GET THE APP

Recent Advances in the Management of Diabetic Kidney Disease
..

Molecular Biomarkers & Diagnosis

ISSN: 2155-9929

Open Access

Short Communication - (2024) Volume 15, Issue 1

Recent Advances in the Management of Diabetic Kidney Disease

Kesav Bhargava*
*Correspondence: Kesav Bhargava, Department of Nephrology and Microbiology, M. S. Ramaiah University of Applied Sciences, Bengaluru, India, Email:
Department of Nephrology and Microbiology, M. S. Ramaiah University of Applied Sciences, Bengaluru, India

Received: 27-Jan-2024, Manuscript No. jmbd-24-130657; Editor assigned: 30-Jan-2024, Pre QC No. P-130657; Reviewed: 13-Feb-2024, QC No. Q-130657; Revised: 19-Feb-2024, Manuscript No. R-130657; Published: 29-Feb-2024 , DOI: 10.37421/2155-9929.2024.15.630
Citation: Ahmad, Waheed. “Genetic Optimization in Uncovering Biologically Meaningful Gene Biomarkers.” J Mol Biomark Diagn 15 (2024): 629.
Copyright: © 2024 Bhargava K. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Introduction

Diabetic Kidney Disease (DKD) represents a significant complication of diabetes mellitus and is a leading cause of End-Stage Renal Disease (ESRD) globally. It is characterized by progressive renal dysfunction and structural abnormalities, posing a considerable burden on healthcare systems worldwide. Over the years, research has led to significant advances in understanding the pathophysiology of DKD and the development of innovative therapeutic approaches aimed at delaying its progression and improving patient outcomes. This article explores recent breakthroughs in the management of DKD, focusing on novel pharmacotherapies, precision medicine approaches and emerging treatment strategies [1].

Description

DKD is a multifactorial condition influenced by various mechanisms, including hyperglycemia-induced oxidative stress, inflammation, hemodynamic alterations and genetic predisposition. Glomerular hypertrophy, thickening of the glomerular basement membrane and mesangial expansion are common histopathological features observed in DKD. The Renin-Angiotensin- Aldosterone System (RAAS) plays a pivotal role in the pathogenesis of DKD, contributing to glomerular hyperfiltration, hypertension and renal fibrosis. SGLT2 inhibitors represent a novel class of antidiabetic agents that exert their effects by inhibiting renal glucose reabsorption, leading to glycosuria and lowering blood glucose levels. Beyond glycemic control, clinical trials have demonstrated the renoprotective benefits of SGLT2 inhibitors in patients with type 2 diabetes and DKD. Drugs such as empagliflozin, dapagliflozin and canagliflozin have shown significant reductions in albuminuria, preservation of estimated Glomerular Filtration Rate (eGFR) and reduced risk of adverse renal outcomes.

GLP-1 receptor agonists are another class of antidiabetic medications that enhance glucose-dependent insulin secretion, suppress glucagon secretion and promote weight loss. Recent trials have demonstrated the cardiovascular and renal benefits of GLP-1 receptor agonists in patients with type 2 diabetes, including those with DKD. Drugs such as liraglutide, semaglutide and dulaglutide have shown reductions in albuminuria and slowed progression of renal decline, independent of glycemic control. MRAs, such as spironolactone and eplerenone, have emerged as promising therapeutic options for DKD due to their ability to antagonize the effects of aldosterone, thereby reducing proteinuria, inflammation and fibrosis. Recent trials, including the BEACON trial, have demonstrated the efficacy of MRAs in reducing albuminuria and preserving renal function in patients with DKD, particularly in those with resistant hypertension and persistent albuminuria despite RAAS blockade.

Advancements in precision medicine have revolutionized the management of DKD by enabling tailored therapeutic interventions based on individual patient characteristics, including genetic predisposition, biomarker profiles and comorbidities. Genome-Wide Association Studies (GWAS) have identified several genetic variants associated with DKD susceptibility, providing insights into its underlying pathogenesis and potential targets for therapeutic intervention. Additionally, biomarkers such as urinary Kidney Injury Molecule-1 (KIM-1), Neutrophil Gelatinase-Associated Lipocalin (NGAL) and soluble urokinase Plasminogen Activator Receptor (suPAR) hold promise for early detection of DKD progression and risk stratification. Chronic low-grade inflammation plays a crucial role in the pathogenesis of DKD, contributing to endothelial dysfunction, glomerular injury and fibrosis. Emerging therapies targeting inflammatory pathways, such as Interleukin-1β (IL-1β) inhibitors, Tumor Necrosis Factor-α (TNF-α) inhibitors and Janus Kinase (JAK) inhibitors, have shown potential in preclinical and early clinical studies for mitigating renal inflammation and slowing DKD progression.

Epigenetic modifications, including DNA methylation, histone acetylation and microRNA dysregulation, have been implicated in the pathogenesis of DKD. Targeting epigenetic regulators holds promise for modulating gene expression patterns associated with renal fibrosis, inflammation and oxidative stress. Histone Deacetylase (HDAC) inhibitors, DNA methyltransferase inhibitors and microRNA-based therapies represent emerging treatment modalities that warrant further investigation in DKD [2-5].

Conclusion

Recent advances in the management of DKD have expanded therapeutic options and improved outcomes for patients with diabetes-associated renal complications. Novel pharmacotherapies targeting SGLT2 inhibitors, GLP- 1 receptor agonists and MRAs have demonstrated renoprotective benefits beyond glycemic control. Precision medicine approaches based on genetic profiling and biomarker assessment enable personalized treatment strategies tailored to individual patient needs. Emerging treatment strategies focusing on anti-inflammatory therapies and epigenetic modulation hold promise for further enhancing renal outcomes in DKD. Continued research efforts aimed at unraveling the complex pathophysiology of DKD and identifying novel therapeutic targets are essential for addressing the growing global burden of diabetic kidney disease.

Acknowledgement

None.

Conflict of Interest

There are no conflicts of interest by author.

References

  1. Alicic, Radica Z., Michele T. Rooney and Katherine R. Tuttle. "Diabetic kidney disease: Challenges, progress and possibilities." Clin J Am Soc Nephrol 12 (2017): 2032-2045.

    Google Scholar, Crossref, Indexed at

  2. Sher, Emina Karahmet, Amina DžidiÄ? KriviÄ?, Alma Karahmet and Merima Beca-Zeco, et al. "Novel therapeutical approaches based on neurobiological and genetic strategies for diabetic polyneuropathyâ??A review." Diabetes Metab Syndr: Clin Res Rev (2023): 102901.

    Google Scholar, Crossref, Indexed at

  3. Köttgen, Anna, Shih-Jen Hwang, Martin G. Larson and Jennifer E. Van Eyk, et al. "Uromodulin levels associate with a common UMOD variant and risk for incident CKD." J Am Soc Nephrol 21 (2010): 337-344.

    Google Scholar, Crossref, Indexed at

  4. Freeman, Roy. "Not all neuropathy in diabetes is of diabetic etiology: Differential diagnosis of diabetic neuropathy." Curr Diabetes Rep 9 (2009): 423-431.

    Google Scholar, Crossref, Indexed at

  5. Krishnan, Arun V. and Matthew C. Kiernan. "Uremic neuropathy: Clinical features and new pathophysiological insights." Muscle Nerve 35 (2007): 273-290.

    Google Scholar, Crossref, Indexed at

Google Scholar citation report
Citations: 2054

Molecular Biomarkers & Diagnosis received 2054 citations as per Google Scholar report

Molecular Biomarkers & Diagnosis peer review process verified at publons

Indexed In

 
arrow_upward arrow_upward