Next Article in Journal
PD-L1-Mediated Immunosuppression in Hepatocellular Carcinoma: Relationship with Macrophages Infiltration and Inflammatory Response Activity
Next Article in Special Issue
Oxidative Stress and NRF2/KEAP1/ARE Pathway in Diabetic Kidney Disease (DKD): New Perspectives
Previous Article in Journal
Regenerative Potential of A Bovine ECM-Derived Hydrogel for Biomedical Applications
Previous Article in Special Issue
Role of Arginase-II in Podocyte Injury under Hypoxic Conditions
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Nicotinamide/Streptozotocin Rodent Model of Type 2 Diabetes: Renal Pathophysiology and Redox Imbalance Features

Department of Pharmaceutical Sciences, College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
Biomolecules 2022, 12(9), 1225; https://doi.org/10.3390/biom12091225
Submission received: 5 August 2022 / Revised: 30 August 2022 / Accepted: 31 August 2022 / Published: 2 September 2022
(This article belongs to the Special Issue Redox Imbalance and Mitochondrial Abnormalities in Kidney Disease II)

Abstract

:
Diabetic nephropathy (DN) is a common complication of diabetes mellitus. While there has been a great advance in our understanding of the pathogenesis of DN, no effective managements of this chronic kidney disease are currently available. Therefore, continuing to elucidate the underlying biochemical and molecular mechanisms of DN remains a constant need. In this regard, animal models of diabetes are indispensable tools. This review article highlights a widely used rodent model of non-obese type 2 diabetes induced by nicotinamide (NA) and streptozotocin (STZ). The mechanism underlying diabetes induction by combining the two chemicals involves blunting the toxic effect of STZ by NA so that only a percentage of β cells are destroyed and the remaining viable β cells can still respond to glucose stimulation. This NA-STZ animal model, as a platform for the testing of numerous antidiabetic and renoprotective materials, is also discussed. In comparison with other type 2 diabetic animal models, such as high-fat-diet/STZ models and genetically engineered rodent models, the NA-STZ model is non-obese and is less time-consuming and less expensive to create. Given that this unique model mimics certain pathological features of human DN, this model should continue to find its applications in the field of diabetes research.

1. Introduction

Diabetic nephropathy (DN), also known as diabetic kidney disease (DKD) [1,2,3], is a severe complication of diabetes mellitus [4,5]. Approximately 30% of diabetic patients can develop DN [6,7,8], which is also a chronic kidney disease and can progress to end-stage renal failure [9,10,11]. The hallmarks of DN are kidney hypertrophy [12,13,14], mesangial cell proliferation and mesangial matrix accumulation [15,16,17], glomerulosclerosis, and persistent levels of proteinuria [18,19]. Despite the great advancement in our understanding of the pathogenesis of DN and numerous approaches that have been tested to slow down DN development and progression, no effective therapeutics are currently available for the treatment of DN. This is because the specific mechanisms underlying the pathogenesis of DN are yet to be fully elucidated. Therefore, there are unmet needs in treating or halting DN. In this regard, animal models of diabetes [20], be they genetically engineered or chemically or dietary induced, are indispensable tools in DN research.
In this brief review, I focus on one particular type 2 diabetes animal model, which is created by using nicotinamide (NA, Figure 1A) and streptozotocin (STZ, Figure 1B) [21,22,23]. This non-obese type 2 diabetes animal model was initially established in rats [24] but has since been extended to include mice with modifications of the original protocol. In comparison with the high-fat-diet (HFD)-STZ-induced type 2 diabetes animal models [25,26,27,28] and genetically engineered diabetic animal models, such as db/db mice [29] and zsf1 obese rats [30], the NA/STZ model is time-saving and less expensive. Therefore, this model can be equally used as a platform for not only exploring the pathogenesis of DN but also screening and testing potential antidiabetic agents [31] or renoprotective compounds for their therapeutic effects [21].

2. Mechanisms Underlying NA/STZ Diabetes Induction

Masiello P. et al. initially developed this non-obese type 2 diabetes in rats in 1998 [24]. Since then, this model has been widely used to test a variety of antidiabetic materials for their beneficial effects on diabetes and diabetic complications, including DN. The establishment of this model takes advantage of the contradictory effects of the two chemicals on β cells as STZ is β cell cytotoxic while NAD is globally cytoprotective. Therefore, STZ-induced β cell damage can be blunted by nicotinamide [23]. Consequently, a certain percentage of β cells are viable and respond to glucose stimulation to release insulin [24]. It should be noted that the percentage of β cells that can survive really depends on the doses of the two chemicals. For a fixed dose of STZ, if the NA is too low, there will be no blunting effect from the NA and all β cells can be destroyed by STZ. On the other hand, if the NA is too high, the blunting or protective effects of the NA could be too high. In fact, the blunting or protective effects of the NA could reach 100% and no diabetes would be induced. For NA to play a protective role, NA is often given before STZ administration. Nonetheless, that NA is given shortly after STZ ingestion has also been reported in the literature [32,33,34,35]. In these cases, however, whether there are any differences between NA being given first or STZ being given first in diabetes induction and the severity of kidney injury has not been investigated. Notwithstanding, based on the observation that NA given immediately after STZ is equally protective [36], any difference should be minimal when the NA is administered right after STZ administration.
STZ is a nitrosourea compound that has a component similar to glucose (Figure 1B) [37]. Hence, STZ is also known as a glucose analog [37]. Because of this structural similarity to glucose, the STZ enters into β cells via the glucose transporter-2 (Glut2) [37] that is abundantly expressed on the β cell surface [38]. Once inside the β cells, the nitrosoamide moiety of STZ can attack DNA and causes DNA alkylation and is thus responsible for STZ genotoxicity and cytotoxicity [23]. STZ-caused DNA damage can activate poly (ADP-ribose) polymerase-1 (PARP-1) that can then repair damaged DNA using NAD+ as a substrate [39]. As a result, NAD+ could be potentially depleted by the activated PARP-1 [23], thereby leading to cell death. When NA is administered prior to STZ administration, the damaging effect of STZ is greatly mitigated. This mitigating effect is thought to be due to two establishments. One is that NA is a direct inhibitor of PARP-1 [23], the other is that NA is a precursor of NAD+ [23]. Hence, the STZ cytotoxic effects can be greatly blunted by NA and the blunting magnitude is known to be NA-concentration-dependent [23]. In rats, although different investigators would use a different dosage combination of NA and STZ, the initially established dosages of NA and STZ (230 mg/kg and 65 mg/kg, respectively) still seem to prevail in the literature (Figure 2), though the application of lower concentrations of NA and STZ has been reported. In mice, the ingested concentrations of the two chemicals also vary widely. Nevertheless, it should be noted that the concentration of STZ for mice can be higher than that for rats. It appears that the use of 240 mg/kg NA and 100 mg/kg STZ in a mouse model is a prevailing approach in the literature [40]. It should also be noted that when a mouse is used as a model, multiple daily injections of NA and STZ (up to a week) may be conducted [41]. Certain investigators have also reported using high-fat-diet (HFD) feeding followed by NA and STZ ingestions [40]. Regardless of whether rats or mice are being modeled, the key point is that a given investigator should stick to their own protocol of NA and STZ administrations, such as the dosages and routes of chemical ingestions [21,23], so that a reproducibility and data comparison could potentially be achieved. It should also be noted that the severity of the diabetic disorders depends on how long the animals are kept after diabetes induction by the two chemicals. In the absence of any interventions, diabetic disorders will progress, mimicking various stages of clinical practice in humans.
Additionally, based on a modified mouse model of type 2 diabetes induced by combining the HFD-NA-STZ treatments [42], investigations of DN created by HFD feeding in conjunction with NA and STZ administrations have also been reported [43,44]. It should be noted that when an HFD is involved, the creation of such a model would take longer than when only NA and STZ are used.

3. Renal Pathophysiology in this NA/STZ Animal Model

It has been reported that when diabetes was induced in mice by injection of 230 mg/kg NAD along with 50 mg/kg or 65 mg/kg STZ, the kidney organ index (kidney weight vs. body weight) for both STZ doses showed an increase when compared with that of the controls [45]. For a six-week duration of testing, urinary and serum parameters, such as creatinine, urea, and uric acid, were enhanced in the NA-STZ diabetic animals. In the presence of NAD, mice lived longer than those that received only STZ administration [45]. Such a result further demonstrates the blunting effects of NA on STZ cytotoxicity.
It should be noted that while DN can be classified into five stages (Table 1), none of the NA-STZ-involved animal studies published so far have systematically addressed the five stages of diabetic kidney injury. Therefore, future studies on the progression of DN from stage 1 to stage 5 in the NA-STZ animal model need to be conducted. Moreover, numerous diabetic kidney injury biomarkers, such as those recently reported by Pelle et al. [46] and Natesan et al. [11], have also not been systematically and comprehensively evaluated in this NA-STZ animal model. Most studies use popular kidney injury parameters [47] such as blood urea nitrogen (BUN), serum cystatin C, creatinine, uric acid, or/and estimated glomerular flow rate (eGFR) for the evaluation of diabetic kidney injury after NA-STZ injections. The histopathological staining of kidney is also frequently used for the analysis of kidney injury in this NA-STZ animal model. Figure 3 and Figure 4 [48,49] show a typical staining of the kidney tissues by the periodic acid–Schiff and Masson trichrome, respectively. As can be observed from these histochemical stainings, the pathophysiological changes are obvious in the NA-STZ diabetic kidneys. Table 2 summarizes the renal pathophysiological measurements in the NA-STZ diabetic animal models in the absence of any interventions.

