1
|
Rodríguez-Rodríguez DR, Mendoza-Hernández OH, Cordero-Pérez P, Rivas-Galindo VM, Moreno-Peña DP, Tijerina-Márquez R, Garza-Villarreal AM, Alarcón-Galván G, Muñoz-Espinosa LE, Zapata-Chavira HA, Hernández-Guedea MA, Solis-Cruz GY, Torres-González L. Nephroprotective and Antioxidant Effects of Jatropha dioica Extract Against Ischemia-Reperfusion Injury in Wistar Rats. Int J Mol Sci 2025; 26:1838. [PMID: 40076464 PMCID: PMC11899379 DOI: 10.3390/ijms26051838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/19/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Plant extracts with antioxidant activities have shown nephroprotection against IR injury. Jatropha dioica (Jd) possesses antioxidant activity. Our aim was to evaluate the effects of a hydroalcoholic Jd extract against IR injury in Wistar rats. Rats were divided into groups (n = 6): sham (SH); no toxicity (JdTox; 300 mg/kg/day of extract for 7 days); IR (on day 7 [I: 45 min/R: 24 h]); and Jd+IR (same treatment as JdTox; same surgical procedure as IR). AST and LDH were significantly lower in the JdTox. IR exhibited significantly higher CrS, BUN, and MDA compared with SH; Jd+IR showed reductions in these markers. GSH and SOD levels were significantly lower in IR compared with SH; an increase in these markers was observed in the Jd+IR. Histologically, IR showed significant increases in medullary tubular necrosis, medullary protein casts, and medullary vascular congestion compared with SH and JdTox. In Jd+IR, a significant decrease was observed only in medullary tubular necrosis. Therefore, the evaluated hydroalcoholic Jd extract dose showed no nephrotoxicity and hepatotoxicity. Jd extract pretreatment attenuated IR-induced renal injury, as evidenced by the improved serum markers of renal damage and oxidative stress.
Collapse
Affiliation(s)
- Diana Raquel Rodríguez-Rodríguez
- Liver Unit, Department of Internal Medicine, University Hospital “Dr. José E. González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (D.R.R.-R.); (O.H.M.-H.); (D.P.M.-P.); (R.T.-M.); (A.M.G.-V.); (L.E.M.-E.)
| | - Oscar Humberto Mendoza-Hernández
- Liver Unit, Department of Internal Medicine, University Hospital “Dr. José E. González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (D.R.R.-R.); (O.H.M.-H.); (D.P.M.-P.); (R.T.-M.); (A.M.G.-V.); (L.E.M.-E.)
| | - Paula Cordero-Pérez
- Liver Unit, Department of Internal Medicine, University Hospital “Dr. José E. González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (D.R.R.-R.); (O.H.M.-H.); (D.P.M.-P.); (R.T.-M.); (A.M.G.-V.); (L.E.M.-E.)
| | - Verónica Mayela Rivas-Galindo
- Department of Analytical Chemistry, School of Medicine, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (V.M.R.-G.); (G.Y.S.-C.)
| | - Diana Patricia Moreno-Peña
- Liver Unit, Department of Internal Medicine, University Hospital “Dr. José E. González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (D.R.R.-R.); (O.H.M.-H.); (D.P.M.-P.); (R.T.-M.); (A.M.G.-V.); (L.E.M.-E.)
| | - Ramiro Tijerina-Márquez
- Liver Unit, Department of Internal Medicine, University Hospital “Dr. José E. González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (D.R.R.-R.); (O.H.M.-H.); (D.P.M.-P.); (R.T.-M.); (A.M.G.-V.); (L.E.M.-E.)
| | - Alondra Michelle Garza-Villarreal
- Liver Unit, Department of Internal Medicine, University Hospital “Dr. José E. González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (D.R.R.-R.); (O.H.M.-H.); (D.P.M.-P.); (R.T.-M.); (A.M.G.-V.); (L.E.M.-E.)
| | - Gabriela Alarcón-Galván
- Basic Science Department, School of Medicine, Universidad de Monterrey, Monterrey 66238, Nuevo León, Mexico;
| | - Linda Elsa Muñoz-Espinosa
- Liver Unit, Department of Internal Medicine, University Hospital “Dr. José E. González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (D.R.R.-R.); (O.H.M.-H.); (D.P.M.-P.); (R.T.-M.); (A.M.G.-V.); (L.E.M.-E.)
| | - Homero Arturo Zapata-Chavira
- Transplant Service, University Hospital “Dr. José E. González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (H.A.Z.-C.); (M.A.H.-G.)
| | - Marco Antonio Hernández-Guedea
- Transplant Service, University Hospital “Dr. José E. González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (H.A.Z.-C.); (M.A.H.-G.)
| | - Guadalupe Yazmín Solis-Cruz
- Department of Analytical Chemistry, School of Medicine, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (V.M.R.-G.); (G.Y.S.-C.)
| | - Liliana Torres-González
- Liver Unit, Department of Internal Medicine, University Hospital “Dr. José E. González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (D.R.R.-R.); (O.H.M.-H.); (D.P.M.-P.); (R.T.-M.); (A.M.G.-V.); (L.E.M.-E.)
| |
Collapse
|
2
|
Zhang T, Widdop RE, Ricardo SD. Transition from acute kidney injury to chronic kidney disease: mechanisms, models, and biomarkers. Am J Physiol Renal Physiol 2024; 327:F788-F805. [PMID: 39298548 DOI: 10.1152/ajprenal.00184.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/15/2024] [Accepted: 09/01/2024] [Indexed: 09/22/2024] Open
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are increasingly recognized as interconnected conditions with overlapping pathophysiological mechanisms. This review examines the transition from AKI to CKD, focusing on the molecular mechanisms, animal models, and biomarkers essential for understanding and managing this progression. AKI often progresses to CKD due to maladaptive repair processes, persistent inflammation, and fibrosis, with both conditions sharing common pathways involving cell death, inflammation, and extracellular matrix (ECM) deposition. Current animal models, including ischemia-reperfusion injury (IRI) and nephrotoxic damage, help elucidate these mechanisms but have limitations in replicating the complexity of human disease. Emerging biomarkers such as kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), and soluble tumor necrosis factor receptors (TNFRs) show promise in early detection and monitoring of disease progression. This review highlights the need for improved animal models and biomarker validation to better mimic human disease and enhance clinical translation. Advancing our understanding of the AKI-to-CKD transition through targeted therapies and refined research approaches holds the potential to significantly improve patient outcomes.
Collapse
Affiliation(s)
- Tingfang Zhang
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Robert E Widdop
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Sharon D Ricardo
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
3
|
Liu Y, Sun D, Huang Y, Shen Y, Chen T, Chen W, Zhu L, Wang F, Hong G, Luo Y, Long S, Zou H. Bibliometric analysis of research on retinoic acid in the field of kidney disorders. Front Pharmacol 2024; 15:1435889. [PMID: 39211779 PMCID: PMC11357955 DOI: 10.3389/fphar.2024.1435889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Retinoic acid is an active metabolite with significant physiological functions in human development, immunity, vision, and skin health. In recent years, research on retinoic acid in the field of kidney disorders has been increasing gradually. Yet, there is a lack of systematic bibliometric analysis of retinoic acid research in the kidney domain. This study included 1,368 articles published between 1998 and 2023 on treating kidney diseases with retinoic acid. Using the bibliometric analysis software VOSviewer and CiteSpace, we analyzed data on publication trends, contributing countries and institutions, journals and cocited journals, authors and cocited authors, cocited references, research hotspots, and frontiers. On the basis of the results of the bibliometric analysis, we identified the research efforts and their developmental trends, providing the groundwork for future research on retinoic acid.
