Back to Journals » Diabetes, Metabolic Syndrome and Obesity » Volume 17

Autophagy, Pyroptosis and Ferroptosis are Rising Stars in the Pathogenesis of Diabetic Nephropathy

Authors Li X, Gao L, Li X, Xia J, Pan Y, Bai C

Received 19 November 2023

Accepted for publication 1 March 2024

Published 13 March 2024 Volume 2024:17 Pages 1289—1299

DOI https://doi.org/10.2147/DMSO.S450695

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Dr Konstantinos Tziomalos



Xiudan Li,1,2 Lifeng Gao,2 Xuyang Li,2 Jingdong Xia,1 Yurong Pan,1 Chunying Bai2

1Department of Endocrinology, Affiliated Hospital of Chifeng University, Chifeng, 024000, China; 2Inner Mongolia Key Laboratory of Human Genetic Disease Research, Chifeng University, Chifeng, 024000, China

Correspondence: Chunying Bai, Email [email protected]

Abstract: Diabetic nephropathy (DN) is one of the most common microvascular complications in diabetes and can potentially develop into end-stage renal disease. Its pathogenesis is complex and not fully understood. Podocytes, glomerular endothelial cells (GECs), glomerular mesangial cells (GMCs) and renal tubular epithelial cells (TECs) play important roles in the normal function of glomerulus and renal tubules, and their injury is involved in the progression of DN. Although our understanding of the mechanisms leading to DN has substantially improved, we still need to find more effective therapeutic targets. Autophagy, pyroptosis and ferroptosis are programmed cell death processes that are associated with inflammation and are closely related to a variety of diseases. Recently, a growing number of studies have reported that autophagy, pyroptosis and ferroptosis regulate the function of podocytes, GECs, GMCs and TECs. This review highlights the contributions of autophagy, pyroptosis, and ferroptosis to DN injury in these cells, offering potential therapeutic targets for DN treatment.

Keywords: diabetic nephropathy, autophagy, pyroptosis, ferroptosis, inflammation

Introduction

With the development of society and changes in lifestyle, the prevalence of diabetes has been steadily rising in recent years.1,2 The statistics from the International Diabetes Federation show that in 2021, the number of diabetic patients worldwide reached 536 million.3 The prevalence of diabetic complications is also increasing. As one of the most common microvascular complications of diabetes, the prevalence of diabetic nephropathy (DN) in the late stages of diabetes is almost 50%.4,5 Due to these trends, DN has become a serious public health problem in the world.6

DN is a complex and chronic disease characterized by glomerular hyperfiltration, hyperperfusion, thickening of the glomerular basement membrane, glomerular capillary injury, mesangial expansion, and microalbuminuria.7,8 In the early stages of DN, the volume of the kidney increases and the glomerular filtration rate increases. As the disease progresses to the middle stages, the basement membrane thickens and the mesangial matrix increases. This leads to glomerular nodular and diffuse lesions, arteriolar hyaline degeneration, further thickening of the glomerular basement membrane, and progressive stenosis of the glomerular capillary cavity. In the late stages, glomerular and tubulointerstitial fibrosis occurs, resulting in glomerular atrophy and ultimately renal failure. Multiple studies have shown that the regulation of inflammation, endoplasmic reticulum stress (ERS), oxidative stress and other related proteins plays crucial roles in the progression of DN.8–13 Furthermore, certain regulatory factors can promote the transformation of endothelial cells into mesenchymal cells, a process known as epithelial-mesenchymal transition (EMT). This process plays a significant role in the development of renal fibrosis in DN. Additionally, recent studies have highlighted the dysfunction of cell junction proteins in podocytes, glomerular endothelial cells (GECs), glomerular mesangial cells (GMCs), and renal tubular epithelial cells (TECs) as key players in DN pathogenesis.14–17 Despite ongoing research, the pathogenesis of DN remains incompletely understood due to its complexity. Therefore, elucidating the underlying mechanisms of DN and identifying therapeutic targets have both emerged as crucial strategies for effective DN treatment.

Autophagy, an intracellular degradation process, plays a pivotal role in maintaining cellular homeostasis.18,19 It is involved in the pathogenesis of numerous diseases, including diabetes,20 DN,9 and cardiovascular disease.19 Pyroptosis, a distinct form of programmed cell death, differs from apoptosis both morphologically and mechanistically. It is characterized by the formation of inflammasomes and the production of pro-inflammatory caspases.21–23 Pyroptosis has been implicated in a range of diseases, including atherosclerosis, diabetes, and DN.10,24 Ferroptosis, another type of programmed cell death, is characterized by iron-dependent accumulation of lipid hydroperoxides to lethal levels.25

The role of inflammation in the onset and progression of DN is well-documented.7,12 Inflammation markers such as C-reactive protein, kidney injury molecule-1, serum uric acid, monocyte/lymphocyte ratio in hemogram, systemic inflammatory index, and uric acid/HDL cholesterol ratio have been associated with DN.25–29 Autophagy, a cellular process that maintains homeostasis, has been shown to suppress inflammation, thereby protecting the kidney.30 On the other hand, both pyroptosis and ferroptosis are forms of programmed cell death that are associated with inflammation.31 Diabetes mellitus and its microvascular complications are associated with high burden of inflammation.32,33 Given the central role of inflammation in DN, it is logical to investigate autophagy, pyroptosis, and ferroptosis in the context of this disease. Elucidating the contributions of these cellular processes in DN may offer valuable insights into potential therapeutic strategies for managing this condition. Besides, recent studies indicate that autophagy, pyroptosis, and ferroptosis of podocytes, GECs, GMCs, and TECs are involved in the occurrence and development of DN. A thorough examination of the uniqueness and correlation among autophagy, pyroptosis, and ferroptosis in the injury pathways of these cell types could lead to the identification of novel therapeutic targets for the prevention and treatment of DN.

Autophagy and DN

The autophagy process is essential for maintaining the homeostatic balance of cellular components, as it regulates their synthesis, degradation, and recirculation. This multistep regulation is orchestrated by various signaling pathways, including phosphatidylinositol 3-kinase (PI3K) -AKT-mTOR signal pathways, AMP-activated protein kinase (AMPK) signal pathway and nuclear factor kappa-B (NF-κB) signal cascades.18 Under physiological conditions, an appropriate amount of autophagy maintains the stability of its structure and function. However under pathological conditions, autophagy dysregulation can lead to cellular dysfunction and contribute to the development of various diseases, including DN,9 cardiovascular disease,19 diabetes20 and others.

The main pathological features of DN are podocytes, GECs, GMCs, TECs injury, which eventually develops into glomerular and tubulointerstitial fibrosis.10 These injuries are driven by various mechanisms such as glucolipotoxicity, inflammation, apoptosis, oxidative stress, and mitochondrial dysfunction.7,10 Autophagy plays a crucial role in maintaining intracellular homeostasis, and its dysfunction has been implicated in the pathogenesis of DN. Notably, glucagon like peptide-1 (GLP-1) has been shown to alleviate kidney damage in DN rats by promoting autophagy through the activation of the PI3K/AKT/mTOR pathway.34 Similarly, oral butyrate has been found to alleviate damage in diabetic rats, possibly by enhancing autophagy via the AMPK/mTOR signaling pathway.35 These studies provide evidence that autophagy plays a key role in the progression of DN and that modulating autophagy may offer therapeutic potential for reducing renal damage. Therefore, in this section, we aim to review and discuss the recent research progress on autophagy in podocytes, GECs, GMCs, and TECs. We focus on elucidating the role of autophagy in the progression of DN and exploring potential therapeutic targets.

