The Role of Autophagy in Diabetic Kidney Disease: Emerging Insights and Therapeutic Potential

  1. Home
  2. Articles

The Role of Autophagy in Diabetic Kidney Disease: Emerging Insights and Therapeutic Potential

   

Sadaf Ghaderzadeh, PhD1,2*

1Division of Endocrinology and Metabolism, Department of Medicine, Georgetown University, Washington, DC 20057, USA

2Division of Endocrinology and Metabolism, Department of Medicine, Howard University College of Medicine, 520 W St, NW, Washington, DC 20059, USA

*Corresponding author: Sadaf Ghaderzadeh, PhD, Division of Endocrinology and Metabolism, Department of Medicine, Georgetown University, Washington, DC 20057, USA

Citation: Ghaderzadeh S. The Role of Autophagy in Diabetic Kidney Disease: Emerging Insights and Therapeutic Potential. J Clin Pract Med Case Rep. 1(2):1-4.

Received: June 22, 2024 | Published: July 10, 2024.

Copyright© 2024 genesis pub by Ghaderzadeh S.. CC BY-NC-ND 4.0 DEED. This is an open-access article distributed under the terms of the Creative Commons Attribution-Non Commercial-No Derivatives 4.0  International License. This allows others distribute, remix, tweak, and build upon the work, even commercially, as long as they credit the authors for the original creation.

Introduction

Diabetic Kidney Disease (DKD) is a common and severe complication of diabetes mellitus, contributing significantly to the burden of end-stage renal disease (ESRD) worldwide. The pathogenesis of DKD is complex, involving hyperglycemia-induced oxidative stress, inflammation, and fibrosis. These processes collectively lead to progressive renal damage, characterized by albuminuria, glomerulosclerosis, and ultimately, a decline in renal function. Despite advances in diabetes management and renal care, DKD remains a leading cause of morbidity and mortality among diabetic patients, highlighting the need for novel therapeutic approaches.

Recent research has identified autophagy as a critical cellular process implicated in the maintenance of renal homeostasis and the pathophysiology of DKD. Autophagy is a conserved lysosomal degradation pathway that removes damaged organelles, misfolded proteins, and other intracellular debris, thereby maintaining cellular integrity and function. It plays a crucial role in responding to cellular stress, preventing the accumulation of toxic materials, and promoting cellular survival. Dysregulation of autophagy has been linked to various kidney diseases, including DKD, where impaired autophagic flux can exacerbate renal injury.

Emerging evidence suggests that enhancing autophagy could provide protective effects against DKD by mitigating hyperglycemia-induced cellular stress and inflammation. This editorial explores the current understanding of autophagy in DKD, its regulation in renal cells, and the potential therapeutic strategies targeting autophagy for the treatment of DKD. By delving into the mechanisms by which autophagy influences renal pathology, we aim to shed light on the promising avenues for future research and clinical interventions.

Current Understanding of Autophagy in DKD

Autophagy is a cellular recycling mechanism that degrades and recycles damaged organelles, proteins, and other macromolecules to maintain cellular homeostasis. It plays a crucial role in renal physiology by preventing the accumulation of damaged cellular components and mitigating stress responses. In DKD, autophagy appears to have a dual role, potentially protecting against renal damage while its deregulation may exacerbate disease progression.

Autophagy in Renal Cells

  • Podocytes: Podocytes are specialized cells in the glomerulus that play a vital role in maintaining the filtration barrier. Autophagy in podocytes is essential for their survival and function. Studies have shown that impaired autophagy in podocytes leads to their loss and contribute to glomerulosclerosis, a hallmark of DKD. For instance, a study demonstrated that podocyte-specific knockout of autophagy-related genes in mice resulted in proteinuria and glomerular injury, mimicking features of human DKD [1].
  • Proximal Tubular Epithelial Cells: Proximal tubular cells are crucial for reabsorption and secretion functions in the kidney. Autophagy in these cells helps to remove damaged mitochondria and proteins, thus preventing tubular injury. Research indicates that high glucose levels and other diabetic conditions impair autophagy in proximal tubular cells, leading to cellular apoptosis and fibrosis [1]. Enhancing autophagy in these cells has shown protective effects against diabetic-induced tubular damage [2].

Mechanisms Regulating Autophagy in DKD

The regulation of autophagy involves several signaling pathways that are often disrupted in diabetes. Key regulators include:

  • mTOR Pathway: The mammalian target of rapamycin (mTOR) is a central inhibitor of autophagy. Hyperglycemia and insulin resistance, common in diabetes, activate the mTOR pathway, thus inhibiting autophagy. Pharmacological inhibition of mTOR using agents like rapamycin has been shown to restore autophagy and protect against DKD in experimental models [3]
  • AMPK Pathway: AMP-activated protein kinase (AMPK) is an energy sensor that promotes autophagy. In diabetic conditions, reduced AMPK activity has been linked to decreased autophagy. Activators of AMPK, such as metformin, have demonstrated potential in enhancing autophagy and offering renal protection in DKD [4].
  • SIRT1 Pathway: Sirtuin 1 (SIRT1) is a NAD+-dependent deacetylase that promotes autophagy. Its expression is reduced in diabetic kidneys. Activation of SIRT1 has been shown to induce autophagy and ameliorate renal damage in diabetic models [5].

