Deepthi R, Suhasin G. A Review on Animal Models of Chronic Kidney Disease- An Update. Biomed Pharmacol J 2023;16(3).
Manuscript received on :04-12-2021
Manuscript accepted on :23-01-2023
Published online on: 14-07-2023
Plagiarism Check: Yes
Reviewed by: Dr. Vega-Crespo, Agustin
Second Review by: Dr. Rajkishor Gogoi
Final Approval by: Dr. Ian James Martin

How to Cite    |   Publication History
Views Views: (Visited 489 times, 1 visits today)   Downloads PDF Downloads: 219

Deepthi R* and  Suhasin Ganta

Department of Pharmacology, GITAM Institute of Pharmacy, GITAM (Deemed to be University) Visakhapatnam 530045, Andhra Pradesh, India

Corresponding Author E-mail: deepthirayilla@gmail.com

DOI : https://dx.doi.org/10.13005/bpj/2711

Abstract

Chronic kidney disease is a global health burden and is an independent risk factor for morbidity and mortality for many diseases. The estimated prevalence of CKD worldwide is 9.1% (697.5 million cases) in all the age groups and about 1·2 million people died from CKD in 2017. Proteinuria and decreased glomerular filtration rate are the major indicators of the kidney damage.The need for reliable models for increasing prevalence is apparent. Animal models allow analysis of complex disease pathophysiology, for introducing new drugs and interventions in CKD. Studying various models will help in selection of appropriate model suitable for the cause of CKD.

Keywords

Alport Syndrome; CKD; Calcification; GFR; Nephrectomy; Polycystic Kidney

Download this article as: 
Copy the following to cite this article:

Deepthi R, Suhasin G. A Review on Animal Models of Chronic Kidney Disease- An Update. Biomed Pharmacol J 2023;16(3).

Copy the following to cite this URL:

Deepthi R, Suhasin G. A Review on Animal Models of Chronic Kidney Disease- An Update. Biomed Pharmacol J 2023;16(3). Available from: https://bit.ly/3XXDQkI

Introduction

Chronic kidney disease is defined as abnormalities of kidney structure or function, present for greater than 3 months. This is defined as a GFR less than 60 mL/min/1.73 m2 or one or more markers of kidney dysfunction including albuminuria.The pathophysiological mechanism underlying CKD includes initial trigger mediated through inflammation or immunological response or a toxicant. Hyper filtration and hypertrophy occurs later that contributes to progression of kidney damage (1).All these eventslead to secondary complications like diabetes, cardiac diseases, stroke, etc. (2). The increase in number of CKD cases is linked with the aging, increased prevalence of diabetes mellitus and hypertension,diabetesmellitus being the leading cause(3,4).CKD is believed to affect 10% to 15% of the population and is estimated to contribute to 5 to 10 million deaths annually(5). It is also observed that CKD imparts cardiovascular burden in the patients that may be because of intimal, medial and valvular calcification of arteries(5,6).The abnormal toxins accumulation in CKD patients can cause imbalances in renin angiotensin aldosterone system causing increased blood pressure, increased coagulation that in turn may pose a threat of myocardial infarction culminating in heart failure. This pathway is known as Cardio-renal syndrome(7).The findings from several studies also prove the linkage between CKD, small blood vessel diseases of cerebrum and impairment of cognition but the pathological mechanisms remain unclear(8). The non-specific interventions in established CKD result in reduction of the disease progression. Also, Early investigations through imaging and biopsy techniques and Targeting Specific underlying cause is also crucial for delivering proper care in the patients with CKD. Renoprotective agents- Angiotensin-converting enzyme II inhibitors and angiotensin II receptor blockers are considered as first line drugs in CKD irrespective of the underlying causes(both diabetic and non-diabetic). Drugs targeting renal fibrosis are still under investigations that can be promising agents for CKD treatment. Dietary sodium restriction and diuretic therapy reduce the fluid overload. Oral alkali therapy with sodium bicarbonate (1.5 to 3.0 g/day) slows the rate of progression(9). Impairment of immune response in CKD either leads to increased risk of infections where as exaggeration leads toinflammation along with its after effects. Targeting these immune abnormalities can also be an effective strategy in advanced CKD(10).

Understanding the underlying mechanisms for the development and progression of kidney diseases is important for finding the new treatments in this context. For achieving these goals both rodent and non rodent models are proven to be valuable investigating tools. Rodents, especially Rats are able to replicate the human histological manifestations of kidney diseases to larger extent making them as useful options(11). The present review focuses on animal models of CKD

Hypertension model of CKD

Hypertension is considered to be a cause and outcome of CKD, characterized by proteins in urine and sclerosis of glomeruli. This leads to reduced glomerular filtration. The histology of hypertensive CKD ischaracterized by renal inflammation and interstitial fibrosis (12).