Autophagy and Podocytes

Podocytes, as highly specialized glomerular epithelial cells, play a crucial role in maintaining glomerular structure and function. The initial pathological manifestation of DN often involves a decrease in podocyte number, followed by their detachment, which further exacerbates glomerular injury. It is reported that autophagy dysfunction is one of the important mechanisms causing homeostatic imbalance in podocytes. Regulating podocyte autophagy can protect against glomerular injury and improve renal function.36,37 SPAG5-AS1 gene knockout can inhibit apoptosis and induce autophagy via suppressing AKT/mTOR pathway, reducing podocyte injury induced by high glucose (HG).38 Adipose-derived stem cell-derived exosomes enhance the expression of miR-486, thereby inhibiting the Smad1/mTOR signaling pathway, and enhancing autophagy flux to reduce podocyte damage.39 Besides, studies have shown that multiple drugs can improve DN by promoting podocyte autophagy. Exogenous spermine ameliorates podocyte autophagy by regulating the AMPK/mTOR signaling pathway, alleviates podocyte injury, and improves serum creatinine, urea and urinary albumin excretion in diabetic rats.36 Curcumin and ursolic acid induce podocyte autophagy and alleviate renal oxidative stress through PI3K/AKT/mTOR pathway, thereby improving renal function in diabetic rats.37,40 Astragaloside-IV via the SIRT1/NF-κB pathway induces autophagy to inhibit glucose-induced EMT of podocytes and to improve function in DN mice.41 Collectively these studies suggest that improving podocyte autophagy can serve as an important target for the treatment of DN.36,37

Autophagy and Glomerular Endothelial Cells

GECs, as a crucial component of the glomerular filtration barrier, play a pivotal role in maintaining renal health. Upon injury, GECs exhibit an elevated level of proteinuria, indicating compromised glomerular function. This injury further exacerbates autophagy and senescence, leading to dysfunction of GECs. Such dysfunction has been implicated in the pathogenesis of renal microvascular disease and may promote the development of DN.

The electron microscopy findings indicate that in streptozotocin-induced diabetic rats, there is a significant decrease in both the volume and numerical densities of autophagic vacuoles in proximal tubular cells compared to the control group.42 Yoshibayashi et al suggest that autophagy-deficient mice can lead to GECs injury and severe proteinuria.43 In ATG5-deficient diabetic mice, proteinuria levels are significantly elevated compared to controls. The electron microscope results show that the cytoplasmic structure of GECs is disordered and vacuolized, which further proves that GECs autophagy deficiency aggravated the DN process.44 Inhibition of autophagy diminishes GEC viability. Additionally, heat-aggregated gamma globulin inhibits GECs autophagy through the AKT/mTOR pathway, further compromising cell survival and exacerbating renal injury.45 Cinacalcet elevates intracellular Ca2+ concentration and activates Ca2+/calmodulin-dependent protein kinase β, leading to the phosphorylation of liver kinase B1 and downstream signaling. This cascade ultimately enhances autophagy, reduces oxidative stress and apoptosis, and protects GECs and DN in mice exposed to HG.46 Spironolactone mitigates podocyte adhesive capacity damage induced by mechanical stress by blocking the PI3K/AKT/mTOR pathway and restoring autophagy activity.47 These findings collectively underscore the crucial role of autophagy in maintaining the structural and functional integrity of GECs.

Autophagy and Glomerular Mesangial Cells

The death and aging of GMCs indeed exacerbate the progression of DN. HG can stimulate human GMCs proliferation, extracellular matrix accumulation, and oxidative stress.48 Astragaloside-IV, a compound that targeting the silent information regulator 1 (SIRT1)-NF-κB pathway, has been shown to induce autophagy and enhance GMCs viability, improving glomerular morphology and renal fibrosis in DN mice.49 On the other hand, aging can inhibit autophagy and promote GMCs senescence through the RAGE/STAT5 pathway.50 Metformin effectively ameliorates glycolipid metabolism and renal damage in diabetic rats by activating the AMPK/SIRT1-FOXO1 pathway. This activation not only alleviates oxidative stress but also enhances autophagy, slowing down the abnormal proliferation of HG cultured GMCs.51 Fibroblast growth factor 21 ameliorates autophagy levels, thus preventing mesangial cells overproliferation via the AMPK/mTOR pathway.52 Trigonelline induces autophagy to protect mesangial cells in response to HG via activating the miR-5189-5p-AMPK pathway.53 Ginkgetin alleviates HG-induced GMCs oxidative stress injury, inflammation, and extracellular matrix deposition through AMPK/mTOR signal pathway mediated autophagy.54 Collectively, these findings suggest that targeting autophagy and its associated signaling pathways may provide therapeutic strategies for mitigating GMCs death and the progression of DN.

Autophagy and Renal Tubular Epithelial Cells

Oxidative stress, inflammation, ERS and hypoxia can cause TECs damage, leading to renal interstitial fibrosis and the decline of renal function.55–60 This damage is further exacerbated by the dysregulation of autophagy, as observed in the proximal tubules of streptozotocin-induced diabetic models and HG-induced Human renal glomerular endothelial cells (HRGECs) HK-2 cells.61 ATG7 knockout mice, which exhibit defective autophagy in renal proximal epithelial tubular cells, result in increased renal cell damage.62 In HG-cultured NRK-52E cells and DN mice, the inhibition of ATF4 can ameliorate DN tubulointerstitial fibrosis by improving autophagy flux and decreasing collagen type-IV levels.14 Cotreatment with molybdenum and autophagy inhibitor 3-methyladenine (3-MA) significantly increase lactate dehydrogenase release, reactive oxygen species (ROS) level, and cell injury, indicating that autophagy inhibition can exacerbate TECs damage and further worsen renal function.63 Metformin promotes autophagy of TECs through AMPK, thereby reducing tubular interstitial fibrosis.64 Besides, traditional medicine Red Ginseng has been shown to reduce the expression of TGF-β1 and kidney injury molecule-1 by inducing autophagy, and then alleviate renal inflammation and fibrosis caused by hyperglycemia.65 Collectively, these findings suggest that autophagy plays a crucial role in maintaining TEC health and preventing renal fibrosis. Therapeutic strategies aimed at restoring or enhancing autophagy may offer new avenues for treating renal damage and dysfunction.

Podocytes, GECs, GMCs and TECs damage are all involved in DN progression. Damage to these cell types plays a significant role in the development and progression of DN. These studies indicate that inhibition of autophagy aggravates renal injury and promotes the progression of DN. Fortunately, some drugs have shown that regulating autophagy can improve renal function in DN. For instance, Metformin can improve renal function by promoting autophagy.51,64 GLP-1 analog liraglutide can upregulate autophagy in vivo and in vitro through the AMPK/mTOR signaling pathway.66 Subcutaneous injection of insulin with liraglutide alleviates oxidative stress and activates autophagy, which not only restored renal morphology but also significantly improved renal hemodynamics.67 Given the substantial interest in developing therapies that enhance autophagy as means of treating DN, further study of autophagy in this setting may lead to fruitful results.

