A NOVEL ANTIOXIDANT-DAPAGLIFLOZIN COMBINATION: COMBATING DIABETIC KIDNEY DISEASE AT THE CELLULAR LEVEL
HTML Full TextA NOVEL ANTIOXIDANT-DAPAGLIFLOZIN COMBINATION: COMBATING DIABETIC KIDNEY DISEASE AT THE CELLULAR LEVEL
B. Dharani * and A. Suba
A. C. S. Medical College and Hospital, Dr. M. G. R. Educational and Research Institute, Chennai, Tamil Nadu, India.
ABSTRACT: At present, the significant cause of chronic kidney disease (CKD) is said to be Diabetic Kidney Disease (DKD). This condition considerably impacts cost of healthcare, mortality and morbidity worldwide. Current treatment options for DKD basically focusses on Blood Pressure (BP) regulation, glycemic control along with suppression of Renin-Angiotensin-Aldosterone System (RAAS). The advancing nature of pathophysiology of DKD demands additional treatment options to reduce the ongoing chronic inflammation and oxidative stress as they are the key operators of renal dysfunction. In the current review, we put forward a novel, rationally designed antioxidant-Dapagliflozin combination as a single tablet formulation incorporating N-Acetyl Cysteine (NAC), Taurine, Alpha-Lipoic Acid (ALA) and Coenzyme Q10 (CoQ10) with Dapagliflozin to boost renoprotection in DKD. In this review, we have explored the pathophysiological rationale underlying this novel drug combination, its mechanism, synergistic potential, efficacy in preventing development of DKD at the cellular level and challenges in drug formulation. This is the first article to propose this novel combination by incorporating SGLT-2 inhibitor with antioxidants based on scientific rationale to target multidimensional cellular mechanism in DKD progression. By addressing challenges in the formulation through innovative pharmaceutical techniques and rigorous clinical approval, this formulation might have a great success in clinical practice as it has an immense potential to improve patient outcome by reducing the stress of polypharmacy while simultaneously preventing the onset and progression of DKD in diabetic patients.
Keywords: Dapagliflozin, Diabetic Kidney Disease, Diabetic Nephropathy, Antioxidant, Novel drug, Alpha-Lipoic Acid, Coenzyme Q10, N-Acetyl Cysteine, Taurine
INTRODUCTION: At present, the significant cause of chronic kidney disease (CKD) is said to be Diabetic Kidney Disease (DKD). This condition considerably impacts cost of healthcare, mortality and morbidity worldwide 1. Current treatment options for DKD basically focusses on Blood Pressure (BP) regulation, glycemic control along with suppression of Renin-Angiotensin-Aldosterone System (RAAS) 2. While oral hypoglycemic agents like Dapagliflozin have shown significant nephroprotective effects apart from blood glucose regulation, the advancing nature of pathophysiology of DKD demands additional treatment options to reduce the ongoing chronic inflammation and oxidative stress as they are the key operators of renal dysfunction.
The central pathophysiological mechanism in DKD is oxidative stress that causes endothelial injury, mitochondrial injury and podocyte dysfunction causing tubular and glomerular injury. Nevertheless, using multiple drugs often leads to polypharmacy which potentially decreases treatment compliance, efficacy and quality of life. In the current review, we put forward a novel, rationally designed antioxidant-Dapagliflozin combination as a single tablet formulation incorporating N-Acetyl Cysteine (NAC), Taurine, Alpha-Lipoic Acid (ALA) and Coenzyme Q10 (CoQ10) with Dapagliflozin to boost reno-protection in DKD 3-5. This proposed formulation seeks to combat synergistically the ongoing inflammation, oxidative stress and mitochondrial dysfunction while concurrently harnessing the reno-protective effects of Dapagliflozin. The underlying aim of this combination is to target multiple pathways involved in pathophysiology of DKD with one rationally designed synergistic therapy. Implementing such therapy could decrease the pill burden, improve the patient treatment compliance and ultimately enhance the patient’s quality of life.
In this review, we explore the pathophysiological rationale underlying this novel drug combination, its synergistic potential and efficacy in preventing development of DKD at the cellular level. Also, we will discuss its clinical applications, formulation, pharmaceutical challenges and future directions, in order to convert this proposed formulation into therapeutical practical drug.
METHODOLOGY:
Literature Search: A thorough search of literature was conducted by using various databases like SCOPUS, PubMed, Google Scholar, Cochrane and Web of Science to collect studies related to DKD, nephroprotective drugs and oxidative stress.
The key words used for search are “Diabetes”, “Diabetic Kidney Disease”, “antioxidants”, “Nephroprotective drugs”, “Oxidative stress”, “Alpha-Lipoic Acid”, “N-Acetlycysteine”, “Taurine”, “Coenzyme Q10” and “Dapagliflozin”. Inclusion criteria consist of clinical trial, meta-analysis, peer-reviewed article and experimental studies that are published in English. Those studies with lack of data and poor relevance were excluded.
Criteria for Selection: Studies that are relevant to DKD pathophysiology, effect of oxidative stress and therapeutic efficacy of nephroprotective antioxidant drugs and Dapagliflozin were selected.
Those articles which highlights mechanism, clinical efficacy and safety profile of these drugs were given priority. Studies which provide in-depth mechanistic evidence on renal protection and modulation of oxidative stress were considered.
Approach for Formulation Development: The rationale behind this formulation is combining nephroprotective anti-diabetic drug, Dapagliflozin with nephroprotective antioxidants like NAC, Taurine, CoQ10 and ALA. With this formulation, we aim to target different pathways and cellular mechanism that contribute to development and progression of DKD. Hence, these drugs were also selected based on their capability to reduce the diabetes-related oxidative stress and inflammation.
Extraction and Analysis of Data: The extraction of data was done focussing on key parameters like mitochondrial dysfunction, oxidative stress, renal structural changes and clinical efficacy associated with development and progression of DKD. An evidence-based critical analysis was performed to assess the combined effects these drugs on these factors. All the relevant clinical and experimental studies were reviewed systematically to substantiate rationally this proposed formulation and to assess its therapeutic potential in preventing DKD at cellular level.
Pathophysiology of DKD: Diabetes is the major cause of chronic kidney disease (CKD) worldwide. Diabetic Kidney Disease (DKD) is exemplified by progressive form of renal dysfunction developing into CKD and ultimately to End Stage Renal Disease (ESRD). The Patho-mechanism of DKD is combinatorial and complex, encompassing hyperglycemia-induced inflammation, oxidative stress, endothelial dysfunction, mitochondrial damage, unregulated autophagy and podocyte injury 6.
The key element in pathophysiology of DKD is persistent hyperglycemia. This can lead to a state of glucotoxicity that evokes hemodynamic and metabolic changes, which cause kidney dysfunction. Chronic hyperglycemia enhances the polyol pathway by increasing aldose reductase activity. This causes accumulation of sorbitol leading to osmotic and oxidative stress in renal cells. It also causes increased formation of Advanced Glycation End Products (AGEs) that adhere to RAGE receptors fostering oxidative stress, inflammation and finally fibrosis. This also causes deviant post-translational changes due to enhanced hexosamine biosynthetic pathway causing renal fibrosis.