Spontaneously hypertensive rat model

8-week-old male spontaneously hypertensive rats(SHR) can serve as better tool for understanding hypertension induced kidney injury (13). The uninephrectemised SHR rats are observed over a period of 40 weeks in presence or absence of drug treatments and at the end of 40th week, the urine samples are collected.The animals are then sacrificed; blood samples are collected followed by dissection of kidneys. Biochemical, histological and immunohistochemistry studies are performed. It is believed that in these models microinflammation is the cause of renal injury(14). Proteinuria, glomerulosclerosis, and interstitial fibrosis are observed. The pathology can be exaggerated by introducing high salt diet especially in male SHR rats (15). Oxidative stress appears in the beginning followed by inflammation(16).

5/6thNephrectomy model

Often called as sub total nephrectomy, is the best model for studying progressive renal failure with loss of renal mass(17). Tissue ablation or ligation of renal tissue induces renal failure(18). 12- weeks after establishing the model, drastic increase in blood urea nitrogen and proteinuria can be observed(19). This method requires expertise in surgical procedures.Nephrectomy-induces systolic arterial hypertension and thereby cause changes in remnant kidney tissue both structurally and functionally. Structural changes include glomerular hypertrophy, mesangial expansion, glomerular sclerosis, interstitial fibrosis and tubule-interstitial atrophy(20). This model is considered as an established model for glomerulonephritis and renal fibrosis especially in female wistar rats (21).

Deoxycorticosterone acetate salt hypertension induced model of CKD

Deoxycorticosterone acetate salt induces hypertension by increasing expression of TNF alpha there by increasing inflammation in renal tissue (22). This is a model of primary aldosteronism that also induces oxidative stress and renal fibrosis (23).Uninephrectomy performed 8 weeks old Sprague Dawley rats are given access to NaCl; and DOCA salt is administered subcutaneously that develop hypertension and fibrosis (24). This model lasts only for 8-12 weeks.so not a popular choice for modelling CKD.

Diabetic Nephropathy model of CKD

Diabetic nephropathy is the leading cause of CKD worldwide. It is characterized by renal inflammation and fibrosis, tissue remodeling along with oxidative stress (25,26). Glomerulosclerosis is also a characteristic feature of diabetic nephropathy, reflecting the accumulation of α-smooth muscle actin  and the epithelialmesenchymal transition. Diabetes can be induced by single intraperitoneal injection of 45mg/kg of Streptozotocin in 8-week old SD rats. Increased weight of kidneys, blood glucose, alanine transaminase, aspartate transaminase, catalase, superoxide dismutase and glutathione are the parameters to be assessed(27). Transforming Growth Factor-β1 is deemed to take a vital part in the deposition of Extracellular matrix, serum creatinine and blood urea nitrogen (BUN) levels in Diabetes(28). Addition of high fat diet to Steptozotocin can mimic human type 2 diabetes(insulin resistance). SD rats are fed with high fat diet containing 50 % fat in food for 9 weeks. Streptozotocin (35mg/kg) is injected i.p. at 4th and 6th week then plasma and renal parameters can beevaluated(29).

The AkitaIns2+ mice that has spontaneous point mutation in preproinsulin gene produces Type 1 Diabetes by direct pancreatic  beta cell toxicity is a model for studying progressive events of kidney disease including mesangial expansion and albuminuria. Non obese diabetic mice is characterized by autoimmune destruction of beta cells of pancreas thus produces renal injury in 40days due to mesangial expansion and podocyte loss. New Zealand Obese mouse (Type 2 diabetes along with obesityrelated to leptin resistance),Ob/ob, db/db and Zucker fatty rat models can be useful for studying Diabetic Nephropathy (30).

Drug induced Chronic Kidney disease

As surgical methods possess risk of mortality among the animals in the study, the use of inducing agents to produce chronic nephrotoxicity is considered as desired and beneficial option.

Adriamycin induced CKD

Adriamycin induces nephropathy similar to renal injury in humans. Glomerular damage, increased proteins in urine, segmental sclerosis and tubular interstitial fibrosis are the sequeale of renal events associated with adriamycin. All these events occur 6weeks after a single intravenous injection of Adriamycin (5mg/kg) in male albino rats (31, 32) or 20mg/kg i.p. in wistar rats(33). Adriamycin causes podocyte injury followed by expression of transforming growth factor beta-1 associated with glomerulosclerosis, depletion of podocytes and decreased renal function (34).

Adenine-induced model of CKD

This model is first explained in 1986 by Yokozawa et al. and is very much adopted in recent studies. Adenine when given either with diet or vehicle causes occlusion of renal tubules, ischemia and finally fibrosis leading to CKD progression and retardation of growth which is more relevant to CKD in human (35). In initial models, 0.75%w/w of adenine was given with diet, this is later modified to 0.5 or 0.25% w/w. Diet containing 0.25% adenine when fed for 35days in 9-10 weeks old Sprague Dawley rats, produces progressive CKD. Adenine increased plasma concentrations of inflammatory cytokines and decreased antioxidant levels(36,37) serum blood urea nitrogen, creatinine and uric acid are found to be increased with adenine diet(38). Adenine model is more advantageous over surgical model of CKD as it reduces mortality and inter-species difference during experimentation(39).