Pyroptosis and DN

Pyroptosis can be activated by Caspase-1 and induce an inflammatory response, leading to cell membrane rupture and dissolution. The molecular mechanisms of pyroptosis21 mainly include classical cell pyroptosis mediated by Caspase-1, non-classical pyroptosis mediated by Caspase-11/-4/-5 and by Caspase-3. The activation of Caspase-1 is the core of the classical pyroptosis pathway, a defence mechanism against pathogenic microbial infection, and an important part of the natural immune system. Caspase-11/-4/-5 can directly recognize and bind lipopolysaccharide (LPS) and act on GSDMD (gasdermin d) to release active N-terminal domain (GSDMD-N); In addition, Caspase-11 also activates the inflammasomes of nod like receptor protein-3 (NLRP3) and downstream caspase-1, resulting in the release of inflammatory factors and promoting pyroptosis. Caspase-1 mediates pyroptosis by activating GSDMD. Inflammasomes are produced during pyroptosis, which can induce tumor pyroptosis and inhibit tumor cell proliferation. Pyroptosis is a physiological state that can promote the immune response against infection, but excessively activate pyroptosis leads to GSDMD activation and numerous inflammatory cytokines such as interleukin (IL)-1β and IL-18 release, which can cause cell death, tissue damage, organ failure, and even cause diabetes, cause irreversible damage to the body.11,21–24

Pyroptosis has been implicated in a wide range of diseases such as diabetes, DN and infectious diseases.11,21,24 The expression of Caspase-1 and GSDMD in renal tissue of DN mice increased significantly.68 A series of researches have indicated that inhibiting pyroptosis can reduce the damage to podocytes,68 GECs,69 GMCs70 and TECs.71

Pyroptosis and Podocytes

Podocyte injury plays a crucial role in the early stages of DN. The expression of Caspase-11, Caspase-4 and GSDMD-N in podocytes is increased significantly in DN mice, the loss and fusion of podocyte foot processes, and elevated inflammatory cytokines IL-1β and IL-18, suggesting increased podocyte pyroptosis in DN.72,73 Similar results are obtained in vitro models of DN.72 Knockout of Caspase-11, Caspase-4 or GSDMD gene can attenuate the above changes in diabetic mice, proposing that inhibiting pyroptosis can ameliorate podocyte damage in DN.73 Moreover, studies have shown that multiple drugs can improve DN not only by promoting podocyte autophagy but also by alleviating podocyte pyroptosis. High-glucose can induce mouse podocyte line MPC5 cells pyroptosis and oxidative stress.74 Further, atorvastatin protects MPC-5 cells from pyroptosis and downregulates the level of renal oxidative stress through MALAT1/miR-200c/NRF2 signal pathway.75 Carnosine can significantly reverse the albuminuria and histopathological lesions damage of the DN mouse model and reduce renal inflammation and pyroptosis via the targeting of Caspase-1.68 The administration of geniposide to DN mice through AMPK/SIRT1/NF-κB pathway inhibits podocyte pyroptosis, oxidative stress and inflammation, and reduces glomerular basement membrane thickening and inflammatory cell infiltration, thereby inhibiting the development of DN.75 In vitro studies have shown that high fat can induce NLRP3 dependent pyroptosis of podocytes. Ginsenoside Rg1 inhibits pyroptosis through the mTOR/NF-κB/NLRP3 axis and protects podocytes from damage induced by hyperlipidemia.76 Total flavones of Abelmoschus Manihot (TFA) have been widely used to treat DN. Further research reveals that TFA can ameliorate podocytes pyroptosis under diabetic conditions by regulating METTL3-dependent methyladenosine modification and downregulating NLRP3-inflammasome formation and PTEN/PI3K/AKT axis.77 All these results suggest that podocyte pyroptosis is involved in the development of DN.

Pyroptosis and Glomerular Endothelial Cells

The whole process of DN is accompanied by GECs injury. Hyperglycemia, inflammation, ERS and oxidative stress can lead to endothelial cell injury. High-glucose can induce pyroptosis of human endothelial cells, accompanied by an increase in NLRP3 inflammatory bodies.78 This finding is further supported by another study, which demonstrates that high-glucose/LPS and ATP enhance the release of pyroptosis-related factors IL-18 and IL-1β in GECs.69 Besides, the deletion of GSDMD reduces pyroptosis and kidney injury both in-vivo and in-vitro.69 Gu et al reported that high-glucose increases GSDMD-N and IL-1β, IL-18, which induces GECs pyroptosis.79 And sodium butyrate can improve GECs pyroptosis under diabetic conditions through Caspase1-GSDMD typical pyroptosis pathway.79 Hirudin has been found to ameliorate renal injury in DN. Further studies show that Hirudin significantly ameliorates the GECs injury by inhibiting GSDMD-mediated pyroptosis.69 Collectively, these findings suggest that GEC pyroptosis plays a pivotal role in the pathogenesis of DN. Therapeutic strategies that target GEC pyroptosis may offer promising avenues for the treatment of this debilitating disease.

Pyroptosis and Glomerular Mesangial Cells

Another study shows that streptozotocin-induced diabetic rats exhibit increased expression of Caspase-1, NLRP3, and IL-1β.70 This is consistent with the latest findings that high-glucose promotes the expression of Caspase-1, NLRP3, and IL-1β in GMCs.70,80 Sodium butyrate, through the NF-κB signaling pathway, reduces LPS and high-glucose-induced IL-1β expression in GMCs.79 These results suggest that GMCs pyroptosis is involved in the development of DN.

Pyroptosis and Renal Tubular Epithelial Cells

Hyperglycemia, proteinuria and oxidative stress can aggravate renal tubular injury.7,81,82 Changes such as apoptosis and interstitial fibrosis of TECs further lead to the decline of renal function and aggravate the process of DN.10 In diabetic rats, renal function is significantly impaired, accompanied by NLRP3-dependent pyroptosis, while high-glucose-induced expression of TECs pyroptosis-related proteins is increased.11,83 Reducing the expression of pyroptosis-associated protein can reduce the damage to renal function.11 High-glucose increases the expression of NLRP3, Caspase-1 and IL-1β and IL-18 in HK-2 cells, promotes TECs pyroptosis and induces renal tubular injury.71 Another study is verified that high-glucose could induce NLRP3, Caspase-1, and IL-1β expression in HK-2 cells. In addition, high-glucose also induced the expression of pyroptosis-related executive protein GSDMD.84 Inhibition of ROS/NLRP3/Caspase-1 mediated pyroptosis can reduce cadmium-induced apoptosis of duck TECs, suggesting that inhibition of pyroptosis can reduce TECs injury.85 Selective Caspase-1 inhibitors VX-765 ameliorates ballooned cell membrane, decreases the expression of GSDMD cleavage, and the release of inflammatory cytokines in high-glucose induced HK-2 cells. On the other hand, VX-765 treatment can improve renal function, inhibit inflammatory cell infiltration and pyroptosis-related protein expression. Therefore VX-765 can downregulate collagen I and fibronectin deposition in DN mice to alleviate tubulointerstitial fibrosis.15 Pyroptosis upregulated TECs adhesion protein 1 (VCAM1) expression under high-glucose, while disulfiram treatment abrogating pyroptosis inhibited VCAM1 expression, inflammation and fibrosis in HK-2 cells. Disulfiram might improve fibrosis in DN by targeting renal tubular pyroptosis and VCAM1 expression.16 The above studies show that pyroptosis is closely related to TECs injury, and inhibition of TECs pyroptosis is expected to become an important target for reducing DN, suggesting that pyroptosis can be an important target for treating DN.