Therapeutic Implications and Future Direction

Given the protective role of autophagy in renal cells, targeting autophagy presents a promising therapeutic strategy for DKD. Several potential approaches include:

Pharmacological agents:

  • Rapamycin: An mTOR inhibitor that enhances autophagy. Studies have shown that rapamycin treatment reduces proteinuria and glomerular injury in diabetic mice [6].
  • Metformin: An AMPK activator widely used as an anti-diabetic drug. It has been reported to enhance autophagy and protect against renal fibrosis in DKD models [7].
  • Resveratrol: A natural compound that activates SIRT1, promoting autophagy. Resveratrol administration has shown beneficial effects in reducing renal inflammation and fibrosis in diabetes [8].

Gene Therapy: Gene therapy approaches to enhance the expression of autophagy-related genes in renal cells are being explored. For instance, overexpression of Atg5 in podocytes has been shown to prevent diabetic nephropathy in mouse models [9].

Lifestyle Interventions

  • Dietary Restriction: Caloric restriction and intermittent fasting are known to induce autophagy. These interventions have shown potential in reducing renal injury and improving kidney function in diabetic conditions [10].

Exercise: Regular physical activity enhances autophagy through the activation of AMPK and other pathways. Exercise has been shown to improve renal outcomes in diabetic patients [11].

Challenges and Considerations

While enhancing autophagy presents a promising therapeutic avenue, several challenges and considerations must be addressed:

  • Selective Targeting: Enhancing autophagy systemically may have unintended effects on other organs. Developing targeted delivery systems to the kidneys is crucial.
  • Stage-Specific Interventions: The role of autophagy may vary at different stages of DKD. Interventions need to be tailored based on the disease stage to maximize benefits and minimize potential adverse effects.
  • Clinical Trials: Rigorous clinical trials are needed to evaluate the safety and efficacy of autophagy-enhancing therapies in DKD patients. Current evidence is primarily based on preclinical studies.

Conclusion

Autophagy plays a vital role in maintaining renal homeostasis, and its dysregulation is a key contributor to the pathogenesis of DKD. Targeting autophagy offers a novel and promising therapeutic strategy for DKD. Continued research into the mechanisms regulating autophagy and the development of targeted therapies is essential to combat this debilitating disease and improve outcomes for diabetic patients.

References

  1. Ding Y, Choi ME. (2015) Autophagy in diabetic nephropathy. J Endocrinol. 224(1):R15-30.
  2. Yang D, Livingston MJ, Liu Z, Dong G, Zhang M, et.al (2018) Autophagy in diabetic kidney disease: regulation, mechanisms and therapeutic potential. Cell Mol Life Sci. 75(4):669-88.
  3. Gödel M, Hartleben B, Herbach N, Liu S, Zschiedrich S, et al (2011) Role of mTOR in podocyte function and diabetic nephropathy in humans and mice. J Clin Invest. 121(6):2197-209.
  4. Lee SH, Kim KR, Kim JH, Moon MK, Chung J, et al (2012) Metformin ameliorates diabetic nephropathy through the suppression of renal fibrosis and inflammation via AMPK and the activation of Nrf2.Exp Diabetes Res. 2012:438596.
  5. Zhangn J, Xiang H, Liu J, Chen Y, He RR, (2018) SIRT1 Deacetylates RIP1 and Protects Neurons against Ischemic Stroke-Induced Cell Death. Oxid Med Cell Longev. 2018: 9457326
  6. Lee SH, Kim KR, Kim JH, Moon MK, Chung JH, et al (2012) Metformin ameliorates diabetic nephropathy through the suppression of renal
  7. Das L, Vinayak M, (2015) Long term effect of curcumin down regulates expression of TNF-α and iNOS in liver of lymphoma bearing mice. Med Chem. 11(1):52-58.
  8. Wang S, Zhao Z, Feng X, Cheng H, Zhou X, (2017) Autophagy-related gene Atg5 protects against podocyte injury in diabetic nephropathy. Sci Rep. 7: 14664.
  9. Gödel M, Hartleben B, Herbach N, Liu S, Zschiedrich S, et al (2011) Role of mTOR in podocyte function and diabetic nephropathy in humans and mice. J Clin Invest. 121(6):2197-209.
  10. Wang S, Zhao Z, Feng X, Cheng H, Zhou X. Autophagy-related gene Atg5 protects against podocyte injury in diabetic nephropathy. Scientific Reports. 2017;7:14664.
  11. Das L, Vinayak M, (2015) Long term effect of curcumin down regulates expression of TNF-α and iNOS in liver of lymphoma bearing mice. Med Chem. 11(1):52-58.
whatsapp