4. Application of this Model in DN Research

As mentioned above, this NA-STZ diabetes animal model is a non-obese type 2 diabetes model [23]. The pathogenesis of diabetes in this model may be different from that of HFD/STZ or genetically engineered models, such as the db/db mouse model and the zsf1 obese rat model [30,94,95,96]. Nevertheless, the NA-STZ model may provide a unique platform for the study of non-obese diabetes and diabetic complications [43,97]. With respect to DN research, this model has been widely used for testing the therapeutic effects of numerous antidiabetic or renoprotective materials (Table 3). Most of these materials are natural products derived from plants, such as herbs, trees, teas, and vegetables. Table 3 lists the representative materials that exhibit renoprotective effects in DN in the NA-STZ rodent model of type 2 diabetes. It should be noted that as oxidative stress has been established as one of the major mechanisms underlying DN, many of the listed materials in Table 3 thus have antioxidant properties. Figure 5 summarizes the major mechanisms of the renoprotective materials tested by this NA-STZ animal model. It should also be noted that the renoprotective effects of many of the tested materials are in a dose-dependent manner, such as reported in reference [98] and others.

5. Redox-Related Mechanisms That Remain to Be Elucidated in this NA-STZ Model

The non-tissue-specific mechanisms involved in cellular injury are thought to be implicated in the development of diabetic nephropathy [5,50]. These mechanisms, as shown in Figure 6, include the activation of the polyol pathway [105] and protein kinase C signaling, the hexosamine pathway, and the increased formation of the advanced glycation products [5,106]. However, what has been lacking is the underlying pathological mechanisms of DN in this unique NA-STZ model, in particular, redox signaling and the mitochondrial mechanisms of NA-STZ-induced DN. In fact, numerous aspects remain to be investigated in detail. These include mitochondrial redox imbalance [39]; sources of mitochondrial reactive oxygen species [107]; proteomics of mitochondrial protein oxidation [108,109]; mitochondrial abnormalities such as the derangement of mitochondrial metabolic pathways, including the Krebs cycle and electron transport chain [29]; fatty acid oxidation [110,111]; mitochondrial fusion and fission [112,113]; and mitophagy and the mitochondrial unfolded protein response [114,115,116,117,118,119] (Figure 6). The changes in redox signaling during the progression of DN in this animal model also remain to be comprehensively studied. Nephron segment-specific investigations of targeted genes [120,121] as well as the role of epigenetics [122,123] in this DN model also remain to be fully conducted. Delineating the mechanisms of these biological processes in the diabetic kidney may provide comprehensive insights into the underpinnings of DN. Additionally, this model could also provide a platform for testing the therapeutic effects of stem cells and gene therapy on DN [11]. For studying multiple kidney disease-causing risk factors, this model could also be combined with other kidney disease animal models, such as those induced by folic acid [47,124,125,126], cisplatin [127,128], cadmium [129,130,131], lipopolysaccharide [132,133], and hypoxia or ischemia reperfusion [134,135,136,137,138,139,140,141].
Finally, this NA-STZ diabetes animal model may also be used to evaluate any potential renoprotective effects of caloric restriction [142,143,144], intermittent caloric restriction [145,146], exercise [147,148,149,150], and ketone bodies [151,152,153,154], which all have been demonstrated to provide beneficial effects on the kidney in a variety of pathological conditions [155,156]. Indeed, the underlying mechanisms of renoprotection conferred by these approaches in this non-obese type 2 diabetes model remain to be comprehensively elucidated.

6. Summary

The NA-STZ induction of a type 2 diabetic animal model is a useful tool for both studying the mechanisms of DN and screening renoprotective materials for diabetic kidney disease. The model is less time-consuming and less expensive than that created by genetic engineering or high-fat-diet feeding. The establishment of this model is based on the fact that NA can partially protect pancreatic β cells against STZ cytotoxicity, leading to the incomplete destruction of β cells and thus development of non-insulin-dependent type 2 diabetes mellitus [21,23,24]. This unique animal model should continue to serve as a utility for studying the non-obese type 2 diabetes that is highly prevalent in East Asian diabetic patients [157].