It could activate Protein Kinase C (PKC) that changes the normal renal hemodynamic, expands endothelial dysfunction and facilitates renal fibrosis 7, 8. The second step is the mitochondrial dysfunction leading to enhanced production of Reactive Oxygen Species (ROS), compromised antioxidant system such as glutathione peroxidase and superoxide dismutase and eventually Endoplasmic Reticulum (ER) stress. The key attribute of DKD is ongoing chronic inflammation in diabetes. The inflammatory pathway involves excess release of pro-inflammatory cytokines like IL-6, TNF-α and IL-1β and induction of NLRP3 inflammasome which further facilitate the inflammation. Ultimately, this leads to renal tissue infiltration by macrophages, mediated by TGF- β1 and connective tissue growth factor (CTGF). This intensifies tubulointerstitial fibrosis 9.
The marked hemodynamic alteration is glomerular hyperfiltration caused by high intraglomerular pressure due to dilation of afferent arteriole and constriction of efferent arteriole. It also causes defective nitric oxide (NO) production and enhanced levels of endothelin-1 (ET-1) levels. This leads to vasodilation further causing tissue hypoxia and damage. In addition, Angiotensin II causes glomerular hypertension. The chronic oxidative stress causes cytoskeletal disruption, diminution of Podocin and nephrin and dysregulation of autophagy and apoptosis. These effects lead to disrupted filtration barrier, detachment of podocyte which eventually leads to glomerular damage causing Proteinuria 10. The cumulative effect of these combined alterations like pro-inflammatory cytokines, tubular atrophy and proteinuria-induced fibrotic pathways is tubulointerstitial damage. Due to chronic hyperglycemia, the normal autophagy flux is decreased leading to buildup of damaged proteins and organelles. This induces pro-inflammatory secretions which further exacerbates the kidney damage. Apart from these mechanisms, there occurs epigenetic modifications like histone alterations, DNA methylation and alterations in MicroRNAs (miRNAs), that causes progression of inflammatory and fibrotic pathways in DKD 11.
Studies have shown that dysbiosis of gut microbiota causes further activation of systemic inflammation due to defective production of Short-Chain Fatty Acid (SCFA) that aggravates DKD 12.
FIG. 1: PATHOPHYSIOLOGY OF DKD
Proposed Combination of Dapagliflozin-Antioxidants: The rationally designed novel formulation consists of Dapagliflozin with nephropotective antioxidants like NAC, Taurine, ALA and CoQ10. The basis of this formulation is to target specifically different cellular mechanisms related to oxidative stress that is involved in development of DKD. Each component of this combination plays a crucial role in alleviating diabetes-related oxidative stress, enhancing mitochondrial activity and thereby safeguarding renal function and structure.
Dapagliflozin: Dapagliflozin is a nephroprotective anti-diabetic drug. It inhibits Sodium-Glucose co-Transporter-2 (SGLT2), thereby reduces reabsorption of glucose in the proximal tubule of kidney. This decreases chronic kidney damage caused by hyperglycemia in diabetes. This helps to attain adequate glycemic control, mitigates glomerular hyperfiltration. Additionally, it decreases chronic inflammation and oxidative stress related to progression of DKD. Also, this effect has been connected to enhanced mitochondrial efficiency and autophagy that are important in maintaining renal cell homeostasis 13-18.
Alpha-Lipoic Acid (ALA): ALA is said to be a dithiol compound that occurs naturally with potent anti-inflammatory and anti-oxidant capacity. It has a direct ROS scavenging property and helps in regeneration of other vital antioxidants like vitamin C and Glutathione. It also improves the mitochondrial function, hinders lipid peroxidation and decreases formation of AGE. All of which together promotes renal protection from oxidative stress in diabetes 19-21.
N-Acetylcysteine (NAC): It acts as an important precursor for synthesis of glutathione which is known as the body’s master antioxidant. It safely decreases the level of oxidative stress, inhibits the release of pro-inflammatory cytokines and reduces apoptosis of renal tubular cells. Also, NAC has proven to alleviate renal fibrosis by decreasing the activation of TGF-β which is said to be the key driver of DKD progression 22-24.
Taurine: Taurine is a sufur-containing amino acid that plays a crucial role in osmoregulation of cells and defense against oxidative stress. It shields against development of diabetic nephropathy by steadying mitochondrial membranes, alleviating oxidative damage and thereby mitigating inflammation. It also balances NO availability by which enhancing the function of endothelium and renal microcirculation 25.
Coenzyme Q10 (CoQ10): CoQ10 is an essential component in the process of ATP production as it belongs to mitochondrial electron transport chain. Hence, it plays critical role in energy metabolism of cells. It executes a dual role in prevention of DKD by enhancing the mitochondrial energy production, thereby hold the role of potential lipid-soluble antioxidant. It can also alleviate the podocyte injury, decrease proteinuria and improve the renal cells function in DKD 26.
FIG. 2: PROPOSED COMBINATION OF DAPAGLIFLOZIN AND ANTIOXIDANTS
Mechanism of Action of Dapagliflozin-Antioxidants Combination: DKA is basically fuelled by chronic hyperglycemia in diabetes, that causes various metabolic and hemodynamic modifications. This leads to chronic oxidative stress, mitochondrial insufficiency, chronic inflammation and ultimately ending in renal fibrosis. The current study formulates a novel combination of Dapagliflozin, ALA, NAC, Taurine and CoQ10 in such a way that this combination targets specifically the pathological pathways, thereby prevents the progression of DKD synergistically.
Reduction of Hyperglycemia-Induced Oxidative Stress and Mitochondrial Dysfunction: In diabetes, uncontrolled hyperglycemia increases the polyol pathway, causing excess accumulation of sorbitol and high generation of mitochondrial ROS. Taurine, NAC, ALA and CoQ10 are efficient antioxidants that scavenges these excess ROS. Also, they improve the endogenous antioxidant enzymes, by which they help to decrease ER stress and mitochondrial injury. Addition of Dapagliflozin to this antioxidant combination can enhance their effect by its glucose lowering effect, thereby reducing mitochondrial overload induced oxidative stress 27-49.
Protection against Glomerular Injury and Endothelial Dysfunction: Chronic hyperglycemia induces AGE-RAGE signaling and PKC, which leads to glomerular hypertension and endothelial dysfunction.ALA and CoQ10 enhance the mitochondrial function and decreased AGE-related oxidative stress. Taurine prevents glomerular injury by controlling CTGF and TGF-β pathways. Addition of Dapagliflozin with this combination might help to preserve nephrin, protect expression of podocin, safegaurd podocytes and thereby maintains the glomerular filtration barrier 50-64.
Anti-inflammatory and Anti-fibrogenic Effects: Diabetes is a chronic condition characterized by chronic inflammation. It is marked by increased activity of inflammatory cytokines like IL-1β, IL-6, TNF-α and activation of NLRP3 inflammasome which accelerates the process of renal fibrosis. NAC, ALA and Taurine are antioxidants that alleviates the inflammation by supressing these inflammatory cytokines and thereby curbing fibrotic pathways. Addition of Dapagliflozin with CoQ10 helps to regulate the inflammatory signalling, thereby decreasing the mesangial expansion and alleviates interstitial fibrosis 65-86.
