perseghin G: Association between plasma monocyte chemoattractant protein-1 concentration and cardiovascular disease mortality in middle-aged diabetic and nondiabetic individuals. Diabetes Care 2009, 32:2105–2110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Radhakrishnan P, Srikanth P, Seshadri KG, Barani R, Samanta M: Serum monocyte chemoattractant protein-1 is a biomarker in patients with diabetes and periodontitis. Indian J Endocrinol Metab 2014, 18:505–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Karadeniz M, Erdogan M, Cetinkalp S, Berdeli A, Eroglu Z, Ozgen AG: Monocyte chemoattractant protein-1 (MCP-1) 2518G/A gene polymorphism in Turkish type 2 diabetes patients with nephropathy. Endocrine 2010, 37:513–517. [DOI] [PubMed] [Google Scholar]
  • 31.Jing Y, Zhu D, Bi Y, Yang D, Hu Y, Shen S: Monocyte chemoattractant protein 1–2518 A/G polymorphism and susceptibility to type 2 diabetes in a Chinese population. Clin Chim Acta 2011, 412:466–469. [DOI] [PubMed] [Google Scholar]
  • 32.Kouyama K, Miyake K, Zenibayashi M, Hirota Y, Teranishi T, Tamori Y, Kanda H, Sakaguchi K, Ohara T, Kasuga M: Association of serum MCP-1 concentration and MCP-1 polymorphism with insulin resistance in Japanese individuals with obese type 2 diabetes. Kobe J Med Sci 2008, 53:345–354. [PubMed] [Google Scholar]
  • 33.Liu ZH, Chen LL, Deng XL, Song HJ, Liao YF, Zeng TS, Zheng J, Li HQ: Methylation status of CpG sites in the MCP-1 promoter is correlated to serum MCP-1 in Type 2 diabetes. Journal of Endocrinological Investigation 2012, 35:585–589. [DOI] [PubMed] [Google Scholar]
  • 34.Wada T, Furuichi K, Sakai N, Iwata Y, Kitagawa K, Ishida Y, Kondo T, Hashimoto H, Ishiwata Y, Mukaida N, et al. : Gene therapy via blockade of monocyte chemoattractant protein-1 for renal fibrosis. J Am Soc Nephrol 2004, 15:940–948. [DOI] [PubMed] [Google Scholar]
  • 35.Lin J, Glynn RJ, Rifai N, Manson JE, Ridker PM, Nathan DM, Schaumberg DA: Inflammation and progressive nephropathy in type 1 diabetes in the diabetes control and complications trial. Diabetes Care 2008, 31:2338–2343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Karimi Z, Kahe F, Jamil A, Marszalek J, Ghanbari A, Afarideh M, Khajeh E, Noshad S, Esteghamati A, Chi G: Intercellular adhesion molecule-1 in diabetic patients with and without microalbuminuria. Diabetes Metab Syndr 2018, 12:365–368. [DOI] [PubMed] [Google Scholar]
  • 37.Hegazy GA, Awan Z, Hashem E, Al-Ama N, Abunaji AB: Levels of soluble cell adhesion molecules in type 2 diabetes mellitus patients with macrovascular complications. Journal of International Medical Research 2019:0300060519893858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Boulbou MS, Koukoulis GN, Makri ED, Petinaki EA, Gourgoulianis KI, Germenis AE: Circulating adhesion molecules levels in type 2 diabetes mellitus and hypertension. Int J Cardiol 2005, 98:39–44. [DOI] [PubMed] [Google Scholar]
  • 39.Kulkarni H, Mamtani M, Peralta J, Almeida M, Dyer TD, Goring HH, Johnson MP, Duggirala R, Mahaney MC, Olvera RL, et al. : Soluble Forms of Intercellular and Vascular Cell Adhesion Molecules Independently Predict Progression to Type 2 Diabetes in Mexican American Families. PLoS One 2016, 11:e0151177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Chung FM, Tsai JC, Chang DM, Shin SJ, Lee YJ: Peripheral total and differential leukocyte count in diabetic nephropathy: the relationship of plasma leptin to leukocytosis. Diabetes Care 2005, 28:1710–1717. [DOI] [PubMed] [Google Scholar]