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Lodhi, A.H.; Ahmad, F.-U.; Furwa, K.; Madni, A. Role of oxidative stress and reduced endogenous hydrogen sulfide in diabetic nephropathy. Drug Des. Dev. Ther. 2021, 15, 1031–1043. [Google Scholar]
  2. Ji, J.; Tao, P.; Wang, Q.; Li, L.; Xu, Y. Sirt1: Mechanism and protective effect in diabetic nephropathy. Endocr. Metab. Immune Disord.-Drug Targets 2021, 21, 835–842. [Google Scholar]
  3. Zoja, C.; Xinaris, C.; Macconi, D. Diabetic nephropathy: Novel molecular mechanisms and therapeutic targets. Front. Pharmacol. 2020, 11, 586892. [Google Scholar]
  4. Chowdhury, T.A.; Ali, O. Diabetes and the kidney. Clin. Med. Lond. 2021, 21, e318–e322. [Google Scholar]
  5. Yan, L.J. Nadh/nad (+) redox imbalance and diabetic kidney disease. Biomolecules 2021, 11, 730. [Google Scholar]
  6. Nakhoul, F.; Abassi, Z.; Morgan, M.; Sussan, S.; Mirsky, N. Inhibition of diabetic nephropathy in rats by an oral antidiabetic material extracted from yeast. J. Am. Soc. Nephrol. 2006, 17, S127–S131. [Google Scholar]
  7. Machado, D.I.; Silva, E.D.O.; Ventura, S.; Vattimo, M.D.F.F. The effect of curcumin on renal ischemia/reperfusion injury in diabetic rats. Nutrients 2022, 14, 2798. [Google Scholar]
  8. Hernandez, L.F.; Eguchi, N.; Whaley, D.; Alexander, M.; Tantisattamo, E.; Ichii, H. Anti-Oxidative therapy in diabetic nephropathy. Front. Biosci. 2022, 14, 14. [Google Scholar]
  9. Eboh, C.; Chowdhury, T.A. Management of diabetic renal disease. Ann. Transl. Med. 2015, 3, 154. [Google Scholar]
  10. Sheng, X.; Dong, Y.; Cheng, D.; Wang, N.; Guo, Y. Efficacy and safety of bailing capsules in the treatment of type 2 diabetic nephropathy: A meta-Analysis. Ann. Palliat. Med. 2020, 9, 3885–3898. [Google Scholar]
  11. Natesan, V.; Kim, S.J. Diabetic nephropathy—A review of risk factors, progression, mechanism, and dietary management. Biomol. Ther. 2021, 29, 365–372. [Google Scholar]
  12. Fabris, B.; Candido, R.; Armini, L.; Fischetti, F.; Calci, M.; Bardelli, M.; Fazio, M.; Campanacci, L.; Carretta, R. Control of glomerular hyperfiltration and renal hypertrophy by an angiotensin converting enzyme inhibitor prevents the progression of renal damage in hypertensive diabetic rats. J. Hypertens. 1999, 17, 1925–1931. [Google Scholar]
  13. Li, Z.; Guo, H.; Li, J.; Ma, T.; Zhou, S.; Zhang, Z.; Miao, L.; Cai, L. Sulforaphane prevents type 2 diabetes-Induced nephropathy via ampk-Mediated activation of lipid metabolic pathways and nrf2 antioxidative function. Clin. Sci. 2020, 134, 2469–2487. [Google Scholar]
  14. Palygin, O.; Spires, D.; Levchenko, V.; Bohovyk, R.; Fedoriuk, M.; Klemens, C.A.; Sykes, O.; Bukowy, J.D.; Cowley, A.W.; Lazar, J.; et al. Progression of diabetic kidney disease in t2dn rats. Am. J. Physiol. Ren. Physiol. 2019, 317, F1450–F1461. [Google Scholar]
  15. Thomas, H.Y.; Versypt, A.N.F. Pathophysiology of mesangial expansion in diabetic nephropathy: Mesangial structure, glomerular biomechanics, and biochemical signaling and regulation. J. Biol. Eng. 2022, 16, 19. [Google Scholar]
  16. Ma, J.; Zhao, N.; Du, L.; Wang, Y. Downregulation of lncrna neat1 inhibits mouse mesangial cell proliferation, fibrosis, and inflammation but promotes apoptosis in diabetic nephropathy. Int. J. Clin. Exp. Pathol. 2019, 12, 1174–1183. [Google Scholar]
  17. Zang, X.J.; Li, L.; Du, X.; Yang, B.; Mei, C.L. Lncrna tug1 inhibits the proliferation and fibrosis of mesangial cells in diabetic nephropathy via inhibiting the pi3k/akt pathway. Eur. Rev. Med. Pharm. Sci. 2019, 23, 7519–7525. [Google Scholar]
  18. Zheng, S.; Powell, D.W.; Zheng, F.; Kantharidis, P.; Gnudi, L. Diabetic nephropathy: Proteinuria, inflammation, and fibrosis. J. Diabetes Res. 2016, 2016, 5241549. [Google Scholar]
  19. Rai, U.; Kosuru, R.; Prakash, S.; Tiwari, V.; Singh, S. Tetramethylpyrazine alleviates diabetic nephropathy through the activation of akt signalling pathway in rats. Eur. J. Pharmacol. 2019, 865, 172763. [Google Scholar]
  20. Joost, H.-G.; Al-Hasani, H.; Schurmann, A. Animal Models in Diabetes Research; Humana Press: New York, NY, USA, 2012; Volume 933, p. 325. [Google Scholar]
  21. Ghasemi, A.; Khalifi, S.; Jedi, S. Streptozotocin-Nicotinamide-Induced rat model of type 2 diabetes (review). Acta Physiol. Hung. 2014, 101, 408–420. [Google Scholar]
  22. Sathaye, S.; Kaikini, A.A.; Dhodi, D.; Muke, S.; Peshattiwar, V.; Bagle, S.; Korde, A.; Sarnaik, J.; Kadwad, V.; Sachdev, S. Standardization of type 1 and type 2 diabetic nephropathy models in rats: Assessment and characterization of metabolic features and renal injury. J. Pharm. Bioallied Sci. 2020, 12, 295–307. [Google Scholar]
  23. Szkudelski, T. Streptozotocin-Nicotinamide-Induced diabetes in the rat. Characteristics of the experimental model. Exp. Biol. Med. 2012, 237, 481–490. [Google Scholar]
  24. Masiello, P.; Broca, C.; Gross, R.; Roye, M.; Manteghetti, M.; Hillaire-Buys, D.; Novelli, M.; Ribes, G. Experimental niddm: Development of a new model in adult rats administered streptozotocin and nicotinamide. Diabetes 1998, 47, 224–229. [Google Scholar]
  25. Gheibi, S.; Jeddi, S.; Carlstrom, M.; Kashfi, K.; Ghasemi, A. Hydrogen sulfide potentiates the favorable metabolic effects of inorganic nitrite in type 2 diabetic rats. Nitric Oxide 2019, 92, 60–72. [Google Scholar]
  26. Jeddi, S.; Gheibi, S.; Kashfi, K.; Ghasemi, A. Sodium hydrosulfide has no additive effects on nitrite-Inhibited renal gluconeogenesis in type 2 diabetic rats. Life Sci. 2021, 283, 119870. [Google Scholar]
  27. Javrushyan, H.; Nadiryan, E.; Grigoryan, A.; Avtandilyan, N.; Maloyan, A. Antihyperglycemic activity of l-Norvaline and l-Arginine in high-Fat diet and streptozotocin-Treated male rats. Exp. Mol. Pathol. 2022, 126, 104763. [Google Scholar]
  28. Liu, P.; Zhang, Z.; Li, Y. Relevance of the pyroptosis-Related inflammasome pathway in the pathogenesis of diabetic kidney disease. Front. Immunol. 2021, 12, 603416. [Google Scholar]
  29. Wu, J.; Luo, X.; Thangthaeng, N.; Sumien, N.; Chen, Z.; Rutledge, M.A.; Jing, S.; Forster, M.J.; Yan, L.J. Pancreatic mitochondrial complex i exhibits aberrant hyperactivity in diabetes. Biochem. Biophys. Rep. 2017, 11, 119–129. [Google Scholar]
  30. Li, C.Y.; Ma, W.X.; Yan, L.J. 5-Methoxyindole-2-Carboxylic acid (mica) fails to retard development and progression of type ii diabetes in zsf1 diabetic rats. React. Oxyg. Species Apex NC 2020, 9, 144–147. [Google Scholar]
  31. Dugbartey, G.J.; Wonje, Q.L.; Alornyo, K.K.; Adams, I.; Diaba, D.E. Alpha-Lipoic acid treatment improves adverse cardiac remodelling in the diabetic heart—The role of cardiac hydrogen sulfide-Synthesizing enzymes. Biochem. Pharmacol. 2022, 203, 115179. [Google Scholar]
  32. Qasem, M.A.; Noordin, M.I.; Arya, A.; Alsalahi, A.; Jayash, S.N. Evaluation of the glycemic effect of ceratonia siliqua pods (carob) on a streptozotocin-Nicotinamide induced diabetic rat model. PeerJ 2018, 6, e4788. [Google Scholar]
  33. Patra, S.; Bhattacharya, S.; Bala, A.; Haldar, P.K. Antidiabetic effect of drymaria cordata leaf against streptozotocin-Nicotinamide-Induced diabetic albino rats. J. Adv. Pharm. Technol. Res. 2020, 11, 44–52. [Google Scholar]
  34. Kumar, E.K.; Janardhana, G.R. Antidiabetic activity of alcoholic stem extract of nervilia plicata in streptozotocin-Nicotinamide induced type 2 diabetic rats. J. Ethnopharmacol. 2011, 133, 480–483. [Google Scholar]
  35. Balaji, P.; Madhanraj, R.; Rameshkumar, K.; Veeramanikandan, V.; Eyini, M.; Arun, A.; Thulasinathan, B.; Al Farraj, D.; Elshikh, M.; Alokda, A.; et al. Evaluation of antidiabetic activity of pleurotus pulmonarius against streptozotocin-Nicotinamide induced diabetic wistar albino rats. Saudi J. Biol. Sci. 2020, 27, 913–924. [Google Scholar]
  36. Szkudelski, T. The mechanism of alloxan and streptozotocin action in b cells of the rat pancreas. Physiol. Res. 2001, 50, 537–546. [Google Scholar]
  37. Wu, J.; Yan, L.J. Streptozotocin-Induced type 1 diabetes in rodents as a model for studying mitochondrial mechanisms of diabetic beta cell glucotoxicity. Diabetes Metab. Syndr. Obes. Targets Ther. 2015, 8, 181–188. [Google Scholar]
  38. Lenzen, S. The mechanisms of alloxan- and streptozotocin-Induced diabetes. Diabetologia 2008, 51, 216–226. [Google Scholar]
  39. Wu, J.; Jin, Z.; Zheng, H.; Yan, L.J. Sources and implications of nadh/nad (+) redox imbalance in diabetes and its complications. Diabetes Metab. Syndr. Obes. 2016, 9, 145–153. [Google Scholar]