Collapse
Affiliation(s)
- Yu Liu
- Department of Nephrology, South China Hospital of Shenzhen University, Shenzhen, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Dongxuan Sun
- Department of Nephrology, South China Hospital of Shenzhen University, Shenzhen, China
| | - Youqun Huang
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuli Shen
- Nephrology Depariment of The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen, China
| | - Tong Chen
- Department of Nephrology, South China Hospital of Shenzhen University, Shenzhen, China
| | - Wenya Chen
- Department of Nephrology, South China Hospital of Shenzhen University, Shenzhen, China
| | - Liangjun Zhu
- Department of Oncology, The Third Affiliated Hospital of Guizhou Medical University, Duyun, China
| | - Fang Wang
- Department of Nephrology, South China Hospital of Shenzhen University, Shenzhen, China
| | - Guoai Hong
- Department of Nephrology, South China Hospital of Shenzhen University, Shenzhen, China
| | - Yuechan Luo
- Department of Nephrology, South China Hospital of Shenzhen University, Shenzhen, China
| | - Siyu Long
- Department of Nephrology, South China Hospital of Shenzhen University, Shenzhen, China
| | - Hequn Zou
- Medical School, The Chinese University of Hong Kong, Shenzhen, China
| |
Collapse
|
4
|
Goyal A, Dubey N, Agrawal A, Sharma R, Verma A. An Insight into the Promising Therapeutic Potential of Chicoric Acid. Curr Pharm Biotechnol 2024; 25:1708-1718. [PMID: 38083896 DOI: 10.2174/0113892010280616231127075921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/09/2023] [Accepted: 11/21/2023] [Indexed: 09/04/2024]
Abstract
The pharmacological treatments that are now recommended for the therapy of chronic illnesses are examined in a great number of studies to determine whether or not they are both safe and effective. Therefore, it is important to investigate various alternative therapeutic assistance, such as natural remedies derived from medicinal plants. In this context, chicoric acid, classified as a hydroxycinnamic acid, has been documented to exhibit a range of health advantages. These include antiviral, antioxidant, anti-inflammatory, obesity-preventing, and neuroprotective effects. Due to its considerable pharmacological properties, chicoric acid has found extensive applications in food, pharmaceuticals, animal husbandry, and various other commercial sectors. This article provides a comprehensive overview of in vitro and in vivo investigations on chicoric acid, highlighting its beneficial effects and therapeutic activity when used as a preventative and management aid for public health conditions, including diabetes, cardiovascular disease, and hepatic illnesses like non-alcoholic steatohepatitis. Moreover, further investigation of this compound can lead to its development as a potential phytopharmaceutical candidate.
Collapse
Affiliation(s)
- Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Nandini Dubey
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Anant Agrawal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Rashmi Sharma
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Aanchal Verma
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| |
Collapse
|
5
|
Feng YL, Yang Y, Chen H. Small molecules as a source for acute kidney injury therapy. Pharmacol Ther 2022; 237:108169. [DOI: 10.1016/j.pharmthera.2022.108169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/28/2022] [Accepted: 03/07/2022] [Indexed: 10/18/2022]
|
6
|
Martín‐Saiz L, Guerrero‐Mauvecin J, Martín‐Sanchez D, Fresnedo O, Gómez MJ, Carrasco S, Cannata‐Ortiz P, Ortiz A, Fernandez JA, Sanz AB. Ferrostatin‐1 modulates dysregulated kidney lipids in acute kidney injury. J Pathol 2022; 257:285-299. [DOI: 10.1002/path.5882] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 01/18/2022] [Accepted: 02/11/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Lucía Martín‐Saiz
- Department of Physical Chemistry, Faculty of Science and Technology University of the Basque Country (UPV/EHU) Leioa Spain
| | - Juan Guerrero‐Mauvecin
- Laboratory of Experimental Nephrology. Research Institute‐Fundacion Jimenez Diaz, Universidad Autonoma de Madrid Madrid Spain
| | - Diego Martín‐Sanchez
- Laboratory of Experimental Nephrology. Research Institute‐Fundacion Jimenez Diaz, Universidad Autonoma de Madrid Madrid Spain
| | - Olatz Fresnedo
- Department of Physiology, Faculty of Medicine and Nursing University of the Basque Country (UPV/EHU) Leioa Spain
| | - Manuel J. Gómez
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid Spain
| | - Susana Carrasco
- Laboratory of Experimental Nephrology. Research Institute‐Fundacion Jimenez Diaz, Universidad Autonoma de Madrid Madrid Spain
| | - Pablo Cannata‐Ortiz
- Department of Pathology Research Institute ‐ Fundación Jiménez Díaz, Universidad Autonoma de Madrid Madrid Spain
| | - Alberto Ortiz
- Laboratory of Experimental Nephrology. Research Institute‐Fundacion Jimenez Diaz, Universidad Autonoma de Madrid Madrid Spain
- REDINREN Madrid Spain
- Department of Medicine Universidad Autonoma de Madrid Madrid 28049 Spain
- IRSIN Madrid Spain
| | - José A. Fernandez
- Department of Physical Chemistry, Faculty of Science and Technology University of the Basque Country (UPV/EHU) Leioa Spain
| | - Ana B Sanz
- Laboratory of Experimental Nephrology. Research Institute‐Fundacion Jimenez Diaz, Universidad Autonoma de Madrid Madrid Spain
- REDINREN Madrid Spain
| |
Collapse
|
7
|
Franco ML, Beyerstedt S, Rangel ÉB. Klotho and Mesenchymal Stem Cells: A Review on Cell and Gene Therapy for Chronic Kidney Disease and Acute Kidney Disease. Pharmaceutics 2021; 14:11. [PMID: 35056905 PMCID: PMC8778857 DOI: 10.3390/pharmaceutics14010011] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 02/07/2023] Open
Abstract
Chronic kidney disease (CKD) and acute kidney injury (AKI) are public health problems, and their prevalence rates have increased with the aging of the population. They are associated with the presence of comorbidities, in particular diabetes mellitus and hypertension, resulting in a high financial burden for the health system. Studies have indicated Klotho as a promising therapeutic approach for these conditions. Klotho reduces inflammation, oxidative stress and fibrosis and counter-regulates the renin-angiotensin-aldosterone system. In CKD and AKI, Klotho expression is downregulated from early stages and correlates with disease progression. Therefore, the restoration of its levels, through exogenous or endogenous pathways, has renoprotective effects. An important strategy for administering Klotho is through mesenchymal stem cells (MSCs). In summary, this review comprises in vitro and in vivo studies on the therapeutic potential of Klotho for the treatment of CKD and AKI through the administration of MSCs.
Collapse
Affiliation(s)
- Marcella Liciani Franco
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, Sao Paulo 05652-900, Brazil; (M.L.F.); (S.B.)
| | - Stephany Beyerstedt
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, Sao Paulo 05652-900, Brazil; (M.L.F.); (S.B.)
| | - Érika Bevilaqua Rangel
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, Sao Paulo 05652-900, Brazil; (M.L.F.); (S.B.)
- Nephrology Division, Federal University of São Paulo, Sao Paulo 04038-901, Brazil
| |
Collapse
|
8
|
Miricescu D, Balan DG, Tulin A, Stiru O, Vacaroiu IA, Mihai DA, Popa CC, Enyedi M, Nedelea AS, Nica AE, Stefani C. Impact of adipose tissue in chronic kidney disease development (Review). Exp Ther Med 2021; 21:539. [PMID: 33815612 PMCID: PMC8014972 DOI: 10.3892/etm.2021.9969] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 01/14/2021] [Indexed: 12/13/2022] Open
Abstract
Obesity is a worldwide pandemic health issue. Obesity is associated with the pathogenesis of type 2 diabetes, hypertension, dyslipidemia, cardiovascular diseases, cancer, and kidney diseases. This systemic disease can affect the kidneys by two mechanisms: Indirectly through diabetes mellitus (DM) and hypertension and directly through adipokines secreted by adipose tissue. Obesity is a risk factor for chronic kidney disease (CKD), which is associated with an increased risk of morbidity and mortality among the adult population. Increased visceral adipose tissue leads to renal glomerular hyperfiltration and hyperperfusion, which may lead to glomerular hypertrophy, proteinuria, and CKD development. Adipokines are hormones produced by fat tissue. They are involved in energy homeostasis, sugar and fat metabolism, reproduction, immunity, and thermogenesis control. Hormones and cytokines secreted by adipose tissue contribute to the development and progression of CKD. Decreased serum or urinary adiponectin levels are specific in diabetic and non-diabetic CKD patients, while leptin presents increased levels, and both are associated with the development of glomerulopathy. Excessive adipose tissue is associated with inflammation, oxidative stress (OS), insulin resistance and activation of the renin angiotensin-aldosterone system (RAAS). Therefore, adipose tissue dysfunction plays an important role in the development of CKD.