  • 41.Cavalot F, Massucco P, Perna P, Traversa M, Anfossi G, Trovati M: White blood cell count is positively correlated with albumin excretion rate in subjects with type 2 diabetes. Diabetes Care 2002, 25:2354–2355. [DOI] [PubMed] [Google Scholar]
  • 42.Hojs R, Ekart R, Bevc S, Hojs N: Markers of Inflammation and Oxidative Stress in the Development and Progression of Renal Disease in Diabetic Patients. Nephron 2016, 133:159–162. [DOI] [PubMed] [Google Scholar]
  • 43.Nawaz SS, Joy SS, Al Farsi Y, George TP, Siddiqui K: Potential role of serum fetuin-A in relation with pro-inflammatory, chemokine and adhesion molecules in diabetic kidney disease: a case-control study. 2019, 46:1239–1246. [DOI] [PubMed] [Google Scholar]
  • 44.Liu L, He D, Fang L, Yan X: Association between E469K polymorphism in the ICAM1 gene and the risk of diabetic nephropathy: a meta-analysis. Lipids Health Dis 2018, 17:293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Mueller PW, Rogus JJ, Cleary PA, Zhao Y, Smiles AM, Steffes MW, Bucksa J, Gibson TB, Cordovado SK, Krolewski AS, et al. : Genetics of Kidneys in Diabetes (GoKinD) study: a genetics collection available for identifying genetic susceptibility factors for diabetic nephropathy in type 1 diabetes. J Am Soc Nephrol 2006, 17:1782–1790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Avci E, Uzeli S: The role of adhesion molecules and cytokines in patients with diabetic nephropathy. Biomedical Research (India) 2016, 2016:S343–S348. [Google Scholar]
  • 47.Wan RJ, Li YH: MicroRNA146a/NAPDH oxidase4 decreases reactive oxygen species generation and inflammation in a diabetic nephropathy model. Mol Med Rep 2018, 17:4759–4766. [DOI] [PubMed] [Google Scholar]
  • 48.Okada S, Shikata K, Matsuda M, Ogawa D, Usui H, Kido Y, Nagase R, Wada J, Shikata Y, Makino H: Intercellular adhesion molecule-1-deficient mice are resistant against renal injury after induction of diabetes. Diabetes 2003, 52:2586–2593. [DOI] [PubMed] [Google Scholar]
  • 49.Chow FY, Nikolic-Paterson DJ, Ozols E, Atkins RC, Tesch GH: Intercellular adhesion molecule-1 deficiency is protective against nephropathy in type 2 diabetic db/db mice. J Am Soc Nephrol 2005, 16:1711–1722. [DOI] [PubMed] [Google Scholar]
  • 50.Gohda T, Niewczas MA, Ficociello LH, Walker WH, Skupien J, Rosetti F, Cullere X, Johnson AC, Crabtree G, Smiles AM, et al. : Circulating TNF receptors 1 and 2 predict stage 3 CKD in type 1 diabetes. J Am Soc Nephrol 2012, 23:516–524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. *.Lopes-Virella MF, Baker NL, Hunt KJ, Cleary PA, Klein R, Virella G: Baseline markers of inflammation are associated with progression to macroalbuminuria in type 1 diabetic subjects. Diabetes Care 2013, 36:2317–2323. This study explores the association of TNF receptors 1 and 2 with progression to macroalbuminuria in the Diabetes Control and Complications Trial (DCCT)/Epidemiology of Diabetes Interventions and Complications cohort.