Protection against Tubular Hypoxia and Dysfunction: The progression of DKD is characterized by impaired autophagy, tubular apoptosis and proteinuria-driven tubular toxicity. Taurine and NAC improve the detoxification mechanisms of cells, thereby alleviating the oxidative damage of renal tubules. Addition of Dapagliflozin along with CoQ10, reduces the peritubular capillary depletion, maintaining renal oxygenation and slowing the process of hypoxia-induced tubular damage 87-97.
Gut-Kidney Axis and Epigenetic Modulation: Diabetes is also characterized by SCFA deficiency and gut dysbiosis, further aggravating the ongoing systemic inflammation and renal insult. CoQ10 and ALA are capable of influencing miRNA expression and DNA methylation, thereby protecting against persistent renal injury. NAC and Taurine helps to regain the gut microbiota balance, thereby decreasing the inflammatory overload on kidneys 98-107.
Hence, this combination consisting of Dapagliflozin, ALA, NAC, CoQ10 and Taurine offers a multidimensional nephroprotection that addresses the underlying inflammation, oxidative load, mitochondrial injury and fibrosis at the cellular level. This novel, rationally-designed treatment strategy provides a synergistic effect and offers a promising effect to halt the development and progression of DKD, thereby prevents ESRD.
TABLE 1: EVIDENCE-BASED CRITICAL ANALYSIS OF THE FORMULATION
S. no. | Mechanism of action | Drugs | Individual effects | References |
1. | Reduction of hyperglycemia-induced oxidative stress and mitochondrial dysfunction | Dapagliflozin, ALA, Taurine, NAC, CoQ10 | Dapagliflozin: glucose lowering effect, thereby reducing mitochondrial overload- induced oxidative stress | 27-31 |
ALA: reduce ROS, improves antioxidant enzymes | 32-34 | |||
NAC: mitigates ER stress and mitochondrial injury | 35-39 | |||
Taurine: improves mitochondrial function and decrease oxidative load | 40-44 | |||
CoQ10: enhances mitochondrial bioenergetics and prevents oxidative injury | 45-49 | |||
2. | Protection against glomerular injury and endothelial dysfunction | Dapagliflozin, ALA, CoQ 10, Taurine | Dapagliflozin: maintains podocin and nephrin expression amd preserves glomerular filtration barrier. | 50,51 |
CoQ10: Alleviates AGE-related oxidative injury and enhances mitochondrial function |
52-54 |
|||
ALA: Supresses oxidative stress and enhances endothelial function | 55-62 | |||
Taurine: Modulate CTGF and TGF-β and prevents glomerular injury. | 63,64 | |||
3. | Anti-inflammatory and anti-fibrogenic effects | Dapagliflozin, ALA, NAC, CoQ10, Taurine | Dapagliflozin: Controls inflammatory signalling and decreases interstitial fibrosis | 65-70 |
ALA: reduces pro-inflammatory cytokines | 71-74 | |||
NAC: supresses fibrosis-associated pathways | 75-80 | |||
Taurine: Decreases NLRP3activation | 81-84 | |||
CoQ10: reduces mesangial expansion and fibrosis | 85,86 | |||
4. | Protection against tubular hypoxia and dysfunction | Dapagliflozin, Taurine, CoQ10, NAC | Dapagliflozin: Reduces peritubular capillary depletion and slows down the hypoxia-induced damage | 87-89 |
NAC: improves cellular detoxification and protects renal tubules | 90,91 | |||
Taurine: Decreases proteinuria associated renal toxicity | 92-94 | |||
CoQ10: Preserves renal oxygenation and decreases oxidative stress | 95-97 | |||
5. | Gut-Kidney Axis and Epigenetic modulation | NAC, ALA, Taurine, CoQ10 | ALA: controls DNA methylation and prevents underlying renal injury | 98-100 |
CoQ10: modulates miRNA expression and decreases inflammation | 101,102 | |||
Taurine: Regains gut microbiota balance and decreases systemic inflammation | 103-105 | |||
NAC: preserves the healthy gut-kidney axis | 106,107 |
FIG. 3: MECHANISM OF DAPAGLIFLOZIN-ANTIOXIDANT COMBINATION IN COMBATING DKD
Figure Legend: This figure depicts the multidimensional pathophysiology of DKD, highlighting the key pathways such as inflammatory, metabolic and hemodynamic pathways that are involved in development and progression of DKD. Hyperglycemia activated metabolic alterations causes mitochondrial dysfunction, oxidative stress, endothelial dysfunction and glomerular injury, that contribute to podocyte damage, proteinuria and ultimately renal fibrosis. The complex interplay of AGE-RAGE signalling, activation of polyol pathway, excess mitochondrial ROS formation and release of inflammatory cytokines exacerbates renal dysfunction. The synergistic effect of this novel combination (Dapagliflozin, NAC, Taurine, ALA and CoQ10) is shown by targeting various pathological pathways to reduce oxidative stress, chronic inflammation and prevent fibrosis, thereby aiming prevent DKD development and progression.
Challenges: These are few challenges in development of a combination consisting of Dapagliflozin with nephroprotective antioxidants namely NAC, Taurine, CoQ10 and ALA as a single-tablet formulation. This includes pharmaceutical compatibility, optimal drug release profile, absorption and bioavailability, dosing, manufacturing possibility, adverse effects, drug regulatory approval, cost and accessibility. These challenges must be addressed effectively for a successful clinical validation and practical formulation.
Future Perspectives: The future directions in this formulation could be incorporating innovative drug delivery technologies, fixed-ratio dosing strategies, integrating other anti-fibrotic and anti-diabetic drugs, large-scale studies to prove its efficacy, scalability and commercialization.
CONCLUSION: The present study on a rationally-designed single tablet formulation, by combining nephroprotective anti-diabetic drug, Dapagliflozin with nephroprotective antioxidants provides an innovative and novel strategy to prevent DKD at cellular level. This article underscores the evidence-based potential of this combination to mitigate the oxidative stress and mitochondrial dysfunction, which are the key driving factors for development and progression of DKD along with achieving adequate glycemic control. This is the first article to propose this novel combination by incorporating SGLT-2 inhibitor with antioxidants based on scientific rationale to target multidimensional cellular mechanism in DKD progression. This points its strength and credibility for a successful formulation. If this formulation is clinically validated and successfully developed, it has an immense potential to create a prominent impact in the diabetic population by enhancing the renal outcomes, improving the treatment compliance and thereby improving the overall quality of life. Also, it has a greater potential to pave way for formulation of similar drugs in future. By addressing challenges in the formulation through innovative pharmaceutical techniques and rigorous clinical approval, this formulation might have a great success in clinical practice as it has an immense potential to improve patient outcome by reducing the stress of polypharmacy while simultaneously preventing the onset and progression of DKD in diabetic patients.
ACKNOWLEDGEMENT: The authors sincerely acknowledge and express their gratitude to all the scientists and researchers whose pioneering work in the fields of SGLT2 inhibitors, antioxidants and diabetic kidney disease has laid the foundation for this novel drug formulation. Their invaluable contributions to scientific literature and innovation have been instrumental in shaping the rationale behind this combination therapy.