CKD in Aging rats

Aging is characterized by proteinuria, lesions in tubularinterstitium and cell damage. Thus is an important factor for development of end stage renal disease (40). Endoplasmic Reticulum stress and apoptosis are considered as the contributing factor for tubular cell injury. Rodents older than 20 to 24 months are considered to be aged and are suitable for the study (41). Short term high fat diet fed aged Sprague Dawley rats for 15 days can produce renal inflammation and fibrosis (42).

Autoimmune Chronic Kidney disease

Kidney can be target of autoimmunity resulting in nephritis caused by systemic lupus erythematosus in 35-55% of patients because of abnormal glomerular inflammation (43). Lupus Nephritis, that is one of the serious complication of Systemic Lupus Erythematosus causes activation of inflammatory cells and proliferation of local tissue that stimulate chemokine and cytokine release(44). Nephritis induced by IgA causes hematuria and glomerulonephritis involving mesangial cells. IgG anti-IgA formation induces inflammation that causes kidney failure (45). Heymann Nephritis is another form of immune mediated injury of glomerulus often termed as membraneousglomerulo-nephropathy. Its active model is produced by injection of isolated brush border components of rat’s proximal tubules into Lewis or fisher rats. Within 3 to 4 weeks, IgG deposition occur in glomerulus and proteinuria develops in 8-weeks(46). Whereas in passive model, antisera produced from antigen of another animal is injected to exhibit immune response (47). Injection of rabbit or mouse thymocyte serum through tail vein induces membrano proliferative glomerulonephritis that causes proliferation of mesangial cells and proteinuria with in 1-week. This model repairs on its own after 3 weeks whereas repeated injections cause progressive chronic kidney disease (12). Anti- Glomerular Basement Membrane model is another modelof glomerulonephritis induced by active immunization with isolated or recombinant collagen IV or by injection of anti GBM antibodies passively in Wistar Kyoto Rats (48,49).

Hereditary or genetic models of CKD

Alport syndrome is an inherited genetic disease that occurs due to mutation in genes encoding collagen IV α5 chain(COL4A5). Accumulation of these chains occurs in glomerular basement membrane and alters its function. The development of nephropathy is same in both sexes(50). Initially it is manifested as hematuria. Further collagen signallings causes albuminuria that further contribute to renal fibrosis(51). Polycystic kidney disease is hereditary disorder  characterized by abnormal cell proliferation, fluid accumulation, inflammation and renal fibrosis. Two types of Poly cystic kidney diseases include, Autosomal dominant polycystic kidney diseases and Autosomal recessive polycystic kidney diseases (52). Autosomal dominant polycystic kidney diseases is one of the common genetic cause of renal disease. It is associated with mutations in genes of polycystic kidney disease (PKD1 & PKD2). The Han:SPRD-Cy rat strain is one of the spontaneous hereditary model for PKD characterized by large number of cysts formed by missense mutations. Another rodent model includes, PCK rats that are discovered from Sprague Dawley rats outbreeding, Pkhd1 being the responsible gene. Pcy(polycystic) mouse derived from KK strain produces missense mutation in gene similar to human Nphp3 is also one of the hereditary model for PKD(53). Crj:CD/SD is another homozygous mutant model for PKD that develop renal cysts within one week after birth where asbpk (BALB/c polycystic kidney) mice model, homozygous mutants develop renal cysts and die within 4 weeks after birth. Although many models of PKD share similar pathological features of end stage renal diseases, understanding molecular mechanisms and identifying novel drug targets still remain as a challenge.(54)Podocyte-specific genetic model for focal segmental glomerular sclerosis(Nep 25 mice) is produced by administration of immunotoxin specific to podocytes under nephrin promoter in mouse causing intraglomerular injury. Human immunodeficient virus associated Nephropathy, is produced in Tg26 mice (it has replication deficient HIV transgene) characterized by severe proteinuria, ascites, low levels of urinary albumin and mesangial hyperplasia especially on FVB/N, C57BL/6, 129/Sv  strain backgrounds (55).

Recently role of Sirtuins in development of renal diseases has gained attention. Various body organs especially kidneys are vulnerable to age related damage and also injuries occurring due to toxic substances. Silent information regulators are NAD+ dependent deacetylases( consisting of 7 isoforms-SIRT1 to SIRT7). SIRT1 gene is highly expressed in nucleus and cytoplasm of fetal and adult tissues like Liver, Kidney, Brain.  High glucose concentrations can increase expression of p53 and cleaved caspase-3 in renal epithelium. This stimulates expression of SIRT1 by medullary mesenchymal cells that inturn reverses the p53 levels. Thus SIRT1 inhibits podocyte apoptosis.Thus SIRT1 has a significant role in Diabetic kidney disease. Additionally it regulates the TGFbeta/Smad pathway thereby inhibits kidney fibrosis. It also has role in regulation of renal inflammation in diabetic nephropathy through TNF alpha and COX-2 gene downregulation. Thus upregulation of SIRT1 activity can inhibit renal cell apoptosis.(63,64,65) Podocyte-specific Sirt1 knockout mice model is useful to study the role of sirtuins in Diabetic nephropathy where it inactivates p65 subunit of NF-kB and STAT3 and suppresses podocyte dysfunction.(66)