Collapse
Affiliation(s)
- Daniela Miricescu
- Department of Biochemistry, Faculty of Dental Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Daniela Gabriela Balan
- Discipline of Physiology, Faculty of Dental Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Adrian Tulin
- Department of Anatomy, Faculty of Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of General Surgery, ‘Prof. Dr. Agrippa Ionescu’ Clinical Emergency Hospital, 011356 Bucharest, Romania
| | - Ovidiu Stiru
- Department of Cardiovascular Surgery, Faculty of Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Cardiovascular Surgery, ‘Prof. Dr. C.C. Iliescu’ Emergency Institute for Cardiovascular Diseases, 022322 Bucharest, Romania
| | - Ileana Adela Vacaroiu
- Department of Nephrology and Dialysis, ‘Sf. Ioan’ Emergency Clinical Hospital, 042122 Bucharest, Romania
- Department of Nephrology, Nutrition and Metabolic Diseases, Faculty of Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Doina Andrada Mihai
- Discipline of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department II of Diabetes, ‘Prof. N. Paulescu’ National Institute of Diabetes, Nutrition and Metabolic Diseases, 020474 Bucharest, Romania
| | - Cristian Constantin Popa
- Department of Surgery, Faculty of Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Surgery, Bucharest Emergency University Hospital, 050098 Bucharest, Romania
| | - Mihaly Enyedi
- Department of Anatomy, Faculty of Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Radiology, ‘Victor Babes’ Private Medical Clinic, 030303 Bucharest, Romania
| | - Andrei Sorin Nedelea
- Department of Urology, ‘Prof. Dr. Agrippa Ionescu’ Clinical Emergency Hospital, 011356 Bucharest, Romania
| | - Adriana Elena Nica
- Department of Orthopedics, Anesthesia Intensive Care Unit, Faculty of Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Constantin Stefani
- Department of Family Medicine and Clinical Base, Dr. Carol Davila Central Military Emergency University Hospital, 010825 Bucharest, Romania
| |
Collapse
|
9
|
Saini S, Rani L, Shukla N, Banerjee M, Chowdhuri DK, Gautam NK. Development of a Drosophila melanogaster based model for the assessment of cadmium and mercury mediated renal tubular toxicity. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 201:110811. [PMID: 32544744 DOI: 10.1016/j.ecoenv.2020.110811] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 05/18/2020] [Accepted: 05/22/2020] [Indexed: 06/11/2023]
Abstract
Xenobiotic mediated renal toxicity is one of the major health concerns to the organisms, including humans. New chemicals with nephrotoxic potential are continuously being added to the list of existing nephrotoxicants. To predict the nephrotoxicity of these new chemicals, reliable and cost-effective alternative animal models are required. It is a prerequisite for the identification and assessment of these compounds as potential nephrotoxicants to prevent renal toxicity in the exposed population. Drosophila melanogaster, a genetically tractable invertebrate animal model, has a renal system functionally analogous to humans. The Malpighian tubules (MTs) of D. melanogaster are similar to the tubular part of nephron of the human kidney. Besides, it recapitulates the renal toxicity hallmark with mammals when exposed to known nephrotoxicants. In this study, first instar larvae of D. melanogaster (Oregon R) were exposed to different concentrations of two well-known nephrotoxicants, cadmium (Cd) and mercury (Hg). Akin to higher organisms, Cd and Hg exposure to D. melanogaster produce similar phenotypes. MTs of exposed D. melanogaster larvae exhibited increased oxidative stress, activated cellular antioxidant defense mechanism, GSH depletion, increased cleaved caspase-3 expression, increased DEVDase activity and increased cell death. The functional status of MTs was assessed by fluid secretion rate (FSR), efflux activity of transporter protein, mitochondrial membrane potential (MMP), ATP level and expression of junctional protein (Dlg). All the phenotypes observed in MTs of D. melanogaster larvae recapitulate the phenotypes observed in higher organisms. Increased uric acid level, the hallmark of renal dysfunction, was also observed in exposed larvae. Taken together, the study suggests that MTs of D. melanogaster may be used as a functional model to evaluate xenobiotic mediated nephrotoxicity.
Collapse
Affiliation(s)
- Sanjay Saini
- Embryotoxicology Laboratory, Environmental Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31 Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Molecular and Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow, 226007, India
| | - Lavi Rani
- Embryotoxicology Laboratory, Environmental Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31 Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Neha Shukla
- Department of Urology and Renal Transplantation, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Raebareli Road, Lucknow, 226014, Uttar Pradesh, India; Embryotoxicology Laboratory, Environmental Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31 Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India
| | - Monisha Banerjee
- Molecular and Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow, 226007, India
| | - Debapratim Kar Chowdhuri
- Embryotoxicology Laboratory, Environmental Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31 Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Naveen Kumar Gautam
- Department of Urology and Renal Transplantation, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Raebareli Road, Lucknow, 226014, Uttar Pradesh, India; Embryotoxicology Laboratory, Environmental Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31 Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India.
| |
Collapse
|
10
|
Georgiadis G, Zisis IE, Docea AO, Tsarouhas K, Fragkiadoulaki I, Mavridis C, Karavitakis M, Stratakis S, Stylianou K, Tsitsimpikou C, Calina D, Sofikitis N, Tsatsakis A, Mamoulakis C. Current Concepts on the Reno-Protective Effects of Phosphodiesterase 5 Inhibitors in Acute Kidney Injury: Systematic Search and Review. J Clin Med 2020; 9:jcm9051284. [PMID: 32365529 PMCID: PMC7287956 DOI: 10.3390/jcm9051284] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/16/2020] [Accepted: 04/24/2020] [Indexed: 12/11/2022] Open
Abstract
Acute kidney injury (AKI) is associated with increased morbidity, prolonged hospitalization, and mortality, especially in high risk patients. Phosphodiesterase 5 inhibitors (PDE5Is), currently available as first-line therapy of erectile dysfunction in humans, have shown a beneficial potential of reno-protection through various reno-protective mechanisms. The aim of this work is to provide a comprehensive overview of the available literature on the reno-protective properties of PDE5Is in the various forms of AKI. Medline was systematically searched from 1946 to November 2019 to detect all relevant animal and human studies in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) statement. In total, 83 studies were included for qualitative synthesis. Sildenafil is the most widely investigated compound (42 studies), followed by tadalafil (20 studies), icariin (10 studies), vardenafil (7 studies), zaprinast (4 studies), and udenafil (2 studies). Even though data are limited, especially in humans with inconclusive or negative results of only two clinically relevant studies available at present, the results of animal studies are promising. The reno-protective action of PDE5Is was evident in the vast majority of studies, independently of the AKI type and the agent applied. PDE5Is appear to improve the renal functional/histopathological alternations of AKI through various mechanisms, mainly by affecting regional hemodynamics, cell expression, and mitochondrial response to oxidative stress and inflammation.
Collapse
Affiliation(s)
- Georgios Georgiadis
- Department of Urology, University General Hospital of Heraklion, University of Crete, Medical School, Heraklion, Crete, Greece; (G.G.); (I.-E.Z.); (I.F.); (C.M.); (M.K.)
| | - Ioannis-Erineos Zisis
- Department of Urology, University General Hospital of Heraklion, University of Crete, Medical School, Heraklion, Crete, Greece; (G.G.); (I.-E.Z.); (I.F.); (C.M.); (M.K.)