Author Contribution: Dr. B. Dharani conceptualized the study, conducted the literature review, and drafted the manuscript. Dr. Suba. A contributed to data analysis, manuscript revision and final approval. All authors have read the manuscript and approved its final version.
Funding: No specific grant from any funding agency in the public, commercial, or not-for-profit sectors was obtained for this study.
CONFLICT OF INTEREST: The authors declare no conflicts of interest.
REFERENCES:
- Folkerts K, Petruski-Ivleva N, Kelly A, Fried L, Blankenburg M, Gay A and Kovesdy CP: Annual health care resource utilization and cost among type 2 diabetes patients with newly recognized chronic kidney disease within a large U.S. administrative claims database. J Manag Care Spec Pharm 2020; 26: 1506–16. https://doi.org/10.18553/jmcp.2020.26.12.1506.
- Hoogeveen EK: The epidemiology of diabetic kidney disease. Kidney Dial 2022; 2: 433–42. https://doi.org/10.3390/kidneydial2030038.
- Manojkumar SM, Chandrashekhar DU, Aman BU and Vishal SG: Renoprotective effect of co-enzyme Q10 and N-acetylcysteine on streptozotocin-induced diabetic nephropathy in rats. Int J Diabetes Clin Res 2020; 7. https://doi.org/10.23937/2377-3634/1410123.
- Golbidi S, Badran M and Laher I: Diabetes and alpha lipoic Acid. Front Pharmacol 2011; 2: 69. https://doi.org/10.3389/fphar.2011.00069.
- Tippairote T, Bjørklund G, Gasmi A, Semenova Y, Peana M, Chirumbolo S and Hangan T: Combined supplementation of coenzyme Q10 and other nutrients in specific medical conditions. Nutrients 2022; 14: 4383. https://doi.org/10.3390/nu14204383
- Jha R, Lopez-Trevino S, Kankanamalage HR and Jha JC: Diabetes and renal complications: An overview on pathophysiology, biomarkers and therapeutic interventions. Biomedicines 2024; 12: 1098. https://doi.org/10.3390/biomedicines12051098.
- Rout P and Jialal I: Diabetic nephropathy. StatPearls, Treasure Island (FL): StatPearls Publishing 2025.
- Agarwal R: Pathogenesis of Diabetic Nephropathy. Compendia 2021; 2021: 2–7. https://doi.org/10.2337/db20211-2.
- Mora-Fernández C, Domínguez-Pimentel V, de Fuentes MM, Górriz JL, Martínez-Castelao A and Navarro-González JF: Diabetic kidney disease: from physiology to therapeutics. J Physiol 2014; 592: 3997–4012. https://doi.org/10.1113/jphysiol.2014.272328.
- Wang N and Zhang C: Oxidative stress: A culprit in the progression of diabetic kidney disease. Antioxidants (Basel) 2024; 13: 455. https://doi.org/10.3390/antiox13040455.
- Akhouri V, Majumder S and Gaikwad AB: Targeting DNA methylation in diabetic kidney disease: A new perspective. Life Sci 2023; 335: 122256. https://doi.org/10.1016/j.lfs.2023.122256
- Lin JR, Wang ZT, Sun JJ, Yang YY, Li XX, Wang XR, Shi Y, Zhu YY, Wang RT, Wang MN, Xie FY, Wei P and Liao ZH: Gut microbiota and diabetic kidney diseases: Pathogenesis and therapeutic perspectives. World J Diabetes 2022; 13: 308–18. https://doi.org/10.4239/wjd.v13.i4.308.
- Heerspink HJL, Stefánsson BV, Correa-Rotter R, Chertow GM, Greene T, Hou FF, Mann JFE, McMurray JJV, Lindberg M, Rossing P, Sjöström CD, Toto RD, Langkilde AM and Wheeler DC: Dapagliflozin in patients with chronic kidney disease. N Engl J Med 2020; 383: 1436–46. https://doi.org/10.1056/NEJMoa2024816.
- Yang H, Mei Z, Chen W, Pan Y, Liu L, Zhao R, Ni W, Wang Y and Fei C: Therapeutic efficacy of dapagliflozin on diabetic kidney disease in rats. Int Immunopharmacol 2022; 113: 109272. https://doi.org/10.1016/j.intimp.2022.109272.
- Wheeler DC, Stefansson BV, Batiushin M, Bilchenko O, Cherney DZI, Chertow GM, Douthat W, Dwyer JP, Escudero E, Pecoits-Filho R, Furuland H, Górriz JL, Greene T, Haller H, Hou FF, Kang S-W, Isidto R, Khullar D, Mark PB, McMurray JJV, Kashihara N, Nowicki M, Persson F, Correa-Rotter R, Rossing P, Toto RD, Umanath K, Van Bui P, Wittmann I, Lindberg M, Sjöström CD, Langkilde AM and Heerspink HJL: The dapagliflozin and prevention of adverse outcomes in chronic kidney disease (DAPA-CKD) trial: baseline characteristics. Nephrol Dial Transplant 2020; 35: 1700–11. https://doi.org/10.1093/ndt/gfaa234.
- Chertow GM, Correa-Rotter R, Vart P, Jongs N, McMurray JJV, Rossing P, Langkilde AM, Sjöström CD, Toto RD, Wheeler DC and Heerspink HJL: Effects of dapagliflozin in Chronic Kidney Disease, with and without other cardiovascular medications: DAPA-CKD trial. J Am Heart Assoc 2023; 12: 028739. https://doi.org/10.1161/JAHA.122.028739.
- Heerspink HJL, Jongs N, Chertow GM, Langkilde AM, McMurray JJV, Correa-Rotter R, Rossing P, Sjöström CD, Stefansson BV, Toto RD, Wheeler DC and Greene T: Effect of dapagliflozin on the rate of decline in kidney function in patients with chronic kidney disease with and without type 2 diabetes: a prespecified analysis from the DAPA-CKD trial. Lancet Diabetes Endocrinol 2021; 9: 743–54. https://doi.org/10.1016/S2213-8587(21)00242-4.
- Cai A, Shen J, Yang X, Shao X, Gu L, Mou S and Che X: Dapagliflozin alleviates renal inflammation and protects against diabetic kidney diseases, both dependent and independent of blood glucose levels. Front Immunol 2023; 14: 1205834. https://doi.org/10.3389/fimmu.2023.1205834
- Bell DSH and Jerkins T: The potential for improved outcomes in the prevention and therapy of diabetic kidney disease through “stacking” of drugs from different classes. Diabetes Obes Metab 2024; 26: 2046–53. https://doi.org/10.1111/dom.15559.
- Dugbartey GJ, Alornyo KK, Diaba DE and Adams I: Activation of renal CSE/H2S pathway by alpha-lipoic acid protects against histological and functional changes in the diabetic kidney. Biomed Pharmacother 2022; 153: 113386. https://doi.org/10.1016/j.biopha.2022.113386.