In addition, Klotho, an antiageing gene has been involved with cellular senescence. Deficiency of klotho gene induces oxidative stress, associated with multiple disorders like atherosclerosis, infertility, osteoporosis, cognitive decline etc. Klotho deficient mice can be a useful tool for studying premature ageing like syndrome-altered glucose, lipid and amyloid beta metabolism.(67) Low klotho expression is associated with raised Fibroblast growth factor(FGF23) that causes advanced CKD associated with cardiovascular complications such as vascular calcification, Left ventricular atrophy and cardiac fibrosis.(68)

Table 1: Animal models for chronic kidney diseases.

 

MODEL

ANIMAL/STRAIN

CHANGES IN HISTOLOGY

REFERENCE

1

Hypertension models of CKD

 

 

 

A

Spontaneously hypertensive rat model:

7 to 8 weeks old male SHR rats

Glomerulosclerosis, and interstitial fibrosis 

15,55,56

B

5/6 Nephrectomy model:

Male Sprague–Dawley rats, female wistar rats

Mesangial expansion, glomerular sclerosis, interstitial fibrosis and tubule-interstitial atrophy.

20,21,57

C

Deoxycorticosterone acetate salt hypertension-induced model

8- weeks old Sprague Dawley rats

renal fibrosis

23,24

2

Diabetic Nephropathy model of CKD

 

 

 

 

 

 8-week old SD rats(STZ 45mg/kg I.P)

Glomerulosclerosis

27

 

 

Zucker fatty rat

Renal interstitial fibrosis and glomerulosclerosis 

58

 

 

db/db

Glomerulosclerosis,

59

 

 

Ob/ob

Podocyte apoptosis

60

 

 

Non Obese mouse

Glomerular lesions

61

3

Drug-induced Chronic Kidney disease

 

 

 

A

Adriamycin induced CKD

Adriamycin (5mg/kg) in male albino rats. 20mg/kg i.p. in wistar rats.

Podocyte injury, glomerulosclerosis

32,33

B

 Adenine-induced model of CKD

9-10 weeks old Sprague Dawley rats

Renal fibrosis

34

4

CKD in Aging rats

24-months old male Sprague Dawley rats

Tubularinterstitial lesions,

Glomerulosclerosis

40,55

5

Autoimmune Chronic Kidney disease

 

 

 

A

Lupus Nephritis

MRL/lpr and NZB/W mice

Renal fibrosis

55

B

IgA  induced Nephritis

 

Mesangioproliferative glomerulonephritis

55

C

Heymann Nephritis

Lewis or fisher rats

Membranous nephropathy 

62

D

Anti GBM Nephritis

Wistar Kyoto Rats

Membranous nephropathy 

49

6

Hereditary or genetic models of CKD

 

 

 

A

Alport syndrome

Alport mice

Tubularinterstitial lesions,

Glomerulosclerosis

55

B

Polycystic kidney disease

Han:SPRD-Cy rat

Intraglomerular injury

53,54

 

 

 PCK rats

Intraglomerular injury

53,54

 

 

Pcy mouse

Intraglomerular injury

53,54

 

 

bpk (BALB/c polycystic kidney) mice

Intraglomerular injury

53,54

Conclusion

Chronic kidney disease is becoming a common disease with greater prevalence associated various etiologies, majorly with Diabetes and Hypertension. Immune mediated responses also account for many cases of CKD worldwide. Hence there is an immediate need for modeling kidney diseases that approximate to human pathology. Animal models, especially mice and rats are convenient for evaluating the novel drugs in chronic kidney diseases. Even after the discovery of various models for CKD, no single model exactly reflects human CKD. Strain, genetics and non-detectable effects of CKD can be limitations of animal studies, Thus progressive efforts are put forward in this direction to create new models or improve existing ones. Presence of co-morbidities, age, causative factors of kidney disease are to be taken into consideration while designing new models. Further advancement in molecular biology techniques are useful in understanding CKD molecular level pathogenesis and its complications which further aid in developing new transgenic models. The present review provides concise information on animal models of CKD along with their histological data for discovering new leads for CKD.

Conflict of Interest

There are no conflicts of Interest.

Funding Sources

There is no conflict of interest.