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, Heraklion, Crete 71003, Greece;
| | - Anca Oana Docea
- Department of Toxicology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | | | - Irene Fragkiadoulaki
- Department of Urology, University General Hospital of Heraklion, University of Crete, Medical School, Heraklion, Crete, Greece; (G.G.); (I.-E.Z.); (I.F.); (C.M.); (M.K.)
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, Heraklion, Crete 71003, Greece;
| | - Charalampos Mavridis
- Department of Urology, University General Hospital of Heraklion, University of Crete, Medical School, Heraklion, Crete, Greece; (G.G.); (I.-E.Z.); (I.F.); (C.M.); (M.K.)
| | - Markos Karavitakis
- Department of Urology, University General Hospital of Heraklion, University of Crete, Medical School, Heraklion, Crete, Greece; (G.G.); (I.-E.Z.); (I.F.); (C.M.); (M.K.)
| | - Stavros Stratakis
- Department of Nephrology, University General Hospital of Heraklion, University of Crete, Medical School, Heraklion, Crete, Greece; (S.S.); (K.S.)
| | - Kostas Stylianou
- Department of Nephrology, University General Hospital of Heraklion, University of Crete, Medical School, Heraklion, Crete, Greece; (S.S.); (K.S.)
| | - Christina Tsitsimpikou
- Department of Hazardous Substances, Mixtures and Articles, General Chemical State Laboratory of Greece, Ampelokipi, Athens, Greece;
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Nikolaos Sofikitis
- Department of Urology, School of Medicine, Ioannina University, Ioannina, Greece;
| | - Aristidis Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, Heraklion, Crete 71003, Greece;
| | - Charalampos Mamoulakis
- Department of Urology, University General Hospital of Heraklion, University of Crete, Medical School, Heraklion, Crete, Greece; (G.G.); (I.-E.Z.); (I.F.); (C.M.); (M.K.)
- Correspondence:
| |
Collapse
|
11
|
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are worldwide public health problems affecting millions of people and have rapidly increased in prevalence in recent years. Due to the multiple causes of renal failure, many animal models have been developed to advance our understanding of human nephropathy. Among these experimental models, rodents have been extensively used to enable mechanistic understanding of kidney disease induction and progression, as well as to identify potential targets for therapy. In this review, we discuss AKI models induced by surgical operation and drugs or toxins, as well as a variety of CKD models (mainly genetically modified mouse models). Results from recent and ongoing clinical trials and conceptual advances derived from animal models are also explored.
Collapse
Affiliation(s)
- Yin-Wu Bao
- Kidney Disease Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou Zhejiang 310058, China. .,Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou Zhejiang 310058, China
| | - Yuan Yuan
- Kidney Disease Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou Zhejiang 310058, China. .,Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou Zhejiang 310058, China
| | - Jiang-Hua Chen
- Kidney Disease Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou Zhejiang 310058, China.
| | - Wei-Qiang Lin
- Kidney Disease Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou Zhejiang 310058, China. .,Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou Zhejiang 310058, China
| |
Collapse
|
12
|
Brightbill HD, Suto E, Blaquiere N, Ramamoorthi N, Sujatha-Bhaskar S, Gogol EB, Castanedo GM, Jackson BT, Kwon YC, Haller S, Lesch J, Bents K, Everett C, Kohli PB, Linge S, Christian L, Barrett K, Jaochico A, Berezhkovskiy LM, Fan PW, Modrusan Z, Veliz K, Townsend MJ, DeVoss J, Johnson AR, Godemann R, Lee WP, Austin CD, McKenzie BS, Hackney JA, Crawford JJ, Staben ST, Alaoui Ismaili MH, Wu LC, Ghilardi N. NF-κB inducing kinase is a therapeutic target for systemic lupus erythematosus. Nat Commun 2018; 9:179. [PMID: 29330524 PMCID: PMC5766581 DOI: 10.1038/s41467-017-02672-0] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 12/18/2017] [Indexed: 02/06/2023] Open
Abstract
NF-κB-inducing kinase (NIK) mediates non-canonical NF-κB signaling downstream of multiple TNF family members, including BAFF, TWEAK, CD40, and OX40, which are implicated in the pathogenesis of systemic lupus erythematosus (SLE). Here, we show that experimental lupus in NZB/W F1 mice can be treated with a highly selective and potent NIK small molecule inhibitor. Both in vitro as well as in vivo, NIK inhibition recapitulates the pharmacological effects of BAFF blockade, which is clinically efficacious in SLE. Furthermore, NIK inhibition also affects T cell parameters in the spleen and proinflammatory gene expression in the kidney, which may be attributable to inhibition of OX40 and TWEAK signaling, respectively. As a consequence, NIK inhibition results in improved survival, reduced renal pathology, and lower proteinuria scores. Collectively, our data suggest that NIK inhibition is a potential therapeutic approach for SLE.
Collapse
Affiliation(s)
- Hans D Brightbill
- Department of Immunology Discovery, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Eric Suto
- Department of Translational Immunology, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Nicole Blaquiere
- Department of Discovery Chemistry, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Nandhini Ramamoorthi
- Department of Biomarker Discovery, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Swathi Sujatha-Bhaskar
- Department of Immunology Discovery, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Emily B Gogol
- Department of Immunology Discovery, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Georgette M Castanedo
- Department of Discovery Chemistry, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Benjamin T Jackson
- Department of Immunology Discovery, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Youngsu C Kwon
- Department of Translational Immunology, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Susan Haller
- Department of Pathology, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Justin Lesch
- Department of Translational Immunology, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Karin Bents
- Evotec, Inc., Essener Bogen 7, Hamburg, 22419, Germany
| | - Christine Everett
- Department of Biochemical and Cellular Pharmacology, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Pawan Bir Kohli
- Department of Biochemical and Cellular Pharmacology, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Sandra Linge
- Evotec, Inc., Essener Bogen 7, Hamburg, 22419, Germany
| | - Laura Christian
- Department of Immunology Discovery, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Kathy Barrett
- Department of Biochemical and Cellular Pharmacology, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Allan Jaochico
- Department of Drug Metabolism and Pharmacokinetics, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Leonid M Berezhkovskiy
- Department of Drug Metabolism and Pharmacokinetics, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Peter W Fan
- Department of Drug Metabolism and Pharmacokinetics, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Zora Modrusan
- Department of Molecular Biology, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Kelli Veliz
- Department of Laboratory Animal Resources, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Michael J Townsend
- Department of Biomarker Discovery, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Jason DeVoss
- Department of Translational Immunology, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Adam R Johnson
- Department of Biochemical and Cellular Pharmacology, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | | | - Wyne P Lee
- Department of Translational Immunology, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Cary D Austin
- Department of Pathology, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Brent S McKenzie
- Department of Translational Immunology, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Jason A Hackney
- Department of Bioinformatics and Computational Biology, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - James J Crawford
- Department of Discovery Chemistry, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Steven T Staben
- Department of Discovery Chemistry, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Moulay H Alaoui Ismaili
- Department of Biochemical and Cellular Pharmacology, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Lawren C Wu
- Department of Immunology Discovery, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA
| | - Nico Ghilardi
- Department of Immunology Discovery, Genentech, 1 DNA Way, South San Francisco, CA-94080, USA.