  40. Wu, C.C.; Hung, C.N.; Shin, Y.C.; Wang, C.J.; Huang, H.P. Myrciaria cauliflora extracts attenuate diabetic nephropathy involving the ras signaling pathway in streptozotocin/nicotinamide mice on a high fat diet. J. Food Drug Anal. 2016, 24, 136–146. [Google Scholar]
  41. Yu, W.C.; Huang, R.Y.; Chou, T.C. Oligo-Fucoidan improves diabetes-Induced renal fibrosis via activation of sirt-1, glp-1r, and nrf2/ho-1: An in vitro and in vivo study. Nutrients 2020, 12, 3068. [Google Scholar]
  42. Nakamura, T.; Terajima, T.; Ogata, T.; Ueno, K.; Hashimoto, N.; Ono, K.; Yano, S. Establishment and pathophysiological characterization of type 2 diabetic mouse model produced by streptozotocin and nicotinamide. Biol. Pharm. Bull. 2006, 29, 1167–1174. [Google Scholar]
  43. Weng, Y.; Yu, L.; Cui, J.; Zhu, Y.-R.; Guo, C.; Wei, G.; Duan, J.-L.; Yin, Y.; Guan, Y.; Wang, Y.-H.; et al. Antihyperglycemic, hypolipidemic and antioxidant activities of total saponins extracted from aralia taibaiensis in experimental type 2 diabetic rats. J. Ethnopharmacol. 2014, 152, 553–560. [Google Scholar]
  44. Bayrasheva, V.K.; Babenko, A.Y.; Dobronravov, V.A.; Dmitriev, Y.V.; Chefu, S.G.; Pchelin, I.Y.; Ivanova, A.N.; Bairamov, A.A.; Alexeyeva, N.P.; Shatalov, I.S.; et al. Uninephrectomized high-Fat-Fed nicotinamide-Streptozotocin-Induced diabetic rats: A model for the investigation of diabetic nephropathy in type 2 diabetes. J. Diabetes Res. 2016, 2016, 8317850. [Google Scholar]
  45. Sasongko, H.; Nurrochmad, A.; Rohman, A.; Nugroho, A.E. Characteristic of Streptozotocin-Nicotinamide-Induced Inflammation in A Rat Model of Diabetes-Associated Renal Injury. Open Access Maced. J. Med. Sci. 2022, 10, 16–22. [Google Scholar]
  46. Pelle, M.C.; Provenzano, M.; Busutti, M.; Porcu, C.V.; Zaffina, I.; Stanga, L.; Arturi, F. Up-Date on diabetic nephropathy. Life 2022, 12, 1202. [Google Scholar]
  47. Yan, L.J. Folic acid-Induced animal model of kidney disease. Animal. Model. Exp. Med. 2021, 4, 329–342. [Google Scholar]
  48. Corremans, R.; D’Haese, P.C.; Vervaet, B.A.; Verhulst, A. L-Name administration enhances diabetic kidney disease development in an stz/nad rat model. Int. J. Mol. Sci. 2021, 22, 12767. [Google Scholar]
  49. Arigela, C.S.; Nelli, G.; Gan, S.H.; Sirajudeen, K.N.S.; Krishnan, K.; Abdul Rahman, N.; Pasupuleti, V.R. Bitter gourd honey ameliorates hepatic and renal diabetic complications on type 2 diabetes rat models by antioxidant, anti-Inflammatory, and anti-Apoptotic mechanisms. Foods 2021, 10, 2872. [Google Scholar]
  50. Agarwal, R. Pathogenesis of diabetic nephropathy. In Chronic Kidney Disease and Type 2 Diabetes; American Diabetes Association: Arlington, VA, USA, 2021; pp. 2–7. [Google Scholar]
  51. Mahdavifard, S.; Nakhjavani, M. 1,8 cineole protects type 2 diabetic rats against diabetic nephropathy via inducing the activity of glyoxalase-I and lowering the level of transforming growth factor-1beta. J. Diabetes Metab. Disord. 2022, 21, 567–572. [Google Scholar]
  52. Khanra, R.; Dewanjee, S.; Dua, T.K.; Sahu, R.; Gangopadhyay, M.; De Feo, V.; Zia-Ul-Haq, M. Abroma augusta l. (malvaceae) leaf extract attenuates diabetes induced nephropathy and cardiomyopathy via inhibition of oxidative stress and inflammatory response. J. Transl. Med. 2015, 13, 6. [Google Scholar]
  53. Olaniyi, K.S.; Amusa, O.A.; Akinnagbe, N.T.; Ajadi, I.O.; Ajadi, M.B.; Agunbiade, T.B.; Michael, O.S. Acetate ameliorates nephrotoxicity in streptozotocin-Nicotinamide-Induced diabetic rats: Involvement of xanthine oxidase activity. Cytokine 2021, 142, 155501. [Google Scholar]
  54. Wu, W.T.; Hsu, T.H.; Lee, C.H.; Lo, H.C. Fruiting bodies of chinese caterpillar mushroom, ophiocordyceps sinensis (ascomycetes) alleviate diabetes-Associated oxidative stress. Int. J. Med. Mushrooms 2020, 22, 15–29. [Google Scholar]
  55. Kishore, L.; Kaur, N.; Singh, R. Renoprotective effect of bacopa monnieri via inhibition of advanced glycation end products and oxidative stress in stz-Nicotinamide-Induced diabetic nephropathy. Ren. Fail. 2016, 38, 1528–1544. [Google Scholar]
  56. Pourfarjam, Y.; Rezagholizadeh, L.; Nowrouzi, A.; Meysamie, A.; Ghaseminejad, S.; Ziamajidi, N.; Norouzi, D. Effect of cichorium intybus l. Seed extract on renal parameters in experimentally induced early and late diabetes type 2 in rats. Ren. Fail. 2017, 39, 211–221. [Google Scholar]
  57. Ali, A.M.; Gabbar, M.A.; Abdel-Twab, S.M.; Fahmy, E.M.; Ebaid, H.; Alhazza, I.M.; Ahmed, O.M. Antidiabetic potency, antioxidant effects, and mode of actions of citrus reticulata fruit peel hydroethanolic extract, hesperidin, and quercetin in nicotinamide/streptozotocin-Induced wistar diabetic rats. Oxidative Med. Cell. Longev. 2020, 2020, 1730492. [Google Scholar]
  58. Kpemissi, M.; Potârniche, A.-V.; Lawson-Evi, P.; Metowogo, K.; Melila, M.; Dramane, P.; Taulescu, M.; Chandramohan, V.; Suhas, D.S.; Puneeth, T.A.; et al. Nephroprotective effect of combretum micranthum g. Don in nicotinamide-Streptozotocin induced diabetic nephropathy in rats: In-Vivo and in-Silico experiments. J. Ethnopharmacol. 2020, 261, 113133. [Google Scholar]
  59. Maheshwari, R.A.; Balaraman, R.; Sen, A.K.; Seth, A.K. Effect of coenzyme q10 alone and its combination with metformin on streptozotocin-Nicotinamide-Induced diabetic nephropathy in rats. Indian J. Pharmacol. 2014, 46, 627–632. [Google Scholar]
  60. Maheshwari, R.; Balaraman, R.; Sen, A.K.; Shukla, D.; Seth, A. Effect of concomitant administration of coenzyme q10 with sitagliptin on experimentally induced diabetic nephropathy in rats. Ren. Fail. 2017, 39, 130–139. [Google Scholar]
  61. Yu, S.H.; Dubey, N.K.; Li, W.S.; Liu, M.C.; Chiang, H.S.; Leu, S.J.; Shieh, Y.H.; Tsai, F.C.; Deng, W.P. Cordyceps militaris treatment preserves renal function in type 2 diabetic nephropathy mice. PLoS ONE 2016, 11, e0166342. [Google Scholar]
  62. Margaritis, I.; Angelopoulou, K.; Lavrentiadou, S.; Mavrovouniotis, I.C.; Tsantarliotou, M.; Taitzoglou, I.; Theodoridis, A.; Veskoukis, A.; Kerasioti, E.; Kouretas, D.; et al. Effect of crocin on antioxidant gene expression, fibrinolytic parameters, redox status and blood biochemistry in nicotinamide-Streptozotocin-Induced diabetic rats. J. Biol. Res. 2020, 27, 4. [Google Scholar]
  63. Singla, K.; Singh, R. Nephroprotective effect of curculigo orchiodies in streptozotocin-Nicotinamide induced diabetic nephropathy in wistar rats. J. Ayurveda Integr. Med. 2020, 11, 399–404. [Google Scholar]
  64. El Medany, A.M.H.; Hammadi, S.H.M.; Khalifa, H.M.; Ghazala, R.A.; Mohammed, H.S.Z. The vascular impact of dapagliflozin, liraglutide, and atorvastatin alone or in combinations in type 2 diabetic rat model. Fundam. Clin. Pharmacol. 2022, 36, 731–741. [Google Scholar]
  65. Abdel-Wahab, A.F.; Bamagous, G.A.; Al-Harizy, R.M.; ElSawy, N.A.; Shahzad, N.; Ibrahim, I.A.; Ghamdi, S.S.A. Renal protective effect of sglt2 inhibitor dapagliflozin alone and in combination with irbesartan in a rat model of diabetic nephropathy. Biomed. Pharm. 2018, 103, 59–66. [Google Scholar]
  66. Jangale, N.M.; Devarshi, P.P.; Bansode, S.B.; Kulkarni, M.J.; Harsulkar, A.M. Dietary flaxseed oil and fish oil ameliorates renal oxidative stress, protein glycation, and inflammation in streptozotocin-Nicotinamide-Induced diabetic rats. J. Physiol. Biochem. 2016, 72, 327–336. [Google Scholar]
  67. Kaur, N.; Kishore, L.; Singh, R. Dillenia indica l. Attenuates diabetic nephropathy via inhibition of advanced glycation end products accumulation in stz-Nicotinamide induced diabetic rats. J. Tradit. Complement Med. 2018, 8, 226–238. [Google Scholar]
  68. Srinivasan, S.; Pari, L. Ameliorative effect of diosmin, a citrus flavonoid against streptozotocin-Nicotinamide generated oxidative stress induced diabetic rats. Chem.-Biol. Interact. 2012, 195, 43–51. [Google Scholar]
  69. El-Kader, M.A.; Hashish, H.A. Potential role of empagliflozin in prevention of nephropathy in streptozotocin-Nicotinamideinduced type 2 diabetes: An ultrastructural study. Anatomy 2019, 13, 137–148. [Google Scholar]
  70. Gutierrez, R.M.P.; Campoy, A.H.G.; Carrera, S.P.P.; Ramirez, A.M.; Flores, J.M.M.; Valle, S.O.F. 3′-O-Beta-D-Glucopyranosyl-Alpha,4,2′,4′,6′-Pentahydroxy-Dihydrochalcone, from bark of eysenhardtia polystachya prevents diabetic nephropathy via inhibiting protein glycation in stz-Nicotinamide induced diabetic mice. Molecules 2019, 24, 1214. [Google Scholar]
  71. Ziamajidi, N.; Nasiri, A.; Abbasalipourkabir, R.; Sadeghi Moheb, S. Effects of garlic extract on tnf-Alpha expression and oxidative stress status in the kidneys of rats with stz + nicotinamide-Induced diabetes. Pharm. Biol. 2017, 55, 526–531. [Google Scholar]