References

  1. Cornelia Charles, Allison H. Ferris. Chronic Kidney Disease, Primary Care: Clinics in Office Practice. 2020;47(8):585-595.
    CrossRef
  2. Xinling Song, Hui Pang, Weijun Cui, Jianjun Zhang, Jian Li, Le Jia. Renoprotective effects of enzyme-hydrolyzed polysaccharides from Auriculariapolytricha on adenine-induced chronic kidney diseases in mice. Biomedicine & Pharmacotherapy. 2021;135: 111004.
    CrossRef
  3. Yen-Cheng Chen, Chung-Yi Cheng, Chung-Te Liu, Yuh-Mou Sue, Tso-Hsiao Chen, Yung-Ho Hsu, Nai-Jen Huang, Cheng-Hsien Chen. Combined protective effects of oligo-fucoidan, fucoxanthin, and L-carnitine on the kidneys of chronic kidney disease mice. European Journal of Pharmacology. 2021; 892:173708.
    CrossRef
  4. SidarCopur, Emine M. Onal, BarisAfsar, Alberto Ortiz, Daniel H. van Raalte, David Z. Cherney, Peter Rossing, Mehmet Kanbay. Diabetes mellitus in chronic kidney disease: Biomarkers beyond HbA1c to estimate glycemic control and diabetes-dependent morbidity and mortality. Journal of Diabetes and its Complications. 2020; 34(11):107707.
    CrossRef
  5. Adam J. Nelson, Paolo Raggi, Myles Wolf, Alexander M. Gold, Glenn M. Chertow, Matthew T. Roe.Targeting Vascular Calcification in Chronic Kidney Disease. JACC: Basic to Translational Science. 2020;5(4):398-412
    CrossRef
  6. Palit S, Kendrick J. Vascular calcification in chronic kidney disease: role of disordered mineral metabolism. Curr Pharm Des. 2014;20(37):5829-5833.
    CrossRef
  7. Xiaorong Han, Shuai Zhang, Zhongbo Chen, Binay Kumar Adhikari, Ying Zhang, Jin Zhang, Jian Sun, Yonggang Wang. Cardiac biomarkers of heart failure in chronic kidney disease. ClinicaChimicaActa. 2020; 510:298-310.
    CrossRef
  8. Mark Fisher,Mechanisms of Cerebral Microvascular Disease in Chronic Kidney Disease. Journal of Stroke and Cerebrovascular Diseases. 2020;105404.
    CrossRef
  9. Charlie Tomson, Samuel Duffy. Management of chronic kidney disease. Medicine. 2019;47(9): 567-575.
    CrossRef
  10. Maaz Syed-Ahmed, MohanramNarayanan,Immune Dysfunction and Risk of Infection in Chronic Kidney Disease. Advances in Chronic Kidney Disease. 2019;26(1): 8-15.
    CrossRef
  11. Dai, C., Kiss, L. P., & Liu, Y. Animal Models of Kidney Diseases. Sourcebook of Models for Biomedical Research. 2008;657–664.
    CrossRef
  12. Zahraa Mohammed-Ali, Rachel E. Carlisle, SameraNademi, Jeffrey G. Dickhout, Chapter 16 – Animal Models of Kidney Disease. 2017;379-417.
    CrossRef
  13. Luo W-m, Kong J, Gong Y, Liu X-q, Yang R-x, Zhao Y-x. Tongxinluo Protects against Hypertensive Kidney Injury in Spontaneously-Hypertensive Rats by Inhibiting Oxidative Stress and Activating Forkhead Box O1 Signaling. PLoS ONE.2015; 10(12): e0145130.
    CrossRef
  14. Hai-Yan Xue, Li Yuan, Ying-Jie Cao, Ya-Ping Fan, Xiao-Lan Chen, Xin-ZhongHuang.Resveratrol ameliorates renal injury in spontaneously hypertensive rats by inhibiting renal micro-inflammation. Biosci Rep. 2016; 36 (3): e00339.
    CrossRef
  15. David A. Blizard, Wanda N. Peterson, Samy S. Iskandar, Z. K. Shihabi & Nelson Adams. The Effect of a High Salt Diet and Gender on Blood Pressure, Urinary Protein Excretion and Renal Pathology in Shr Rats. Clinical and Experimental Hypertension Part A: Theory and Practice. 1991;13(5):687-697.
    CrossRef
  16. Subrata K. Biswas, Jose B. Lopes De Faria, Subrata K. Biswas & Jose B. Lopes De Faria. Which comes first: Renal inflammation or oxidative stress in spontaneously hypertensive rats?. Free Radical Research. 2007;41(2):216-224.
    CrossRef
  17. Hewitson TD, Ono T, Becker GJ. Small animal models of kidney disease: a review. Methods Mol Biol. 2009;466:41-57.
    CrossRef
  18. Mumna Al Banchaabouchi, Bart Marescau, Rudi D’Hooge, Eric Van Marck, Andre Van Daele, Olivier Levillain, Peter Paul De Deyn. Biochemical and histopathological changes in nephrectomized mice.Metabolism.1998;47(3):355-361.