| |
Collapse
|
13
|
Cantley LG. Introduction: Understanding Human AKI. Semin Nephrol 2018; 38:1-2. [DOI: 10.1016/j.semnephrol.2017.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
14
|
Martin-Sanchez D, Poveda J, Fontecha-Barriuso M, Ruiz-Andres O, Sanchez-Niño MD, Ruiz-Ortega M, Ortiz A, Sanz AB. Targeting of regulated necrosis in kidney disease. Nefrologia 2017. [PMID: 28647049 DOI: 10.1016/j.nefro.2017.04.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The term acute tubular necrosis was thought to represent a misnomer derived from morphological studies of human necropsies and necrosis was thought to represent an unregulated passive form of cell death which was not amenable to therapeutic manipulation. Recent advances have improved our understanding of cell death in acute kidney injury. First, apoptosis results in cell loss, but does not trigger an inflammatory response. However, clumsy attempts at interfering with apoptosis (e.g. certain caspase inhibitors) may trigger necrosis and, thus, inflammation-mediated kidney injury. Second, and most revolutionary, the concept of regulated necrosis emerged. Several modalities of regulated necrosis were described, such as necroptosis, ferroptosis, pyroptosis and mitochondria permeability transition regulated necrosis. Similar to apoptosis, regulated necrosis is modulated by specific molecules that behave as therapeutic targets. Contrary to apoptosis, regulated necrosis may be extremely pro-inflammatory and, importantly for kidney transplantation, immunogenic. Furthermore, regulated necrosis may trigger synchronized necrosis, in which all cells within a given tubule die in a synchronized manner. We now review the different modalities of regulated necrosis, the evidence for a role in diverse forms of kidney injury and the new opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Diego Martin-Sanchez
- Research Institute-Fundación Jiménez Díaz, Autónoma University, Madrid, Spain; IRSIN, Madrid, Spain; REDINREN, Madrid, Spain
| | - Jonay Poveda
- Research Institute-Fundación Jiménez Díaz, Autónoma University, Madrid, Spain; IRSIN, Madrid, Spain; REDINREN, Madrid, Spain
| | - Miguel Fontecha-Barriuso
- Research Institute-Fundación Jiménez Díaz, Autónoma University, Madrid, Spain; IRSIN, Madrid, Spain; REDINREN, Madrid, Spain
| | - Olga Ruiz-Andres
- Research Institute-Fundación Jiménez Díaz, Autónoma University, Madrid, Spain; IRSIN, Madrid, Spain; REDINREN, Madrid, Spain
| | - María Dolores Sanchez-Niño
- Research Institute-Fundación Jiménez Díaz, Autónoma University, Madrid, Spain; IRSIN, Madrid, Spain; REDINREN, Madrid, Spain
| | - Marta Ruiz-Ortega
- Research Institute-Fundación Jiménez Díaz, Autónoma University, Madrid, Spain; IRSIN, Madrid, Spain; REDINREN, Madrid, Spain
| | - Alberto Ortiz
- Research Institute-Fundación Jiménez Díaz, Autónoma University, Madrid, Spain; IRSIN, Madrid, Spain; REDINREN, Madrid, Spain
| | - Ana Belén Sanz
- Research Institute-Fundación Jiménez Díaz, Autónoma University, Madrid, Spain; IRSIN, Madrid, Spain; REDINREN, Madrid, Spain.
| |
Collapse
|
15
|
Gallagher KM, O'neill S, Harrison EM, Ross JA, Wigmore SJ, Hughes J. Recent early clinical drug development for acute kidney injury. Expert Opin Investig Drugs 2016; 26:141-154. [PMID: 27997816 DOI: 10.1080/13543784.2017.1274730] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Despite significant need and historical trials, there are no effective drugs in use for the prevention or treatment of acute kidney injury (AKI). There are several promising agents in early clinical development for AKI and two trials have recently been terminated. There are also exciting new findings in pre-clinical AKI research. There is a need to take stock of current progress in the field to guide future drug development for AKI. Areas covered: The main clinical trial registries, PubMed and pharmaceutical company website searches were used to extract the most recent clinical trials for sterile, transplant and sepsis-associated AKI. We summarise the development of the agents recently in clinical trial, update on their trial progress, consider reasons for failed efficacy of two agents, and discuss new paradigms in pre-clinical targets for AKI. Agents covered include- QPI-1002, THR-184, BB-3, heme arginate, human recombinant alkaline phosphatase (recAP), ciclosporin A, AB103, levosimendan, AC607 and ABT-719. Expert opinion: Due to the heterogenous nature of AKI, agents with the widest pleiotropic effects on multiple pathophysiological pathways are likely to be most effective. Linking preclinical models to clinical indication and improving AKI definition and diagnosis are key areas for improvement in future clinical trials.
Collapse
Affiliation(s)
- Kevin M Gallagher
- a MRC Centre for Inflammation Research, Royal Infirmary of Edinburgh , University of Edinburgh , Edinburgh , UK
| | - Stephen O'neill
- a MRC Centre for Inflammation Research, Royal Infirmary of Edinburgh , University of Edinburgh , Edinburgh , UK
| | - Ewen M Harrison
- a MRC Centre for Inflammation Research, Royal Infirmary of Edinburgh , University of Edinburgh , Edinburgh , UK
| | - James A Ross
- b MRC Centre for Regenerative Medicine, Royal Infirmary of Edinburgh , University of Edinburgh , Edinburgh , UK
| | - Stephen J Wigmore
- a MRC Centre for Inflammation Research, Royal Infirmary of Edinburgh , University of Edinburgh , Edinburgh , UK
| | - Jeremy Hughes
- a MRC Centre for Inflammation Research, Royal Infirmary of Edinburgh , University of Edinburgh , Edinburgh , UK
| |
Collapse
|
16
|
McKee RA, Wingert RA. Nephrotoxin Microinjection in Zebrafish to Model Acute Kidney Injury. J Vis Exp 2016. [PMID: 27500823 DOI: 10.3791/54241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The kidneys are susceptible to harm from exposure to chemicals they filter from the bloodstream. This can lead to organ injury associated with a rapid decline in renal function and development of the clinical syndrome known as acute kidney injury (AKI). Pharmacological agents used to treat medical circumstances ranging from bacterial infection to cancer, when administered individually or in combination with other drugs, can initiate AKI. Zebrafish are a useful animal model to study the chemical effects on renal function in vivo, as they form an embryonic kidney comprised of nephron functional units that are conserved with higher vertebrates, including humans. Further, zebrafish can be utilized to perform genetic and chemical screens, which provide opportunities to elucidate the cellular and molecular facets of AKI and develop therapeutic strategies such as the identification of nephroprotective molecules. Here, we demonstrate how microinjection into the zebrafish embryo can be utilized as a paradigm for nephrotoxin studies.
Collapse
Affiliation(s)
- Robert A McKee
- Center for Zebrafish Research, Department of Biological Sciences, University of Notre Dame; Center for Stem Cells and Regenerative Medicine, Department of Biological Sciences, University of Notre Dame
| | - Rebecca A Wingert
- Center for Zebrafish Research, Department of Biological Sciences, University of Notre Dame; Center for Stem Cells and Regenerative Medicine, Department of Biological Sciences, University of Notre Dame;
| |
Collapse
|
17
|
Tumor Necrosis Factor-Like Weak Inducer of Apoptosis Accelerates the Progression of Renal Fibrosis in Lupus Nephritis by Activating SMAD and p38 MAPK in TGF-β1 Signaling Pathway. Mediators Inflamm 2016; 2016:8986451. [PMID: 27365897 PMCID: PMC4913011 DOI: 10.1155/2016/8986451] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 04/21/2016] [Accepted: 05/05/2016] [Indexed: 01/07/2023] Open
Abstract
This study aim was to explore the effects of tumor necrosis factor-like weak inducer of apoptosis (TWEAK) in lupus nephritis and its potential underlying mechanisms. MRL/lpr mice were used for in vivo experiments and human proximal tubular cells (HK2 cells) were used for in vitro experiments. Results showed that MRL/lpr mice treated with vehicle solution or LV-Control shRNA displayed significant proteinuria and severe renal histopathological changes. LV-TWEAK-shRNA treatment reversed these changes and decreased renal expressions of TWEAK, TGF-β1, p-p38 MAPK, p-Smad2, COL-1, and α-SMA proteins. In vitro, hTWEAK treatment upregulated the expressions of TGF-β1, p-p38 MAPK, p-SMAD2, α-SMA, and COL-1 proteins in HK2 cells and downregulated the expressions of E-cadherin protein, which were reversed by cotreatment with anti-TWEAK mAb or SB431542 treatment. These findings suggest that TWEAK may contribute to chronic renal changes and renal fibrosis by activating TGF-β1 signaling pathway, and phosphorylation of Smad2 and p38 MAPK proteins was also involved in this signaling pathway.