  • 52.Lee CP, Nithiyanantham S, Hsu HT, Yeh KT, Kuo TM, Ko YC: ALPK1 regulates streptozotocin-induced nephropathy through CCL2 and CCL5 expressions. J Cell Mol Med 2019, 23:7699–7708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Nadkarni GN, Rao V, Ismail-Beigi F, Fonseca VA, Shah SV, Simonson MS, Cantley L, Devarajan P, Parikh CR, Coca SG: Association of Urinary Biomarkers of Inflammation, Injury, and Fibrosis with Renal Function Decline: The ACCORD Trial. Clin J Am Soc Nephrol 2016, 11:1343–1352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Satirapoj B, Dispan R, Radinahamed P, Kitiyakara C: Urinary epidermal growth factor, monocyte chemoattractant protein-1 or their ratio as predictors for rapid loss of renal function in type 2 diabetic patients with diabetic kidney disease. 2018, 19:246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Yahya MJ, Ismail PB, Nordin NB, Akim ABM, Yusuf W, Adam NLB, Yusoff MJ: Association of CCL2, CCR5, ELMO1, and IL8 Polymorphism with Diabetic Nephropathy in Malaysian Type 2 Diabetic Patients. Int J Chronic Dis 2019, 2019:2053015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Su N, Zhao N, Wang G, Wang L, Zhang Y, Li R, Liu Y, Yang X, Li C, Hou M: Association of MCP-1 rs1024611 polymorphism with diabetic foot ulcers. Medicine (Baltimore) 2018, 97:e11232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Ahluwalia TS, Khullar M, Ahuja M, Kohli HS, Bhansali A, Mohan V, Venkatesan R, Rai TS, Sud K, Singal PK: Common variants of inflammatory cytokine genes are associated with risk of nephropathy in type 2 diabetes among Asian Indians. PLoS One 2009, 4:e5168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Moon JY, Jeong L, Lee S, Jeong K, Lee T, Ihm CG, Suh J, Kim J, Jung YY, Chung JH: Association of polymorphisms in monocyte chemoattractant protein-1 promoter with diabetic kidney failure in Korean patients with type 2 diabetes mellitus. J Korean Med Sci 2007, 22:810–814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Wu L, Li XQ, Chang DY, Zhang H, Li JJ, Wu SL, Zhang LX, Chen M, Zhao MH: Associations of urinary epidermal growth factor and monocyte chemotactic protein-1 with kidney involvement in patients with diabetic kidney disease. Nephrol Dial Transplant 2020, 35:291–297. [DOI] [PubMed] [Google Scholar]
  • 60.Cha DR, Kang YS, Han SY, Jee YH, Han KH, Kim HK, Han JY, Kim YS: Role of aldosterone in diabetic nephropathy. Nephrology (Carlton) 2005, 10 Suppl:S37–39. [DOI] [PubMed] [Google Scholar]
  • 61.Meng X-M, Tang PM-K, Li J, Lan HY: TGF-β/Smad signaling in renal fibrosis. 2015, 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Aroune D, Libdiri F, Leboucher S, Maouche B, Marco S, El-Aoufi S: Changes in the NFkappaB and E-cadherin expression are associated to diabetic nephropathy inPsammomys obesus. Saudi J Biol Sci 2017, 24:843–850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Wu C, Lv C, Chen F, Ma X, Shao Y, Wang Q: The function of miR-199a-5p/Klotho regulating TLR4/NF-kappaB p65/NGAL pathways in rat mesangial cells cultured with high glucose and the mechanism. Mol Cell Endocrinol 2015, 417:84–93. [DOI] [PubMed] [Google Scholar]
  • 64.Ruiz S, Pergola PE, Zager RA, Vaziri ND: Targeting the transcription factor Nrf2 to ameliorate oxidative stress and inflammation in chronic kidney disease. Kidney Int 2013, 83:1029–1041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Chin MP, Bakris GL, Block GA, Chertow GM, Goldsberry A, Inker LA, Heerspink HJL, O’Grady M, Pergola PE, Wanner C, et al. : Bardoxolone Methyl Improves Kidney Function in Patients with Chronic Kidney Disease Stage 4 and Type 2 Diabetes: Post-Hoc Analyses from Bardoxolone Methyl Evaluation in Patients with Chronic Kidney Disease and Type 2 Diabetes Study. Am J Nephrol 2018, 47:40–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Chertow GM, Pergola PE, Chen F, Kirby BJ, Sundy JS, Patel UD: Effects of Selonsertib in Patients with Diabetic Kidney Disease. Journal of the American Society of Nephrology 2019, 30:1980–1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Han KH, Han SY, Kim HS, Kang YS, Cha DR: Prolonged administration enhances the renoprotective effect of pentoxifylline via anti-inflammatory activity in streptozotocin-induced diabetic nephropathy. Inflammation 2010, 33:137–143. [DOI] [PubMed] [Google Scholar]