- Dugbartey GJ, Alornyo KK, N’guessan BB, Atule S, Mensah SD and Adjei S: Supplementation of conventional anti-diabetic therapy with alpha-lipoic acid prevents early development and progression of diabetic nephropathy. Biomed Pharmacother 2022; 149: 112818. https://doi.org/10.1016/j.biopha.2022.112818
- Hernández-Cruz EY, Aparicio-Trejo OE, Hammami FA, Bar-Shalom D, Tepel M, Pedraza-Chaverri J and Scholze A: N-acetylcysteine in kidney disease: Molecular mechanisms, pharmacokinetics, and clinical effectiveness. Kidney Int Rep 2024; 9: 2883–903. https://doi.org/10.1016/j.ekir.2024.07.020.
- Ye M, Lin W, Zheng J and Lin S: N-acetylcysteine for chronic kidney disease: a systematic review and meta-analysis. Am J Transl Res 2021; 13: 2472–85.
- Chiu AH, Wang CJ, Lin YL, Wang CL and Chiang TI: N-acetylcysteine alleviates the progression of chronic kidney disease: A three-year cohort study. Medicina (Kaunas) 2023; 59: 1983. https://doi.org/10.3390/medicina59111983.
- Ahmed K, Choi HN and Yim JE: The impact of taurine on obesity-induced diabetes mellitus: Mechanisms underlying its effect. Endocrinol Metab (Seoul) 2023; 38: 482–92. https://doi.org/10.3803/EnM.2023.1776.
- Samimi F, Namiranian N, Sharifi-Rigi A, Siri M, Abazari O and Dastghaib S: Coenzyme Q10: A key antioxidant in the management of diabetes-induced cardiovascular complications-an overview of mechanisms and clinical evidence. Int J Endocrinol 2024; 2024: 2247748. https://doi.org/10.1155/2024/2247748
- Albarrán OG, Ampudia-Blasco FJ: Dapagliflozin, the first SGLT-2 inhibitor in the treatment of type 2 diabetes. Med Clin (Barc) 2013; 141 Suppl 2: 36–43. https://doi.org/10.1016/S0025-7753(13)70062-9.
- Veelen A, Andriessen C, Op den Kamp Y, Erazo-Tapia E, de Ligt M, Mevenkamp J, örgensen JA, Moonen-Kornips E, Schaart G, Esterline R, Havekes B, Oscarsson J, Schrauwen-Hinderling VB, Phielix E and Schrauwen P: Effects of the sodium-glucose cotransporter 2 inhibitor dapagliflozin on substrate metabolism in prediabetic insulin resistant individuals: A randomized, double-blind crossover trial. Metabolism 2023; 140: 155396. https://doi.org/10.1016/j.metabol.2022.155396.
- Oraby MA, El-Yamany MF, Safar MM, Assaf N and Ghoneim HA: Dapagliflozin attenuates early markers of diabetic nephropathy in fructose-streptozotocin-induced diabetes in rats. Biomed Pharmacother 2019; 109: 910–20. https://doi.org/10.1016/j.biopha.2018.10.100.
- Zaibi N, Li P and Xu SZ: Protective effects of dapagliflozin against oxidative stress-induced cell injury in human proximal tubular cells. PLoS One 2021; 16: 0247234. https://doi.org/10.1371/journal.pone.0247234.
- Eleftheriadis T, Pissas G, Filippidis G, Efthymiadi M, Liakopoulos V and Stefanidis I: Dapagliflozin prevents high-glucose-induced cellular senescence in renal tubular epithelial cells. Int J Mol Sci 2022; 23: 16107. https://doi.org/10.3390/ijms232416107.
- Ashok A, Andrabi SS, Mansoor S, Kuang Y, Kwon BK and Labhasetwar V: Antioxidant therapy in oxidative stress-induced neurodegenerative diseases: Role of nanoparticle-based drug delivery systems in clinical translation. Antioxidants (Basel) 2022; 11: 408. https://doi.org/10.3390/antiox11020408.
- Liu J, Han X, Zhang T, Tian K, Li Z and Luo F: Reactive oxygen species (ROS) scavenging biomaterials for anti-inflammatory diseases: from mechanism to therapy. J Hematol Oncol 2023; 16: 116. https://doi.org/10.1186/s13045-023-01512-7.
- Jena AB, Samal RR, Bhol NK and Duttaroy AK: Cellular Red-Ox system in health and disease: The latest update. Biomed Pharmacother 2023; 162: 114606. https://doi.org/10.1016/j.biopha.2023.114606
- Zhang B, Li M, Yang W, Loor JJ, Liang Y, Wang S, Zhao Y, Guo H, Ma X, Yu L and Xu C: Mitochondrial dysfunction and endoplasmic reticulum stress in calf hepatocytes are associated with fatty acid-induced ORAI calcium release-activated calcium modulator 1 signaling. J Dairy Sci 2020; 103: 11945–56. https://doi.org/10.3168/jds.2020-18684.
- Sun Y: N-acetylcysteine attenuates reactive-oxygen-species-mediated endoplasmic reticulum stress during liver ischemia-reperfusion injury. World J Gastroenterol 2014; 20: 15289. https://doi.org/10.3748/wjg.v20.i41.15289.
- Ali M, Tabassum H, Alam MM and Parvez S: N-acetyl-L-cysteine ameliorates mitochondrial dysfunction in ischemia/reperfusion injury via attenuating Drp-1 mediated mitochondrial autophagy. Life Sci 2022; 293: 120338. https://doi.org/10.1016/j.lfs.2022.120338.
- Zhou J, Terluk MR, Orchard PJ, Cloyd JC and Kartha RV: N-acetylcysteine reverses the mitochondrial dysfunction induced by very long-chain fatty acids in Murine oligodendrocyte model of adrenoleukodystrophy. Biomedicines 2021; 9: 1826. https://doi.org/10.3390/biomedicines9121826.
- Wright DJ, Renoir T, Smith ZM, Frazier AE, Francis PS, Thorburn DR, McGee SL, Hannan AJ and Gray LJ: N-Acetylcysteine improves mitochondrial function and ameliorates behavioral deficits in the R6/1 mouse model of Huntington’s disease. Transl Psychiatry 2015; 5: 492–e492. https://doi.org/10.1038/tp.2014.131
- Hansen SH, Andersen ML, Cornett C, Gradinaru R, Grunnet N: A role for taurine in mitochondrial function. J Biomed Sci 2010; 17(1): 23. https://doi.org/10.1186/1423-0127-17-S1-S23.
- Seneff S and Kyriakopoulos AM: Taurine prevents mitochondrial dysfunction and protects mitochondria from reactive oxygen species and deuterium toxicity. Amino Acids 2025; 57: 6. https://doi.org/10.1007/s00726-024-03440-3.
- Jong CJ, Sandal P and Schaffer SW: The role of taurine in mitochondria health: More than just an antioxidant. Molecules 2021; 26: 4913. https://doi.org/10.3390/molecules26164913.
- Surai PF, Earle-Payne K and Kidd MT: Taurine as a natural antioxidant: From direct antioxidant effects to protective action in various toxicological models. Antioxidants (Basel) 2021; 10: 1876. https://doi.org/10.3390/antiox10121876.