    CrossRef
  19. Rui-Zhi Tan, Xia Zhong, Jian-Chun Li, Yu-Wei Zhang, Ying Yan, Yuan Liao, Dan Wen, HuiDiao, Li Wang & Hong-Chun Shen.An optimized 5/6 nephrectomy mouse model based on unilateral kidney ligation and its application in renal fibrosis research.Renal Failure.2019;41(1): 555-566.
    CrossRef
  20. Jorge Montes-Rivera, Mónica Arellano-Mendoza, NayelliNájera, Leonardo Del Valle-Mondragón, Francisco Villarreal, Ivan Rubio-Gayosso, Javier Perez-Duran, Eduardo Meaney, Guillermo Ceballos.Effect of (-)-epicatechin on the modulation of progression markers of chronic renal damage in a 5/6 nephrectomy experimental model.Heliyon.2019;5(4):e01512.
    CrossRef
  21. Christian Fleck, Dorothea Appenroth, Patrick Jonas, Mark Koch, GüntherKundt, Horst Nizze, Günter Stein.Suitability of 5/6 nephrectomy (5/6NX) for the induction of interstitial renal fibrosis in rats – Influence of sex, strain, and surgical procedure. Experimental and Toxicologic Pathology.2006;57(3):195-205.
    CrossRef
  22. Ashok Jadhav, EminaTorlakovic, and Joseph FomusiNdisang. Hemin therapy attenuates kidney injury in deoxycorticosterone acetate-salt hypertensive rats. American Journal of Physiology-Renal Physiology.2009;296(3):F521-F534.
    CrossRef
  23. Ndisang JF, Lane N, Jadhav A. Crosstalk between the hemeoxygenase system, aldosterone, and phospholipase C in hypertension. J Hypertens. 2008;26(6):1188-99. 
    CrossRef
  24. Jadhav A, Torlakovic E, Ndisang JF. Interaction among hemeoxygenase, nuclear factor-κB, and transcription activating factors in cardiac hypertrophy in hypertension. Hypertension.2008;52: 910–917.
    CrossRef
  25. Lu Jiandong, Yilong Yang, JintingPeng, Min Xiang, Dongcai Wang, GuoliangXiong, ShunminLi,Trichosantheskirilowii.lectin ameliorates streptozocin-induced kidney injury via modulation of the balance between M1/M2 phenotype macrophage.Biomedicine& Pharmacotherapy.2019;109:93-102.
    CrossRef
  26. Benhong Zhou, Qiaoling Li, Jing Wang, Peng Chen, Shan Jiang.Ellagic acid attenuates streptozocin induced diabetic nephropathy via the regulation of oxidative stress and inflammatory signaling,Food and Chemical Toxicology.2019;123:16-27
    CrossRef
  27. Amit Kundu, PrasantaDey, Pradipta Sarkar, SanmoyKarmakar, In Hwan Tae, KyeongSeok Kim, Jae Hyeon Park, Su Hyun Lee, Byung Mu Lee, LalmuanawmiRenthlei, ZothanPuia, HyungSik Kim. Protective effects of Croton hookeri on streptozotocin-induced diabetic nephropathy.Food and Chemical Toxicology.2020;135:110873.
    CrossRef
  28. Li Jie, QiuPengcheng, He Qiaoyan, Bi Linlin, Zhang Meng, Wang Fang, Jia Min, Yan Li, Zhang Ya, Yang Qian, Wang Siwang.Dencichine ameliorates kidney injury in induced type II diabetic nephropathy via the TGF-β/Smadsignalling pathway.European Journal of Pharmacology.2017; 812:196-205.
    CrossRef
  29. Solomon OladapoRotimi, OluwakemiAnuoluwapoRotimi, Isaacson BababodeAdelani, ChinonyeOnuzulu, Patience Obi, RotimiOkungbaye.Stevioside modulates oxidative damage in the liver and kidney of high fat/low streptozocin diabetic rats. Heliyon.2018;4(5):e00640.
    CrossRef
  30. Anna Giralt-Lopez ,Mireia Molina, Van den Bosch. Revisiting Experimental Models of Diabetic Nephropathy. Int. J. Mol. Sci. 2020; 21:3587.
    CrossRef
  31. EmanMostafaSadek, Nagla Mohamed Salama, Dalia Ibrahim Ismail, Asmaa Ahmed Elshafei.Histological study on the protective effect of endogenous stem-cell mobilization in Adriamycin-induced chronic nephropathy in rats.Journal of Microscopy and Ultrastructure.2016;4(3):133-142.
    CrossRef
  32. Minggang Wei, Wei Sun, Weiming He, Li Ni, XiaofengCai, Zongqi Cheng, Kun Gao, Fengling Li, Lin Chen, XinpingZhang.Qiguiyishen decoction reduced the accumulation of extracellular matrix in the kidneys of rats with adriamycin-induced nephropathy.Journal of Traditional Chinese Medicine.2014;34(3):351-356.