  • 68.An Z-m, Dong X-g, Guo Y, Zhou J-l, Qin T: Effects and clinical significance of pentoxifylline on the oxidative stress of rats with diabetic nephropathy. Journal of Huazhong University of Science and Technology [Medical Sciences] 2015, 35:356–361. [DOI] [PubMed] [Google Scholar]
  • 69.Garcia FAdO, Rebouças JF, Balbino TQ, da Silva TG, de Carvalho-Júnior CHR, Cerqueira GS, Brito GAdC, Viana GSdB: Pentoxifylline reduces the inflammatory process in diabetic rats: relationship with decreases of pro-inflammatory cytokines and inducible nitric oxide synthase. Journal of Inflammation 2015, 12:33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Navarro-González JF, Mora-Fernández C, Muros de Fuentes M, Chahin J, Méndez ML, Gallego E, Macía M, del Castillo N, Rivero A, Getino MA, et al. : Effect of Pentoxifylline on Renal Function and Urinary Albumin Excretion in Patients with Diabetic Kidney Disease: The PREDIAN Trial. Journal of the American Society of Nephrology 2015, 26:220–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Mohammad MK, Morran M, Slotterbeck B, Leaman DW, Sun Y, Grafenstein H, Hong SC, McInerney MF: Dysregulated Toll-like receptor expression and signaling in bone marrow-derived macrophages at the onset of diabetes in the non-obese diabetic mouse. Int Immunol 2006, 18:1101–1113. [DOI] [PubMed] [Google Scholar]
  • 72.Devaraj S, Dasu MR, Rockwood J, Winter W, Griffen SC, Jialal I: Increased toll-like receptor (TLR) 2 and TLR4 expression in monocytes from patients with type 1 diabetes: further evidence of a proinflammatory state. J Clin Endocrinol Metab 2008, 93:578–583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Lee AS, Ghoreishi M, Cheng WK, Chang TY, Zhang YQ, Dutz JP: Toll-like receptor 7 stimulation promotes autoimmune diabetes in the NOD mouse. Diabetologia 2011, 54:1407–1416. [DOI] [PubMed] [Google Scholar]
  • 74.Assmann TS, Brondani Lde A, Boucas AP, Canani LH, Crispim D: Toll-like receptor 3 (TLR3) and the development of type 1 diabetes mellitus. Arch Endocrinol Metab 2015, 59:4–12. [DOI] [PubMed] [Google Scholar]
  • 75.Vallois D, Grimm CH, Avner P, Boitard C, Rogner UC: The type 1 diabetes locus Idd6 controls TLR1 expression. J Immunol 2007, 179:3896–3903. [DOI] [PubMed] [Google Scholar]
  • 76.Dong B, Qi D, Yang L, Huang Y, Xiao X, Tai N, Wen L, Wong FS: TLR4 regulates cardiac lipid accumulation and diabetic heart disease in the nonobese diabetic mouse model of type 1 diabetes. Am J Physiol Heart Circ Physiol 2012, 303:H732–742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Gulden E, Ihira M, Ohashi A, Reinbeck AL, Freudenberg MA, Kolb H, Burkart V: Toll-like receptor 4 deficiency accelerates the development of insulin-deficient diabetes in non-obese diabetic mice. PLoS One 2013, 8:e75385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Dasu MR, Devaraj S, Zhao L, Hwang DH, Jialal I: High Glucose Induces Toll-Like Receptor Expression in Human Monocytes. Mechanism of Activation 2008, 57:3090–3098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Chavarría-Smith J, Mitchell PS, Ho AM, Daugherty MD, Vance RE: Functional and Evolutionary Analyses Identify Proteolysis as a General Mechanism for NLRP1 Inflammasome Activation. PLoS Pathog 2016, 12:e1006052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Salazar-Leon J, Valdez-Hernandez AL, Garcia-Jimenez S, Roman-Dominguez L, Huanosta-Murillo E, Bonifaz LC, Perez-Martinez L, Pedraza-Alva G: Nlrp1b1 negatively modulates obesity-induced inflammation by promoting IL-18 production. 2019, 9:13815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Grandemange S, Sanchez E, Louis-Plence P, Tran Mau-Them F, Bessis D, Coubes C, Frouin E, Seyger M, Girard M, Puechberty J, et al. : A new autoinflammatory and autoimmune syndrome associated with NLRP1 mutations: NAIAD (NLRP1-associated autoinflammation with arthritis and dyskeratosis). Ann Rheum Dis 2017, 76:1191–1198. [DOI] [PubMed] [Google Scholar]