- Schaffer S and Kim HW: Effects and mechanisms of taurine as a therapeutic agent. Biomol Ther (Seoul) 2018; 26: 225–41. https://doi.org/10.4062/biomolther.2017.251
- Noh YH, Kim KY, Shim MS, Choi SH, Choi S, Ellisman MH, Weinreb RN, Perkins GA and Ju WK: Inhibition of oxidative stress by coenzyme Q10 increases mitochondrial mass and improves bioenergetic function in optic nerve head astrocytes. Cell Death Dis 2013; 4: 820. https://doi.org/10.1038/cddis.2013.341.
- Suárez-Rivero JM, Pastor-Maldonado CJ, Povea-Cabello S, Álvarez-Córdoba M, Villalón-García I, Munuera-Cabeza M, Suárez-Carrillo A, Talaverón-Rey M and Sánchez-Alcázar JA: Coenzyme Q10 analogues: Benefits and challenges for therapeutics. Antioxidants (Basel) 2021; 10: 236. https://doi.org/10.3390/antiox10020236.
- Luo K, Yu JH, Quan Y, Shin YJ, Lee KE, Kim HL, Ko EJ, Chung BH, Lim SW and Yang CW: Therapeutic potential of coenzyme Q10 in mitochondrial dysfunction during tacrolimus-induced beta cell injury. Sci Rep 2019; 9: 7995. https://doi.org/10.1038/s41598-019-44475-x.
- Dasgupta A and Klein K: Herbal and other dietary supplements that are antioxidants. Antioxidants in Food, Vitamins and Supplements, Elsevier 2014; 295–315.
- Marcheggiani F, Cirilli I, Orlando P, Silvestri S, Vogelsang A, Knott A, Blatt T, Weise JM and Tiano L: Modulation of Coenzyme Q10 content and oxidative status in human dermal fibroblasts using HMG-CoA reductase inhibitor over a broad range of concentrations. From mitohormesis to mitochondrial dysfunction and accelerated aging. Aging (Albany NY) 2019; 11: 2565–82. https://doi.org/10.18632/aging.101926
- Cassis P, Locatelli M, Cerullo D, Corna D, Buelli S, Zanchi C, Villa S, Morigi M, Remuzzi G, Benigni A and Zoja C: SGLT2 inhibitor dapagliflozin limits podocyte damage in proteinuric nondiabetic nephropathy. JCI Insight 2018; 3. https://doi.org/10.1172/jci.insight.98720.
- Chen X, Wang J, Lin Y, Liu Y and Zhou T: Signaling pathways of podocyte injury in diabetic kidney disease and the effect of sodium-glucose cotransporter 2 inhibitors. Cells 2022; 11: 3913. https://doi.org/10.3390/cells11233913
- Sood B, Patel P and Keenaghan M: Coenzyme Q10. StatPearls, Treasure Island (FL): StatPearls Publishing; 2025.
- Hernández-Camacho JD, Bernier M, López-Lluch G, Navas P: Coenzyme Q10 supplementation in aging and disease. Front Physiol 2018; 9: 44. https://doi.org/10.3389/fphys.2018.00044.
- Barcelos IP de, Haas RH: CoQ10 and aging. Biology (Basel) 2019; 8: 28. https://doi.org/10.3390/biology8020028
- Badran M, Abuyassin B, Golbidi S, Ayas N and Laher I: Alpha lipoic acid improves endothelial function and oxidative stress in mice exposed to chronic intermittent hypoxia. Oxid Med Cell Longev 2019; 2019: 4093018. https://doi.org/10.1155/2019/4093018.
- Wollin SD and Jones PJH: Alpha-lipoic acid and cardiovascular disease. J Nutr 2003; 133: 3327–30. https://doi.org/10.1093/jn/133.11.3327.
- Hajizadeh-Sharafabad F and Sharifi Zahabi E: Role of alpha-lipoic acid in vascular function: A systematic review of human intervention studies. Crit Rev Food Sci Nutr 2022; 62: 2928–41. https://doi.org/10.1080/10408398.2020.1861425.
- Wang W, An LP, Li YF, An R, Bian Z, Liu WZ, Song Q-H and Li AY: Alpha-lipoic acid ameliorates H2O2-induced human vein endothelial cells injury via suppression of inflammation and oxidative stress. Biosci Biotechnol Biochem 2020; 84: 2253–63. https://doi.org/10.1080/09168451.2020.1802221.
- Tromba L, Perla FM, Carbotta G, Chiesa C and Pacifico L: Effect of alpha-lipoic acid supplementation on endothelial function and cardiovascular risk factors in overweight/obese youths: A double-blind, placebo-controlled randomized trial. Nutrients 2019; 11: 375. https://doi.org/10.3390/nu11020375.
- Sharman JE, Gunaruwan P, Knez WL, Schmitt M, Marsh SA, Wilson GR, Cockcroft JR and Coombes JS: Alpha-lipoic acid does not acutely affect resistance and conduit artery function or oxidative stress in healthy men. Br J Clin Pharmacol 2004; 58: 243–8. https://doi.org/10.1111/j.1365-2125.2004.02146.x.
- Skibska B and Goraca A: The protective effect of lipoic acid on selected cardiovascular diseases caused by age-related oxidative stress. Oxid Med Cell Longev 2015; 2015: 313021. https://doi.org/10.1155/2015/313021.
- Superti F and Russo R: Alpha-lipoic acid: Biological mechanisms and health benefits. Antioxidants (Basel) 2024; 13: 1228. https://doi.org/10.3390/antiox13101228
- Ural C, Celik A, Ozbal S, Guneli E, Arslan S, Ergur BU, Cavdar C, Akdoğan G and Cavdar Z: The renoprotective effects of taurine against diabetic nephropathy via the p38 MAPK and TGF-β/Smad2/3 signaling pathways. Amino Acids 2023; 55: 1665–77. https://doi.org/10.1007/s00726-023-03342-w.
- Yuan LQ, Lu Y, Luo XH, Xie H, Wu XP and Liao EY: Taurine promotes connective tissue growth factor (CTGF) expression in osteoblasts through the ERK signal pathway. Amino Acids 2007; 32: 425–30. https://doi.org/10.1007/s00726-006-0380-4
- Liu Y, Wang Y, Chen S, Bai L, Xie X, Zhang L and Wang X: Investigation into the effect and mechanism of dapagliflozin against renal interstitial fibrosis based on transcriptome and network pharmacology. Int Immunopharmacol 2022; 112: 109195. https://doi.org/10.1016/j.intimp.2022.109195.
- Zeng J, Huang H, Zhang Y, Lv X, Cheng J, Zou SJ, Han Y, Wang S, Gong L and Peng Z: Dapagliflozin alleviates renal fibrosis in a mouse model of adenine-induced renal injury by inhibiting TGF-β1/MAPK mediated mitochondrial damage. Front Pharmacol 2023; 14: 1095487. https://doi.org/10.3389/fphar.2023.1095487.
- Xuan MY, Piao SG, Ding J, Nan QY, Piao MH, Jiang YJ, Zheng HL, Jin JZ and Li C: Dapagliflozin alleviates renal fibrosis by inhibiting RIP1-RIP3-MLKL-mediated necroinflammation in unilateral ureteral obstruction. Front Pharmacol 2021; 12: 798381. https://doi.org/10.3389/fphar.2021.798381.