    CrossRef
  33. Sachinthi S. Amarasiri, Anoja P. Attanayake, Liyanagae D.A.M. Arawwawala, Kamani A.P.W. Jayatilaka, Lakmini K.B. Mudduwa.Protective effects of three selected standardized medicinal plant extracts used in Sri Lankan traditional medicine in adriamycin induced nephrotoxic Wistarrats.Journal of Ethnopharmacology.2020;259:112933.
    CrossRef
  34. Yitian Dou, Yichun Shang, YongmeiShen, JingtianQu, Chunliu Liu, JiasongCao.Baicalin alleviates adriamycin-induced focal segmental glomerulosclerosis and proteinuria by inhibiting the Notch1-Snail axis mediated podocyteEMT.Life Sciences.2020;257:118010.
    CrossRef
  35. Badreldin H. Ali, Mohammed Al Za’abi, Sirin A. Adham, Yousuf Al Suleimani, TuranKaraca, PriyadarsiniManoj, Jamila Al Kalbani, JavidYasin, AbderrahimNemmar.The effect of sildenafil on rats with adenine—Induced chronic kidney disease.Biomedicine& Pharmacotherapy.2018;108:391-402.
    CrossRef
  36. Diwan, V., Brown, L. and Gobe, G.C..Adenine‐induced chronic kidney disease in rats. Nephrology.2018; 23: 5-11.
    CrossRef
  37. Aly M. Abdelrahman, Yousuf Al Suleimani, Mohammed Al Za’abi, Mohammed Ashique, PriyadarsiniManoj, Christina Hartmann, AbderrahimNemmar, Nicole Schupp, Badreldin H. Ali.Therenoprotective effect of the dipeptidyl peptidase-4 inhibitor sitagliptin on adenine-induced kidney disease in rats. Biomedicine & Pharmacotherapy.2019;110:667-676.
    CrossRef
  38. Anshuk Sharma, Richa Thakur, Madhu C. Lingaraju, Dhirendra Kumar, KarikalanMathesh, Avinash G. Telang, Thakur Uttam Singh, Dinesh Kumar.Betulinic acid attenuates renal fibrosis in rat chronic kidney disease model.Biomedicine& Pharmacotherapy.2017;89:796-804.
    CrossRef
  39. Amarasiri S S, Attanayake A P, Jayatilaka K A, Mudduwa L K. Animal models of chronic kidney disease: Screening tool to investigate nephroprotective effects of natural products. Int J Pharm Chem Anal.2018;5(2):52-58
    CrossRef
  40. Naoko Takeda, Shinji Kume, Yuki Tanaka, YoshikataMorita,Masami Chin-Kanasaki, Hisazumi Araki, KeijiIsshiki,Shin-ichiAraki,MasakazuHaneda,y Daisuke Koya,zAtsunoriKashiwagi,HiroshiMaegawa, and Takashi Uzu.Altered Unfolded Protein Response Is Implicated in the Age-Related Exacerbation of Proteinuria-Induced Proximal Tubular Cell Damage.American journal of pathology.2013;183:774-785.
    CrossRef
  41. Shixin Ding, Han Zhang, Zhenghao Sun, Yuli Han, Yan Li, Xianan Dong, Yanyan Yin, Weiping Li, WeizuLi.Ginsenoside Rg1 protects against aging-induced renal interstitial fibrosis due to inhibition of tubular epithelial cells endoplasmic reticulum stress in SAMP8 mice.Journal of Functional Foods.2020;72:104049.
    CrossRef
  42. Sugyeong Ha, Min Jo Kim, Dae Hyun Kim, Byeong Moo Kim, Ki Wung Chung, Hae Young Chung.Short-term intake of high fat diet aggravates renal fibrosis in aged Sprague-Dawleyrats.Experimental Gerontology.2020;142:111108.
    CrossRef
  43. Jing Gong, Ami Tamhaney, MohanrajSadasivam, Hamid Rabb, Abdel Rahim A.Hamad.Chapter 68 – Autoimmune Diseases in the Kidney,Editor(s): Noel R. Rose, Ian R. Mackay.The Autoimmune Diseases (Sixth Edition),Academic Press.2020;1355-1366.
    CrossRef
  44. MårtenSegelmark, Thomas Hellmark. Autoimmune kidney diseases. Autoimmunity Reviews.2010;9:A366–A371.
    CrossRef
  45. Alberto de Zubiria Salgado, Catalina Herrera-Diaz. Lupus Nephritis: An Overview of Recent Findings.Autoimmune Diseases.2012;
    CrossRef
  46. J.P. Gaut. Immune-Mediated Glomerular Injury. Pathobiology of Human Disease. Academic Press:2014; 2788-2801.
    CrossRef
  47. Jefferson JA, Pippin JW, Shankland SJ. Experimental Models of Membranous Nephropathy. Drug Discov Today Dis Models. 2010;7(1-2):27-33.
    CrossRef
  48. Mary H. Foster.Optimizing the translational value of animal models of glomerulonephritis: insights from recent murine prototypes, Am J Physiol Renal Physiol.2016; 311: F487–F495.