In summary, the pyroptosis of podocytes, GECs, GMCs and TECs is significantly activated in the diabetic state, and inhibiting pyroptosis can prevent the progression of DN, it can effectively inhibit the development of DN. The specific targets of action and related pathways have yet to be further explored, and the mechanism of pyroptosis itself also needs to be explored. In the future, we may be able to find a cure for diabetic nephropathy with pyroptosis as the target.

Ferroptosis and DN

Ferroptosis is a new type of programmed cell death which has attracted widespread attention in recent years. It mainly refers to cell death caused by excessive accumulation of lipid peroxides under iron-dependent overload. Multiple factors are involved in the occurrence of ferroptosis including iron metabolism disorders, Cystine/glutamate reverse transporter/glutathione (GSH)/glutathione peroxidase 4 (GPX4) pathway abnormality, oxidative stress, lipid peroxidation, tumor suppressor p53 targeted induction.

Ferroptosis is involved in many diseases such as diabetes,86 diabetic cardiomyopathy,87 DN88 and diabetic retinopathy.89 HG and palmitate trigger ferroptosis in mouse pancreatic β cells and mouse islets through decreased the level of expression of GPX4 and increased the level of expression of anti-acyl-CoA synthetase long-chain family member 4(ACSL4) while metformin is effective in inhibiting pancreatic β cells and mouse islets ferroptosis through regulation of the GPX4/ACSL4 axis.86 Palmitate induces cardiomyocytes ferroptosis, astragaloside IV ameliorated myocardial injury and improved contractile function, attenuated lipid deposition, and decreased the expression level of ferroptosis-related factors in DCM rats and cellular ferroptosis.87 Ferroptosis is increased in HG-treated HK-2 cells. ACSL4 is increased in kidney tissues of DN mice.88 HG stimulation increased those of MDA, ROS, GSSG, and Fe in HRECs.89 Mounting studies suggest that ferroptosis plays an important role in kidney cells injury and is one of the key mechanisms to induce DN, supporting that ferroptosis could be a potential therapeutic target for DN.90,91 The marker of ferroptosis, GPX4, is decreased in db/db mice and T2D patients, which means ferroptosis plays a pathophysiologic role in DN,68 which means ferroptosis might serve as a target for treating DN. In this section, we mainly separately discuss the role of ferroptosis in podocytes, GECs, GMCs and TECs.

Ferroptosis and Podocytes

HG induces podocytes injury by stimulating podocyte ferroptosis.92 Besides, high fructose feeding and intraperitoneal injection of STZ is helpful for establishing DN model.93 High fructose consumption induces podocyte ferroptosis in glomerular injury (main features include low-expression of ferroptosis protein markers SLC7A11 and GPX4, lipid peroxidation accumulation and cell death).94 Accumulating evidence suggests that some medicines can modulate ferroptosis in podocytes and show great potential for improving DN. Ginkgolide B alleviates ferroptosis in palmitic acid-high-glucose induced podocytes and DN mice through inhibiting GPX4 ubiquitination to improve DN.95 Germacrone possesses anti-inflammation, anti-apoptotic and antioxidative effects. Germacrone plays a pivotal role in the progression of DN. Recent study reveals that germacrone mediates the improvement of DN through regulating ferroptosis by targeting miR-188- 3p/GPX4 signaling axis.96

Ferroptosis and Glomerular Endothelial Cells

More and more studies have shown that HG induces ferroptosis of endothelial cells participates in diabetes retinopathy,89 diabetes atherosclerosis,97 diabetic wounds,98 and diabetes-related limb ischemia.99 Previous studies have shown that glomerular endothelial cells ferroptosis plays an important role in the pathophysiological process of DN.100–102 HG induced ferroptosis in an in vitro model of DN by increasing iron concentration in HRGECs. Traditional Chinese medicine, Schisandrin A attenuates ferroptosis in HG induced HRGECs.100 Another study also suggests that high glucose-induces ferroptosis in HUVECs, as evidenced by the protective effect of the ferroptosis inhibitors, deferoxamine and ferrostatin-1, resulting in increased lipid ROS and decreased cell viability.101

Ferroptosis and Glomerular Mesangial Cells

Previous study has shown that ferroptosis activated in DN patients and in mesangial cells in response to HG.102 The research results indicate that the release of serum ferritin, LDH and ferroptosis-related proteins ACSL4, GPX4 are elevated in DN patients and HG-induced renal mesangial cells.102 Besides, galactose deficient IgA1 leads to ferroptosis in human mesangial cells by inhibiting PPARα and FABP1 expression. Levels of GPX4 were decreased in immunoglobulin A nephropathy renal tissue, which means ferroptosis plays a key role in the processes of IgA nephropathy.103 Senescence is closely related to DN.104,105 Aging causes iron overload in renal tubules. Salidroside is a phenylpropanoid glycoside and acts as an anti-oxidative, anti-inflammatory, anti-aging, and alleviates renal interstitial fibrosis. Studies have suggested that salidroside delays renal aging and inhibits aging-related glomerular fibrosis by inhibiting ferroptosis in SAMP8 mice.106 Ferroptosis inhibitor ferrostatin-1 increased the expression of GPX4 in the mouse kidneys.106

Ferroptosis and Renal Tubular Epithelial Cells

HG injures rat renal tubular epithelial NRK-52E cells by triggering ferroptosis as demonstrated by the accumulation of iron content and down-regulation of GPX4, SLC7A11, indicators of ferroptosis.107 Another study also suggests that the GPX4 protein expression and GSH activity levels are decreased in HG induced HK-2 cells. Circular ASAP2 through SLC7A11 decreases the inflammation and ferroptosis in DN.108 Traditional Chinese Medicine glabridin ameliorates DN via suppressing ferroptosis in diabetic rats’ kidney and HG-induced NRK-52E cells.107

The above studies have explained the important role of ferroptosis regulated by related signaling pathways in the occurrence and development of DN at different cellular levels, indicating that the interaction between different cells in kidney tissue may provide clues to the pathogenesis of DN and targeting ferroptosis could provide a new way to prevent and treat DN.

Conclusions

In recent years, autophagy, pyroptosis and ferroptosis have attracted increasing attention, and have become a research focus in DN and other diseases. Besides, some drugs such as metformin and a large number of Chinese herbal medicines can improve DN by regulating autophagy, pyroptosis and ferroptosis, further proving that autophagy, pyroptosis and ferroptosis can be used as a star target for treating DN. However, the specific mechanism still needs to be further studied. Therefore, seeking targets that can act on autophagy and pyroptosis of podocytes, GECs, GMCs and TECs at the same time is expected to become a breakthrough in the treatment of DN.

Abbreviations

AMPK, AMP-activated protein kinase; DN, diabetic nephropathy; EMT, epithelial-mesenchymal transition; ERS, endoplasmic reticulum stress; GECs, glomerular endothelial cells; GMCs, glomerular mesangial cells; GSDMD, gasdermin d; HK-2, human renal tubular cell lines; IL, interleukin; PI3K, phosphatidylinositol 3-kinase; TECs, renal tubular epithelial cells; NLRP3, nod like receptor protein-3; ROS, reactive oxygen species; SIRT1, silent information regulator 1; VCAM1, vascular cell adhesion protein 1; 3-MA, 3-methyladenine.

Funding

The present review was supported by the Natural Science Foundation of Inner Mongolia Autonomous Region (grant number. 2023QN08008); College Student Innovation Program (grant number. S202310138035); Natural Science Foundation of ChiFeng city (grant number. SZR2023026).