- Quagliariello V, Canale ML, Bisceglia I, Iovine M, Paccone A, Maurea C, Scherillo M, Merola A, Giordano V, Palma G, Luciano A, Bruzzese F, Zito Marino F, Montella M, Franco R, Berretta M, Gabrielli D, Gallucci G and Maurea N: Sodium-glucose cotransporter 2 inhibitor dapagliflozin prevents ejection fraction reduction, reduces myocardial and renal NF-κB expression and systemic pro-inflammatory biomarkers in models of short-term doxorubicin cardiotoxicity. Front Cardiovasc Med 2024; 11: 1289663. https://doi.org/10.3389/fcvm.2024.1289663.
- Feng L, Chen Y, Li N, Yang X, Zhou L, Li H, Wang T, Xie M and Liu H: Dapagliflozin delays renal fibrosis in diabetic kidney disease by inhibiting YAP/TAZ activation. Life Sci 2023; 322: 121671. https://doi.org/10.1016/j.lfs.2023.121671.
- Ke Q, Shi C, Lv Y, Wang L, Luo J, Jiang L, Yang J and Zhou Y: SGLT2 inhibitor counteracts NLRP3 inflammasome via tubular metabolite itaconate in fibrosis kidney. FASEB J 2022; 36: 22078. https://doi.org/10.1096/fj.202100909RR
- Dharani B, Sebastian S and Suba: Alpha-Lipoic Acid in type 2 Diabetes Mellitus: Mechanisms, clinical benefits, and implementation in therapy. Asian J Pharm Clin Res 2024; 16–21. https://doi.org/10.22159/ajpcr.2024v17i12.51657.
- Zhao G, Etherton TD, Martin KR, Gillies PJ, West SG and Kris-Etherton PM: Dietary alpha-linolenic acid inhibits proinflammatory cytokine production by peripheral blood mononuclear cells in hypercholesterolemic subjects. Am J Clin Nutr 2007; 85: 385–91. https://doi.org/10.1093/ajcn/85.2.385.
- Schuerwegh AJ, Dombrecht EJ, Stevens WJ, Van Offel JF, Bridts CH and De Clerck LS: Influence of pro-inflammatory (IL-1 alpha, IL-6, TNF-alpha, IFN-gamma) and anti-inflammatory (IL-4) cytokines on chondrocyte function. Osteoarthritis Cartilage 2003; 11: 681–7. https://doi.org/10.1016/s1063-4584(03)00156-0.
- Altunina NV, Lizogub VG and Bondarchuk OM: Alpha-lipoic acid as a means of influence on systemic inflammation in type 2 diabetes mellitus patients with prior myocardial infarction. J Med Life 2020; 13: 32–6. https://doi.org/10.25122/jml-2020-0018
- Liu N, Li G, Guan Y, Wang R, Ma Z, Zhao L and Yao S: N-acetylcysteine alleviates pulmonary alveolar proteinosis induced by indium-tin oxide nanoparticles in male rats: involvement of the NF-κB signaling pathway. Ecotoxicol Environ Saf 2022; 241: 113812. https://doi.org/10.1016/j.ecoenv.2022.113812.
- Li S, Yang X, Li W, Li J, Su X, Chen L and Yan G: N-acetylcysteine downregulation of lysyl oxidase activity alleviating bleomycin-induced pulmonary fibrosis in rats. Respiration 2012; 84: 509–17. https://doi.org/10.1159/000340041.
- Ryu C-M, Shin JH, Yu HY, Ju H, Kim S, Lim J, Heo J, Lee S, Shin DM and Choo MS: N-acetylcysteine prevents bladder tissue fibrosis in a lipopolysaccharide-induced cystitis rat model. Sci Rep 2019; 9: 8134. https://doi.org/10.1038/s41598-019-44631-3.
- Sahasrabudhe SA, Terluk MR and Kartha RV: N-acetylcysteine pharmacology and applications in rare diseases-repurposing an old antioxidant. Antioxidants (Basel) 2023; 12. https://doi.org/10.3390/antiox12071316.
- Maghsadi Z, Azadmehr A, Moghadamnia AA, Feizi F and Hamidi N: N-Acetylcysteine attenuated pulmonary fibrosis induced by bleomycin via immunomodulation responses. Res Pharm Sci 2023; 18: 177–84. https://doi.org/10.4103/1735-5362.367796.
- Rodriguez LR, Bui SN, Beuschel RT, Ellis E, Liberti EM, Chhina MK, Cannon B, Lemma M, Nathan SD and Grant GM: Curcumin induced oxidative stress attenuation by N-acetylcysteine co-treatment: a fibroblast and epithelial cell in-vitro study in idiopathic pulmonary fibrosis. Mol Med 2019; 5: 27. https://doi.org/10.1186/s10020-019-0096-z
- Lee CC, Chen WT, Chen SY and Lee TM: Taurine alleviates sympathetic innervation by inhibiting NLRP3 inflammasome in postinfarcted rats. J Cardiovasc Pharmacol 2021; 77: 745–55. https://doi.org/10.1097/FJC.0000000000001005.
- Swiderski J, Sakkal S, Apostolopoulos V, Zulli A and Gadanec LK: Combination of taurine and black pepper extract as a treatment for cardiovascular and coronary artery diseases. Nutrients 2023; 15: 2562. https://doi.org/10.3390/nu15112562.
- Qiu T, Pei P, Yao X, Jiang L, Wei S, Wang Z, Bai J, Yang G, Gao N, Yang L, Qi S, Yan R, Liu X and Sun X: Taurine attenuates arsenic-induced pyroptosis and nonalcoholic steatohepatitis by inhibiting the autophagic-inflammasomal pathway. Cell Death Dis 2018; 9: 946. https://doi.org/10.1038/s41419-018-1004-0.
- Ouyang G, Wang N, Tong J, Sun W, Yang J and Wu G: Alleviation of taurine on liver injury of type 2 diabetic rats by improving antioxidant and anti-inflammatory capacity. Heliyon 2024; 10: 28400. https://doi.org/10.1016/j.heliyon.2024.e28400
- Maheshwari RA, Sen AK, Balaraman R, Shah NV, Shah UH, Solanki N and Sen DB: Co-administration of Coenzyme Q10 and HMG-CoA Reductase Inhibitor Attenuates Oxidative Stress, TGF-β, TNF-α, Nitrite Content and MPO Levels against Experimentally-induced Diabetic Nephropathy in Rats. Indian Journal of Pharmaceutical Education and Research 2022; 56: 1091–8.
- Quadri MM, Fatima SS, Che RC and Zhang AH: Mitochondria and renal fibrosis. Adv Exp Med Biol 2019; 1165: 501–24. https://doi.org/10.1007/978-981-13-8871-2_25
- Papaetis GS: SGLT2 inhibitors, intrarenal hypoxia and the diabetic kidney: insights into pathophysiological concepts and current evidence. Arch Med Sci Atheroscler Dis 2023; 8: 155–68. https://doi.org/10.5114/amsad/176658.
- Papaetis G: SGLT2 inhibitors and diabetic kidney disease: Targeting multiple and interrelated signaling pathways for renal protection. Curr Mol Pharmacol 2023. https://doi.org/10.2174/0118761429261105231011101200.