Collapse
|
18
|
Skrypnyk NI, Siskind LJ, Faubel S, de Caestecker MP. Bridging translation for acute kidney injury with better preclinical modeling of human disease. Am J Physiol Renal Physiol 2016; 310:F972-84. [PMID: 26962107 PMCID: PMC4889323 DOI: 10.1152/ajprenal.00552.2015] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 03/04/2016] [Indexed: 12/14/2022] Open
Abstract
The current lack of effective therapeutics for patients with acute kidney injury (AKI) represents an important and unmet medical need. Given the importance of the clinical problem, it is time for us to take a few steps back and reexamine current practices. The focus of this review is to explore the extent to which failure of therapeutic translation from animal studies to human studies stems from deficiencies in the preclinical models of AKI. We will evaluate whether the preclinical models of AKI that are commonly used recapitulate the known pathophysiologies of AKI that are being modeled in humans, focusing on four common scenarios that are studied in clinical therapeutic intervention trials: cardiac surgery-induced AKI; contrast-induced AKI; cisplatin-induced AKI; and sepsis associated AKI. Based on our observations, we have identified a number of common limitations in current preclinical modeling of AKI that could be addressed. In the long term, we suggest that progress in developing better preclinical models of AKI will depend on developing a better understanding of human AKI. To this this end, we suggest that there is a need to develop greater in-depth molecular analyses of kidney biopsy tissues coupled with improved clinical and molecular classification of patients with AKI.
Collapse
Affiliation(s)
- Nataliya I Skrypnyk
- Division of Nephology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Leah J Siskind
- Department of Pharmacology and Toxicology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky; and
| | - Sarah Faubel
- Renal Division, University of Colorado Denver and Denver Veterans Affairs Medical Center, Aurora, Colorado
| | - Mark P de Caestecker
- Division of Nephology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee;
| |
Collapse
|
19
|
Drummond BE, Wingert RA. Insights into kidney stem cell development and regeneration using zebrafish. World J Stem Cells 2016; 8:22-31. [PMID: 26981168 PMCID: PMC4766248 DOI: 10.4252/wjsc.v8.i2.22] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 11/28/2015] [Accepted: 01/11/2016] [Indexed: 02/06/2023] Open
Abstract
Kidney disease is an escalating global health problem, for which the formulation of therapeutic approaches using stem cells has received increasing research attention. The complexity of kidney anatomy and function, which includes the diversity of renal cell types, poses formidable challenges in the identification of methods to generate replacement structures. Recent work using the zebrafish has revealed their high capacity to regenerate the integral working units of the kidney, known as nephrons, following acute injury. Here, we discuss these findings and explore the ways that zebrafish can be further utilized to gain a deeper molecular appreciation of renal stem cell biology, which may uncover important clues for regenerative medicine.
Collapse
|
20
|
Cano-Peñalver JL, Griera M, García-Jerez A, Hatem-Vaquero M, Ruiz-Torres MP, Rodríguez-Puyol D, Frutos SD, Rodríguez-Puyol M. Renal Integrin-Linked Kinase Depletion Induces Kidney cGMP-Axis Upregulation: Consequences on Basal and Acutely Damaged Renal Function. Mol Med 2015; 21:873-885. [PMID: 26562149 DOI: 10.2119/molmed.2015.00059] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 11/09/2015] [Indexed: 12/12/2022] Open
Abstract
Soluble guanylyl cyclase (sGC) is activated by nitric oxide (NO) and produces cGMP, which activates cGMP-dependent protein kinases (PKG) and is hydrolyzed by specific phosphodiesterases (PDE). The vasodilatory and cytoprotective capacity of cGMP-axis activation results in a therapeutic strategy for several pathologies. Integrin-linked kinase (ILK), a major scaffold protein between the extracellular matrix and intracellular signaling pathways, may modulate the expression and functionality of the cGMP-axis-related proteins. We introduce ILK as a novel modulator in renal homeostasis as well as a potential target for cisplatin (CIS)-induced acute kidney injury (AKI) improvement. We used an adult mice model of depletion of ILK (cKD-ILK), which showed basal increase of sGC and PKG expressions and activities in renal cortex when compared with wildtype (WT) littermates. Twenty-four h activation of sGC activation with NO enhanced the filtration rate in cKD-ILK. During AKI, cKD-ILK maintained the cGMP-axis upregulation with consequent filtration rates enhancement and ameliorated CIS-dependent tubular epithelial-to-mesenchymal transition and inflammation and markers. To emphasize the role of cGMP-axis upregulation due to ILK depletion, we modulated the cGMP axis under AKI in vivo and in renal cultured cells. A suboptimal dose of the PDE inhibitor ZAP enhanced the beneficial effects of the ILK depletion in AKI mice. On the other hand, CIS increased contractility-related events in cultured glomerular mesangial cells and necrosis rates in cultured tubular cells; ILK depletion protected the cells while sGC blockade with ODQ fully recovered the damage.
Collapse
Affiliation(s)
- José Luis Cano-Peñalver
- Department of Systems Biology, Physiology Unit, Universidad de Alcalà, Alcalà de Henares, Madrid, Spain.,Instituto Reina Sofia de Investigaciόn Renal and REDinREN from Instituto de Salud Carlos III, Madrid, Spain
| | - Mercedes Griera
- Department of Systems Biology, Physiology Unit, Universidad de Alcalà, Alcalà de Henares, Madrid, Spain.,Instituto Reina Sofia de Investigaciόn Renal and REDinREN from Instituto de Salud Carlos III, Madrid, Spain
| | - Andrea García-Jerez
- Department of Systems Biology, Physiology Unit, Universidad de Alcalà, Alcalà de Henares, Madrid, Spain.,Instituto Reina Sofia de Investigaciόn Renal and REDinREN from Instituto de Salud Carlos III, Madrid, Spain
| | - Marco Hatem-Vaquero
- Department of Systems Biology, Physiology Unit, Universidad de Alcalà, Alcalà de Henares, Madrid, Spain.,Instituto Reina Sofia de Investigaciόn Renal and REDinREN from Instituto de Salud Carlos III, Madrid, Spain
| | - María Piedad Ruiz-Torres
- Department of Systems Biology, Physiology Unit, Universidad de Alcalà, Alcalà de Henares, Madrid, Spain.,Instituto Reina Sofia de Investigaciόn Renal and REDinREN from Instituto de Salud Carlos III, Madrid, Spain
| | - Diego Rodríguez-Puyol
- Instituto Reina Sofia de Investigaciόn Renal and REDinREN from Instituto de Salud Carlos III, Madrid, Spain.,Biomedical Research Foundation and Nephrology Department, Hospital Príncipe de Asturias, Alcalà de Henares, Madrid, Spain
| | - Sergio de Frutos
- Department of Systems Biology, Physiology Unit, Universidad de Alcalà, Alcalà de Henares, Madrid, Spain.,Instituto Reina Sofia de Investigaciόn Renal and REDinREN from Instituto de Salud Carlos III, Madrid, Spain
| | - Manuel Rodríguez-Puyol
- Department of Systems Biology, Physiology Unit, Universidad de Alcalà, Alcalà de Henares, Madrid, Spain.,Instituto Reina Sofia de Investigaciόn Renal and REDinREN from Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
21
|
de Caestecker M, Humphreys BD, Liu KD, Fissell WH, Cerda J, Nolin TD, Askenazi D, Mour G, Harrell FE, Pullen N, Okusa MD, Faubel S. Bridging Translation by Improving Preclinical Study Design in AKI. J Am Soc Nephrol 2015; 26:2905-16. [PMID: 26538634 DOI: 10.1681/asn.2015070832] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Despite extensive research, no therapeutic interventions have been shown to prevent AKI, accelerate recovery of AKI, or reduce progression of AKI to CKD in patients. This failure in translation has led investigators to speculate that the animal models being used do not predict therapeutic responses in humans. Although this issue continues to be debated, an important concern that has not been addressed is whether improvements in preclinical study design can be identified that might also increase the likelihood of translating basic AKI research into clinical practice using the current models. In this review, we have taken an evidence-based approach to identify common weaknesses in study design and reporting in preclinical AKI research that may contribute to the poor translatability of the findings. We focused on use of N-acetylcysteine or sodium bicarbonate for the prevention of contrast-induced AKI and use of erythropoietin for the prevention of AKI, two therapeutic approaches that have been extensively studied in clinical trials. On the basis of our findings, we identified five areas for improvement in preclinical study design and reporting. These suggested and preliminary guidelines may help improve the quality of preclinical research for AKI drug development.