Disclosure

The authors report no conflicts of interest in this work.

References

1. Demir Y, Ceylan H, Türkeş C, et al. Molecular docking and inhibition studies of vulpinic, carnosic and usnic acids on polyol pathway enzymes. J Biomol Struct Dyn. 2022;40(22):12008–12021. doi:10.1080/07391102.2021.1967195

2. Sever B, Altıntop MD, Demir Y, et al. Design, synthesis, in vitro and in silico investigation of aldose reductase inhibitory effects of new thiazole-based compounds. Bioorg Chem. 2020;102:104110. doi:10.1016/j.bioorg.2020.104110

3. Sun H, Saeedi P, Karuranga S, et al. IDF diabetes Atlas: global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract. 2021:109119. doi:10.1016/j.diabres.2021.109119

4. Bjornstad P, Drews KL; TODAY Study Group. Long-term complications in youth-onset type 2 diabetes. N Engl J Med. 2021;385(5):416–426. doi:10.1056/NEJMoa2100165

5. Sever B, Altıntop MD, Demir Y, et al. Identification of a new class of potent aldose reductase inhibitors: design, microwave-assisted synthesis, in vitro and in silico evaluation of 2-pyrazolines. Chem Biol Interact. 2021;345:109576. doi:10.1016/j.cbi.2021.109576

6. Sinha N, Kumar V, Puri V, et al. Urinary exosomes: potential biomarkers for diabetic nephropathy. Nephrology. 2020;25(12):881–887. doi:10.1111/nep.13720

7. Dong C, Liu S, Cui Y, et al. 12-Lipoxygenase as a key pharmacological target in the pathogenesis of diabetic nephropathy. Eur J Pharmacol. 2020;879:173122. doi:10.1016/j.ejphar.2020.173122

8. Tokalı FS, Demir Y, Türkeş C, et al. Novel acetic acid derivatives containing quinazolin-4(3H)-one ring: synthesis, in vitro, and in silico evaluation of potent aldose reductase inhibitors. Drug Dev Res. 2023;84(2):275–295. doi:10.1002/ddr.22031

9. Koch EAT, Nakhoul R, Nakhoul F, et al. Autophagy in diabetic nephropathy: a review. Int Urol Nephrol. 2020;52(9):1705–1712. doi:10.1007/s11255-020-02545-4

10. Sun H, Yuan Y, Sun Z. Update on mechanisms of renal tubule injury caused by advanced glycation end products. Biomed Res Int. 2016;2016:5475120. doi:10.1155/2016/5475120

11. Ke R, Wang Y, Hong S, Xiao L. Endoplasmic reticulum stress related factor IRE1α regulates TXNIP/NLRP3-mediated pyroptosis in diabetic nephropathy. Exp Cell Res. 2020;396(2):112293. doi:10.1016/j.yexcr.2020.112293

12. Wu X, Li H, Wan Z, et al. The combination of ursolic acid and empagliflozin relieves diabetic nephropathy by reducing inflammation, oxidative stress and renal fibrosis. Biomed Pharmacother. 2021;144:112267. doi:10.1016/j.biopha.2021.112267

13. Demir Y, Tokalı FS, Kalay E, et al. Synthesis and characterization of novel acyl hydrazones derived from vanillin as potential aldose reductase inhibitors. Mol Divers. 2023;27(4):1713–1733. doi:10.1007/s11030-022-10526-1

14. Liang Q, Liu T, Guo T, et al. ATF4 promotes renal tubulointerstitial fibrosis by suppressing autophagy in diabetic nephropathy. Life Sci. 2021;264:118686. doi:10.1016/j.lfs.2020.118686

15. Wen S, Deng F, Li L, et al. VX-765 ameliorates renal injury and fibrosis in diabetes by regulating caspase-1-mediated pyroptosis and inflammation. J Diabetes Investig. 2022;13(1):22–33. doi:10.1111/jdi.13660

16. Jia Y, Xu H, Yu Q, et al. Identification and verification of vascular cell adhesion protein 1 as an immune-related hub gene associated with the tubulointerstitial injury in diabetic kidney disease. Bioengineered. 2021;12(1):6655–6673. doi:10.1080/21655979.2021.1976540

17. Eftekhari A, Vahed SZ, Kavetskyy T, et al. Cell junction proteins: crossing the glomerular filtration barrier in diabetic nephropathy. Int J Biol Macromol. 2020;148:475–482. doi:10.1016/j.ijbiomac.2020.01.168

18. Lee YH, Kim J, Park K, et al. β-cell autophagy: mechanism and role in β-cell dysfunction. Mol Metab. 2019;27S(Suppl):S92–S103. doi:10.1016/j.molmet.2019.06.014

19. New J, Thomas SM. Autophagy-dependent secretion: mechanism, factors secreted, and disease implications. Autophagy. 2019;15(10):1682–1693. doi:10.1080/15548627.2019.1596479

20. Li XD, He SS, Wan TT, et al. Liraglutide protects palmitate-induced INS-1 cell injury by enhancing autophagy mediated via FoxO1. Mol Med Rep. 2021;23(2):147. doi:10.3892/mmr.2020.11786

21. Gong W, Shi Y, Ren J. Research progresses of molecular mechanism of pyroptosis and its related diseases. Immunobiology. 2020;225(2):151884. doi:10.1016/j.imbio.2019.11.019

22. Kesavardhana S, Malireddi RKS, Kanneganti TD. Caspases in Cell Death, Inflammation, and Pyroptosis. Annu Rev Immunol. 2020;38:567–595. doi:10.1146/annurev-immunol-073119-095439

23. Feng S, Fox D, Man SM. Mechanisms of gasdermin family members in inflammasome signaling and cell death. J Mol Biol. 2018;430(18 Pt B):3068–3080. doi:10.1016/j.jmb.2018.07.002

24. Sharma BR, Kanneganti TD. NLRP3 inflammasome in cancer and metabolic diseases. Nat Immunol. 2021;22(5):550–559. doi:10.1038/s41590-021-00886-5

25. Li Z, Lange M, Dixon SJ, et al. Lipid quality control and ferroptosis: from concept to mechanism. Annu Rev Biochem. 2023. doi:10.1146/annurev-biochem-052521-033527

26. Xia D, Zang J. Telmisartan combined with calcitriol enhances therapeutic efficacy for diabetic nephropathy while inhibiting inflammation and renal interstitial fibrosis. Am J Transl Res. 2023;15(11):6543–6550.