    CrossRef
  49. Dorin-Bogdan Borza and Billy G. Hudson.Of mice and men: Murine models of anti-GBM antibody nephritis. Kidney International.2002; 61:1905–1906.
    CrossRef
  50. Kim M, Piaia A, Shenoy N, Kagan D, Gapp B, Kueng B, Delphine Weber, William Dietrich, IwonaKsiazek. Progression of Alport Kidney Disease in Col4a3 Knock Out Mice Is Independent of Sex or Macrophage Depletion by Clodronate Treatment. PLoS ONE.2015;10(11): e0141231.
    CrossRef
  51. Kashtan C. Alport syndrome: facts and opinions. F1000Res.2017;6:50.
    CrossRef
  52. Shizuko Nagao, Masanori Kugita, Daisuke Yoshihara, and Tamio Yamaguchi. Animal Models for Human Polycystic Kidney Disease.Exp. Anim. 2012; 61(5):477–488.
    CrossRef
  53. Emilie Cornec-Le Gall, Vicente E. Torres, Peter C. Harris.Genetic Complexity of Autosomal Dominant Polycystic Kidney and Liver Diseases.JASN.2018;29 (1):13-23. 
    CrossRef
  54. Evelyne Fischer, Lionel Gresh, Andreas Reimann, Marco Pontoglio.Cystic kidney diseases: learning from animal models. Nephrology Dialysis Transplantation.2004;19(11):2700–2702. 
    CrossRef
  55. Yang HC, Zuo Y, Fogo AB. Models of chronic kidney disease. Drug Discov Today Dis Models. 2010;7(1-2):13-19.
    CrossRef
  56. Jane F. Reckelhoff, Huimin Zhang, Joey P. Granger.Decline in Renal Hemodynamic Function in Aging SHR.Hypertension. 1997;30:677–681.
    CrossRef
  57. Kohei Hayashi, TakaomiShimokawa, Masayo Yamagata, Kozo Yoneda.Inhibition of α2-adrenoceptor is renoprotective in 5/6 nephrectomy-induced chronic kidney injury rats.Journal of Pharmacological Sciences.2021;145(1): 79-87.
    CrossRef
  58. Zijian Wang, Qingbo Liu, Wendi Dai, Bing Hua, Hongwei Li, WeipingLi.Pioglitazonedownregulates Twist-1 expression in the kidney and protects renal function of Zucker diabetic fatty rats.Biomedicine& Pharmacotherapy.2019;118:109346.
    CrossRef
  59. YanliGuo, Zheng Ran, Yongwei Zhang, Zhipeng Song, Lifeng Wang, Lan Yao, Minfang Zhang, JialiangXin, XinminMao.Marein ameliorates diabetic nephropathy by inhibiting renal sodium glucose transporter 2 and activating the AMPK signaling pathway in db/db mice and high glucose–treated HK-2 cells.Biomedicine& Pharmacotherapy. 2020;131:110684.
    CrossRef
  60. Zhi-Wei Dai, Ke-Dan Cai, Ling-CangXu, Lai-Liang Wang.Perilipin2 inhibits diabetic nephropathy-induced podocyte apoptosis by activating the PPARγ signaling pathway.Molecular and Cellular Probes.2020;53: 101584.
    CrossRef
  61. Segev Y, Landau D, Marbach M, Shehadeh N, Flyvbjerg A, Phillip M. Renal hypertrophy in hyperglycemic non-obese diabetic mice is associated with persistent renal accumulation of insulin-like growth factor I. J Am SocNephrol. 1997 Mar;8(3):436-44.
    CrossRef
  62. J.P. Gaut,Immune-Mediated Glomerular Injury.Academic Press.2014;2788-2801.
    CrossRef
  63. Yi-jun Dong, Nian Liu, Zhi Xiao, Tao Sun, Shu-hui Wu, Wei-xia Sun, Zhong-gao Xu, Hang Yuan, “Renal Protective Effect of Sirtuin 1”, Journal of Diabetes Research, vol. 2014.
    CrossRef
  64. Guan Y, Hao C, -M: SIRT1 and Kidney Function. Kidney Dis 2015;1:258-265.
    CrossRef
  65. Hong, Yu Ah & Kim, Ji Eun & Jo, Minjee. (2020). The Role of Sirtuins in Kidney Diseases. International journal of molecular sciences. 21. 10.3390/ijms21186686.
    CrossRef
  66. Shu Wakino, Kazuhiro Hasegawa, Hiroshi Itoh,Sirtuin and metabolic kidney disease,Kidney International,Volume 88, Issue 4,2015,Pages 691-698.
    CrossRef
  67. Anti-Aging Genes Improve Appetite Regulation and Reverse Cell Senescence and Apoptosis in Global Populations. Advances in Aging Research, 2016, 5, 9-26.
    CrossRef
  68. Lu X, Hu M, C: Klotho/FGF23 Axis in Chronic Kidney Disease and Cardiovascular Disease. Kidney Dis 2017;3:15-23.
    CrossRef
Share Button
(Visited 489 times, 1 visits today)

Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 International License.