Collapse
Affiliation(s)
- Mark de Caestecker
- Division of Nephology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee;
| | - Ben D Humphreys
- Division of Renal Diseases, Washington University School of Medicine, St. Louis, Missouri
| | - Kathleen D Liu
- Division of Nephrology, Department of Medicine, University of California, San Francisco, California
| | - William H Fissell
- Division of Nephology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jorge Cerda
- Division of Nephrology and Hypertension, Albany Medical College, Albany, New York
| | - Thomas D Nolin
- Renal-Electrolyte Division, Department of Medicine and Center for Critical Care Nephrology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - David Askenazi
- Department of Pediatrics, Division of Nephrology, University of Alabama, Birmingham, Alabama
| | - Girish Mour
- Renal-Electrolyte Division, Department of Medicine and Center for Critical Care Nephrology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Frank E Harrell
- Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Nick Pullen
- Pfizer Global Research and Development, Inflammation & Immunology Research Unit, Cambridge, Massachusetts
| | - Mark D Okusa
- Division of Nephrology, Department of Medicine, University of California, San Francisco, California; Division of Nephrology, Department of Medicine, University of California, San Francisco, California
| | - Sarah Faubel
- Renal Division, University of Colorado Denver and Denver Veterans Affairs Medical Center, Aurora, Colorado
| | | |
Collapse
|
22
|
McKee RA, Wingert RA. Zebrafish Renal Pathology: Emerging Models of Acute Kidney Injury. CURRENT PATHOBIOLOGY REPORTS 2015; 3:171-181. [PMID: 25973344 PMCID: PMC4419198 DOI: 10.1007/s40139-015-0082-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The renal system is vital to maintain homeostasis in the body, where the kidneys contain nephron functional units that remove metabolic waste from the bloodstream, regulate fluids, and balance electrolytes. Severe organ damage from toxins or ischemia that occurs abruptly can cause acute kidney injury (AKI) in which there is a rapid, life-threatening loss of these activities. Humans have a limited but poorly understood ability to regenerate damaged nephrons after AKI. However, researchers studying AKI in vertebrate animal models such as mammals, and more recently the zebrafish, have documented robust regeneration within the nephron blood filter and tubule following injury. Further, zebrafish kidneys contain progenitors that create new nephrons after AKI. Here, we review investigations in zebrafish which have established a series of exciting renal pathology paradigms that complement existing AKI models and can be implemented to discover insights into kidney regeneration and the roles of stem cells.
Collapse
Affiliation(s)
- Robert A. McKee
- Department of Biological Sciences, Center for Zebrafish Research, Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN 46556 USA
| | - Rebecca A. Wingert
- Department of Biological Sciences, Center for Zebrafish Research, Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN 46556 USA
| |
Collapse
|
23
|
Ramos AM, González-Guerrero C, Sanz A, Sanchez-Niño MD, Rodríguez-Osorio L, Martín-Cleary C, Fernández-Fernández B, Ruiz-Ortega M, Ortiz A. Designing drugs that combat kidney damage. Expert Opin Drug Discov 2015; 10:541-56. [PMID: 25840605 DOI: 10.1517/17460441.2015.1033394] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Kidney disease remains one of the last worldwide frontiers in the field of non-communicable human disease. From 1990 to 2013, chronic kidney disease (CKD) was the top non-communicable cause of death with a greatest increase in global years of life lost while mortality of acute kidney injury (AKI) still hovers around 50%. This reflects the paucity (for CKD) or lack of (for AKI) therapeutic approaches beyond replacing renal function. Understanding what the barriers are and what potential pathways may facilitate the design of new drugs to combat kidney disease is a key public health priority. AREAS COVERED The authors discuss the hurdles and opportunities for future drug development for kidney disease in light of experience accumulated with drugs that made it to clinical trials. EXPERT OPINION Inflammation, cell death and fibrosis are key therapeutic targets to combat kidney damage. While the specific targeting of drugs to kidney cells would be desirable, the technology is only working at the preclinical stage and with mixed success. Nanomedicines hold promise in this respect. Most drugs undergoing clinical trials for kidney disease have been repurposed from other indications. Currently, the chemokine receptor inhibitor CCX140 holds promise for CKD and the p53 inhibitor QPI-1002 for AKI.
Collapse
Affiliation(s)
- Adrián M Ramos
- Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD), Laboratory of Renal and Vascular Pathology and Diabetes , Av. Reyes Católicos 2, 28040, Madrid , Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Ortiz A, Sanchez-Niño MD, Izquierdo MC, Martin-Cleary C, Garcia-Bermejo L, Moreno JA, Ruiz-Ortega M, Draibe J, Cruzado JM, Garcia-Gonzalez MA, Lopez-Novoa JM, Soler MJ, Sanz AB. Translational value of animal models of kidney failure. Eur J Pharmacol 2015; 759:205-20. [PMID: 25814248 DOI: 10.1016/j.ejphar.2015.03.026] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 02/08/2015] [Accepted: 03/12/2015] [Indexed: 11/28/2022]
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are associated with decreased renal function and increased mortality risk, while the therapeutic armamentarium is unsatisfactory. The availability of adequate animal models may speed up the discovery of biomarkers for disease staging and therapy individualization as well as design and testing of novel therapeutic strategies. Some longstanding animal models have failed to result in therapeutic advances in the clinical setting, such as kidney ischemia-reperfusion injury and diabetic nephropathy models. In this regard, most models for diabetic nephropathy are unsatisfactory in that they do not evolve to renal failure. Satisfactory models for additional nephropathies are needed. These include anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis, IgA nephropathy, anti-phospholipase-A2-receptor (PLA2R) membranous nephropathy and Fabry nephropathy. However, recent novel models hold promise for clinical translation. Thus, the AKI to CKD translation has been modeled, in some cases with toxins of interest for human CKD such as aristolochic acid. Genetically modified mice provide models for Alport syndrome evolving to renal failure that have resulted in clinical recommendations, polycystic kidney disease models that have provided clues for the development of tolvaptan, that was recently approved for the human disease in Japan; and animal models also contributed to target C5 with eculizumab in hemolytic uremic syndrome. Some ongoing trials explore novel concepts derived from models, such TWEAK targeting as tissue protection for lupus nephritis. We now review animal models reproducing diverse, genetic and acquired, causes of AKI and CKD evolving to kidney failure and discuss the contribution to clinical translation and prospects for the future.