27. Kocak MZ, Aktas G, Duman TT, et al. Monocyte lymphocyte ratio as a predictor of diabetic kidney injury in type 2 diabetes mellitus; the MADKID Study. J Diabetes Metab Disord. 2020;19(2):997–1002. doi:10.1007/s40200-020-00595-0

28. Kocak MZ, Aktas G, Duman TT, et al. Is Uric Acid elevation a random finding or a causative agent of diabetic nephropathy? Rev Assoc Med Bras. 2019;65(9):1155–1160. doi:10.1590/1806-9282.65.9.1156

29. Taslamacioglu Duman T, Ozkul FN, Balci B. Could systemic inflammatory index predict diabetic kidney injury in type 2 diabetes mellitus? Diagnostics. 2023;13(12):2063. doi:10.3390/diagnostics13122063

30. Kimura T, Isaka Y, Yoshimori T. Autophagy and kidney inflammation. Autophagy. 2017;13(6):997–1003. doi:10.1080/15548627.2017.1309485

31. Lin L, Zhang MX, Zhang L, et al. Autophagy, pyroptosis, and ferroptosis: new regulatory mechanisms for atherosclerosis. Front Cell Dev Biol. 2022;9:809955. doi:10.3389/fcell.2021.809955

32. Aktas G, Atak Tel BM, Tel R, et al. Treatment of type 2 diabetes patients with heart conditions. Expert Rev Endocrinol Metab. 2023;18(3):255–265. doi:10.1080/17446651.2023.2204941;

33. Aktas G. Association between the prognostic nutritional index and chronic microvascular complications in patients with type 2 diabetes mellitus. J Clin Med. 2023;12(18):5952. doi:10.3390/jcm12185952

34. Hu Y, Wang SX, Wu FY, et al. Effects and mechanism of ganoderma lucidum polysaccharides in the treatment of diabetic nephropathy in streptozotocin-induced diabetic rats. Biomed Res Int. 2022;2022:4314415. doi:10.1155/2022/4314415

35. Zhong Y, Liu J, Sun D, et al. Dioscin relieves diabetic nephropathy via suppressing oxidative stress and apoptosis, and improving mitochondrial quality and quantity control. Food Funct. 2022;13(6):3660–3673. doi:10.1039/d1fo02733f

36. Zhang X, Zhang L, Chen Z, et al. Exogenous spermine attenuates diabetic kidney injury in rats by inhibiting AMPK/mTOR signaling pathway. Int J Mol Med. 2021;47(3):27. doi:10.3892/ijmm.2021.4860

37. Di Tu Q, Jin J, Hu X, et al. Curcumin improves the renal autophagy in rat experimental membranous nephropathy via regulating the PI3K/AKT/mTOR and Nrf2/HO-1 signaling pathways. Biomed Res Int. 2020;2020:7069052. doi:10.1155/2020/7069052

38. Xu J, Deng Y, Wang Y, et al. SPAG5-AS1 inhibited autophagy and aggravated apoptosis of podocytes via SPAG5/AKT/mTOR pathway. Cell Prolif. 2020;53(2):e12738. doi:10.1111/cpr.12738

39. Jin J, Shi Y, Gong J, et al. Exosome secreted from adipose-derived stem cells attenuates diabetic nephropathy by promoting autophagy flux and inhibiting apoptosis in podocyte. Stem Cell Res Ther. 2019;10(1):95. doi:10.1186/s13287-019-1177-1

40. Xu L, Fan Q, Wang X, et al. Ursolic acid improves podocyte injury caused by high glucose. Nephrol Dial Transplant. 2017;32(8):1285–1293. doi:10.1093/ndt/gfv382

41. Wang X, Gao Y, Tian N, et al. Astragaloside IV inhibits glucose-induced epithelial-mesenchymal transition of podocytes through autophagy enhancement via the SIRT-NF-κB p65 axis. Sci Rep. 2019;9(1):323. doi:10.1038/s41598-018-36911-1

42. Barbosa AA, Zhou H, Hültenschmidt D, et al. Inhibition of cellular autophagy in proximal tubular cells of the kidney in streptozotocin-diabetic and uninephrectomized rats. Virchows Arch B Cell Pathol Incl Mol Pathol. 1992;61(6):359–366. doi:10.1007/BF02890439

43. Yoshibayashi M, Kume S, Yasuda-Yamahara M, et al. Protective role of podocyte autophagy against glomerular endothelial dysfunction in diabetes. Biochem Biophys Res Commun. 2020;525(2):319–325. doi:10.1016/j.bbrc.2020.02.088

44. Lenoir O, Jasiek M, Hénique C, et al. Endothelial cell and podocyte autophagy synergistically protect from diabetes-induced glomerulosclerosis. Autophagy. 2015;11(7):1130–1145. doi:10.1080/15548627.2015.1049799

45. Wang L, Law HKW. Immune complexes suppressed autophagy in glomerular endothelial cells. Cell Immunol. 2018;328:1–8. doi:10.1016/j.cellimm.2018.02.013

46. Lim JH, Kim HW, Kim MY, et al. Cinacalcet-mediated activation of the CaMKKβ-LKB1-AMPK pathway attenuates diabetic nephropathy in db/db mice by modulation of apoptosis and autophagy. Cell Death Dis. 2018;9(3):270. doi:10.1038/s41419-018-0324-4

47. Li D, Lu Z, Xu Z, et al. Spironolactone promotes autophagy via inhibiting PI3K/AKT/mTOR signalling pathway and reduce adhesive capacity damage in podocytes under mechanical stress. Biosci Rep. 2016;36(4):e00355. doi:10.1042/BSR20160086

48. Yun J, Ren J, Liu Y, et al. Circ-ACTR2 aggravates the high glucose-induced cell dysfunction of human renal mesangial cells through mediating the miR-205-5p/HMGA2 axis in diabetic nephropathy. Diabetol Metab Syndr. 2021;13(1):72. doi:10.1186/s13098-021-00692-x

49. Wang X, Gao Y, Tian N, et al. Astragaloside IV represses high glucose-induced mesangial cells activation by enhancing autophagy via SIRT1 deacetylation of NF-κB p65 subunit. Drug Des Devel Ther. 2018;12:2971–2980. doi:10.2147/DDDT.S174058

50. Shi M, Yang S, Zhu X, et al. The RAGE/STAT5/autophagy axis regulates senescence in mesangial cells. Cell Signal. 2019;62:109334. doi:10.1016/j.cellsig.2019.05.019

51. Ren H, Shao Y, Wu C, et al. Metformin alleviates oxidative stress and enhances autophagy in diabetic kidney disease via AMPK/SIRT1-FoxO1 pathway. Mol Cell Endocrinol. 2020;500:110628. doi:10.1016/j.mce.2019.110628

52. Meng F, Cao Y, Khoso MH, et al. Therapeutic effect and mechanism of combined use of FGF21 and insulin on diabetic nephropathy. Arch Biochem Biophys. 2021;713:109063. doi:10.1016/j.abb.2021.109063

53. Chen C, Ma J, Miao CS, et al. Trigonelline induces autophagy to protect mesangial cells in response to high glucose via activating the miR-5189-5p-AMPK pathway. Phytomedicine. 2021;92:153614. doi:10.1016/j.phymed.2021.153614

54. Wei L, Jian P, Erjiong H, et al. Ginkgetin alleviates high glucose-evoked mesangial cell oxidative stress injury, inflammation, and extracellular matrix (ECM) deposition in an AMPK/mTOR-mediated autophagy axis. Chem Biol Drug Des. 2021;98(4):620–630. doi:10.1111/cbdd.13915

55. Palabıyık E, Sulumer AN, Uguz H, et al. Assessment of hypolipidemic and anti-inflammatory properties of walnut (Juglans regia) seed coat extract and modulates some metabolic enzymes activity in triton WR-1339-induced hyperlipidemia in rat kidney, liver, and heart. J Mol Recognit. 2023;36(3):e3004. doi:10.1002/jmr.3004

56. Demir Y, Türkeş C, Çavuş MS, et al. Enzyme inhibition, molecular docking, and density functional theory studies of new thiosemicarbazones incorporating the 4-hydroxy-3,5-dimethoxy benzaldehyde motif. Arch Pharm. 2023;356(4):e2200554. doi:10.1002/ardp.202200554