  72. Kasozi, K.I.; Namubiru, S.; Safiriyu, A.A.; Ninsiima, H.I.; Nakimbugwe, D.; Namayanja, M.; Valladares, M.B. Grain amaranth is associated with improved hepatic and renal calcium metabolism in type 2 diabetes mellitus of male wistar rats. Evid.-Based Complement. Altern. Med. 2018, 2018, 4098942. [Google Scholar]
  73. Selvaraj, G.; Kaliamurthi, S.; Thirugnasambandan, R. Effect of glycosin alkaloid from rhizophora apiculata in non-Insulin dependent diabetic rats and its mechanism of action: In vivo and in silico studies. Phytomedicine 2016, 23, 632–640. [Google Scholar]
  74. Abd El Motteleb, D.M.; Abd El Aleem, D.I. Renoprotective effect of hypericum perforatum against diabetic nephropathy in rats: Insights in the underlying mechanisms. Clin. Exp. Pharmacol. Physiol. 2017, 44, 509–521. [Google Scholar]
  75. Dugbartey, G.J.; Alornyo, K.K.; N’Guessan, B.B.; Atule, S.; Mensah, S.D.; 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. [Google Scholar]
  76. Dugbartey, G.J.; Alornyo, K.K.; Diaba, D.E.; 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. [Google Scholar]
  77. Deshmukh, A.; Manjalkar, P. Synergistic effect of micronutrients and metformin in alleviating diabetic nephropathy and cardiovascular dysfunctioning in diabetic rat. J. Diabetes Metab. Disord. 2021, 20, 533–541. [Google Scholar]
  78. Hsu, J.D.; Wu, C.C.; Hung, C.N.; Wang, C.J.; Huang, H.P. Myrciaria cauliflora extract improves diabetic nephropathy via suppression of oxidative stress and inflammation in streptozotocin-Nicotinamide mice. J. Food Drug Anal. 2016, 24, 730–737. [Google Scholar]
  79. Kishore, L.; Kaur, N.; Singh, R. Nephroprotective effect of paeonia emodi via inhibition of advanced glycation end products and oxidative stress in streptozotocin-Nicotinamide induced diabetic nephropathy. J. Food Drug Anal. 2017, 25, 576–588. [Google Scholar]
  80. Giribabu, N.; Karim, K.; Kilari, E.K.; Salleh, N. Phyllanthus niruri leaves aqueous extract improves kidney functions, ameliorates kidney oxidative stress, inflammation, fibrosis and apoptosis and enhances kidney cell proliferation in adult male rats with diabetes mellitus. J. Ethnopharmacol. 2017, 205, 123–137. [Google Scholar]
  81. Afzal, H.R.; Khan, N.U.H.; Sultana, K.; Mobashar, A.; Lareb, A.; Khan, A.; Gull, A.; Afzaal, H.; Khan, M.T.; Rizwan, M.; et al. Schiff bases of pioglitazone provide better antidiabetic and potent antioxidant effect in a streptozotocin-Nicotinamide-Induced diabetic rodent model. ACS Omega 2021, 6, 4470–4479. [Google Scholar]
  82. Lin, C.F.; Kuo, Y.T.; Chen, T.Y.; Chien, C.T. Quercetin-Rich guava (psidium guajava) juice in combination with trehalose reduces autophagy, apoptosis and pyroptosis formation in the kidney and pancreas of type ii diabetic rats. Molecules 2016, 21, 334. [Google Scholar]
  83. Palsamy, P.; Subramanian, S. Resveratrol protects diabetic kidney by attenuating hyperglycemia-Mediated oxidative stress and renal inflammatory cytokines via nrf2-Keap1 signaling. Biochim. Biophys. Acta-Mol. Basis Dis. 2011, 1812, 719–731. [Google Scholar]
  84. Zhao, L.L.; Makinde, E.A.; Shah, M.A.; Olatunji, O.J.; Panichayupakaranant, P. Rhinacanthins-Rich extract and rhinacanthin c ameliorate oxidative stress and inflammation in streptozotocin-Nicotinamide-Induced diabetic nephropathy. J. Food Biochem. 2019, 43, e12812. [Google Scholar]
  85. Uddandrao, V.V.S.; Brahmanaidu, P.; Ravindarnaik, R.; Suresh, P.; Vadivukkarasi, S.; Saravanan, G. Restorative potentiality of s-Allylcysteine against diabetic nephropathy through attenuation of oxidative stress and inflammation in streptozotocin-Nicotinamide-Induced diabetic rats. Eur. J. Nutr. 2019, 58, 2425–2437. [Google Scholar]
  86. Inoue, M.-K.; Matsunaga, Y.; Nakatsu, Y.; Yamamotoya, T.; Ueda, K.; Kushiyama, A.; Sakoda, H.; Fujishiro, M.; Ono, H.; Iwashita, M.; et al. Possible involvement of normalized pin1 expression level and ampk activation in the molecular mechanisms underlying renal protective effects of sglt2 inhibitors in mice. Diabetol. Metab. Syndr. 2019, 11, 57. [Google Scholar]
  87. Sheela, N.; Jose, M.A.; Sathyamurthy, D.; Kumar, B.N. Effect of silymarin on streptozotocin-Nicotinamide-Induced type 2 diabetic nephropathy in rats. Iran. J. Kidney Dis. 2013, 7, 117–123. [Google Scholar]
  88. Mandave, P.; Khadke, S.; Karandikar, M.; Pandit, V.; Ranjekar, P.; Kuvalekar, A.; Mantri, N. Antidiabetic, lipid normalizing, and nephroprotective actions of the strawberry: A potent supplementary fruit. Int. J. Mol. Sci. 2017, 18, 124. [Google Scholar]
  89. Chandran, R.; Parimelazhagan, T.; Shanmugam, S.; Thankarajan, S. Antidiabetic activity of syzygium calophyllifolium in streptozotocin-Nicotinamide induced type-2 diabetic rats. Biomed. Pharmacother. 2016, 82, 547–554. [Google Scholar]
  90. Murugan, P.; Pari, L. Protective role of tetrahydrocurcumin on changes in the fatty acid composition in streptozotocin-Nicotinamide induced type 2 diabetic rats. J. Appl. Biomed. 2007, 5, 31–38. [Google Scholar]
  91. Singh, B.; Kumar, A.; Singh, H.; Kaur, S.; Arora, S.; Singh, B. Protective effect of vanillic acid against diabetes and diabetic nephropathy by attenuating oxidative stress and upregulation of nf-Kappab, tnf-Alpha and cox-2 proteins in rats. Phytother. Res. 2022, 36, 1338–1352. [Google Scholar]
  92. Zhang, R.; Lu, M.; Zhang, S.; Liu, J. Renoprotective effects of tilianin in diabetic rats through modulation of oxidative stress via nrf2-Keap1 pathway and inflammation via tlr4/mapk/nf-Kappab pathways. Int. Immunopharmacol. 2020, 88, 106967. [Google Scholar]
  93. Kyei-Barffour, I.; Kwarkoh, R.K.B.; Arthur, O.D.; Akwetey, S.A.; Acheampong, D.O.; Aboagye, B.; Brah, A.S.; Amponsah, I.K.; Adokoh, C.K. Alkaloidal extract from zanthoxylum zanthoxyloides stimulates insulin secretion in normoglycemic and nicotinamide/streptozotocin-Induced diabetic rats. Heliyon 2021, 7, e07452. [Google Scholar]
  94. Homer, B.L.; Dower, K. 41-Week study of progressive diabetic nephropathy in the zsf1 fa/fa(cp) rat model. Toxicol. Pathol. 2018, 46, 976–977. [Google Scholar]
  95. Zhao, Y.; Yan, T.; Xiong, C.; Chang, M.; Gao, Q.; Yao, S.; Wu, W.; Yi, X.; Xu, G. Overexpression of lipoic acid synthase gene alleviates diabetic nephropathy of lepr(db/db) mice. BMJ Open Diabetes Res. Care 2021, 9, e002260. [Google Scholar]
  96. Zhang, B.; Zhang, X.; Zhang, C.; Sun, G.; Sun, X. Berberine improves the protective effects of metformin on diabetic nephropathy in db/db mice through trib1-Dependent inhibiting inflammation. Pharm. Res. 2021, 38, 1807–1820. [Google Scholar]
  97. Khan, M.F.; Mathur, A.; Pandey, V.K.; Kakkar, P. Endoplasmic reticulum stress-Dependent activation of trb3-Foxo1 signaling pathway exacerbates hyperglycemic nephrotoxicity: Protection accorded by naringenin. Eur. J. Pharmacol. 2022, 917, 174745. [Google Scholar]
  98. Boye, A.; Acheampong, D.O.; Gyamerah, E.O.; Asiamah, E.A.; Addo, J.K.; Mensah, D.A.; Brah, A.S.; Ayiku, P.J. Glucose lowering and pancreato-Protective effects of abrus precatorius (l.) leaf extract in normoglycemic and stz/nicotinamide-Induced diabetic rats. J. Ethnopharmacol. 2020, 258, 112918. [Google Scholar]
  99. Indumathi, D.; Sujithra, K.; Srinivasan, S.; Vinothkumar, V. Protective effect of betanin against streptozotocin-Nicotinamide induced liver, kidney and pancreas damage by attenuating lipid byproducts and improving renal biomarkers in wistar rats. Int. J. Adv. Res. Biol. Sci. 2017, 4, 160–170. [Google Scholar]
  100. Ablat, A.; Halabi, M.F.; Mohamad, J.; Hasnan, M.H.; Hazni, H.; Teh, S.H.; Shilpi, J.A.; Mohamed, Z.; Awang, K. Antidiabetic effects of brucea javanica seeds in type 2 diabetic rats. BMC Complement. Altern. Med. 2017, 17, 94. [Google Scholar]
  101. Nankar, R.P.; Doble, M. Hybrid drug combination: Anti-Diabetic treatment of type 2 diabetic wistar rats with combination of ellagic acid and pioglitazone. Phytomedicine 2017, 37, 4–9. [Google Scholar]
  102. Karle, P.P.; Dhawale, S.C.; Navghare, V.V. Amelioration of diabetes and its complications by manilkara zapota (l) p. Royen fruit peel extract and its fractions in alloxan and stz-Na induced diabetes in wistar rats. J. Diabetes Metab. Disord. 2022, 21, 493–510. [Google Scholar]
  103. Aboonabi, A.; Rahmat, A.; Othman, F. Antioxidant effect of pomegranate against streptozotocin-Nicotinamide generated oxidative stress induced diabetic rats. Toxicol. Rep. 2014, 1, 915–922. [Google Scholar]
  104. Soufi, F.G.; Vardyani, M.; Sheervalilou, R.; Mohammadi, M.; Somi, M.H. Long-Term treatment with resveratrol attenuates oxidative stress pro-Inflammatory mediators and apoptosis in streptozotocin-Nicotinamide-Induced diabetic rats. Gen. Physiol. Biophys. 2012, 31, 431–438. [Google Scholar]