  • 82.Sun X, Xia Y, Liu Y, Wang Y, Luo S, Lin J, Huang G, Li X, Xie Z, Zhou Z: Polymorphisms in NLRP1 Gene Are Associated with Type 1 Diabetes. J Diabetes Res 2019, 2019:7405120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Liu H, Xu R, Kong Q, Liu J, Yu Z, Zhao C: Downregulated NLRP3 and NLRP1 inflammasomes signaling pathways in the development and progression of type 1 diabetes mellitus. Biomed Pharmacother 2017, 94:619–626. [DOI] [PubMed] [Google Scholar]
  • 84.Murphy AJ, Kraakman MJ, Kammoun HL, Dragoljevic D, Lee MK, Lawlor KE, Wentworth JM, Vasanthakumar A, Gerlic M, Whitehead LW, et al. : IL-18 Production from the NLRP1 Inflammasome Prevents Obesity and Metabolic Syndrome. Cell Metab 2016, 23:155–164. [DOI] [PubMed] [Google Scholar]
  • 85.Shahzad K, Bock F, Al-Dabet MM, Gadi I, Nazir S, Wang H, Kohli S, Ranjan S, Mertens PR, Nawroth PP, et al. : Stabilization of endogenous Nrf2 by minocycline protects against Nlrp3-inflammasome induced diabetic nephropathy. Sci Rep 2016, 6:34228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Xiao J, Fu C, Zhang X, Zhu D, Chen W, Lu Y, Ye Z: Soluble monosodium urate, but not its crystal, induces toll like receptor 4-dependent immune activation in renal mesangial cells. Mol Immunol 2015, 66:310–318. [DOI] [PubMed] [Google Scholar]
  • 87.Chen F, Wei G, Xu J, Ma X, Wang Q: Naringin ameliorates the high glucose-induced rat mesangial cell inflammatory reaction by modulating the NLRP3 Inflammasome. BMC Complement Altern Med 2018, 18:192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Samra YA, Said HS, Elsherbiny NM, Liou GI, El-Shishtawy MM, Eissa LA: Cepharanthine and Piperine ameliorate diabetic nephropathy in rats: role of NF-kappaB and NLRP3 inflammasome. Life Sci 2016, 157:187–199. [DOI] [PubMed] [Google Scholar]
  • 89.Feng H, Gu J, Gou F, Huang W, Gao C, Chen G, Long Y, Zhou X, Yang M, Liu S, et al. : High Glucose and Lipopolysaccharide Prime NLRP3 Inflammasome via ROS/TXNIP Pathway in Mesangial Cells. J Diabetes Res 2016, 2016:6973175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Wang C, Pan Y, Zhang QY, Wang FM, Kong LD: Quercetin and allopurinol ameliorate kidney injury in STZ-treated rats with regulation of renal NLRP3 inflammasome activation and lipid accumulation. PLoS One 2012, 7:e38285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Bhattacharjee CK, Paine SK, Mahanta J, Borphukan S, Borah PK: Expression of inflammasome complex mRNA and its targeted microRNA in type 2 diabetes mellitus: A possible predictor of the severity of diabetic nephropathy. J Diabetes 2019, 11:90–92. [DOI] [PubMed] [Google Scholar]
  • 92.Chung KW, Dhillon P, Huang S, Sheng X, Shrestha R, Qiu C, Kaufman BA, Park J, Pei L, Baur J, et al. : Mitochondrial Damage and Activation of the STING Pathway Lead to Renal Inflammation and Fibrosis. Cell Metab 2019, 30:784–799.e785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Khedr S, Dissanayake LV, Palygin O, Staruschenko A: Potential Role of cGAS-STING Pathway in the Induction of Diabetic Kidney Disease. The FASEB Journal 2020, 34:1–1. [Google Scholar]
  • 94.Tong X, Dai C, Walker JT, Nair GG, Kennedy A, Carr RM, Hebrok M, Powers AC, Stein R: Lipid Droplet Accumulation in Human Pancreatic Islets Is Dependent On Both Donor Age and Health. 2020, 69:342–354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. **.Ducasa GM, Mitrofanova A, Mallela SK, Liu X, Molina J, Sloan A, Pedigo CE, Ge M, Santos JV, Hernandez Y, et al. : ATP-binding cassette A1 deficiency causes cardiolipin-driven mitochondrial dysfunction in podocytes. J Clin Invest 2019, 129:3387–3400. Findings from this study demonstrated that decreased expression of ABCA1 plays a pivotal role in alteration in the OXPHOS complexes and is associated with cardiolipid accumulation in DKD.