57. Thomas K, Zondler L, Ludwig N, et al. Glutamine prevents acute kidney injury by modulating oxidative stress and apoptosis in tubular epithelial cells. JCI Insight. 2022;7(21):e163161. doi:10.1172/jci.insight.163161

58. Widjaja AA, Viswanathan S, Shekeran SG, et al. Targeting endogenous kidney regeneration using anti-IL11 therapy in acute and chronic models of kidney disease. Nat Commun. 2022;13(1):7497. doi:10.1038/s41467-022-35306-1

59. Zheng Y, Zhang Z, Zheng D, et al. METTL14 promotes the development of diabetic kidney disease by regulating m6A modification of TUG1. Acta Diabetol. 2023;60(11):1567–1580. doi:10.1007/s00592-023-02145-5

60. Jiang A, Liu J, Wang Y, Zhang C. cGAS-STING signaling pathway promotes hypoxia-induced renal fibrosis by regulating PFKFB3-mediated glycolysis. Free Radic Biol Med. 2023;208:516–529. doi:10.1016/j.freeradbiomed.2023.09.011

61. Li A, Yi B, Han H, et al. Vitamin D-VDR (vitamin D receptor) regulates defective autophagy in renal tubular epithelial cell in streptozotocin-induced diabetic mice via the AMPK pathway. Autophagy. 2022;18(4):877–890. doi:10.1080/15548627.2021.1962681

62. Suzuki C, Tanida I, Oliva Trejo JA, et al. Autophagy deficiency in renal proximal tubular cells leads to an increase in cellular injury and apoptosis under normal fed conditions. Int J Mol Sci. 2019;21(1):155. doi:10.3390/ijms21010155

63. Zhuang J, Nie G, Hu R, et al. Inhibition of autophagy aggravates molybdenum-induced mitochondrial dysfunction by aggravating oxidative stress in duck renal tubular epithelial cells. Ecotoxicol Environ Saf. 2021;209:111771. doi:10.1016/j.ecoenv.2020.111771

64. Wang F, Sun H, Zuo B, et al. Metformin attenuates renal tubulointerstitial fibrosis via upgrading autophagy in the early stage of diabetic nephropathy. Sci Rep. 2021;11(1):16362. doi:10.1038/s41598-021-95827-5

65. Karunasagara S, Hong GL, Park SR, et al. Korean red ginseng attenuates hyperglycemia-induced renal inflammation and fibrosis via accelerated autophagy and protects against diabetic kidney disease. J Ethnopharmacol. 2020;254:112693. doi:10.1016/j.jep.2020.112693

66. Yang S, Lin C, Zhuo X, et al. Glucagon-like peptide-1 alleviates diabetic kidney disease through activation of autophagy by regulating AMP-activated protein kinase-mammalian target of rapamycin pathway. Am J Physiol Endocrinol Metab. 2020;319(6):E1019–E1030. doi:10.1152/ajpendo.00195.2019

67. Tong MQ, Luo LZ, Xue PP, et al. Glucose-responsive hydrogel enhances the preventive effect of insulin and liraglutide on diabetic nephropathy of rats. Acta Biomater. 2021;122:111–132. doi:10.1016/j.actbio.2021.01.007

68. Zhu W, Li YY, Zeng HX, et al. Carnosine alleviates podocyte injury in diabetic nephropathy by targeting caspase-1-mediated pyroptosis. Int Immunopharmacol. 2021;101(Pt B):108236. doi:10.1016/j.intimp.2021.108236

69. Han J, Zuo Z, Shi X, et al. Hirudin ameliorates diabetic nephropathy by inhibiting Gsdmd-mediated pyroptosis. Cell Biol Toxicol. 2023;39(3):573–589. doi:10.1007/s10565-021-09622-z

70. Zhan JF, Huang HW, Huang C, et al. Long non-coding RNA NEAT1 regulates pyroptosis in diabetic nephropathy via mediating the miR-34c/NLRP3 axis. Kidney Blood Press Res. 2020;45(4):589–602. doi:10.1159/000508372

71. Liu C, Zhuo H, Ye MY, et al. LncRNA MALAT1 promoted high glucose-induced pyroptosis of renal tubular epithelial cell by sponging miR-30c targeting for NLRP3. Kaohsiung J Med Sci. 2020;36(9):682–691. doi:10.1002/kjm2.12226

72. Cheng Q, Pan J, Zhou ZL, et al. Caspase-11/4 and gasdermin D-mediated pyroptosis contributes to podocyte injury in mouse diabetic nephropathy. Acta Pharmacol Sin. 2021;42(6):954–963. doi:10.1038/s41401-020-00525-z

73. Dong W, Jia C, Li J, et al. Fisetin attenuates diabetic nephropathy-induced podocyte injury by inhibiting NLRP3 inflammasome. Front Pharmacol. 2022;13:783706. doi:10.3389/fphar.2022.783706

74. Zuo Y, Chen L, He X, et al. Atorvastatin regulates MALAT1/miR-200c/NRF2 activity to protect against podocyte pyroptosis induced by high glucose. Diabetes Metab Syndr Obes. 2021;14:1631–1645. doi:10.2147/DMSO.S298950

75. Li F, Chen Y, Li Y, et al. Geniposide alleviates diabetic nephropathy of mice through AMPK/SIRT1/NF-κB pathway. Eur J Pharmacol. 2020;886:173449. doi:10.1016/j.ejphar.2020.173449

76. Wang T, Gao Y, Yue R, et al. Ginsenoside Rg1 alleviates podocyte injury induced by hyperlipidemia via targeting the mTOR/NF-κB/NLRP3 axis. Evid Based Complement Alternat Med. 2020;2020:2735714. doi:10.1155/2020/2735714

77. Liu BH, Tu Y, Ni GX, et al. Total flavones of abelmoschus manihot ameliorates podocyte pyroptosis and injury in high glucose conditions by targeting METTL3-dependent m6a modification-mediated NLRP3-inflammasome activation and PTEN/PI3K/Akt signaling. Front Pharmacol. 2021;12:667644. doi:10.3389/fphar.2021.667644

78. Song Y, Yang L, Guo R, et al. Long noncoding RNA MALAT1 promotes high glucose-induced human endothelial cells pyroptosis by affecting NLRP3 expression through competitively binding miR-22. Biochem Biophys Res Commun. 2019;509(2):359–366. doi:10.1016/j.bbrc.2018.12.139

79. Gu J, Huang W, Zhang W, et al. Sodium butyrate alleviates high-glucose-induced renal glomerular endothelial cells damage via inhibiting pyroptosis. Int Immunopharmacol. 2019;75:105832. doi:10.1016/j.intimp.2019.105832

80. Du Y, Feng Y, Cai Y, et al. CircLARP1B promotes pyroptosis of high glucose-induced renal mesangial cells by regulating the miR-578/TLR4 axis. Int Urol Nephrol. 2024;56(1):283–293. doi:10.1007/s11255-023-03672-4

81. Xie Y, J E, Cai H, et al. Reticulon-1A mediates diabetic kidney disease progression through endoplasmic reticulum-mitochondrial contacts in tubular epithelial cells. Kidney Int. 2022;102(2):293–306. doi:10.1016/j.kint.2022.02.038

82. Eleftheriadis T, Pissas G, Golfinopoulos S, et al. Routes of albumin overload toxicity in renal tubular epithelial cells. Int J Mol Sci. 2023;24(11):9640. doi:10.3390/ijms24119640