  105. Yan, L.J. Redox imbalance stress in diabetes mellitus: Role of the polyol pathway. Animal Model. Exp. Med. 2018, 1, 7–13. [Google Scholar]
  106. Luo, X.; Wu, J.; Jing, S.; Yan, L.J. Hyperglycemic stress and carbon stress in diabetic glucotoxicity. Aging Dis. 2016, 7, 90–110. [Google Scholar]
  107. Yan, L.J.; Sumien, N.; Thangthaeng, N.; Forster, M.J. Reversible inactivation of dihydrolipoamide dehydrogenase by mitochondrial hydrogen peroxide. Free Radic. Res. 2013, 47, 123–133. [Google Scholar]
  108. Wu, J.; Luo, X.; Jing, S.; Yan, L.J. Two-Dimensional gel electrophoretic detection of protein carbonyls derivatized with biotin-Hydrazide. J. Chromatogr. B 2016, 1019, 128–131. [Google Scholar]
  109. Zheng, H.; Wu, J.; Jin, Z.; Yan, L.J. Protein modifications as manifestations of hyperglycemic glucotoxicity in diabetes and its complications. Biochem. Insights 2016, 9, BCI-S36141. [Google Scholar]
  110. Murea, M.; Freedman, B.I.; Parks, J.S.; Antinozzi, P.A.; Elbein, S.C.; Ma, L. Lipotoxicity in diabetic nephropathy: The potential role of fatty acid oxidation. Clin. J. Am. Soc. Nephrol. 2010, 5, 2373–2379. [Google Scholar]
  111. Jang, H.S.; Noh, M.R.; Kim, J.; Padanilam, B.J. Defective mitochondrial fatty acid oxidation and lipotoxicity in kidney diseases. Front. Med. 2020, 7, 65. [Google Scholar]
  112. Yang, S.-K.; Li, A.-M.; Han, Y.-C.; Peng, C.-H.; Song, N.; Yang, M.; Zhan, M.; Zeng, L.-F.; Song, P.-A.; Zhang, W.; et al. Mitochondria-Targeted peptide ss31 attenuates renal tubulointerstitial injury via inhibiting mitochondrial fission in diabetic mice. Oxidative Med. Cell. Longev. 2019, 2019, 2346580. [Google Scholar]
  113. Ma, Y.; Chen, Z.; Tao, Y.; Zhu, J.; Yang, H.; Liang, W.; Ding, G. Increased mitochondrial fission of glomerular podocytes in diabetic nephropathy. Endocr. Connect. 2019, 8, 1206–1212. [Google Scholar]
  114. Higgins, G.C.; Coughlan, M.T. Mitochondrial dysfunction and mitophagy: The beginning and end to diabetic nephropathy? Br. J. Pharmacol. 2014, 171, 1917–1942. [Google Scholar]
  115. Sun, J.; Zhu, H.; Wang, X.; Gao, Q.; Li, Z.; Huang, H. Coq10 ameliorates mitochondrial dysfunction in diabetic nephropathy through mitophagy. J. Endocrinol. 2019, 240, 445–465. [Google Scholar]
  116. Cybulsky, A.V. Endoplasmic reticulum stress, the unfolded protein response and autophagy in kidney diseases. Nat. Rev. Nephrol. 2017, 13, 681–696. [Google Scholar]
  117. Wang, H.; Karnati, S.; Madhusudhan, T. Regulation of the homeostatic unfolded protein response in diabetic nephropathy. Pharmaceuticals 2022, 15, 401. [Google Scholar]
  118. Lu, C.; Wu, B.; Liao, Z.; Xue, M.; Zou, Z.; Feng, J.; Sheng, J. Dusp1 overexpression attenuates renal tubular mitochondrial dysfunction by restoring parkin-Mediated mitophagy in diabetic nephropathy. Biochem. Biophys. Res. Commun. 2021, 559, 141–147. [Google Scholar]
  119. Sherkhane, B.; Kalvala, A.K.; Arruri, V.K.; Khatri, D.K.; Singh, S.B. Renoprotective potential of myo-Inositol on diabetic kidney disease: Focus on the role of the pink1/parkin pathway and mitophagy receptors. J. Biochem. Mol. Toxicol. 2022, 36, e23032. [Google Scholar]
  120. Asico, L.D.; Cuevas, S.; Ma, X.; Jose, P.A.; Armando, I.; Konkalmatt, P.R. Nephron segment-Specific gene expression using aav vectors. Biochem. Biophys. Res. Commun. 2018, 497, 19–24. [Google Scholar]
  121. Thallas-Bonke, V.; Tan, S.M.; Lindblom, R.S.; Snelson, M.; Granata, C.; Jha, J.C.; Sourris, K.C.; Laskowski, A.; Watson, A.; Tauc, M.; et al. Targeted deletion of nicotinamide adenine dinucleotide phosphate oxidase 4 from proximal tubules is dispensable for diabetic kidney disease development. Nephrol. Dial. Transpl. 2021, 36, 988–997. [Google Scholar]
  122. Shao, B.Y.; Zhang, S.F.; Li, H.D.; Meng, X.M.; Chen, H.Y. Epigenetics and inflammation in diabetic nephropathy. Front. Physiol. 2021, 12, 649587. [Google Scholar]
  123. Li, X.; Lu, L.; Hou, W.; Huang, T.; Chen, X.; Qi, J.; Zhao, Y.; Zhu, M. Epigenetics in the pathogenesis of diabetic nephropathy. Acta Biochim. Biophys. Sin. 2022, 54, 163–172. [Google Scholar]
  124. Aparicio-Trejo, O.E.; Reyes-Fermin, L.M.; Briones-Herrera, A.; Tapia, E.; Leon-Contreras, J.C.; Hernandez-Pando, R.; Sanchez-Lozada, L.G.; Pedraza-Chaverri, J. Protective effects of n-Acetyl-Cysteine in mitochondria bioenergetics, oxidative stress, dynamics and s-Glutathionylation alterations in acute kidney damage induced by folic acid. Free Radic. Biol. Med. 2019, 130, 379–396. [Google Scholar]
  125. Aparicio-Trejo, O.E.; Avila-Rojas, S.H.; Tapia, E.; Rojas-Morales, P.; Leon-Contreras, J.C.; Martinez-Klimova, E.; Hernandez-Pando, R.; Sanchez-Lozada, L.G.; Pedraza-Chaverri, J. Chronic impairment of mitochondrial bioenergetics and beta-Oxidation promotes experimental aki-To-Ckd transition induced by folic acid. Free Radic. Biol. Med. 2020, 154, 18–32. [Google Scholar]
  126. Gupta, A.; Puri, V.; Sharma, R.; Puri, S. Folic acid induces acute renal failure (arf) by enhancing renal prooxidant state. Exp. Toxicol. Pathol. 2012, 64, 225–232. [Google Scholar]
  127. Perse, M.; Veceric-Haler, Z. Cisplatin-Induced rodent model of kidney injury: Characteristics and challenges. Biomed. Res. Int. 2018, 2018, 1462802. [Google Scholar]
  128. Oh, G.S.; Kim, H.J.; Shen, A.; Lee, S.B.; Yang, S.H.; Shim, H.; Cho, E.Y.; Kwon, K.B.; Kwak, T.H.; So, H.S. New therapeutic concept of nad redox balance for cisplatin nephrotoxicity. Biomed. Res. Int. 2016, 2016, 4048390. [Google Scholar]
  129. Prozialeck, W.C.; Vaidya, V.S.; Liu, J.; Waalkes, M.P.; Edwards, J.R.; Lamar, P.C.; Bernard, A.M.; Dumont, X.; Bonventre, J.V. Kidney injury molecule-1 is an early biomarker of cadmium nephrotoxicity. Kidney Int. 2007, 72, 985–993. [Google Scholar]
  130. Prozialeck, W.C.; Edwards, J.R. Mechanisms of cadmium-Induced proximal tubule injury: New insights with implications for biomonitoring and therapeutic interventions. J. Pharmacol. Exp. Ther. 2012, 343, 2–12. [Google Scholar]
  131. Yan, L.J.; Allen, D.C. Cadmium-Induced kidney injury: Oxidative damage as a unifying mechanism. Biomolecules 2021, 11, 1575. [Google Scholar]
  132. Chi, P.J.; Lee, C.J.; Hsieh, Y.J.; Lu, C.W.; Hsu, B.G. Dapagliflozin ameliorates lipopolysaccharide related acute kidney injury in mice with streptozotocin-Induced diabetes mellitus. Int. J. Med. Sci. 2022, 19, 729–739. [Google Scholar]
  133. Tang, J.L.; Xin, M.; Zhang, L.C. Protective effect of astragalus membranaceus and astragaloside iv in sepsis-Induced acute kidney injury. Aging 2022, 14, 5855. [Google Scholar]
  134. Pabla, N.; Bajwa, A. Role of mitochondrial therapy for ischemic-Reperfusion injury and acute kidney injury. Nephron 2022, 146, 253–258. [Google Scholar]
  135. Hesp, A.C.; Schaub, J.A.; Prasad, P.V.; Vallon, V.; Laverman, G.D.; Bjornstad, P.; van Raalte, D.H. The role of renal hypoxia in the pathogenesis of diabetic kidney disease: A promising target for newer renoprotective agents including sglt2 inhibitors? Kidney Int. 2020, 98, 579–589. [Google Scholar]
  136. Ahmadi, F.; Hajihashemi, S.; Rahbari, A.; Ghanbari, F. Effects of nitroglycerine on renal ischemia-Reperfusion injury in adult male rats. Drug Res. 2019, 69, 612–620. [Google Scholar]
  137. Khbouz, B.; Lallemand, F.; Cirillo, A.; Rowart, P.; Legouis, D.; Sounni, N.E.; Noel, A.; De Tullio, P.; de Seigneux, S.; Jouret, F. Kidney-Targeted irradiation triggers renal ischemic preconditioning in mice. Am. J. Physiol.-Ren. Physiol. 2022, 323, F198–F211. [Google Scholar]
  138. Khbouz, B.; Rowart, P.; Poma, L.; Dahlke, E.; Bottner, M.; Stokes, M.; Bolen, G.; Rahmouni, S.; Theilig, F.; Jouret, F. The genetic deletion of the dual specificity phosphatase 3 (dusp3) attenuates kidney damage and inflammation following ischaemia/reperfusion injury in mouse. Acta Physiol. 2022, 234, e13735. [Google Scholar]
  139. Yoo, Y.C.; Yoo, K.J.; Lim, B.J.; Jun, J.H.; Shim, J.K.; Kwak, Y.L. Propofol attenuates renal ischemia-Reperfusion injury aggravated by hyperglycemia. J. Surg. Res. 2013, 183, 783–791. [Google Scholar]
  140. Sehirli, O.; Sener, E.; Cetinel, S.; Yuksel, M.; Gedik, N.; Sener, G. Alpha-Lipoic acid protects against renal ischaemia-reperfusion injury in rats. Clin. Exp. Pharmacol. Physiol. 2008, 35, 249–255. [Google Scholar]
  141. Gang, G.-T.; Hwang, J.H.; Kim, Y.-H.; Noh, J.-R.; Kim, K.-S.; Jeong, J.Y.; Choi, D.E.; Lee, K.W.; Jung, J.-Y.; Shong, M.; et al. Protection of nad (p) h:Quinone oxidoreductase 1 against renal ischemia/reperfusion injury in mice. Free Radic. Biol. Med. 2014, 67, 139–149. [Google Scholar]