  • 96.Ducasa GM, Mitrofanova A, Fornoni A: Crosstalk Between Lipids and Mitochondria in Diabetic Kidney Disease. Curr Diab Rep 2019, 19:144. [DOI] [PubMed] [Google Scholar]
  • 97.Alkhatatbeh MJ, Enjeti AK, Acharya S, Thorne RF, Lincz LF: The origin of circulating CD36 in type 2 diabetes. Nutrition & Diabetes 2013, 3:e59–e59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Boni-Schnetzler M, Boller S, Debray S, Bouzakri K, Meier DT, Prazak R, Kerr-Conte J, Pattou F, Ehses JA, Schuit FC, et al. : Free fatty acids induce a proinflammatory response in islets via the abundantly expressed interleukin-1 receptor I. Endocrinology 2009, 150:5218–5229. [DOI] [PubMed] [Google Scholar]
  • 99.Wei X, Song H, Yin L, Rizzo MG, Sidhu R, Covey DF, Ory DS, Semenkovich CF: Fatty acid synthesis configures the plasma membrane for inflammation in diabetes. Nature 2016, 539:294–298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Merscher-Gomez S, Guzman J, Pedigo CE, Lehto M, Aguillon-Prada R, Mendez A, Lassenius MI, Forsblom C, Yoo T, Villarreal R, et al. : Cyclodextrin protects podocytes in diabetic kidney disease. Diabetes 2013, 62:3817–3827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Ben Nasr M, D’Addio F, Malvandi AM, Faravelli S, Castillo-Leon E, Usuelli V, Rocchio F, Letizia T, El Essawy AB, Assi E, et al. : Prostaglandin E2 Stimulates the Expansion of Regulatory Hematopoietic Stem and Progenitor Cells in Type 1 Diabetes. 2018, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Amior L, Srivastava R, Nano R, Bertuzzi F, Melloul D: The role of Cox-2 and prostaglandin E2 receptor EP3 in pancreatic beta-cell death. Faseb j 2019, 33:4975–4986. [DOI] [PubMed] [Google Scholar]
  • 103.Jia Z, Sun Y, Liu S, Liu Y, Yang T: COX-2 but not mPGES-1 contributes to renal PGE2 induction and diabetic proteinuria in mice with type-1 diabetes. PLoS One 2014, 9:e93182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Nasrallah R, Xiong H, Hebert RL: Renal prostaglandin E2 receptor (EP) expression profile is altered in streptozotocin and B6-Ins2Akita type I diabetic mice. Am J Physiol Renal Physiol 2007, 292:F278–284. [DOI] [PubMed] [Google Scholar]
  • 105.Makino H, Tanaka I, Mukoyama M, Sugawara A, Mori K, Muro S, Suganami T, Yahata K, Ishibashi R, Ohuchida S, et al. : Prevention of diabetic nephropathy in rats by prostaglandin E receptor EP1-selective antagonist. J Am Soc Nephrol 2002, 13:1757–1765. [DOI] [PubMed] [Google Scholar]
  • 106.Nasrallah R, Robertson SJ, Hebert RL: Chronic COX inhibition reduces diabetes-induced hyperfiltration, proteinuria, and renal pathological markers in 36-week B6-Ins2(Akita) mice. Am J Nephrol 2009, 30:346–353. [DOI] [PubMed] [Google Scholar]
  • 107.Thibodeau JF, Nasrallah R, Carter A, He Y, Touyz R, Hebert RL, Kennedy CRJ: PTGER1 deletion attenuates renal injury in diabetic mouse models. Am J Pathol 2013, 183:1789–1802. [DOI] [PubMed] [Google Scholar]
  • 108.Cherney DZ, Miller JA, Scholey JW, Bradley TJ, Slorach C, Curtis JR, Dekker MG, Nasrallah R, Hebert RL, Sochett EB: The effect of cyclooxygenase-2 inhibition on renal hemodynamic function in humans with type 1 diabetes. Diabetes 2008, 57:688–695. [DOI] [PubMed] [Google Scholar]
  • 109.Behl T, Kaur I, Kotwani A: Role of leukotrienes in diabetic retinopathy. Prostaglandins Other Lipid Mediat 2016, 122:1–9. [DOI] [PubMed] [Google Scholar]