83. El-Lateef AEA, El-Shemi AGA, Alhammady MS, et al. LncRNA NEAT2 modulates pyroptosis of renal tubular cells induced by high glucose in Diabetic Nephropathy (DN) by via miR-206 regulation. Biochem Genet. 2022;60(5):1733–1747. doi:10.1007/s10528-021-10164-6

84. Zhu B, Cheng X, Jiang Y, et al. Silencing of KCNQ1OT1 decreases oxidative stress and pyroptosis of renal tubular epithelial cells. Diabetes Metab Syndr Obes. 2020;13:365–375. doi:10.2147/DMSO.S225791

85. Wei Z, Nie G, Yang F, et al. Inhibition of ROS/NLRP3/Caspase-1 mediated pyroptosis attenuates cadmium-induced apoptosis in duck renal tubular epithelial cells. Environ Pollut. 2020;273:115919. doi:10.1016/j.envpol.2020.115919

86. Sun Y, Guo LQ, Wang DG, et al. Metformin alleviates glucolipotoxicity-induced pancreatic β cell ferroptosis through regulation of the GPX4/ACSL4 axis. Eur J Pharmacol. 2023;956:175967. doi:10.1016/j.ejphar.2023.175967

87. Li X, Li Z, Dong X, et al. Astragaloside IV attenuates myocardial dysfunction in diabetic cardiomyopathy rats through downregulation of CD36-mediated ferroptosis. Phytother Res. 2023;37(7):3042–3056. doi:10.1002/ptr.7798

88. Fang X, Song J, Chen Y, et al. LncRNA SNHG1 knockdown inhibits hyperglycemia induced ferroptosis via miR-16-5p/ACSL4 axis to alleviate diabetic nephropathy. J Diabetes Investig. 2023;14(9):1056–1069. doi:10.1111/jdi.14036

89. Li S, Lu S, Wang L, et al. Effects of amygdalin on ferroptosis and oxidative stress in diabetic retinopathy progression via the NRF2/ARE signaling pathway. Exp Eye Res. 2023;234:109569. doi:10.1016/j.exer.2023.109569

90. Tsai YC, Kuo MC, Huang JC, et al. Single-cell transcriptomic profiles in the pathophysiology within the microenvironment of early diabetic kidney disease. Cell Death Dis. 2023;14(7):442. doi:10.1038/s41419-023-05947-1

91. Wei M, Liu X, Tan Z, et al. Ferroptosis: a new strategy for Chinese herbal medicine treatment of diabetic nephropathy. Front Endocrinol. 2023;14:1188003. doi:10.3389/fendo.2023.1188003

92. Zhang Q, Hu Y, Hu JE, et al. Sp1-mediated upregulation of Prdx6 expression prevents podocyte injury in diabetic nephropathy via mitigation of oxidative stress and ferroptosis. Life Sci. 2021;278:119529. doi:10.1016/j.lfs.2021.119529

93. Cao M, Li Y, Famurewa AC, et al. Antidiabetic and nephroprotective effects of polysaccharide extract from the seaweed Caulerpa racemosa in high fructose-streptozotocin induced diabetic nephropathy. Diabetes Metab Syndr Obes. 2021;14:2121–2131. doi:10.2147/DMSO.S302748

94. Wu WY, Wang ZX, Li TS, et al. SSBP1 drives high fructose-induced glomerular podocyte ferroptosis via activating DNA-PK/p53 pathway. Redox Biol. 2022;52:102303. doi:10.1016/j.redox.2022.102303

95. Chen J, Ou Z, Gao T, et al. Ginkgolide B alleviates oxidative stress and ferroptosis by inhibiting GPX4 ubiquitination to improve diabetic nephropathy. Biomed Pharmacother. 2022;156:113953. doi:10.1016/j.biopha.2022.113953

96. Jin J, Wang Y, Zheng D, et al. A novel identified circular RNA, mmu_mmu_circRNA_0000309, involves in germacrone-mediated improvement of diabetic nephropathy through regulating ferroptosis by targeting miR-188-3p/GPX4 signaling axis. Antioxid Redox Signal. 2022;36(10–12):740–759. doi:10.1089/ars.2021.0063

97. Rong J, Li C, Zhang Q, et al. Hydroxysafflor yellow A inhibits endothelial cell ferroptosis in diabetic atherosclerosis mice by regulating miR-429/SLC7A11. Pharm Biol. 2023;61(1):404–415. doi:10.1080/13880209.2023.2225543

98. Cui S, Liu X, Liu Y, et al. Autophagosomes defeat ferroptosis by decreasing generation and increasing discharge of free Fe2+ in skin repair cells to accelerate diabetic wound healing. Adv Sci. 2023;10(25):e2300414. doi:10.1002/advs.202300414

99. Bai T, Li M, Liu Y, et al. The promotion action of AURKA on post-ischemic angiogenesis in diabetes-related limb ischemia. Mol Med. 2023;29(1):39. doi:10.1186/s10020-023-00635-4

100. Wang X, Li Q, Sui B, et al. Schisandrin A from schisandra chinensis attenuates ferroptosis and NLRP3 inflammasome-mediated pyroptosis in diabetic nephropathy through mitochondrial damage by AdipoR1 ubiquitination. Oxid Med Cell Longev. 2022;2022:5411462. doi:10.1155/2022/5411462

101. Luo EF, Li HX, Qin YH, et al. Role of ferroptosis in the process of diabetes-induced endothelial dysfunction. World J Diabetes. 2021;12(2):124–137. doi:10.4239/wjd.v12.i2.124

102. Wu Y, Zhao Y, Yang HZ, et al. HMGB1 regulates ferroptosis through Nrf2 pathway in mesangial cells in response to high glucose. Biosci Rep. 2021;41(2):BSR20202924. doi:10.1042/BSR20202924

103. Wu J, Shao X, Shen J, et al. Downregulation of PPARα mediates FABP1 expression, contributing to IgA nephropathy by stimulating ferroptosis in human mesangial cells. Int J Biol Sci. 2022;18(14):5438–5458. doi:10.7150/ijbs.74675

104. Shen S, Ji C, Wei K. Cellular senescence and regulated cell death of tubular epithelial cells in diabetic kidney disease. Front Endocrinol. 2022;13:924299. doi:10.3389/fendo.2022.924299

105. Han Y, Su Y, Han M, et al. Ginsenoside Rg1 attenuates glomerular fibrosis by inhibiting CD36/TRPC6/NFAT2 signaling in type 2 diabetes mellitus mice. J Ethnopharmacol. 2023;302(Pt A):115923. doi:10.1016/j.jep.2022.115923

106. Yang S, Pei T, Wang L, et al. Salidroside alleviates renal fibrosis in SAMP8 mice by inhibiting ferroptosis. Molecules. 2022;27(22):8039. doi:10.3390/molecules27228039

107. Tan H, Chen J, Li Y, et al. Glabridin, a bioactive component of licorice, ameliorates diabetic nephropathy by regulating ferroptosis and the VEGF/Akt/ERK pathways. Mol Med. 2022;28(1):58. doi:10.1186/s10020-022-00481-w

108. Li Q, Meng X, Hua Q. Circ ASAP2 decreased inflammation and ferroptosis in diabetic nephropathy through SOX2/SLC7A11 by miR-770-5p. Acta Diabetol. 2023;60(1):29–42. doi:10.1007/s00592-022-01961-5

Creative Commons License © 2024 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.