Collapse
Affiliation(s)
- Alberto Ortiz
- Nephrology, IIS-Fundacion Jimenez Diaz, Madrid, Spain; REDinREN, Madrid, Spain; Universidad Autonoma de Madrid, Madrid, Spain; IRSIN, Madrid, Spain
| | | | - Maria C Izquierdo
- Nephrology, IIS-Fundacion Jimenez Diaz, Madrid, Spain; REDinREN, Madrid, Spain
| | | | - Laura Garcia-Bermejo
- REDinREN, Madrid, Spain; Dpt. of Pathology, Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Madrid, Spain
| | - Juan A Moreno
- Nephrology, IIS-Fundacion Jimenez Diaz, Madrid, Spain
| | - Marta Ruiz-Ortega
- Nephrology, IIS-Fundacion Jimenez Diaz, Madrid, Spain; REDinREN, Madrid, Spain; Universidad Autonoma de Madrid, Madrid, Spain
| | - Juliana Draibe
- REDinREN, Madrid, Spain; Nephrology Department, Hospital Universitari de Bellvitge, IDIBELL, L׳Hospitalet de Llobregat, Barcelona, Spain
| | - Josep M Cruzado
- REDinREN, Madrid, Spain; Nephrology Department, Hospital Universitari de Bellvitge, IDIBELL, L׳Hospitalet de Llobregat, Barcelona, Spain
| | - Miguel A Garcia-Gonzalez
- REDinREN, Madrid, Spain; Laboratorio de Nefrología, Complexo Hospitalario de Santiago de Compostela (CHUS), Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - Jose M Lopez-Novoa
- REDinREN, Madrid, Spain; Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamnca, Spain
| | - Maria J Soler
- REDinREN, Madrid, Spain; Nephrology Department, Hospital del Mar, Barcelona, Spain
| | - Ana B Sanz
- Nephrology, IIS-Fundacion Jimenez Diaz, Madrid, Spain; REDinREN, Madrid, Spain.
| | | |
Collapse
|
25
|
Ortiz A. Translational nephrology: what translational research is and a bird's-eye view on translational research in nephrology. Clin Kidney J 2015; 8:14-22. [PMID: 25713705 PMCID: PMC4310441 DOI: 10.1093/ckj/sfu142] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 12/16/2014] [Indexed: 01/05/2023] Open
Abstract
The ultimate aim of biomedical research is to preserve health and improve patient outcomes. However, by a variety of measures, preservation of kidney health and patient outcomes in kidney disease are suboptimal. Severe acute kidney injury has been treated solely by renal replacement therapy for over 50 years and mortality still hovers at around 50%. Worldwide deaths from chronic kidney disease (CKD) increased by 80% in 20 years--one of the greatest increases among major causes of death. This dramatic data concur with huge advances in the cellular and molecular pathophysiology of kidney disease and its consequences. The gap appears to be the result of sequential roadblocks that impede an adequate flow from basic research to clinical development [translational research type 1 (T1), bench-to-bed and back] and from clinical development to clinical practice and widespread implementation (translational research T2) that supported by healthcare policy-making reaches all levels of society throughout the globe (sometimes called translational research T3). Thus, it is more than 10 years since the introduction of the last new-concept drug for CKD patients, cinacalcet; and 30 years since the introduction of reninangiotensin system (RAS) blockade, the current mainstay to prevent progression of CKD, illustrating the basic science-clinical practice disconnect. Roadblocks from clinical advances to widespread implementation, together with lag time-to-benefit may underlie the 20 years since the description of the antiproteinuric effect of RAS blockade to the observation of decreased age-adjusted incidence of endstage renal disease due to diabetic kidney disease. Only a correct understanding of the roadblocks in translational medicine and a full embracement of a translational research culture will spread the benefits of the biomedical revolution to its ultimate destinatary, the society.
Collapse
Affiliation(s)
- Alberto Ortiz
- IIS-Fundacion Jimenez Diaz, School of Medicine, Universidad Autonoma de Madrid, Madrid, Spain
- Fundacion Renal Iñigo Alvarez de Toledo-IRSIN and REDINREN, Madrid, Spain
| |
Collapse
|
26
|
McCampbell KK, Wingert RA. New tides: using zebrafish to study renal regeneration. Transl Res 2014; 163:109-22. [PMID: 24183931 PMCID: PMC3946610 DOI: 10.1016/j.trsl.2013.10.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Revised: 09/24/2013] [Accepted: 10/08/2013] [Indexed: 12/30/2022]
Abstract
Over the past several decades, the zebrafish has become one of the major vertebrate model organisms used in biomedical research. In this arena, the zebrafish has emerged as an applicable system for the study of kidney diseases and renal regeneration. The relevance of the zebrafish model for nephrology research has been increasingly appreciated as the understanding of zebrafish kidney structure, ontogeny, and the response to damage has steadily expanded. Recent studies have documented the amazing regenerative characteristics of the zebrafish kidney, which include the ability to replace epithelial populations after acute injury and to grow new renal functional units, termed nephrons. Here we discuss how nephron composition is conserved between zebrafish and mammals, and highlight how recent findings from zebrafish studies utilizing transgenic technologies and chemical genetics can complement traditional murine approaches in the effort to dissect how the kidney responds to acute damage and identify therapeutics that enhance human renal regeneration.
Collapse
Affiliation(s)
| | - Rebecca A Wingert
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Ind.
| |
Collapse
|
27
|
Poveda J, Tabara LC, Fernandez-Fernandez B, Martin-Cleary C, Sanz AB, Selgas R, Ortiz A, Sanchez-Niño MD. TWEAK/Fn14 and Non-Canonical NF-kappaB Signaling in Kidney Disease. Front Immunol 2013; 4:447. [PMID: 24339827 PMCID: PMC3857575 DOI: 10.3389/fimmu.2013.00447] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 11/26/2013] [Indexed: 12/27/2022] Open
Abstract
The incidence of acute kidney injury (AKI) and chronic kidney disease (CKD) is increasing. However, there is no effective therapy for AKI and current approaches only slow down, but do not prevent progression of CKD. TWEAK is a TNF superfamily cytokine. A solid base of preclinical data suggests a role of therapies targeting the TWEAK or its receptor Fn14 in AKI and CKD. In particular TWEAK/Fn14 targeting may preserve renal function and decrease cell death, inflammation, proteinuria, and fibrosis in mouse animal models. Furthermore there is clinical evidence for a role of TWEAK in human kidney injury including increased tissue and/or urinary levels of TWEAK and parenchymal renal cell expression of the receptor Fn14. In this regard, clinical trials of TWEAK targeting are ongoing in lupus nephritis. Nuclear factor-kappa B (NF-κB) activation plays a key role in TWEAK-elicited inflammatory responses. Activation of the non-canonical NF-κB pathway is a critical difference between TWEAK and TNF. TWEAK activation of the non-canonical NF-κB pathways promotes inflammatory responses in tubular cells. However, there is an incomplete understanding of the role of non-canonical NF-κB activation in kidney disease and on its contribution to TWEAK actions in vivo.
Collapse
Affiliation(s)
- Jonay Poveda
- Department of Nephrology, IIS-Fundacion Jimenez Diaz, Universidad Autonoma de Madrid and IRSIN , Madrid , Spain
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Poureetezadi SJ, Wingert RA. Congenital and Acute Kidney Disease: Translational Research Insights from Zebrafish Chemical Genetics. ACTA ACUST UNITED AC 2013; 1:112. [PMID: 24653992 DOI: 10.4172/2327-5146.1000112] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Today, acute kidney injury (AKI) and congenital anomalies of the kidney and urinary tract (CAKUT) represent major issues in healthcare. Both AKI and CAKUT can lead to end stage renal disease (ESRD) that requires life-long medical care with renal replacement therapy. Renal replacement by dialysis is intensive, and kidney transplantation is restricted by organ availability. These limitations, along with the growing epidemic of patients affected by kidney disease, highlight the significant need to identify alternative ways to treat renal injury and birth defects. Drug discovery is one promising avenue of current research. Here, we discuss zebrafish chemical genetics and its latent potency as a method to rapidly identify small molecule therapeutics to accelerate recovery after AKI. Specifically, we review two groundbreaking studies that have recently provided a template to screen for compounds that expand the renal progenitor field in development that were capable of treating AKI in both the zebrafish and the mouse. These new findings demonstrate that drug discovery using zebrafish can be used for relevant translational research to identify clinical interventions for renal conditions in humans.
Collapse
Affiliation(s)
| | - Rebecca A Wingert
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|