  • 110.Hardy G, Boizel R, Bessard J, Cracowski JL, Bessard G, Halimi S, Stanke-Labesque F: Urinary leukotriene E4 excretion is increased in type 1 diabetic patients: a quantification by liquid chromatography-tandem mass spectrometry. Prostaglandins Other Lipid Mediat 2005, 78:291–299. [DOI] [PubMed] [Google Scholar]
  • 111.Rafnsson A, Bäck M: Urinary leukotriene E4 is associated with renal function but not with endothelial function in type 2 diabetes. Dis Markers 2013, 35:475–480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Hellmann J, Tang Y, Kosuri M, Bhatnagar A, Spite M: Resolvin D1 decreases adipose tissue macrophage accumulation and improves insulin sensitivity in obese-diabetic mice. Faseb j 2011, 25:2399–2407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Sima C, Montero E, Nguyen D, Freire M, Norris P, Serhan CN, Van Dyke TE: ERV1 Overexpression in Myeloid Cells Protects against High Fat Diet Induced Obesity and Glucose Intolerance. Sci Rep 2017, 7:12848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Lopez-Vicario C, Rius B, Alcaraz-Quiles J, Gonzalez-Periz A, Martinez-Puchol AI, Casulleras M, Duran-Guell M, Ibarzabal A, Corcelles R, Laguna-Fernandez A, et al. : Association of a variant in the gene encoding for ERV1/ChemR23 with reduced inflammation in visceral adipose tissue from morbidly obese individuals. Sci Rep 2017, 7:15724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Freire MO, Dalli J, Serhan CN, Van Dyke TE: Neutrophil Resolvin E1 Receptor Expression and Function in Type 2 Diabetes. 2017, 198:718–728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Brennan EP, Cacace A, Godson C: Specialized pro-resolving mediators in renal fibrosis. Mol Aspects Med 2017, 58:102–113. [DOI] [PubMed] [Google Scholar]
  • 117.Brennan EP, Mohan M, McClelland A, de Gaetano M, Tikellis C, Marai M, Crean D, Dai A, Beuscart O, Derouiche S, et al. : Lipoxins Protect Against Inflammation in Diabetes-Associated Atherosclerosis. 2018, 67:2657–2667. [DOI] [PubMed] [Google Scholar]
  • 118.Rodgers K, McMahon B, Mitchell D, Sadlier D, Godson C: Lipoxin A4 modifies platelet-derived growth factor-induced pro-fibrotic gene expression in human renal mesangial cells. Am J Pathol 2005, 167:683–694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Zador IZ, Deshmukh GD, Kunkel R, Johnson K, Radin NS, Shayman JA: A role for glycosphingolipid accumulation in the renal hypertrophy of streptozotocin-induced diabetes mellitus. J Clin Invest 1993, 91:797–803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Maceyka M, Spiegel S: Sphingolipid metabolites in inflammatory disease. Nature 2014, 510:58–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.de la Maza MP, Rodriguez JM, Hirsch S, Leiva L, Barrera G, Bunout D: Skeletal muscle ceramide species in men with abdominal obesity. J Nutr Health Aging 2015, 19:389–396. [DOI] [PubMed] [Google Scholar]
  • 122.Kowalski GM, Carey AL, Selathurai A, Kingwell BA, Bruce CR: Plasma sphingosine-1-phosphate is elevated in obesity. PLoS One 2013, 8:e72449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Majumdar I, Mastrandrea LD: Serum sphingolipids and inflammatory mediators in adolescents at risk for metabolic syndrome. Endocrine 2012, 41:442–449. [DOI] [PubMed] [Google Scholar]