  142. Xu, X.M.; Cai, G.Y.; Bu, R.; Wang, W.J.; Bai, X.Y.; Sun, X.F.; Chen, X.M. Beneficial effects of caloric restriction on chronic kidney disease in rodent models: A meta-Analysis and systematic review. PLoS ONE 2015, 10, e0144442. [Google Scholar]
  143. Koehler, F.C.; Spath, M.R.; Hoyer-Allo, K.J.R.; Muller, R.U. Mechanisms of caloric restriction-Mediated stress-Resistance in acute kidney injury. Nephron 2022, 146, 234–238. [Google Scholar]
  144. Serna, J.D.C.; Amaral, A.G.; Caldeira da Silva, C.C.; Munhoz, A.C.; Vilas-Boas, E.A.; Menezes-Filho, S.L.; Kowaltowski, A.J. Regulation of kidney mitochondrial function by caloric restriction. Am. J. Physiol. Renal. Physiol. 2022, 323, F92–F106. [Google Scholar]
  145. Bai, M.; Wang, Y.; Han, R.; Xu, L.; Huang, M.; Zhao, J.; Lin, Y.; Song, S.; Chen, Y. Intermittent caloric restriction with a modified fasting-Mimicking diet ameliorates autoimmunity and promotes recovery in a mouse model of multiple sclerosis. J. Nutr. Biochem. 2021, 87, 108493. [Google Scholar]
  146. Bai, M.; Cao, P.; Lin, Y.; Yu, P.; Song, S.; Chen, L.; Wang, L.; Chen, Y. Intermittent caloric restriction promotes erythroid development and ameliorates phenylhydrazine-Induced anemia in mice. Front. Nutr. 2022, 9, 892435. [Google Scholar]
  147. Yang, L.; Li, D.X.; Cao, B.Q.; Liu, S.J.; Xu, D.H.; Zhu, X.Y.; Liu, Y.J. Exercise training ameliorates early diabetic kidney injury by regulating the h2 s/sirt1/p53 pathway. FASEB J. 2021, 35, e21823. [Google Scholar]
  148. Monno, I.; Ogura, Y.; Xu, J.; Koya, D.; Kitada, M. Exercise ameliorates diabetic kidney disease in type 2 diabetic fatty rats. Antioxidants 2021, 10, 1754. [Google Scholar]
  149. Sabet, N.; Soltani, Z.; Khaksari, M. The effects of exercise on kidney injury: The role of sirt1. Mol. Biol. Rep. 2022, 49, 4025–4038. [Google Scholar]
  150. Gajos-Draus, A.; Duda, M.; Beresewicz, A. Exercise and nitrite prevent and nomega-Nitrol-L-Arginine methyl ester reproduces imbalance in the nuclear factor-Kappab/nadph oxidase 2 and nuclear factor erythroid 2-Related factor 2/nadph oxidase 4/endothelial nitric oxide synthase systems in diabetes. J. Physiol. Pharmacol. 2021, 72. [Google Scholar] [CrossRef]
  151. Rojas-Morales, P.; León-Contreras, J.C.; Sánchez-Tapia, M.; Silva-Palacios, A.; Cano-Martínez, A.; González-Reyes, S.; Jiménez-Osorio, A.S.; Hernández-Pando, R.; Osorio-Alonso, H.; Sánchez-Lozada, L.G.; et al. A ketogenic diet attenuates acute and chronic ischemic kidney injury and reduces markers of oxidative stress and inflammation. Life Sci. 2022, 289, 120227. [Google Scholar]
  152. Han, Y.M.; Ramprasath, T.; Zou, M.H. Beta-Hydroxybutyrate and its metabolic effects on age-Associated pathology. Exp. Mol. Med. 2020, 52, 548–555. [Google Scholar]
  153. Kolb, H.; Kempf, K.; Rohling, M.; Lenzen-Schulte, M.; Schloot, N.C.; Martin, S. Ketone bodies: From enemy to friend and guardian angel. BMC Med. 2021, 19, 313. [Google Scholar]
  154. Wei, T.; Tian, W.; Liu, F.; Xie, G. Protective effects of exogenous beta-Hydroxybutyrate on paraquat toxicity in rat kidney. Biochem. Biophys. Res. Commun. 2014, 447, 666–671. [Google Scholar]
  155. Rojas-Morales, P.; Pedraza-Chaverri, J.; Tapia, E. Ketone bodies for kidney injury and disease. Adv. Redox Res. 2021, 2, 100009. [Google Scholar]
  156. Rojas-Morales, P.; Pedraza-Chaverri, J.; Tapia, E. Ketone bodies, stress response, and redox homeostasis. Redox Biol. 2020, 29, 101395. [Google Scholar]
  157. Islam, M.S.; Wilson, R.D. Experimentally induced rodent models of type 2 diabetes. Methods Mol. Biol. 2012, 933, 161–174. [Google Scholar]
Figure 1. Chemical structures of nicotinamide and streptozotocin. (A): nicotinamide; (B): streptozotocin.
Figure 1. Chemical structures of nicotinamide and streptozotocin. (A): nicotinamide; (B): streptozotocin.
Biomolecules 12 01225 g001
Figure 2. Diagrams showing representative flow charts of non-obese type 2 diabetes animal models induced by nicotinamide and streptozotocin. Renoprotective materials can be tested for their beneficial effects by this model, which is also outlined in the diagram. Note that for mice to be used as a model, more than one NA and STZ injection may be performed. Depending on the objective of a given study, the mouse model of NA-STZ diabetes induction may also involve HFD feeding for weeks before NA and STZ administrations (please see the text for a detailed discussion).
Figure 2. Diagrams showing representative flow charts of non-obese type 2 diabetes animal models induced by nicotinamide and streptozotocin. Renoprotective materials can be tested for their beneficial effects by this model, which is also outlined in the diagram. Note that for mice to be used as a model, more than one NA and STZ injection may be performed. Depending on the objective of a given study, the mouse model of NA-STZ diabetes induction may also involve HFD feeding for weeks before NA and STZ administrations (please see the text for a detailed discussion).
Biomolecules 12 01225 g002
Figure 3. Histopathology staining of DN. Periodic acid–Schiff (PAS)-stained renal sections of a non-diabetic control rat (A) and STZ/NAD (B) diabetic rat at week 12. Indicated are tubular epithelial cell necrosis (asterisk), thickening of tubular basement membrane (arrow). This figure was reproduced from Corremans et al. [48].
Figure 3. Histopathology staining of DN. Periodic acid–Schiff (PAS)-stained renal sections of a non-diabetic control rat (A) and STZ/NAD (B) diabetic rat at week 12. Indicated are tubular epithelial cell necrosis (asterisk), thickening of tubular basement membrane (arrow). This figure was reproduced from Corremans et al. [48].
Biomolecules 12 01225 g003
Figure 4. NA-STZ diabetic kidney histopathology stained by Masson trichrome. Kidney tissues were collected and processed for staining after 28 days of diabetes induction. This figure was reproduced from Arigela et al. [49].
Figure 4. NA-STZ diabetic kidney histopathology stained by Masson trichrome. Kidney tissues were collected and processed for staining after 28 days of diabetes induction. This figure was reproduced from Arigela et al. [49].
Biomolecules 12 01225 g004
Figure 5. Diagram summarizing the representative renoprotective mechanisms of the materials listed in Table 3, using the NA-STZ non-obese type 2 diabetes animal models. AGEs stands for “advanced glycation end products”.
Figure 5. Diagram summarizing the representative renoprotective mechanisms of the materials listed in Table 3, using the NA-STZ non-obese type 2 diabetes animal models. AGEs stands for “advanced glycation end products”.
Biomolecules 12 01225 g005
Figure 6. Potential mechanisms underlying diabetic nephropathy in the NA-STZ animal model. While common deleterious mechanisms operate in the kidney upon hyperglycemic challenge, those potential mitochondrial mechanisms underlying kidney injury remain to be elucidated (right side of the figure). These deleterious mechanisms would eventually converge on renal hypertrophy and renal fibrosis, leading to phenotype of diabetic nephropathy and kidney functional decline.
Figure 6. Potential mechanisms underlying diabetic nephropathy in the NA-STZ animal model. While common deleterious mechanisms operate in the kidney upon hyperglycemic challenge, those potential mitochondrial mechanisms underlying kidney injury remain to be elucidated (right side of the figure). These deleterious mechanisms would eventually converge on renal hypertrophy and renal fibrosis, leading to phenotype of diabetic nephropathy and kidney functional decline.
Biomolecules 12 01225 g006
Table 1. Pathophysiological stages of diabetic nephropathy.
Table 1. Pathophysiological stages of diabetic nephropathy.
Stage 1: Glomerular basement membrane thickening, normal GFR *, no urinary albumin; high blood pressure is often observed
Stage 2: Mild to severe mesangial expansion, increased mesangial matrix, normal GFR
Stage 3: Damaged glomerular and increased albuminuria can be observed. This stage is also known as nodular sclerosis
Stage 4: Advanced stage of glomerulosclerosis
Stage 5: Complete kidney failure, GFR is well below 15 mL/min/1.73 m2
* GFR: glomerular flow rate. This table is adapted from Agarwal R. [50] and Natesan V. et al. [11].
Table 2. Renal pathophysiology in the NA-STZ rodent model.
Table 2. Renal pathophysiology in the NA-STZ rodent model.
Rodent Model (NA/STZ, mg/kg)Analysis Time Point after NA/STZ InjectionsMeasured Renal PathophysiologyReference
Rat (200/55)2 monthsIncreased serum Cre and proteinuria, advanced glomerulosclerosis[51]
Rat (110/65)4 weeksIncreased kidney index and BUN, decreased NAD and ATP contents in renal cells, increased oxidative damage[52]