- Wang Y, Jin M, Cheng CK and Li Q: Tubular injury in diabetic kidney disease: molecular mechanisms and potential therapeutic perspectives. Front Endocrinol (Lausanne) 2023; 14: 1238927. https://doi.org/10.3389/fendo.2023.1238927
- Nogueira GB, Punaro GR, Oliveira CS, Maciel FR, Fernandes TO, Lima DY, Rodrigues AM, Mouro MG, Araujo SRR and Higa EMS: N-acetylcysteine protects against diabetic nephropathy through control of oxidative and nitrosative stress by recovery of nitric oxide in rats. Nitric Oxide 2018; 78: 22–31. https://doi.org/10.1016/j.niox.2018.05.003.
- Kobroob A, Peerapanyasut W, Kumfu S, Chattipakorn N and Wongmekiat O: Effectiveness of N-acetylcysteine in the treatment of renal deterioration caused by long-term exposure to bisphenol A. Biomolecules 2021; 11: 655. https://doi.org/10.3390/biom11050655
- Koh JH, Lee ES, Hyun M, Kim HM, Choi YJ, Lee EY, Yadav D and Chung CH: Taurine alleviates the progression of diabetic nephropathy in type 2 diabetic rat model. Int J Endocrinol 2014; 2014: 397307. https://doi.org/10.1155/2014/397307.
- Sarkar P, Basak P, Ghosh S, Kundu M and Sil PC: Prophylactic role of taurine and its derivatives against diabetes mellitus and its related complications. Food Chem Toxicol 2017; 110: 109–21. https://doi.org/10.1016/j.fct.2017.10.022.
- Chesney RW, Han X and Patters AB: Taurine and the renal system. J Biomed Sci 2010; 17(1): 4. https://doi.org/10.1186/1423-0127-17-S1-S4
- Persson MF, Franzén S, Catrina S-B, Dallner G, Hansell P, Brismar K and Palm F: Coenzyme Q10 prevents GDP-sensitive mitochondrial uncoupling, glomerular hyperfiltration and proteinuria in kidneys from db/db mice as a model of type 2 diabetes. Diabetologia 2012; 55: 1535–43. https://doi.org/10.1007/s00125-012-2469-5.
- Sun J, Zhu H, Wang X, Gao Q, Li Z and Huang H: CoQ10 ameliorates mitochondrial dysfunction in diabetic nephropathy through mitophagy. J Endocrinol 2019; 240: 445–65. https://doi.org/10.1530/JOE-18-0578.
- Zhang X, Shi Z, Liu Q, Quan H and Cheng X: Effects of coenzyme Q10 intervention on diabetic kidney disease: A systematic review and meta-analysis: A systematic review and meta-analysis. Medicine (Baltimore) 2019; 98: 15850. https://doi.org/10.1097/MD.0000000000015850
- Grdović N, Rajić J, Arambašić Jovanović J, Dinić S, Tolić A, Đorđević M, Trifunović S, Vidaković M, Uskoković A and Mihailović M: Α-Lipoic acid increases collagen synthesis and deposition in nondiabetic and diabetic rat kidneys. Oxid Med Cell Longev 2021; 2021: 6669352. https://doi.org/10.1155/2021/6669352.
- Dinicola S, Proietti S, Cucina A, Bizzarri M and Fuso A: Alpha-lipoic acid downregulates IL-1β and IL-6 by DNA hypermethylation in SK-N-BE neuroblastoma cells. Antioxidants (Basel) 2017; 6. https://doi.org/10.3390/antiox6040074.
- Martinez-Moreno JM, Fontecha-Barriuso M, Martin-Sanchez D, Guerrero-Mauvecin J, Goma-Garces E, Fernandez-Fernandez B, Carriazo S, Sanchez-Niño MD, Ramos AM, Ruiz-Ortega M, Ortiz A, Sanz AB: Epigenetic modifiers as potential therapeutic targets in diabetic kidney disease. Int J Mol Sci 2020; 21: 4113. https://doi.org/10.3390/ijms21114113
- Wang D, Yan X, Xia M, Yang Y, Li D, Li X, Song F and Ling W: Coenzyme Q10 promotes macrophage cholesterol efflux by regulation of the activator protein-1/miR-378/ATP-binding cassette transporter G1-signaling pathway. Arterioscler Thromb Vasc Biol 2014; 34: 1860–70. https://doi.org/10.1161/ATVBAHA.113.302879.
- Schmelzer C, Kitano M, Rimbach G, Niklowitz P, Menke T, Hosoe K and Döring F: Effects of ubiquinol-10 on microRNA-146a expression in-vitro and in-vivo. Mediators Inflamm 2009; 2009: 415437. https://doi.org/10.1155/2009/415437
- Zhao DD, Gai YD, Li C, Fu ZZ, Yin DQ, Xie M, Dai JY, Wang XX, Li YX, Wu GF, Feng Y, Hu JM, Lin SM and Yang JC: Dietary taurine effect on intestinal barrier function, colonic microbiota and metabolites in weanling piglets induced by LPS. Front Microbiol 2023; 14: 1259133. https://doi.org/10.3389/fmicb.2023.1259133.
- Duszka K: Versatile triad alliance: Bile acid, taurine and Microbiota. Cells 2022; 11: 2337. https://doi.org/10.3390/cells11152337.
- Su Q, Pan XF, Li HB, Xiong LX, Bai J, Wang XM, Qu X-Y, Zhang NR, Zou GQ, Shen Y, Li L, Huang LL, Zhang H and Xu ML: Taurine supplementation alleviates blood pressure via gut-brain communication in spontaneously hypertensive rats. Biomedicines 2024; 12. https://doi.org/10.3390/biomedicines12122711
- Hsu CN, Hou CY, Chang-Chien GP, Lin S and Tain YL: Maternal N-acetylcysteine therapy prevents hypertension in spontaneously hypertensive rat offspring: Implications of hydrogen sulfide-generating pathway and gut Microbiota. Antioxidants (Basel) 2020; 9: 856. https://doi.org/10.3390/antiox9090856.
- Shimizu MHM, Gois PHF, Volpini RA, Canale D, Luchi WM, Froeder L, Heilberg IP and Seguro AC: N-acetylcysteine protects against star fruit-induced acute kidney injury. Ren Fail 2017; 39: 193–202. https://doi.org/10.1080/0886022X.2016.1256315
How to cite this article:
Dharani B and Suba A: A novel antioxidant-dapagliflozin combination: combating diabetic kidney disease at the cellular level. Int J Pharm Sci & Res 2025; 16(9): 2442-53. doi: 10.13040/IJPSR.0975-8232.16(9).2442-53.
All © 2025 are reserved by International Journal of Pharmaceutical Sciences and Research. This Journal licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 3.0 Unported License.
Article Information
2
2442-2453
916 KB
575
English
IJPSR
B. Dharani * and A. Suba
A. C. S Medical College and Hospital, Dr. M. G. R. Educational and Research Institute, Chennai, Tamil Nadu, India.
doctordharanibhaskaran@gmail.com
17 March 2025
05 April 2025
22 April 2025
10.13040/IJPSR.0975-8232.16(9).2442-53
01 September 2025