  • 124.Samad F, Hester KD, Yang G, Hannun YA, Bielawski J: Altered adipose and plasma sphingolipid metabolism in obesity: a potential mechanism for cardiovascular and metabolic risk. Diabetes 2006, 55:2579–2587. [DOI] [PubMed] [Google Scholar]
  • 125.Fang Z, Pyne S, Pyne NJ: Ceramide and sphingosine 1-phosphate in adipose dysfunction. Prog Lipid Res 2019, 74:145–159. [DOI] [PubMed] [Google Scholar]
  • 126.Cantrell Stanford J, Morris AJ, Sunkara M, Popa GJ, Larson KL, Ozcan S: Sphingosine 1-phosphate (S1P) regulates glucose-stimulated insulin secretion in pancreatic beta cells. J Biol Chem 2012, 287:13457–13464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Ishizawa S, Takahashi-Fujigasaki J, Kanazawa Y, Matoba K, Kawanami D, Yokota T, Iwamoto T, Tajima N, Manome Y, Utsunomiya K: Sphingosine-1-phosphate induces differentiation of cultured renal tubular epithelial cells under Rho kinase activation via the S1P2 receptor. Clin Exp Nephrol 2014, 18:844–852. [DOI] [PubMed] [Google Scholar]
  • 128.Awad AS, Rouse MD, Khutsishvili K, Huang L, Bolton WK, Lynch KR, Okusa MD: Chronic sphingosine 1-phosphate 1 receptor activation attenuates early-stage diabetic nephropathy independent of lymphocytes. Kidney Int 2011, 79:1090–1098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. *.Yoo TH, Pedigo CE, Guzman J, Correa-Medina M, Wei C, Villarreal R, Mitrofanova A, Leclercq F, Faul C, Li J, et al. : Sphingomyelinase-like phosphodiesterase 3b expression levels determine podocyte injury phenotypes in glomerular disease. J Am Soc Nephrol 2015, 26:133–147. This study is linking SMPDL3b expression levels to podocyte injury phenotype in association with suPAR-dependent ανβ3 integrin activation.
  • 130.Heinz LX, Baumann CL, Koberlin MS, Snijder B, Gawish R, Shui G, Sharif O, Aspalter IM, Muller AC, Kandasamy RK, et al. : The Lipid-Modifying Enzyme SMPDL3B Negatively Regulates Innate Immunity. Cell Rep 2015, 11:1919–1928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Mallela SK, Mitrofanova A, Merscher S, Fornoni A: Regulation of the amount of ceramide-1-phosphate synthesized in differentiated human podocytes. Biochim Biophys Acta Mol Cell Biol Lipids 2019, 1864:158517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Gohda T, Nishizaki Y, Murakoshi M, Nojiri S, Yanagisawa N, Shibata T, Yamashita M, Tanaka K, Yamashita Y, Suzuki Y, et al. : Clinical predictive biomarkers for normoalbuminuric diabetic kidney disease. Diabetes Res Clin Pract 2018, 141:62–68. [DOI] [PubMed] [Google Scholar]
  • 133.Niewczas MA, Gohda T, Skupien J, Smiles AM, Walker WH, Rosetti F, Cullere X, Eckfeldt JH, Doria A, Mayadas TN, et al. : Circulating TNF receptors 1 and 2 predict ESRD in type 2 diabetes. J Am Soc Nephrol 2012, 23:507–515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Pavkov ME, Nelson RG, Knowler WC, Cheng Y, Krolewski AS, Niewczas MA: Elevation of circulating TNF receptors 1 and 2 increases the risk of end-stage renal disease in American Indians with type 2 diabetes. Kidney Int 2015, 87:812–819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Barr ELM, Barzi F, Hughes JT, Jerums G, Hoy WE, O’Dea K, Jones GRD, Lawton PD, Brown ADH, Thomas M, et al. : High Baseline Levels of Tumor Necrosis Factor Receptor 1 Are Associated With Progression of Kidney Disease in Indigenous Australians With Diabetes: The eGFR Follow-up Study. Diabetes Care 2018, 41:739–747. [DOI] [PubMed] [Google Scholar]

RESOURCES