Rat (110/65)6 weeksIncreased renal triglycerides, enlarged Bowman’s capsule, Congested glomeruli, elevated serum Cre and BUN[53]
Rat (200/65)2 weeksIncreased renal vitamin A and C[54]
Rat (110/55)4 weeksIncreased BUN and serum creatinine, increased renal Oxidative stress, and decreased renal antioxidants[49]
Rat (230/65)30 daysIncreased levels of serum urea, uric acid, creatinine, and BUN[55]
Rat (200/55)21 daysIncreased urinary α1-macroglobulin excretion, increased serum uric acid and BUN, and enlarged glomerular diameter[56]
Rat (120/50)4 weeksElevated serum fructosamine, increased serum creatinine and urea[57]
Rat (100/50)8 weeksIncreased urinary N-acetyl-β-D-glucosaminidase, urea uric acid, and Cre[58]
Rat (110/65)6 weeksIncreased serum Cre, BUN, and uric acid[59]
Rat (110/65)6 weeksIncreased kidney–body index, elevated levels of serum Cre, BUN, uric acid, and urinary protein[60]
Mouse (180/60/HFD)8 weeksIncreased serum Cre and kidney–body index[61]
Rat (110/65)4 weeksIncreased serum Cre and BUN[62]
Rat (230/65)45 daysElevated levels of BUN, serum uric acid, and Cre[63]
Rat (230/65)8 weeksIncreased ratio of urinary albumin to urinary Cre[64]
Rat (230/65)12 weeksIncreased albuminuria and increased serum Cre[65]
Rat (110/65)35 daysIncrease in histological tubular injury[66]
Rat (230/65)30 daysIncreased levels of serum urea, uric acid, Cre, and BUN[67]
Rat (110/45)45 daysMultiple foci of hemorrhage, necrosis, and swelling tubules[68]
Rat (120/40)4 weeksIncreased BUN and serum Cre, increased kidney index and glomerular size[69]
Mouse (120/60)5 weeksIncreased levels of BUN, serum Cre, uric acid, and urea, elevated levels of urine protein[70]
Rat (110/65)40 daysIncreased levels of urea, uric acid, and Cre in the sera[71]
Rat (120/60)5 weeksIncreased renal tubular vacuolation and tubular degeneration[72]
Rat (120/60)45 daysIncreased levels of serum urea, uric acid, and Cre[73]
Rat (85/65)8 weeksIncreased serum glucose, urea, and Cre with albuminuria[74]
Rat (110/55)6 weeksIncrease in: Kim-1, serum Cre, BUN, uric acid, and urine albumin/Cre ratio[75]
Rat (110/55)6 weeksGlomerular and tubular injuries observed histochemically[76]
Rat (230/65)12 weeksIncreased serum Cre and albumin to Cre ratio, glomerular and tubular injury observed histochemically[48]
Rat (230/55)6 weeksIncreased serum Cre and BUN with decreased urine Cre[77]
Mouse (240/100/HFD)8 weeksIncreased microphage infiltration in the kidney[78]
Rat (230/65)45 daysIncreased levels of blood urea, uric acid, BUN, and Cre[79]
Rat (110/55)28 daysDecreased Cre clearance, increased BUN and uric acid, increased urine protein contents[80]
Rat (110/65)21 daysDecrased renal antioxidant power with increased renal oxidative damage[81]
Rat (230/65)12 weeksIncreased hemorrhage and neutrophils gathering in the kidney[82]
Rat (110/50)30 daysDecrease in Cre clearance, tubular lumen dilation, swelling of proximal tubular cells with tubular cell necrosis and intraluminal casts[83]
Rat (100/60) 4 weeksIncreased kidney index, increased urine albumin, thickening of the basement membrane of renal tubule[84]
Rat (110/45)45 daysIncreased levels of Cre and proteinuria, podocyte hypertrophy[85]
Mouse (120/100)4 weeksIncreased fibrotic deposition in the kidney[86]
Rat (120/60)60 daysIncreased urine volume and urine albumin, increased serum uric acid[87]
Rat (110/65)4 weeksTubules with vacuolated cells, glomerulai exhibiting mesangial thickening[88]
Rat (120/60)28 daysIncreased blood urea, glomerular enlargement, and sclerosis[89]
Rat (110/65)45 daysIncreased fatty acid contents in the kidney[90]
Rat (210/55)8 weeksIncreased BUN and serum Cre with elevated proteinuria[19]
Rat (110/50)6 weeksIncreased serum Cre, uric acid, and proteinuria, decrease in creatinine clearance[91]
Rat (110/55)28 daysIncreased BUN, serum creatinine, and uric acid with proteinuria[92]
Rat (100/55)28 daysIncreased serum Cre and urea, glomerular architecture deranged[93]
Abbreviations: BUN, blood urea nitrogen; Cre, creatinine; HFD, high fat diet.
Table 3. Renoprotective materials tested by the NA-STZ type 2 diabetes animal models.
Table 3. Renoprotective materials tested by the NA-STZ type 2 diabetes animal models.
Renoprotective
Materials
Rodent Model
(NA/STZ, mg/kg)
MechanismReference
1,8 CineoleRat (200/55)Glyoxalase-I induction[51]
Abroma augusta L leafRat (110/65)Inhibiting oxidative stress[52]
AcetateRat (110/65)Suppressing xanthine oxidase activity[53]
Abrus precatorius leafRat (110/60)Total antioxidant increase in kidney[98]
AscomycetesRat (200/65)Inhibiting oxidative stress[54]
BetaninRat (110/45)Antioxidative damage[99]
Bitter Gourd HoneyRat (110/55)Antioxidation, anti-inflammation[49]
Bocopa monnieriRat (230/65)Inhibiting AGEs formation[55]
Brucea javanica seedsRat (100/60)Inhibiting alpha-glucosidase[100]
Cichorium intybus L seedRat (200/55)Improving blood and urine parameters[56]
Citrus reticulate fruit peelRat (120/50)Antioxidative stress[57]
Combretum micranthumRat (100/50)Elevating SOD and catalase activities[58]
CoQ-10/metforminRat (110/65)Inhibiting oxidative stress[59]
CoQ-10/sitagliptinRat (110/65)Enhancing antioxidant system[60]
Cordyceps militarisMouse (180/60)Decreasing serum creatinine levels[61]
CrocinRat (110/65)Antioxidation[62]
Curculigo orchiodiesRat (230/65)Antioxidation, anti-hyperlipidemia[63]
DapagliflozinRat (230/65)Normalizing renal corpuscles histology[64]
Dapagliflozin/irbesartanRat (230/65)Inhibiting AGEs formation[65]
Dietary flaxseedRat (110/65)Antioxidative stress[66]
Dillenia Indica LRat (230/65)Inhibiting AGEs formation[67]
DiosminRat (110/45)Inhibiting oxidative stress[68]
Ellagic acid/pioglitazoneRat (175/65)Improving kidney function markers[101]
EmpagliflozinRat (120/40)Decreasing BUN, creatinine, and oxidative stress[69]
Eysenhardtia polystachyaMouse (120/60)Inhibiting glycation[70]
Garlic extractRat (110/65)Inhibiting oxidative stress[71]
Grain amaranthRat (120/60)Improving renal calcium metabolism[72]
GlycosinRat (120/60)Decreasing blood urea and creatinine[73]
Hypericum perforatumRat (85/65)Antioxidative stress[74]
Lipoic acidRat (110/55)Activating CSE/H2S pathway[75,76]
L-NAMERat (230/65)Increasing blood glucose[48]
Manilkara zapota extractRat (120/60)Reversing glomerulosclerosis[102]
MetforminRat (230/55)Decreasing BUN and serum creatinine[77]
Myrciaria caulifloraMouse (240/100)Inhibiting oxidative stress[78]
NaringeninRat (120/60)TRB3-FoxO1 downregulation[97]
Oligo-fucoidanHFD-Mouse (200/50)Activation of Nrf2 and Sirt1[41]
Paeonia emodiRat (230/65)Inhibiting glycation end products[79]
Phyllanthus niruri leavesRat (110/55)Antioxidative stress[80]
PioglitazoneRat (110/65)Antioxidation[81]
PomegranateRat (120/60)Antioxidative stress[103]
QuercetinRat (230/65)Anti-apoptosis[82]
ResveratrolRat (110/50)Attenuating oxidative stress[83,104]
RhinacanthinsRat (100/60)Inhibiting oxidative stress[84]
S-allylcysteineRat (110/45)Attenuating oxidative stress[85]
SGLT2 inhibitorsMouse (120/100)AMPK activation[86]
SilymarinRat (120/60)Lowering serum creatinine and uric acid[87]
StrawberryRat (110/65)Enhancing kidney antioxidant defense[88]
Syzygium calophyllifolium Rat(120/60)Enhancing kidney antioxidant defense[89]
TetrahydrocurcuminRat (110/65)Preventing fatty acid changes in the kidney[90]
TetramethylpyrazineRat (210/55)Akt signaling pathway activation[19]
Vanillic acidRat (110/50)Attenuating oxidative stress[91]
TilianinRat (110/55)Nrf2 signaling pathway activation[92]
Zanthoxylum
Zanthoxyloides extractRat (100/55)Improved kidney histology and biomarkers[93]
Note: This table is not meant to be exhaustive and only shows the materials tested for their renoprotective effects. Therefore, antidiabetic materials screened using this model but not focusing on diabetic nephropathy are not included in this table. AGEs = advanced glycation end products.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Yan, L.-J. The Nicotinamide/Streptozotocin Rodent Model of Type 2 Diabetes: Renal Pathophysiology and Redox Imbalance Features. Biomolecules 2022, 12, 1225. https://doi.org/10.3390/biom12091225

AMA Style

Yan L-J. The Nicotinamide/Streptozotocin Rodent Model of Type 2 Diabetes: Renal Pathophysiology and Redox Imbalance Features. Biomolecules. 2022; 12(9):1225. https://doi.org/10.3390/biom12091225

Chicago/Turabian Style

Yan, Liang-Jun. 2022. "The Nicotinamide/Streptozotocin Rodent Model of Type 2 Diabetes: Renal Pathophysiology and Redox Imbalance Features" Biomolecules 12, no. 9: 1225. https://doi.org/10.3390/biom12091225

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop