1
|
Huang DX, Kang X, Jiang LJ, Zhu DL, Yang L, Luo JY, Yang MM, Li W, Wang GP, Wen Y, Huang Z, Tang LJ. Exploring the impact of high-altitude de-acclimatization on renal function: The roles of oxidative and endoplasmic reticulum stress in rat models. Biochem Biophys Res Commun 2024; 708:149770. [PMID: 38518722 DOI: 10.1016/j.bbrc.2024.149770] [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: 01/18/2024] [Revised: 03/05/2024] [Accepted: 03/11/2024] [Indexed: 03/24/2024]
Abstract
BACKGROUND High-altitude de-acclimatization (HADA) significantly impacts physiological functions when individuals acclimatize to high altitudes return to lower altitudes. This study investigates HADA's effects on renal function and structure in rats, focusing on oxidative and endoplasmic reticulum stress as potential mechanisms of renal injury. OBJECTIVE To elucidate the pathophysiological mechanisms of renal damage in HADA and evaluate the efficacy of antioxidants Vitamin C (Vit C) and tauroursodeoxycholic acid (TUDCA) in mitigating these effects. METHODS 88 male Sprague-Dawley rats were randomly divided into a control group, a high-altitude (HA) group, a high-altitude de-acclimatization (HADA) group, and a treatment group. The control group was housed in a sea level environment (500 m), while the HA, HADA, and treatment groups were placed in a simulated high-altitude chamber (5000 m) for 90 days. After this period, the HA group completed the modeling phase; the HADA group was further subdivided into four subgroups, each continuing to be housed in a sea level environment for 3, 7, 14, and 30 days, respectively. The treatment group was split into the Vit C group, the TUDCA group, and two placebo groups, receiving medication for 3 consecutive days, once daily upon return to the sea level. The Vit C group received 100 mg/kg Vit C solution via intravenous injection, the TUDCA group received 250 mg/kg TUDCA solution via intraperitoneal injection, and the placebo groups received an equivalent volume of saline similarly. Serum, urine, and kidney tissues were collected immediately after the modeling phase. Renal function and oxidative stress levels were assessed using biochemical and ELISA methods. Renal histopathology was observed with H&E, Masson's trichrome, PAS, and PASM staining. Transmission electron microscopy was used to examine the ultrastructure of glomeruli and filtration barrier. TUNEL staining assessed cortical apoptosis in the kidneys. Metabolomics was employed for differential metabolite screening and pathway enrichment analysis. RESULTS Compared to the control and HA groups, the HADA 3-day group (HADA-3D) exhibited elevated renal function indicators, significant pathological damage, observable ultrastructural alterations including endoplasmic reticulum expansion and apoptosis. TUNEL-positive cells significantly increased, indicating heightened oxidative stress levels. Various differential metabolites were enriched in pathways related to oxidative and endoplasmic reticulum stress. Early intervention with Vit C and TUDCA markedly alleviated renal injury in HADA rats, significantly reducing the number of apoptotic cells, mitigating endoplasmic reticulum stress, and substantially lowering oxidative stress levels. CONCLUSION This study elucidates the pivotal roles of oxidative and endoplasmic reticulum stress in the early-stage renal injury in rats undergoing HADA. Early intervention with the Vit C and TUDCA significantly mitigates renal damage caused by HADA. These findings provide insights into the pathophysiological mechanisms of HADA and suggest potential therapeutic strategies for its future management.
Collapse
Affiliation(s)
- Dong-Xin Huang
- College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China; Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China; Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Xia Kang
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China; Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Li-Juan Jiang
- Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China; Department of General Surgery, The General Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China
| | - Dan-Ling Zhu
- Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310000, Zhejiang, China
| | - Lin Yang
- College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China; Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China; Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Jing-Ya Luo
- College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China; Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China; Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Meng-Meng Yang
- Medical Epigenetics Center, Basic Medicine Collage, Chongqing Medical University, Chongqing, 400042, China
| | - Wei Li
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China; Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Guo-Ping Wang
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China; Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Yi Wen
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China; Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China; Department of General Surgery, The General Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China
| | - Zhu Huang
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China; Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China; Department of Hyperbaric Oxygen, The General Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China.
| | - Li-Jun Tang
- College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China; Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China; Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China; Department of General Surgery, The General Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China.
| |
Collapse
|
2
|
Bondi CD, Hartman HL, Rush BM, Tan RJ. Podocyte-Specific Deletion of MCP-1 Fails to Protect against Angiotensin II- or Adriamycin-Induced Glomerular Disease. Int J Mol Sci 2024; 25:4987. [PMID: 38732210 PMCID: PMC11084322 DOI: 10.3390/ijms25094987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/25/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Investigating the role of podocytes in proteinuric disease is imperative to address the increasing global burden of chronic kidney disease (CKD). Studies strongly implicate increased levels of monocyte chemoattractant protein-1 (MCP-1/CCL2) in proteinuric CKD. Since podocytes express the receptor for MCP-1 (i.e., CCR2), we hypothesized that podocyte-specific MCP-1 production in response to stimuli could activate its receptor in an autocrine manner, leading to further podocyte injury. To test this hypothesis, we generated podocyte-specific MCP-1 knockout mice (Podo-Mcp-1fl/fl) and exposed them to proteinuric injury induced by either angiotensin II (Ang II; 1.5 mg/kg/d, osmotic minipump) or Adriamycin (Adr; 18 mg/kg, intravenous bolus). At baseline, there were no between-group differences in body weight, histology, albuminuria, and podocyte markers. After 28 days, there were no between-group differences in survival, change in body weight, albuminuria, kidney function, glomerular injury, and tubulointerstitial fibrosis. The lack of protection in the knockout mice suggests that podocyte-specific MCP-1 production is not a major contributor to either Ang II- or Adr-induced glomerular disease, implicating that another cell type is the source of pathogenic MCP-1 production in CKD.
Collapse
Affiliation(s)
- Corry D. Bondi
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 152671, USA; (H.L.H.); (B.M.R.); (R.J.T.)
| | | | | | | |
Collapse
|
3
|
Alexander A, Herz J, Calvier L. Reelin through the years: From brain development to inflammation. Cell Rep 2023; 42:112669. [PMID: 37339050 PMCID: PMC10592530 DOI: 10.1016/j.celrep.2023.112669] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/09/2023] [Accepted: 06/02/2023] [Indexed: 06/22/2023] Open
Abstract
Reelin was originally identified as a regulator of neuronal migration and synaptic function, but its non-neuronal functions have received far less attention. Reelin participates in organ development and physiological functions in various tissues, but it is also dysregulated in some diseases. In the cardiovascular system, Reelin is abundant in the blood, where it contributes to platelet adhesion and coagulation, as well as vascular adhesion and permeability of leukocytes. It is a pro-inflammatory and pro-thrombotic factor with important implications for autoinflammatory and autoimmune diseases such as multiple sclerosis, Alzheimer's disease, arthritis, atherosclerosis, or cancer. Mechanistically, Reelin is a large secreted glycoprotein that binds to several membrane receptors, including ApoER2, VLDLR, integrins, and ephrins. Reelin signaling depends on the cell type but mostly involves phosphorylation of NF-κB, PI3K, AKT, or JAK/STAT. This review focuses on non-neuronal functions and the therapeutic potential of Reelin, while highlighting secretion, signaling, and functional similarities between cell types.
Collapse
Affiliation(s)
- Anna Alexander
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX, USA
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX, USA; Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA; Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Laurent Calvier
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
4
|
Ha TS, Seong SB, Ha DS, Kim SJ. Upregulation of NADH/NADPH oxidase 4 by angiotensin II induces podocyte apoptosis. Kidney Res Clin Pract 2023; 42:202-215. [PMID: 37037482 PMCID: PMC10085724 DOI: 10.23876/j.krcp.22.198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 01/15/2023] [Indexed: 04/03/2023] Open
Abstract
Background: Angiotensin II induces glomerular and podocyte injury via systemic and local vasoconstrictive or non-hemodynamic effects including oxidative stress. The release of reactive oxygen species (ROS) from podocytes may participate in the development of glomerular injury and proteinuria. We studied the role of oxidative stress in angiotensin II-induced podocyte apoptosis.Methods: Mouse podocytes were incubated in media containing various concentrations of angiotensin II at different incubation times and were transfected with NADH/NADPH oxidase 4 (Nox4) or angiotensin II type 1 receptor for 24 hours. The changes in intracellular and mitochondrial ROS production and podocyte apoptosis were measured according to the presence of angiotensin II.Results: Angiotensin II increased the generation of mitochondrial superoxide anions and ROS levels but suppressed superoxide dismutase activity in a dose- and time-dependent manner that was reversed by probucol, an antioxidant. Angiotensin II increased Nox4 protein and expression by a transcriptional mechanism that was also reversed by probucol. In addition, the suppression of Nox4 by small interfering RNA (siRNA) reduced the oxidative stress induced by angiotensin II. Angiotensin II treatment also upregulated AT1R protein. Furthermore, angiotensin II promoted podocyte apoptosis, which was reduced significantly by probucol and Nox4 siRNA and also recovered by angiotensin II type 1 receptor siRNA.Conclusion: Our findings suggest that angiotensin II increases the generation of mitochondrial superoxide anions and ROS levels via the upregulation of Nox4 and angiotensin II type 1 receptor. This can be prevented by Nox4 inhibition and/or antagonizing angiotensin II type 1 receptor as well as use of antioxidants.
Collapse
Affiliation(s)
- Tae-Sun Ha
- Department of Pediatrics, Chungbuk National University College of Medicine, Cheongju, Republic of Korea
- Department of Pediatrics, Chungbuk National University Hospital, Cheongju, Republic of Korea
- Correspondence: Tae-Sun Ha Department of Pediatrics, Chungbuk National University College of Medicine, 1 Chungdae-ro, Seowon-gu, Cheongju 28644, Republic of Korea. E-mail:
| | - Su-Bin Seong
- Department of Pediatrics, Chungbuk National University College of Medicine, Cheongju, Republic of Korea
| | - Dong-Soo Ha
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Seung Jung Kim
- Department of Pediatrics, Chungbuk National University College of Medicine, Cheongju, Republic of Korea
| |
Collapse
|
5
|
Ma S, Xu J, Zheng Y, Li Y, Wang Y, Li H, Fang Z, Li J. Qian Yang Yu Yin granule improves hypertensive renal damage: A potential role for TRPC6-CaMKKβ-AMPK-mTOR-mediated autophagy. JOURNAL OF ETHNOPHARMACOLOGY 2023; 302:115878. [PMID: 36341814 DOI: 10.1016/j.jep.2022.115878] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/16/2022] [Accepted: 10/24/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Qian Yang Yu Yin granules (QYYYG) have a long history in the treatment of hypertensive renal damage (HRD) in China. Clinical studies have found that QYYYG stabilizes blood pressure and prevents early renal damage. However, the exact mechanism is not entirely clear. AIM OF THE STUDY To evaluate the therapeutic effect and further explore the therapeutic mechanism of QYYYG against HRD. MATERIALS AND METHODS The efficacy of QYYYG in treating HRD was assessed in spontaneous hypertension rats (SHR). Renal autophagy and the TRPC6-CaMKKβ-AMPK pathway in rats were evaluated. The regulatory role of QYYYG in angiotensin II (Ang II) induced abnormal autophagy in rat podocytes was determined by detecting autophagy-related proteins, intracellular Ca2+ content, and the TRPC6-CaMKKβ-AMPK-mTOR pathway expressions. Finally, we established a stable rat podocyte cell line overexpressing TRPC6 and used the cells to verify the regulatory effects of QYYYG. RESULTS QYYYG alleviated HRD and reversed the abnormal expression of autophagy-related genes in the SHR. In vitro, QYYYG protected against Ang II-induced podocyte damage. Furthermore, treatment of podocytes with QYYYG reversed Ang II-induced autophagy and inhibited Ang II-stimulated TRPC6 activation, Ca2+ influx and activation CaMKKβ-AMPK pathway. Overexpression of TRPC6 resulted in pronounced activation of CaMKKβ, AMPK, and autophagy induction in rat podocytes, which were significantly attenuated by QYYYG. CONCLUSIONS The present study suggested that QYYYG may exert its HRD protective effects in part by regulating the abnormal autophagy of podocytes through the TRPC6-CaMKKβ-AMPK-mTOR pathway.
Collapse
Affiliation(s)
- Siqi Ma
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Junyao Xu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Yawei Zheng
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Yin Li
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Yixuan Wang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| | - Haitao Li
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Zhuyuan Fang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| | - Jie Li
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
6
|
Dines V, Suvakov S, Kattah A, Vermunt J, Narang K, Jayachandran M, Abou Hassan C, Norby AM, Garovic VD. Preeclampsia and the Kidney: Pathophysiology and Clinical Implications. Compr Physiol 2023; 13:4231-4267. [PMID: 36715282 DOI: 10.1002/cphy.c210051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Preeclampsia and other hypertensive disorders of pregnancy are major contributors to maternal morbidity and mortality worldwide. This group of disorders includes chronic hypertension, gestational hypertension, preeclampsia, preeclampsia superimposed on chronic hypertension, and eclampsia. The body undergoes important physiological changes during pregnancy to allow for normal placental and fetal development. Several mechanisms have been proposed that may lead to preeclampsia, including abnormal placentation and placental hypoxia, impaired angiogenesis, excessive pro-inflammatory response, immune system imbalance, abnormalities of cellular senescence, alterations in regulation and activity of angiotensin II, and oxidative stress, ultimately resulting in upregulation of multiple mediators of endothelial cell dysfunction leading to maternal disease. The clinical implications of preeclampsia are significant as there are important short-term and long-term health consequences for those affected. Preeclampsia leads to increased risk of preterm delivery and increased morbidity and mortality of both the developing fetus and mother. Preeclampsia also commonly leads to acute kidney injury, and women who experience preeclampsia or another hypertensive disorder of pregnancy are at increased lifetime risk of chronic kidney disease and cardiovascular disease. An understanding of normal pregnancy physiology and the pathophysiology of preeclampsia is essential to develop novel treatment approaches and manage patients with preeclampsia and hypertensive disorders of pregnancy. © 2023 American Physiological Society. Compr Physiol 13:4231-4267, 2023.
Collapse
Affiliation(s)
- Virginia Dines
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Sonja Suvakov
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrea Kattah
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Jane Vermunt
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Kavita Narang
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Coline Abou Hassan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Alexander M Norby
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Vesna D Garovic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA.,Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
7
|
Erichsen L, Kloss LDF, Thimm C, Bohndorf M, Schichel K, Wruck W, Adjaye J. Derivation of the Immortalized Cell Line UM51-PrePodo-hTERT and Its Responsiveness to Angiotensin II and Activation of the RAAS Pathway. Cells 2023; 12:342. [PMID: 36766685 PMCID: PMC9913089 DOI: 10.3390/cells12030342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/12/2023] [Accepted: 01/14/2023] [Indexed: 01/18/2023] Open
Abstract
Recent demographic studies predict there will be a considerable increase in the number of elderly people within the next few decades. Aging has been recognized as one of the main risk factors for the world's most prevalent diseases such as neurodegenerative disorders, cancer, cardiovascular disease, and metabolic diseases. During the process of aging, a gradual loss of tissue volume and organ function is observed, which is partially caused by replicative senescence. The capacity of cellular proliferation and replicative senescence is tightly regulated by their telomere length. When telomere length is critically shortened with progressive cell division, cells become proliferatively arrested, and DNA damage response and cellular senescence are triggered, whereupon the "Hayflick limit" is attained at this stage. Podocytes are a cell type found in the kidney glomerulus where they have major roles in blood filtration. Mature podocytes are terminal differentiated cells that are unable to undergo cell division in vivo. For this reason, the establishment of primary podocyte cell cultures has been very challenging. In our present study, we present the successful immortalization of a human podocyte progenitor cell line, of which the primary cells were isolated directly from the urine of a 51-year-old male. The immortalized cell line was cultured over the course of one year (~100 passages) with high proliferation capacity, endowed with contact inhibition and P53 expression. Furthermore, by immunofluorescence-based expression and quantitative real-time PCR for the podocyte markers CD2AP, LMX1B, NPHS1, SYNPO and WT1, we confirmed the differentiation capacity of the immortalized cells. Finally, we evaluated and confirmed the responsiveness of the immortalized cells on the main mediator angiotensin II (ANGII) of the renin-angiotensin system (RAAS). In conclusion, we have shown that it is possible to bypass cellular replicative senescence (Hayflick limit) by TERT-driven immortalization of human urine-derived pre-podocyte cells from a 51-year-old African male.
Collapse
Affiliation(s)
- Lars Erichsen
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Lea Doris Friedel Kloss
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Chantelle Thimm
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Martina Bohndorf
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Kira Schichel
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Wasco Wruck
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - James Adjaye
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany
- EGA Institute for Women’s Health, University College London, 86-96 Chenies Mews, London WC1E 6HX, UK
| |
Collapse
|
8
|
Erichsen L, Thimm C, Bohndorf M, Rahman MS, Wruck W, Adjaye J. Activation of the Renin–Angiotensin System Disrupts the Cytoskeletal Architecture of Human Urine-Derived Podocytes. Cells 2022; 11:cells11071095. [PMID: 35406662 PMCID: PMC8997628 DOI: 10.3390/cells11071095] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/10/2022] [Accepted: 03/21/2022] [Indexed: 01/27/2023] Open
Abstract
High blood pressure is one of the major public health problems that causes severe disorders in several tissues including the human kidney. One of the most important signaling pathways associated with the regulation of blood pressure is the renin–angiotensin system (RAS), with its main mediator angiotensin II (ANGII). Elevated levels of circulating and intracellular ANGII and aldosterone lead to pro-fibrotic, -inflammatory, and -hypertrophic milieu that causes remodeling and dysfunction in cardiovascular and renal tissues. Furthermore, ANGII has been recognized as a major risk factor for the induction of apoptosis in podocytes, ultimately leading to chronic kidney disease (CKD). In the past, disease modeling of kidney-associated diseases was extremely difficult, as the derivation of kidney originated cells is very challenging. Here we describe a differentiation protocol for reproducible differentiation of sine oculis homeobox homolog 2 (SIX2)-positive urine-derived renal progenitor cells (UdRPCs) into podocytes bearing typical cellular processes. The UdRPCs-derived podocytes show the activation of the renin–angiotensin system by being responsive to ANGII stimulation. Our data reveal the ANGII-dependent downregulation of nephrin (NPHS1) and synaptopodin (SYNPO), resulting in the disruption of the podocyte cytoskeletal architecture, as shown by immunofluorescence-based detection of α-Actinin. Furthermore, we show that the cytoskeletal disruption is mainly mediated through angiotensin II receptor type 1 (AGTR1) signaling and can be rescued by AGTR1 inhibition with the selective, competitive angiotensin II receptor type 1 antagonist, losartan. In the present manuscript we confirm and propose UdRPCs differentiated to podocytes as a unique cell type useful for studying nephrogenesis and associated diseases. Furthermore, the responsiveness of UdRPCs-derived podocytes to ANGII implies potential applications in nephrotoxicity studies and drug screening.
Collapse
|
9
|
Yin L, Yu L, He JC, Chen A. Controversies in Podocyte Loss: Death or Detachment? Front Cell Dev Biol 2021; 9:771931. [PMID: 34881244 PMCID: PMC8645964 DOI: 10.3389/fcell.2021.771931] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/03/2021] [Indexed: 12/21/2022] Open
Abstract
Glomerular podocytes are characterized by terminally differentiated epithelial cells with limited proliferating ability; thus, podocyte loss could not be fully compensated by podocyte regeneration. A large body of clinical studies collectively demonstrated that podocyte loss correlated with glomerular diseases progression. Both podocyte death and podocyte detachment lead to podocyte loss; however, which one is the main cause remains controversial. Up to date, multiple mechanisms are involved in podocyte death, including programmed apoptotic cell death (apoptosis and anoikis), programmed nonapoptotic cell death (autophagy, entosis, and podoptosis), immune-related cell death (pyroptosis), and other types of cell death (necroptosis and mitotic catastrophe-related cell death). Apoptosis is considered a common mechanism of podocyte loss; however, most of the data were generated in vitro and the evidence of in vivo podocyte apoptosis is limited. The isolation of podocytes in the urine and subsequent culture of urinary podocytes in vitro suggest that detachment of viable podocytes could be another important mechanism for podocyte loss. In this review, we summarize recent advances that address this controversial topic on the specific circumstances of podocyte loss.
Collapse
Affiliation(s)
- Lijun Yin
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, China
| | - Lu Yu
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, China.,Department of Health Sciences, Boston University College of Health and Rehabilitation Sciences: Sargent College, Boston University, Boston, MA, United States
| | - John Cijiang He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Renal Program, James J. Peters Veterans Affairs Medical Center at Bronx, New York, NY, United States
| | - Anqun Chen
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, China
| |
Collapse
|
10
|
Corrêa JWN, Boaro KR, Sene LB, Polidoro JZ, Salles TA, Martins FL, Bendhack LM, Girardi ACC. Antiproteinuric and Hyperkalemic Mechanisms Activated by Dual Versus Single Blockade of the RAS in Renovascular Hypertensive Rats. Front Physiol 2021; 12:656460. [PMID: 34177612 PMCID: PMC8221266 DOI: 10.3389/fphys.2021.656460] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/09/2021] [Indexed: 11/24/2022] Open
Abstract
This study aimed to investigate the antiproteinuric and hyperkalemic mechanisms activated by dual renin-angiotensin system (RAS) blockade in renovascular hypertensive rats (2-kidney 1-clip model [2K-1C]). Six weeks after clipping the left renal artery or sham operation (2K), rats were treated with losartan, enalapril, or both drugs for two weeks. We found that 2K-1C rats displayed higher tail-cuff blood pressure (BP), increased non-clipped kidney Ang II concentration, and more pronounced urinary albumin excretion than 2K. BP was decreased by the treatment with either enalapril or losartan, and the combination of both drugs promoted an additional antihypertensive effect in 2K-1C rats. Renal Ang II content and albuminuria were reduced by either enalapril or losartan in monotherapy and restored to control levels by dual RAS blockade. Albuminuria in 2K-1C rats was accompanied by downregulation of the glomerular slit protein podocin, reduction of the endocytic receptors megalin and cubilin, and a marked decrease in the expression of the ClC-5 chloride channel, compared to 2K animals. Treatment with losartan and enalapril in monotherapy or combination increased the expression of podocin, cubilin, and ClC-5. However, only the combined therapy normalized podocin, cubilin, and ClC-5 protein abundance in the non-clipped kidney of 2K-1C rats. Renovascular hypertensive 2K-1C rats had a lower concentration of plasma potassium compared to 2K rats. Single RAS blockade normalized potassium plasma concentration, whereas 2K-1C rats treated with dual RAS blockade exhibited hyperkalemia. Hypokalemia in 2K-1C rats was accompanied by an increase in the cleaved activated forms of α-ENaC and γ-ENaC and the expression of β-ENaC. Combined RAS blockade but not monotherapy significantly reduced the expression of these ENaC subunits in 2K-1C rats. Indeed, double RAS blockade reduced the abundance of cleaved-α-ENaC to levels lower than those of 2K rats. Collectively, these results demonstrate that the antiproteinuric effect of dual RAS blockade in 2K-1C rats is associated with the restored abundance of podocin and cubilin, and ClC-5. Moreover, double RAS blockade-induced hyperkalemia may be due, at least partially, to an exaggerated downregulation of cleaved α-ENaC in the non-clipped kidney of renovascular hypertensive rats.
Collapse
Affiliation(s)
- José Wilson N Corrêa
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor) University of São Paulo Medical School, São Paulo, Brazil.,Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Amazonas, Manaus, Brazil
| | - Karoline R Boaro
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor) University of São Paulo Medical School, São Paulo, Brazil
| | - Letícia B Sene
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor) University of São Paulo Medical School, São Paulo, Brazil
| | - Juliano Z Polidoro
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor) University of São Paulo Medical School, São Paulo, Brazil
| | - Thiago A Salles
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor) University of São Paulo Medical School, São Paulo, Brazil
| | - Flavia L Martins
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor) University of São Paulo Medical School, São Paulo, Brazil
| | - Lusiane M Bendhack
- Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Adriana C C Girardi
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor) University of São Paulo Medical School, São Paulo, Brazil
| |
Collapse
|
11
|
Bensaada I, Robin B, Perez J, Salemkour Y, Chipont A, Camus M, Lemoine M, Guyonnet L, Lazareth H, Letavernier E, Hénique C, Tharaux PL, Lenoir O. Calpastatin prevents Angiotensin II-mediated podocyte injury through maintenance of autophagy. Kidney Int 2021; 100:90-106. [PMID: 33675847 DOI: 10.1016/j.kint.2021.02.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 01/29/2021] [Accepted: 02/10/2021] [Indexed: 12/20/2022]
Abstract
The strong predictive value of proteinuria in chronic glomerulopathies is firmly established as well as the pathogenic role of angiotensin II promoting progression of glomerular disease with an altered glomerular filtration barrier, podocyte injury and scarring of glomeruli. Here we found that chronic angiotensin II-induced hypertension inhibited autophagy flux in mouse glomeruli. Deletion of Atg5 (a gene encoding a protein involved autophagy) specifically in the podocyte resulted in accelerated angiotensin II-induced podocytopathy, accentuated albuminuria and glomerulosclerosis. This indicates that autophagy is a key protective mechanism in the podocyte in this condition. Angiotensin-II induced calpain activity in podocytes inhibits autophagy flux. Podocytes from mice with transgenic expression of the endogenous calpain inhibitor calpastatin displayed higher podocyte autophagy at baseline that was resistant to angiotensin II-dependent inhibition. Also, sustained autophagy with calpastatin limited podocyte damage and albuminuria. These findings suggest that hypertension has pathogenic effects on the glomerular structure and function, in part through activation of calpains leading to blockade of podocyte autophagy. These findings uncover an original mechanism whereby angiotensin II-mediated hypertension inhibits autophagy via calcium-induced recruitment of calpain with pathogenic consequences in case of imbalance by calpastatin activity. Thus, preventing a calpain-mediated decrease in autophagy may be a promising new therapeutic strategy for nephropathies associated with high renin-angiotensin system activity.
Collapse
Affiliation(s)
| | - Blaise Robin
- Université de Paris, PARCC, Inserm, Paris, France
| | - Joëlle Perez
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | | | - Anna Chipont
- Université de Paris, PARCC, Inserm, Paris, France
| | - Marine Camus
- Université de Paris, PARCC, Inserm, Paris, France
| | | | - Lea Guyonnet
- Université de Paris, PARCC, Inserm, Paris, France
| | | | | | | | | | | |
Collapse
|
12
|
Nistala R, Meuth AI, Smith C, An J, Habibi J, Hayden MR, Johnson M, Aroor A, Whaley-Connell A, Sowers JR, McKarns SC, Bender SB. DPP4 inhibition mitigates ANG II-mediated kidney immune activation and injury in male mice. Am J Physiol Renal Physiol 2021; 320:F505-F517. [PMID: 33522410 DOI: 10.1152/ajprenal.00565.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Recent evidence suggests that dipeptidyl peptidase-4 (DPP4) inhibition with saxagliptin (Saxa) is renoprotective under comorbid conditions associated with activation of the renin-angiotensin-aldosterone system (RAAS), such as diabetes, obesity, and hypertension, which confer a high cardiovascular risk. Immune system activation is now recognized as a contributor to RAAS-mediated tissue injury, and, importantly, immunomodulatory effects of DPP4 have been reported. Accordingly, we examined the hypothesis that DPP4 inhibition with Saxa attenuates angiotensin II (ANG II)-induced kidney injury and albuminuria via attenuation of immune activation in the kidney. To this end, male mice were infused with either vehicle or ANG II (1,000 ng/kg/min, s.c.) for 3 wk and received either placebo or Saxa (10 mg/kg/day, p.o.) during the final 2 wk. ANG II infusion increased kidney, but not plasma, DPP4 activity in vivo as well as DPP4 activity in cultured proximal tubule cells. The latter was prevented by angiotensin receptor blockade with olmesartan. Further, ANG II induced hypertension and kidney injury characterized by mesangial expansion, mitochondrial damage, reduced brush border megalin expression, and albuminuria. Saxa inhibited DPP4 activity ∼50% in vivo and attenuated ANG II-mediated kidney injury, independent of blood pressure. Further mechanistic experiments revealed mitigation by Saxa of proinflammatory and profibrotic mediators activated by ANG II in the kidney, including CD8+ T cells, resident macrophages (CD11bhiF4/80loLy6C-), and neutrophils. In addition, Saxa improved ANG II suppressed anti-inflammatory regulatory T cell and T helper 2 lymphocyte activity. Taken together, these results demonstrate, for the first time, blood pressure-independent involvement of renal DPP4 activation contributing to RAAS-dependent kidney injury and immune activation.NEW & NOTEWORTHY This work highlights the role of dipeptidyl peptidase-4 (DPP4) in promoting ANG II-mediated kidney inflammation and injury. Specifically, ANG II infusion in mice led to increases in blood pressure and kidney DPP4 activity, which then led to activation of CD8+ T cells, Ly6C- macrophages, and neutrophils and suppression of anti-inflammatory T helper 2 lymphocytes and regulatory T cells. Collectively, this led to kidney injury, characterized by mesangial expansion, mitochondrial damage, and albuminuria, which were mitigated by DPP4 inhibition independent of blood pressure reduction.
Collapse
Affiliation(s)
- Ravi Nistala
- Divisions of Nephrology and Hypertension, University of Missouri School of Medicine, Columbia, Missouri.,Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, Missouri.,Department of Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri
| | - Alex I Meuth
- Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, Missouri.,Department of Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri.,Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Cassandra Smith
- Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, Missouri.,Department of Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri.,Divisions of Endocrinology and Metabolism, Department of Medicine, University of Missouri School of Medicine, Columbia, Missouri
| | - Jianzhong An
- Divisions of Nephrology and Hypertension, University of Missouri School of Medicine, Columbia, Missouri.,Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, Missouri.,Department of Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri
| | - Javad Habibi
- Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, Missouri.,Department of Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri.,Divisions of Endocrinology and Metabolism, Department of Medicine, University of Missouri School of Medicine, Columbia, Missouri
| | - M R Hayden
- Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, Missouri.,Department of Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri.,Divisions of Endocrinology and Metabolism, Department of Medicine, University of Missouri School of Medicine, Columbia, Missouri
| | - Megan Johnson
- Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, Missouri.,Divisions of Endocrinology and Metabolism, Department of Medicine, University of Missouri School of Medicine, Columbia, Missouri
| | - Annayya Aroor
- Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, Missouri.,Department of Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri.,Divisions of Endocrinology and Metabolism, Department of Medicine, University of Missouri School of Medicine, Columbia, Missouri
| | - Adam Whaley-Connell
- Divisions of Nephrology and Hypertension, University of Missouri School of Medicine, Columbia, Missouri.,Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, Missouri.,Department of Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri
| | - James R Sowers
- Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, Missouri.,Department of Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri.,Divisions of Endocrinology and Metabolism, Department of Medicine, University of Missouri School of Medicine, Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Susan C McKarns
- Departments of Microbiology and Immunology and Surgery, University of Missouri School of Medicine, Columbia, Missouri
| | - Shawn B Bender
- Department of Research, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri.,Department of Biomedical Sciences, University of Missouri, Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| |
Collapse
|
13
|
Arellano-Buendía AS, Castañeda-Lara LG, Loredo-Mendoza ML, García-Arroyo FE, Rojas-Morales P, Argüello-García R, Juárez-Rojas JG, Tapia E, Pedraza-Chaverri J, Sánchez-Lozada LG, Osorio-Alonso H. Effects of Allicin on Pathophysiological Mechanisms during the Progression of Nephropathy Associated to Diabetes. Antioxidants (Basel) 2020; 9:antiox9111134. [PMID: 33203103 PMCID: PMC7697950 DOI: 10.3390/antiox9111134] [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: 10/12/2020] [Revised: 11/03/2020] [Accepted: 11/13/2020] [Indexed: 02/06/2023] Open
Abstract
This study aimed to assess the impact of allicin on the course of diabetic nephropathy. Study groups included control, diabetes, and diabetes-treated rats. Allicin treatment (16 mg/kg day/p.o.) started after 1 month of diabetes onset and was administered for 30 days. In the diabetes group, the systolic blood pressure (SBP) increased, also, the oxidative stress and hypoxia in the kidney cortex were evidenced by alterations in the total antioxidant capacity as well as the expression of nuclear factor (erythroid-derived 2)-like 2/Kelch ECH associating protein 1 (Nrf2/Keap1), hypoxia-inducible factor 1-alpha (HIF-1α), vascular endothelial growth factor (VEGF), erythropoietin (Epo) and its receptor (Epo-R). Moreover, diabetes increased nephrin, and kidney injury molecule-1 (KIM-1) expression that correlated with mesangial matrix, the fibrosis index and with the expression of connective tissue growth factor (CTGF), transforming growth factor-β1 (TGF-β1), and α-smooth muscle actin (α-SMA). The insulin levels and glucose transporter protein type-4 (GLUT4) expression were decreased; otherwise, insulin receptor substrates 1 and 2 (IRS-1 and IRS-2) expression was increased. Allicin increased Nrf2 expression and decreased SBP, Keap1, HIF-1α, and VEGF expression. Concurrently, nephrin, KIM-1, the mesangial matrix, fibrosis index, and the fibrotic proteins were decreased. Additionally, allicin decreased hyperglycemia, improved insulin levels, and prevented changes in (GLUT4) and IRSs expression induced by diabetes. In conclusion, our results demonstrate that allicin has the potential to help in the treatment of diabetic nephropathy. The cellular mechanisms underlying its effects mainly rely on the regulation of antioxidant, antifibrotic, and antidiabetic mechanisms, which can contribute towards delay in the progression of renal disease.
Collapse
Affiliation(s)
- Abraham Said Arellano-Buendía
- Department of Cardio-Renal Physiopathology, Instituto Nacional de Cardiología “Ignacio Chávez”, México City 14080, Mexico; (A.S.A.-B.); (L.G.C.-L.); (F.E.G.-A.); (P.R.-M.); (E.T.); (L.G.S.-L.)
| | - Luis Gerardo Castañeda-Lara
- Department of Cardio-Renal Physiopathology, Instituto Nacional de Cardiología “Ignacio Chávez”, México City 14080, Mexico; (A.S.A.-B.); (L.G.C.-L.); (F.E.G.-A.); (P.R.-M.); (E.T.); (L.G.S.-L.)
| | - María L. Loredo-Mendoza
- Histopathology Laboratory, Research Subdivision, School of Medicine, Universidad Panamericana, Donatello 43, Mexico City 03910, Mexico;
| | - Fernando E. García-Arroyo
- Department of Cardio-Renal Physiopathology, Instituto Nacional de Cardiología “Ignacio Chávez”, México City 14080, Mexico; (A.S.A.-B.); (L.G.C.-L.); (F.E.G.-A.); (P.R.-M.); (E.T.); (L.G.S.-L.)
| | - Pedro Rojas-Morales
- Department of Cardio-Renal Physiopathology, Instituto Nacional de Cardiología “Ignacio Chávez”, México City 14080, Mexico; (A.S.A.-B.); (L.G.C.-L.); (F.E.G.-A.); (P.R.-M.); (E.T.); (L.G.S.-L.)
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Raúl Argüello-García
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico;
| | - Juan G. Juárez-Rojas
- Department of Endocrinology, Instituto Nacional de Cardiología “Ignacio Chávez” México City 14080, Mexico;
| | - Edilia Tapia
- Department of Cardio-Renal Physiopathology, Instituto Nacional de Cardiología “Ignacio Chávez”, México City 14080, Mexico; (A.S.A.-B.); (L.G.C.-L.); (F.E.G.-A.); (P.R.-M.); (E.T.); (L.G.S.-L.)
| | - José Pedraza-Chaverri
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Laura Gabriela Sánchez-Lozada
- Department of Cardio-Renal Physiopathology, Instituto Nacional de Cardiología “Ignacio Chávez”, México City 14080, Mexico; (A.S.A.-B.); (L.G.C.-L.); (F.E.G.-A.); (P.R.-M.); (E.T.); (L.G.S.-L.)
| | - Horacio Osorio-Alonso
- Department of Cardio-Renal Physiopathology, Instituto Nacional de Cardiología “Ignacio Chávez”, México City 14080, Mexico; (A.S.A.-B.); (L.G.C.-L.); (F.E.G.-A.); (P.R.-M.); (E.T.); (L.G.S.-L.)
- Correspondence: or
| |
Collapse
|
14
|
Xu N, Jiang S, Persson PB, Persson EAG, Lai EY, Patzak A. Reactive oxygen species in renal vascular function. Acta Physiol (Oxf) 2020; 229:e13477. [PMID: 32311827 DOI: 10.1111/apha.13477] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/22/2020] [Accepted: 04/14/2020] [Indexed: 12/14/2022]
Abstract
Reactive oxygen species (ROS) are produced by the aerobic metabolism. The imbalance between production of ROS and antioxidant defence in any cell compartment is associated with cell damage and may play an important role in the pathogenesis of renal disease. NADPH oxidase (NOX) family is the major ROS source in the vasculature and modulates renal perfusion. Upregulation of Ang II and adenosine activates NOX via AT1R and A1R in renal microvessels, leading to superoxide production. Oxidative stress in the kidney prompts renal vascular remodelling and increases preglomerular resistance. These are key elements in hypertension, acute and chronic kidney injury, as well as diabetic nephropathy. Renal afferent arterioles (Af), the primary resistance vessel in the kidney, fine tune renal hemodynamics and impact on blood pressure. Vice versa, ROS increase hypertension and diabetes, resulting in upregulation of Af vasoconstriction, enhancement of myogenic responses and change of tubuloglomerular feedback (TGF), which further promotes hypertension and diabetic nephropathy. In the following, we highlight oxidative stress in the function and dysfunction of renal hemodynamics. The renal microcirculatory alterations brought about by ROS importantly contribute to the pathophysiology of kidney injury, hypertension and diabetes.
Collapse
Affiliation(s)
- Nan Xu
- Department of Physiology Zhejiang University School of Medicine Hangzhou China
| | - Shan Jiang
- Department of Physiology Zhejiang University School of Medicine Hangzhou China
| | - Pontus B. Persson
- Charité ‐ Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Institute of Vegetative Physiology Berlin Germany
| | | | - En Yin Lai
- Department of Physiology Zhejiang University School of Medicine Hangzhou China
- Charité ‐ Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Institute of Vegetative Physiology Berlin Germany
| | - Andreas Patzak
- Charité ‐ Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Institute of Vegetative Physiology Berlin Germany
| |
Collapse
|
15
|
Wang L, Chang JH, Buckley AF, Spurney RF. Knockout of TRPC6 promotes insulin resistance and exacerbates glomerular injury in Akita mice. Kidney Int 2020; 95:321-332. [PMID: 30665571 DOI: 10.1016/j.kint.2018.09.026] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 09/22/2018] [Accepted: 09/27/2018] [Indexed: 12/17/2022]
Abstract
Gain-of-function mutations in TRPC6 cause familial focal segmental glomerulosclerosis, and TRPC6 is upregulated in glomerular diseases including diabetic kidney disease. We studied the effect of systemic TRPC6 knockout in the Akita model of type 1 diabetes. Knockout of TRPC6 inhibited albuminuria in Akita mice at 12 and 16 weeks of age, but this difference disappeared by 20 weeks. Knockout of TRPC6 also reduced tubular injury in Akita mice; however, mesangial expansion was significantly increased. Hyperglycemia and blood pressure were similar between TRPC6 knockout and wild-type Akita mice, but knockout mice were more insulin resistant. In cultured podocytes, knockout of TRPC6 inhibited expression of the calcium/calcineurin responsive gene insulin receptor substrate 2 and decreased insulin responsiveness. Insulin resistance is reported to promote diabetic kidney disease independent of blood glucose levels. While the mechanisms are not fully understood, insulin activates both Akt2 and ERK, which inhibits apoptosis signal regulated kinase 1 (ASK1)-p38-induced apoptosis. In cultured podocytes, hyperglycemia stimulated p38 signaling and induced apoptosis, which was reduced by insulin and ASK1 inhibition and enhanced by Akt or ERK inhibition. Glomerular p38 signaling was increased in TRPC6 knockout Akita mice and was associated with enhanced expression of the p38 gene target cyclooxygenase 2. These data suggest that knockout of TRPC6 in Akita mice promotes insulin resistance and exacerbates glomerular disease independent of hyperglycemia.
Collapse
Affiliation(s)
- Liming Wang
- Division of Nephrology, Department of Medicine, Duke University Health System, Durham, North Carolina, USA
| | - Jae-Hyung Chang
- Division of Nephrology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Anne F Buckley
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Robert F Spurney
- Division of Nephrology, Department of Medicine, Duke University Health System, Durham, North Carolina, USA; Durham VA Medical Center, Durham, North Carolina, USA.
| |
Collapse
|
16
|
Inoue K, Tian X, Velazquez H, Soda K, Wang Z, Pedigo CE, Wang Y, Cross E, Groener M, Shin JW, Li W, Hassan H, Yamamoto K, Mundel P, Ishibe S. Inhibition of Endocytosis of Clathrin-Mediated Angiotensin II Receptor Type 1 in Podocytes Augments Glomerular Injury. J Am Soc Nephrol 2019; 30:2307-2320. [PMID: 31511362 PMCID: PMC6900791 DOI: 10.1681/asn.2019010053] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 08/04/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Inhibition of the renin-angiotensin system remains a cornerstone in reducing proteinuria and progression of kidney failure, effects believed to be the result of reduction in BP and glomerular hyperfiltration. However, studies have yielded conflicting results on whether podocyte-specific angiotensin II (AngII) signaling directly induces podocyte injury. Previous research has found that after AngII stimulation, β-arrestin-bound angiotensin II receptor type 1 (AT1R) is internalized in a clathrin- and dynamin-dependent manner, and that Dynamin1 and Dynamin2 double-knockout mice exhibit impaired clathrin-mediated endocytosis. METHODS We used podocyte-specific Dyn double-knockout mice to examine AngII-stimulated AT1R internalization and signaling in primary podocytes and controls. We also examined the in vivo effect of AngII in these double-knockout mice through renin-angiotensin system blockers and through deletion of Agtr1a (which encodes the predominant AT1R isoform expressed in kidney, AT1aR). We tested calcium influx, Rac1 activation, and lamellipodial extension in control and primary podocytes of Dnm double-knockout mice treated with AngII. RESULTS We confirmed augmented AngII-stimulated AT1R signaling in primary Dnm double-knockout podocytes resulting from arrest of clathrin-coated pit turnover. Genetic ablation of podocyte Agtr1a in Dnm double-knockout mice demonstrated improved albuminuria and kidney function compared with the double-knockout mice. Isolation of podocytes from Dnm double-knockout mice revealed abnormal membrane dynamics, with increased Rac1 activation and lamellipodial extension, which was attenuated in Dnm double-knockout podocytes lacking AT1aR. CONCLUSIONS Our results indicate that inhibiting aberrant podocyte-associated AT1aR signaling pathways has a protective effect in maintaining the integrity of the glomerular filtration barrier.
Collapse
Affiliation(s)
- Kazunori Inoue
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Xuefei Tian
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Heino Velazquez
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Keita Soda
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Zhen Wang
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Christopher E Pedigo
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Ying Wang
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Elizabeth Cross
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Marwin Groener
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Jee-Won Shin
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Wei Li
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Hossam Hassan
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Koichi Yamamoto
- Department of Geriatric Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan; and
| | - Peter Mundel
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Shuta Ishibe
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut;
| |
Collapse
|
17
|
Wang J, Fu D, Senouthai S, You Y. Critical roles of PI3K/Akt/NF‑κB survival axis in angiotensin II‑induced podocyte injury. Mol Med Rep 2019; 20:5134-5144. [PMID: 31638199 PMCID: PMC6854545 DOI: 10.3892/mmr.2019.10733] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 08/30/2019] [Indexed: 12/12/2022] Open
Abstract
Numerous studies have reported that angiotensin (Ang) II, nephrin, and podocin serve pivotal roles in podocyte injury, and thus can lead to the occurrence of proteinuria and the progression of kidney diseases. This study aimed to investigate the effects of Ang II on the production of nephrin and podocin, and their relationship with podocyte injury. We also aimed to determine whether nephrin, podocin and caspase-9 production depends on the PI3K/Akt/nuclear factor (NF)-κB signaling pathway in cultured mouse podocytes. We treated mouse podocytes with different doses of Ang II (10−9, 10−8, 10−7 and 10−6 mol/l) for 12, 24, and 48 h to analyse cell viability, and at 10−6 mol/l Ang II for 12, 24, and 48 h to evaluate cell apoptosis. Cells were treated with 10−6 mol/l of Ang II and/or LY294002 (inhibitor of Akt) or 740Y-P (activator of PI3K) for 48 h to detect Akt, phosphorylated (phospho)-Akt, p65 NF-κB, and phospho-p65 NF-κB, nephrin, podocin and caspase-9 expression, and podocyte apoptosis. Treatment with Ang II suppressed the viability and promoted the apoptosis of podocytes in a dose- and time-dependent manner. Ang II decreased phospho-Akt, phospho-p65 NF-κB, nephrin, and podocin and increased caspase-9 expression, while podocyte apoptosis was promoted. LY294002 further enhanced Ang II-induced downregulation of Akt and p65 NF-κB activation, as well as upregulation of caspase-9 mRNA and protein, and promoted the apoptosis of podocytes. Of note, 740Y-P restored Ang II-induced downregulation of Akt and p65 NF-κB activation, and upregulation of caspase-9, and decreased podocyte apoptosis. Interestingly, LY294002 and 740Y-P were determined to have no notable effects on the expression of nephrin and podocin. The data suggested that Ang II could regulate the expression of nephrin, podocin and caspase-9. Collectively, our findings suggested that the PI3K/Akt/NF-κB survival axis may serve a pivotal role in podocyte injury.
Collapse
Affiliation(s)
- Junjie Wang
- Department of Nephrology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Zhuang Autonomous Region 533000, P.R. China
| | - Dongdong Fu
- Department of Nephrology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Zhuang Autonomous Region 533000, P.R. China
| | - Soulixay Senouthai
- Department of Nephrology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Zhuang Autonomous Region 533000, P.R. China
| | - Yanwu You
- Department of Nephrology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Zhuang Autonomous Region 533000, P.R. China
| |
Collapse
|
18
|
Liu Y, Dai E, Yang J. Quercetin suppresses glomerulosclerosis and TGF‑β signaling in a rat model. Mol Med Rep 2019; 19:4589-4596. [PMID: 30942399 PMCID: PMC6522826 DOI: 10.3892/mmr.2019.10118] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 03/06/2019] [Indexed: 12/14/2022] Open
Abstract
The transforming growth factor-β (TGF-β) signaling pathway is an important regulatory pathway in renal fibrosis and is abnormally activated in glomerulosclerosis. Quercetin is a common Chinese herbal medicine and has been reported to inhibit TGF-β signaling pathway activation. In the present study a glomerulosclerosis rat model was constructed and mice were treated with different concentrations of quercetin. Biochemical parameters, pathological indices and expression levels of TGF-β signaling pathway-associated proteins were detected using immunohistochemistry and western blotting. It was demonstrated that quercetin significantly improved physiological indices and altered the expression levels of TGF-β signaling pathway-associated proteins in rats with glomerulosclerosis. In conclusion, quercetin can regulate the TGF-β signaling pathway and reduce the progression of glomerulosclerosis.
Collapse
Affiliation(s)
- Yifan Liu
- Cooperation of Chinese and Western Medicine Department, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, P.R. China
| | - Enlai Dai
- Cooperation of Chinese and Western Medicine Department, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, P.R. China
| | - Jing Yang
- Department of Children Glomerular Disease, The Second Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
19
|
Cardoso VG, Gonçalves GL, Costa-Pessoa JM, Thieme K, Lins BB, Casare FAM, de Ponte MC, Camara NOS, Oliveira-Souza M. Angiotensin II-induced podocyte apoptosis is mediated by endoplasmic reticulum stress/PKC-δ/p38 MAPK pathway activation and trough increased Na +/H + exchanger isoform 1 activity. BMC Nephrol 2018; 19:179. [PMID: 30005635 PMCID: PMC6043975 DOI: 10.1186/s12882-018-0968-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 06/26/2018] [Indexed: 11/12/2022] Open
Abstract
Background Angiotensin II (Ang II) contributes to the progression of renal diseases associated with proteinuria and glomerulosclerosis mainly by inducing podocyte apoptosis. In the present study, we investigated whether the chronic effects of Ang II via AT1 receptor (AT1R) would result in endoplasmic reticulum (ER) stress/PKC-delta/p38 MAPK stimulation, and consequently podocyte apoptosis. Methods Wistar rats were treated with Ang II (200 ng·kg−1·min−1, 42 days) and or losartan (10 mg·kg−1·day−1, 14 days). Immortalized mouse podocyte were treated with 1 μM Ang II and/or losartan (1 μM) or SB203580 (0.1 μM) (AT1 receptor antagonist and p38 MAPK inhibitor) for 24 h. Kidney sections and cultured podocytes were used to evaluate protein expression by immunofluorescence and immunoblotting. Apoptosis was evaluated by flow cytometry and intracellular pH (pHi) was analyzed using microscopy combined with the fluorescent probe BCECF/AM. Results Compared with controls, Ang II via AT1R increased chaperone GRP 78/Bip protein expression in rat glomeruli (p < 0.001) as well as in podocyte culture (p < 0.01); increased phosphorylated eIf2-α (p < 0.05), PKC-delta (p < 0.01) and p38 MAPK (p < 0.001) protein expression. Furthermore, Ang II induced p38 MAPK-mediated late apoptosis and increased the Bax/Bcl-2 ratio (p < 0.001). Simultaneously, Ang II via AT1R induced p38 MAPK-NHE1-mediated increase of pHi recovery rate after acid loading. Conclusion Together, our results indicate that Ang II-induced podocyte apoptosis is associated with AT1R/ER stress/PKC-delta/p38 MAPK axis and enhanced NHE1-mediated pHi recovery rate.
Collapse
Affiliation(s)
- Vanessa Gerolde Cardoso
- Laboratory of Renal Physiology, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-900, Brazil
| | - Guilherme Lopes Gonçalves
- Laboratory of Renal Physiology, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-900, Brazil
| | - Juliana Martins Costa-Pessoa
- Laboratory of Renal Physiology, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-900, Brazil
| | - Karina Thieme
- Laboratory of Carbohydrates and Radioimmunoassays (LIM-18), Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Bruna Bezerra Lins
- Laboratory of Renal Physiology, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-900, Brazil
| | - Fernando Augusto Malavazzi Casare
- Laboratory of Renal Physiology, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-900, Brazil
| | - Mariana Charleaux de Ponte
- Laboratory of Renal Physiology, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-900, Brazil
| | - Niels Olsen Saraiva Camara
- Laboratory for Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Maria Oliveira-Souza
- Laboratory of Renal Physiology, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-900, Brazil.
| |
Collapse
|
20
|
Verma R, Venkatareddy M, Kalinowski A, Li T, Kukla J, Mollin A, Cara-Fuentes G, Patel SR, Garg P. Nephrin is necessary for podocyte recovery following injury in an adult mature glomerulus. PLoS One 2018; 13:e0198013. [PMID: 29924795 PMCID: PMC6010211 DOI: 10.1371/journal.pone.0198013] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 05/12/2018] [Indexed: 11/28/2022] Open
Abstract
Nephrin (Nphs1) is an adhesion protein that is expressed at the podocyte intercellular junction in the glomerulus. Nphs1 mutations in humans or deletion in animal genetic models results in a developmental failure of foot process formation. A number of studies have shown decrease in expression of nephrin in various proteinuric kidney diseases as well as in animal models of glomerular disease. Decrease in nephrin expression has been suggested to precede podocyte loss and linked to the progression of kidney disease. Whether the decrease in expression of nephrin is related to loss of podocytes or lead to podocyte detachment is unclear. To answer this central question we generated an inducible model of nephrin deletion (Nphs1Tam-Cre) in order to lower nephrin expression in healthy adult mice. Following tamoxifen-induction there was a 75% decrease in nephrin expression by 14 days. The Nphs1Tam-Cre mice had normal foot process ultrastructure and intact filtration barriers up to 4-6 weeks post-induction. Despite the loss of nephrin expression, the podocyte number and density remained unchanged during the initial period. Unexpectedly, nephrin expression, albeit at low levels persisted at the slit diaphragm up to 16-20 weeks post-tamoxifen induction. The mice became progressively proteinuric with glomerular hypertrophy and scarring reminiscent of focal and segmental glomerulosclerosis at 20 weeks. Four week-old Nphs1 knockout mice subjected to protamine sulfate model of podocyte injury demonstrated failure to recover from foot process effacement following heparin sulfate. Similarly, Nphs1 knockout mice failed to recover following nephrotoxic serum (NTS) with persistence of proteinuria and foot process effacement. Our results suggest that as in development, nephrin is necessary for maintenance of a healthy glomerular filter. In contrast to the developmental phenotype, lowering nephrin expression in a mature glomerulus resulted in a slowly progressive disease that histologically resembles FSGS a disease linked closely with podocyte depletion. Podocytes with low levels of nephrin expression are both susceptible and unable to recover following perturbation. Our results suggest that decreased nephrin expression independent of podocyte loss occurring as an early event in proteinuric kidney diseases might play a role in disease progression.
Collapse
Affiliation(s)
- Rakesh Verma
- Division of Nephrology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Madhusudan Venkatareddy
- Division of Nephrology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Anne Kalinowski
- Division of Nephrology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Theodore Li
- Division of Nephrology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Joanna Kukla
- Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | | | - Gabriel Cara-Fuentes
- Division of Pediatric Nephrology, Motts Children Hospital, Ann Arbor, Michigan, United States of America
| | - Sanjeevkumar R. Patel
- Division of Nephrology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
- Veterans Administration, VAMC, Ann Arbor, Michigan, United States of America
| | - Puneet Garg
- Division of Nephrology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
- Veterans Administration, VAMC, Ann Arbor, Michigan, United States of America
| |
Collapse
|
21
|
Abstract
Ultimately, the common final pathway of any glomerular disease is podocyte effacement, podocyte loss, and, eventually, glomerular scarring. There has been a long-standing debate on the underlying mechanisms for podocyte depletion, ranging from necrosis and apoptosis to detachment of viable cells from the glomerular basement membrane. However, this debate still continues because additional pathways of programmed cell death have been reported in recent years. Interestingly, viable podocytes can be isolated out of the urine of proteinuric patients easily, emphasizing the importance of podocyte detachment in glomerular diseases. In contrast, detection of apoptosis and other pathways of programmed cell death in podocytes is technically challenging. In fact, we still are lacking direct evidence showing, for example, the presence of apoptotic bodies in podocytes, leaving the question unanswered as to whether podocytes undergo mechanisms of programmed cell death. However, understanding the mechanisms leading to podocyte depletion is of particular interest because future therapeutic strategies might interfere with these to prevent glomerular scarring. In this review, we summarize our current knowledge on podocyte cell death, the different molecular pathways and experimental approaches to study these, and, finally, focus on the mechanisms that prevent the onset of programmed cell death.
Collapse
Affiliation(s)
- Fabian Braun
- Department II of Internal Medicine, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases, University of Cologne, Cologne, Germany
| | - Jan U Becker
- Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - Paul T Brinkkoetter
- Department II of Internal Medicine, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases, University of Cologne, Cologne, Germany.
| |
Collapse
|
22
|
Abu Hamad R, Berman S, Hachmo Y, Stark M, Hasan F, Doenyas-Barak K, Efrati S. Response of Renal Podocytes to Excessive Hydrostatic Pressure: a Pathophysiologic Cascade in a Malignant Hypertension Model. Kidney Blood Press Res 2017; 42:1104-1118. [PMID: 29224013 DOI: 10.1159/000485774] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 11/28/2017] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Renal injuries induced by increased intra-glomerular pressure coincide with podocyte detachment from the glomerular basement membrane (GBM). In previous studies, it was demonstrated that mesangial cells have a crucial role in the pathogenesis of malignant hypertension. However, the exact pathophysiological cascade responsible for podocyte detachment and its relationship with mesangial cells has not been fully elucidated yet and this was the aim of the current study. METHODS Rat renal mesangial or podocytes were exposed to high hydrostatic pressure in an in-vitro model of malignant hypertension. The resulted effects on podocyte detachment, apoptosis and expression of podocin and integrinβ1 in addition to Angiotensin-II and TGF-β1 generation were evaluated. To simulate the paracrine effect podocytes were placed in mesangial cell media pre-exposed to pressure, or in media enriched with Angiotensin-II, TGF-β1 or receptor blockers. RESULTS High pressure resulted in increased Angiotensin-II levels in mesangial and podocyte cells. Angiotensin-II via the AT1 receptors reduced podocin expression and integrinβ1, culminating in detachment of both viable and apoptotic podocytes. Mesangial cells exposed to pressure had a greater increase in Angiotensin-II than pressure-exposed podocytes. The massively increased concentration of Angiotensin-II by mesangial cells, together with increased TGF-β1 production, resulted in increased apoptosis and detachment of non-viable apoptotic podocytes. Unlike the direct effect of pressure on podocytes, the mesangial mediated effects were not related to changes in adhesion proteins expression. CONCLUSIONS Hypertension induces podocyte detachment by autocrine and paracrine effects. In a direct response to pressure, podocytes increase Angiotensin-II levels. This leads, via AT1 receptors, to structural changes in adhesion proteins, culminating in viable podocyte detachment. Paracrine effects of hypertension, mediated by mesangial cells, lead to higher levels of both Angiotensin-II and TGF-β1, culminating in apoptosis and detachment of non-viable podocytes.
Collapse
Affiliation(s)
| | - Sylvia Berman
- Research & Development Unit, Zerifin, Israel.,Nephrology Division, Assaf-Harofeh Medical Center, Zerifin, affiliated to Sackler Faculty of Medicine, Tel Aviv University, Zerifin, Israel
| | | | - Moshe Stark
- Research & Development Unit, Zerifin, Israel
| | - Fadia Hasan
- Research & Development Unit, Zerifin, Israel
| | - Keren Doenyas-Barak
- Nephrology Division, Assaf-Harofeh Medical Center, Zerifin, affiliated to Sackler Faculty of Medicine, Tel Aviv University, Zerifin, Israel
| | - Shai Efrati
- Research & Development Unit, Zerifin, Israel.,Nephrology Division, Assaf-Harofeh Medical Center, Zerifin, affiliated to Sackler Faculty of Medicine, Tel Aviv University, Zerifin, Israel
| |
Collapse
|
23
|
The Beneficial Effects of Allicin in Chronic Kidney Disease Are Comparable to Losartan. Int J Mol Sci 2017; 18:ijms18091980. [PMID: 28926934 PMCID: PMC5618629 DOI: 10.3390/ijms18091980] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 08/24/2017] [Accepted: 09/13/2017] [Indexed: 11/17/2022] Open
Abstract
Recent studies suggest that allicin may play a role in chronic kidney disease (CKD), reducing hypertension and oxidative stress and improving renal dysfunction. In the present study, CKD was induced by 5/6 nephrectomy and the animals were divided into four treatment groups as follows: control (C), CKD, CKD+allicin (40 mg/kg pathway oral) (CKDA), and CKD+Losartan (20 mg/kg) (CKDL). After CKD induction, the rats developed hypertension from week 3 to the end of the study. This was associated with increased creatinine and blood urea nitrogen (BUN) levels in serum, increased albuminuria, increased urinary excretion of N-acetyl-β-d-glucosaminidase (NAG), increased nephrin expression, and incrased histological alterations in the cortex. The levels of angiotensin receptors and endothelial nitric oxide synthase (eNOS) were decreased in the renal cortex from the CKD group. Otherwise, lipid and protein oxidation were higher in the CKD group than in the control group. A disturbance was observed in the expression levels of the nuclear factor erythroid 2-related factor 2/Kelch ECH associating protein 1 system (Nrf2/keap1) and the antioxidant enzymes catalase, superoxide dismutase, and heme oxygenase-1. Allicin or losartan treatments relieved renal dysfunction, hypertension, and oxidative stress. In addition, both treatments showed the same efficacy on the expression of angiotensin receptors, the nephrin, Nrf2/keap1 pathway, and eNOS. Further in silico analyses suggest that allicin and losartan could have a common mechanism involving interaction with AT1 receptors. Allicin showed antihypertensive, antioxidant, and nephroprotective effects. The beneficial effects showed by allicin are similar, or even better, than those of losartan. In fact, the effect of allicin on blood pressure and renal function is comparable to reductions seen with losartan, a prescription drug commonly used as a first-line therapy.
Collapse
|
24
|
Chen Q, Wang Q, Zhu J, Xiao Q, Zhang L. Reactive oxygen species: key regulators in vascular health and diseases. Br J Pharmacol 2017; 175:1279-1292. [PMID: 28430357 DOI: 10.1111/bph.13828] [Citation(s) in RCA: 198] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 03/31/2017] [Accepted: 04/11/2017] [Indexed: 12/18/2022] Open
Abstract
ROS are a group of small reactive molecules that play critical roles in the regulation of various cell functions and biological processes. In the vascular system, physiological levels of ROS are essential for normal vascular functions including endothelial homeostasis and smooth muscle cell contraction. In contrast, uncontrolled overproduction of ROS resulting from an imbalance of ROS generation and elimination leads to the development of vascular diseases. Excessive ROS cause vascular cell damage, the recruitment of inflammatory cells, lipid peroxidation, activation of metalloproteinases and deposition of extracellular matrix, collectively leading to vascular remodelling. Evidence from a large number of studies has revealed that ROS and oxidative stress are involved in the initiation and progression of numerous vascular diseases including hypertension, atherosclerosis, restenosis and abdominal aortic aneurysm. Furthermore, considerable research has been implemented to explore antioxidants that can reduce ROS production and oxidative stress in order to ameliorate vascular diseases. In this review, we will discuss the nature and sources of ROS, their roles in vascular homeostasis and specific vascular diseases and various antioxidants as well as some of the pharmacological agents that are capable of reducing ROS and oxidative stress. The aim of this review is to provide information for developing promising clinical strategies targeting ROS to decrease cardiovascular risks. LINKED ARTICLES This article is part of a themed section on Spotlight on Small Molecules in Cardiovascular Diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.8/issuetoc.
Collapse
Affiliation(s)
- Qishan Chen
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qiwen Wang
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianhua Zhu
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qingzhong Xiao
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Li Zhang
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
25
|
Angiotensin II induces calcium/calcineurin signaling and podocyte injury by downregulating microRNA-30 family members. J Mol Med (Berl) 2017; 95:887-898. [DOI: 10.1007/s00109-017-1547-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 05/04/2017] [Accepted: 05/12/2017] [Indexed: 01/13/2023]
|
26
|
Wang L, Sha Y, Bai J, Eisner W, Sparks MA, Buckley AF, Spurney RF. Podocyte-specific knockout of cyclooxygenase 2 exacerbates diabetic kidney disease. Am J Physiol Renal Physiol 2017; 313:F430-F439. [PMID: 28490532 DOI: 10.1152/ajprenal.00614.2016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 05/02/2017] [Accepted: 05/09/2017] [Indexed: 01/11/2023] Open
Abstract
Enhanced expression of cyclooxygenase 2 (COX2) in podocytes contributes to glomerular injury in diabetic kidney disease, but some basal level of podocyte COX2 expression might be required to promote podocyte attachment and/or survival. To investigate the role of podocyte COX2 expression in diabetic kidney disease, we deleted COX2 specifically in podocytes in a mouse model of Type 1 diabetes mellitus (Akita mice). Podocyte-specific knockout (KO) of COX2 did not affect renal morphology or albuminuria in nondiabetic mice. Albuminuria was significantly increased in wild-type (WT) and KO Akita mice compared with nondiabetic controls, and the increase in albuminuria was significantly greater in KO Akita mice compared with WT Akita mice at both 16 and 20 wk of age. At the 20-wk time point, mesangial expansion was also increased in WT and KO Akita mice compared with nondiabetic animals, and these histologic abnormalities were not improved by KO of COX2. Tubular injury was seen only in diabetic mice, but there were no significant differences between groups. Thus, KO of COX2 enhanced albuminuria and did not improve the histopathologic features of diabetic kidney disease. These data suggest that 1) KO of COX2 in podocytes does not ameliorate diabetic kidney disease in Akita mice, and 2) some basal level of podocyte COX2 expression in podocytes is necessary to attenuate the adverse effects of diabetes on glomerular filtration barrier function.
Collapse
Affiliation(s)
- Liming Wang
- Division of Nephrology, Department of Medicine, Duke University and Durham Veterans Affairs Medical Centers, Durham, North Carolina
| | - Yonggang Sha
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | | | - William Eisner
- Division of Hematological Malignancies, Department of Medicine, Duke University Medical Center, Durham, North Carolina; and
| | - Matthew A Sparks
- Division of Nephrology, Department of Medicine, Duke University and Durham Veterans Affairs Medical Centers, Durham, North Carolina
| | - Anne F Buckley
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Robert F Spurney
- Division of Nephrology, Department of Medicine, Duke University and Durham Veterans Affairs Medical Centers, Durham, North Carolina;
| |
Collapse
|
27
|
Rimpelä JM, Kontula KK, Fyhrquist F, Donner KM, Tuiskula AM, Sarin AP, Mohney RP, Stirdivant SM, Hiltunen TP. Replicated evidence for aminoacylase 3 and nephrin gene variations to predict antihypertensive drug responses. Pharmacogenomics 2017; 18:445-458. [DOI: 10.2217/pgs-2016-0204] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Aim: To replicate the genome-wide associations of the antihypertensive effects of bisoprolol and losartan in GENRES, using the Finnish patients of LIFE study. Patients & methods: We analyzed association of four SNPs with atenolol and three SNPs with losartan response in 927 Finnish LIFE patients (467 for atenolol and 460 for losartan). Results: rs2514036, a variation at a transcription start site of ACY3, was associated with blood pressure response to atenolol in men in LIFE. Response to bisoprolol was correlated to baseline plasma levels of N-acetylphenylalanine and phenylalanine (ACY3 substrate and end product, respectively) in GENRES study. NPHS1 variation rs3814995 was associated with losartan effect in LIFE. Conclusion: We provide support for two pharmacogenomic markers for beta-blockers and angiotensin receptor antagonists.
Collapse
Affiliation(s)
- Jenni M Rimpelä
- Department of Medicine, University of Helsinki, Finland
- Helsinki University Hospital, Helsinki, Finland
| | - Kimmo K Kontula
- Department of Medicine, University of Helsinki, Finland
- Helsinki University Hospital, Helsinki, Finland
| | - Frej Fyhrquist
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Kati M Donner
- Institute for Molecular Medicine Finland, University of Helsinki, Finland
| | | | - Antti-Pekka Sarin
- Institute for Molecular Medicine Finland, University of Helsinki, Finland
| | | | | | - Timo P Hiltunen
- Department of Medicine, University of Helsinki, Finland
- Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
28
|
Velez JCQ, Arif E, Rodgers J, Hicks MP, Arthur JM, Nihalani D, Bruner ET, Budisavljevic MN, Atkinson C, Fitzgibbon WR, Janech MG. Deficiency of the Angiotensinase Aminopeptidase A Increases Susceptibility to Glomerular Injury. J Am Soc Nephrol 2017; 28:2119-2132. [PMID: 28202497 DOI: 10.1681/asn.2016111166] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 01/04/2017] [Indexed: 01/13/2023] Open
Abstract
Aminopeptidase A (APA) is expressed in glomerular podocytes and tubular epithelia and metabolizes angiotensin II (AngII), a peptide known to promote glomerulosclerosis. In this study, we tested whether APA expression changes in response to progressive nephron loss or whether APA exerts a protective role against glomerular damage and during AngII-mediated hypertensive kidney injury. At advanced stages of FSGS, fawn-hooded hypertensive rat kidneys exhibited distinctly increased APA staining in areas of intact glomerular capillary loops. Moreover, BALB/c APA-knockout (KO) mice injected with a nephrotoxic serum showed persistent glomerular hyalinosis and albuminuria 96 hours after injection, whereas wild-type controls achieved virtually full recovery. We then tested the effect of 4-week infusion of AngII (400 ng/kg per minute) in APA-KO and wild-type mice. Although we observed no significant difference in achieved systolic BP, AngII-treated APA-KO mice developed a significant rise in albuminuria not observed in AngII-treated wild-type mice along with increased segmental and global sclerosis and/or collapse of juxtamedullary glomeruli, microcystic tubular dilation, and tubulointerstitial fibrosis. In parallel, AngII treatment significantly increased the kidney AngII content and attenuated the expression of podocyte nephrin in APA-KO mice but not in wild-type controls. These data show that deficiency of APA increases susceptibility to glomerular injury in BALB/c mice. The augmented AngII-mediated kidney injury observed in association with increased intrarenal AngII accumulation in the absence of APA suggests a protective metabolizing role of APA in AngII-mediated glomerular diseases.
Collapse
Affiliation(s)
- Juan Carlos Q Velez
- Department of Nephrology, Ochsner Clinic Foundation, New Orleans, Louisiana;
| | | | | | - Megan P Hicks
- Institute of Public and Preventative Health, Augusta University, Augusta, Georgia; and
| | - John M Arthur
- Division of Nephrology, Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | | | | | | | - Carl Atkinson
- Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | | | | |
Collapse
|
29
|
Abstract
It has become clear that reactive oxygen species (ROS) contribute to the development of hypertension via myriad effects. ROS are essential for normal cell function; however, they mediate pathologic changes in the brain, the kidney, and blood vessels that contribute to the genesis of chronic hypertension. There is also emerging evidence that ROS contribute to immune activation in hypertension. This article discusses these events and how they coordinate to contribute to hypertension and its consequent end-organ damage.
Collapse
Affiliation(s)
- Roxana Loperena
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 2220 Pierce Drive, Room 536 Robinson Research Building, Nashville, TN 37232, USA
| | - David G Harrison
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Vanderbilt University, 2220 Pierce Drive, Room 536 Robinson Research Building, Nashville, TN 37232, USA.
| |
Collapse
|
30
|
CD2-associated protein/phosphoinositide 3-kinase signaling has a preventive role in angiotensin II-induced podocyte apoptosis. Int J Biochem Cell Biol 2016; 79:370-381. [DOI: 10.1016/j.biocel.2016.08.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 08/19/2016] [Accepted: 08/29/2016] [Indexed: 01/13/2023]
|
31
|
Abstract
SIGNIFICANCE A common link between all forms of acute and chronic kidney injuries, regardless of species, is enhanced generation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) during injury/disease progression. While low levels of ROS and RNS are required for prosurvival signaling, cell proliferation and growth, and vasoreactivity regulation, an imbalance of ROS and RNS generation and elimination leads to inflammation, cell death, tissue damage, and disease/injury progression. RECENT ADVANCES Many aspects of renal oxidative stress still require investigation, including clarification of the mechanisms which prompt ROS/RNS generation and subsequent renal damage. However, we currently have a basic understanding of the major features of oxidative stress pathology and its link to kidney injury/disease, which this review summarizes. CRITICAL ISSUES The review summarizes the critical sources of oxidative stress in the kidney during injury/disease, including generation of ROS and RNS from mitochondria, NADPH oxidase, and inducible nitric oxide synthase. The review next summarizes the renal antioxidant systems that protect against oxidative stress, including superoxide dismutase and catalase, the glutathione and thioredoxin systems, and others. Next, we describe how oxidative stress affects kidney function and promotes damage in every nephron segment, including the renal vessels, glomeruli, and tubules. FUTURE DIRECTIONS Despite the limited success associated with the application of antioxidants for treatment of kidney injury/disease thus far, preventing the generation and accumulation of ROS and RNS provides an ideal target for potential therapeutic treatments. The review discusses the shortcomings of antioxidant treatments previously used and the potential promise of new ones. Antioxid. Redox Signal. 25, 119-146.
Collapse
Affiliation(s)
- Brian B Ratliff
- 1 Department of Medicine, Renal Research Institute , New York Medical College, Valhalla, New York.,2 Department of Physiology, Renal Research Institute , New York Medical College, Valhalla, New York
| | - Wasan Abdulmahdi
- 2 Department of Physiology, Renal Research Institute , New York Medical College, Valhalla, New York
| | - Rahul Pawar
- 1 Department of Medicine, Renal Research Institute , New York Medical College, Valhalla, New York
| | - Michael S Wolin
- 2 Department of Physiology, Renal Research Institute , New York Medical College, Valhalla, New York
| |
Collapse
|
32
|
Ha TS, Park HY, Seong SB, Ahn HY. Angiotensin II Modulates p130Cas of Podocytes by the Suppression of AMP-Activated Protein Kinase. J Korean Med Sci 2016; 31:535-41. [PMID: 27051236 PMCID: PMC4810335 DOI: 10.3346/jkms.2016.31.4.535] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 01/07/2016] [Indexed: 01/13/2023] Open
Abstract
Angiotensin II (Ang II) induces the pathological process of vascular structures, including renal glomeruli by hemodynamic and nonhemodynamic direct effects. In kidneys, Ang II plays an important role in the development of proteinuria by the modification of podocyte molecules. We have previously found that Ang II suppressed podocyte AMP-activated protein kinase (AMPK) via Ang II type 1 receptor and MAPK signaling pathway. In the present study, we investigated the roles of AMPK on the changes of p130Cas of podocyte by Ang II. We cultured mouse podocytes and treated them with various concentrations of Ang II and AMPK-modulating agents and analyzed the changes of p130Cas by confocal imaging and western blotting. In immunofluorescence study, Ang II decreased the intensity of p130Cas and changed its localization from peripheral cytoplasm into peri-nuclear areas in a concentrated pattern in podocytes. Ang II also reduced the amount of p130Cas in time and dose-sensitive manners. AMPK activators, metformin and AICAR, restored the suppressed and mal-localized p130Cas significantly, whereas, compound C, an AMPK inhibitor, further aggravated the changes of p130Cas. Losartan, an Ang II type 1 receptor antagonist, recovered the abnormal changes of p130Cas suppressed by Ang II. These results suggest that Ang II induces the relocalization and suppression of podocyte p130Cas by the suppression of AMPK via Ang II type 1 receptor, which would contribute to Ang II-induced podocyte injury.
Collapse
Affiliation(s)
- Tae-Sun Ha
- Department of Pediatrics, College of Medicine, Chungbuk National University, Cheongju, Korea
| | - Hye-Young Park
- Department of Pediatrics, College of Medicine, Chungbuk National University, Cheongju, Korea
| | - Su-Bin Seong
- Department of Pediatrics, College of Medicine, Chungbuk National University, Cheongju, Korea
| | - Hee-Yul Ahn
- Department of Pharmacology, College of Medicine, Chungbuk National University, Cheongju, Korea
| |
Collapse
|
33
|
Cyclin-dependent kinase 2 protects podocytes from apoptosis. Sci Rep 2016; 6:21664. [PMID: 26876672 PMCID: PMC4753499 DOI: 10.1038/srep21664] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 01/14/2016] [Indexed: 12/12/2022] Open
Abstract
Loss of podocytes is an early feature of diabetic nephropathy (DN) and predicts its progression. We found that treatment of podocytes with sera from normoalbuminuric type 1 diabetes patients with high lipopolysaccharide (LPS) activity, known to predict progression of DN, downregulated CDK2 (cyclin-dependent kinase 2). LPS-treatment of mice also reduced CDK2 expression. LPS-induced downregulation of CDK2 was prevented in vitro and in vivo by inhibiting the Toll-like receptor (TLR) pathway using immunomodulatory agent GIT27. We also observed that CDK2 is downregulated in the glomeruli of obese Zucker rats before the onset of proteinuria. Knockdown of CDK2, or inhibiting its activity with roscovitine in podocytes increased apoptosis. CDK2 knockdown also reduced expression of PDK1, an activator of the cell survival kinase Akt, and reduced Akt phosphorylation. This suggests that CDK2 regulates the activity of the cell survival pathway via PDK1. Furthermore, PDK1 knockdown reduced the expression of CDK2 suggesting a regulatory loop between CDK2 and PDK1. Collectively, our data show that CDK2 protects podocytes from apoptosis and that reduced expression of CDK2 associates with the development of DN. Preventing downregulation of CDK2 by blocking the TLR pathway with GIT27 may provide a means to prevent podocyte apoptosis and progression of DN.
Collapse
|
34
|
Abstract
Endothelin-1 (ET-1) is a 21-amino acid peptide with mitogenic and powerful vasoconstricting properties. Under healthy conditions, ET-1 is expressed constitutively in all cells of the glomerulus and participates in homeostasis of glomerular structure and filtration function. Under disease conditions, increases in ET-1 are critically involved in initiating and maintaining glomerular inflammation, glomerular basement membrane hypertrophy, and injury of podocytes (visceral epithelial cells), thereby promoting proteinuria and glomerulosclerosis. Here, we review the role of ET-1 in the function of glomerular endothelial cells, visceral (podocytes) and parietal epithelial cells, mesangial cells, the glomerular basement membrane, stromal cells, inflammatory cells, and mesenchymal stem cells. We also discuss molecular mechanisms by which ET-1, predominantly through activation of the ETA receptor, contributes to injury to glomerular cells, and review preclinical and clinical evidence supporting its pathogenic role in glomerular injury in chronic renal disease. Finally, the therapeutic rationale for endothelin antagonists as a new class of antiproteinuric drugs is discussed.
Collapse
Affiliation(s)
- Matthias Barton
- Molecular Internal Medicine, University of Zurich, Zurich, Switzerland.
| | - Andrey Sorokin
- Department of Medicine, Kidney Disease Center, Division of Nephrology, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
35
|
Abstract
Podocytes are highly specialized cells of the kidney glomerulus that wrap around capillaries and that neighbor cells of the Bowman’s capsule. When it comes to glomerular filtration, podocytes play an active role in preventing plasma proteins from entering the urinary ultrafiltrate by providing a barrier comprising filtration slits between foot processes, which in aggregate represent a dynamic network of cellular extensions. Foot processes interdigitate with foot processes from adjacent podocytes and form a network of narrow and rather uniform gaps. The fenestrated endothelial cells retain blood cells but permit passage of small solutes and an overlying basement membrane less permeable to macromolecules, in particular to albumin. The cytoskeletal dynamics and structural plasticity of podocytes as well as the signaling between each of these distinct layers are essential for an efficient glomerular filtration and thus for proper renal function. The genetic or acquired impairment of podocytes may lead to foot process effacement (podocyte fusion or retraction), a morphological hallmark of proteinuric renal diseases. Here, we briefly discuss aspects of a contemporary view of podocytes in glomerular filtration, the patterns of structural changes in podocytes associated with common glomerular diseases, and the current state of basic and clinical research.
Collapse
Affiliation(s)
- Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Mehmet M Altintas
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
36
|
Leung JCK, Chan LYY, Saleem MA, Mathieson PW, Tang SCW, Lai KN. Combined blockade of angiotensin II and prorenin receptors ameliorates podocytic apoptosis induced by IgA-activated mesangial cells. Apoptosis 2016; 20:907-20. [PMID: 25808596 PMCID: PMC7101871 DOI: 10.1007/s10495-015-1117-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Glomerulo-podocytic communication plays an important role in the podocytic injury in IgA nephropathy (IgAN). In this study, we examine the role of podocytic angiotensin II receptor subtype 1 (AT1R) and prorenin receptor (PRR) in podocytic apoptosis in IgAN. Polymeric IgA (pIgA) was isolated from patients with IgAN and healthy controls. Conditioned media were prepared from growth arrested human mesangial cells (HMC) incubated with pIgA from patients with IgAN (IgA-HMC media) or healthy controls (Ctl-HMC media). A human podocyte cell line was used as a model to examine the regulation of the expression of AT1R, PRR, TNF-α and CTGF by IgA-HMC media. Podocytic nephrin expression, annexin V binding and caspase 3 activity were used as the functional readout of podocytic apoptosis. IgA-HMC media had no effect on AngII release by podocytes. IgA-HMC media significantly up-regulated the expression of AT1R and PRR, down-regulated nephrin expression and induced apoptosis in podocytes. Mono-blockade of AT1R, PRR, TNF-α or CTGF partially reduced podocytic apoptosis. IgA-HMC media activated NFκB, notch1 and HEY1 expression by podocytes and dual blockade of AT1R with PRR, or anti-TNF-α with anti-CTGF, effectively rescued the podocytic apoptosis induced by IgA-HMC media. Our data suggests that pIgA-activated HMC up-regulates the expression of AT1R and PRR expression by podocytes and the associated activation of NFκB and notch signalling pathways play an essential role in the podocytic apoptosis induced by glomerulo-podocytic communication in IgAN. Simultaneously targeting the AT1R and PRR could be a potential therapeutic option to reduce the podocytic injury in IgAN.
Collapse
Affiliation(s)
- Joseph C K Leung
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Room 301, Professorial Block, 102 Pokfulam Road, Pokfulam, Hong Kong, China,
| | | | | | | | | | | |
Collapse
|
37
|
Wen Y, Wang Z, Liu L, Zhang Y, Zhou P, Liang J. Cabin1 localizes in glomerular podocyte and undergoes nuclear translocation during podocyte injury. Ren Fail 2015; 37:1344-8. [DOI: 10.3109/0886022x.2015.1073527] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|
38
|
Sun YH, Yang XL, Li F, Song LJ, Li J. Effect of anti- Helicobacter pylori treatment on early diabetic kidney disease. Shijie Huaren Xiaohua Zazhi 2015; 23:1202-1207. [DOI: 10.11569/wcjd.v23.i7.1202] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To assess the effect of anti-Helicobacter pylori (H. pylori) therapy on chronic kidney disease (CKD) associated with type 2 diabetes mellitus (T2DM).
METHODS: Seventy-five T2DM patients with newly diagnosed CKD and H. pylori infection were randomly divided into an anti-H. pylori therapy group (n = 39) and a control group (n = 36). All of the patients received routine treatment for three months. Patients in the anti-H. pylori group were additionally given anti-H. pylori therapy. Clinical indices including fasting blood glucose (FBG), 2-h plasma glucose (2-h PG), hemoglobin A1c (HbA1c), systolic blood pressure (SBP), diastolic blood pressure (DBP), cholesterol (TC), triglyceride (TG), urinary albumin/creatitine ratio (UAlb/Cr), C-reactive protein (CRP), tumor necrosis factor α (TNF-α), plasma endothelin 1 (ET-1), and homocysteine (HCY) were recorded before and three months after treatment.
RESULTS: No significant differences were observed in all clinical indices between the two groups before treatment (P > 0.05). The eradication rate of H. pylori in the anti-H. pylori group was significantly higher than that in the control group three months after treatment (P < 0.01). There were no differences in FBG, 2-h PG, SBP, DBP, HbA1c, TG, or TC between before and after treatment (P > 0.05). At three months after treatment, UAlb/Cr, CRP, ET-1, TNF-α and HCY decreased significantly in both groups (P < 0.05), and changes were statistically significant different between the two groups (P < 0.05).
CONCLUSION: Anti-H. pylori therapy is beneficial for T2DM patients with CKD, because it can help control UAlb/Cr, CRP, ET-1, TNF-α, and HCY and even play an important role in postponing CKD.
Collapse
|
39
|
Hiltunen TP, Donner KM, Sarin A, Saarela J, Ripatti S, Chapman AB, Gums JG, Gong Y, Cooper‐DeHoff RM, Frau F, Glorioso V, Zaninello R, Salvi E, Glorioso N, Boerwinkle E, Turner ST, Johnson JA, Kontula KK. Pharmacogenomics of hypertension: a genome‐wide, placebo‐controlled cross‐over study, using four classes of antihypertensive drugs. J Am Heart Assoc 2015. [PMID: 25622599 PMCID: PMC4330076 DOI: 10.1161/jaha.114.001521] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Identification of genetic markers of antihypertensive drug responses could assist in individualization of hypertension treatment. METHODS AND RESULTS We conducted a genome-wide association study to identify gene loci influencing the responsiveness of 228 male patients to 4 classes of antihypertensive drugs. The Genetics of Drug Responsiveness in Essential Hypertension (GENRES) study is a double-blind, placebo-controlled cross-over study where each subject received amlodipine, bisoprolol,hydrochlorothiazide, and losartan, each as a monotherapy, in a randomized order. Replication analyses were performed in 4 studies with patients of European ancestry (PEAR Study, N=386; GERA I and II Studies, N=196 and N=198; SOPHIA Study, N=372). We identified 3 single-nucleotide polymorphisms within the ACY3 gene that showed associations with bisoprolol response reaching genome-wide significance (P<5x10(-8))however, this could not be replicated in the PEAR Study using atenolol. In addition, 39 single-nucleotide polymorphisms showed P values of 10(-5) to 10(-7). The 20 top-associated single-nucleotide polymorphisms were different for each antihypertensive drug. None of these top single-nucleotide polymorphisms co-localized with the panel of >40 genes identified in genome-wide association studies of hypertension. Replication analyses of GENRES results provided suggestive evidence for a missense variant (rs3814995) in the NPHS1 (nephrin) gene influencing losartan response, and for 2 variants influencing hydrochlorothiazide response, located within or close to the ALDH1A3 (rs3825926) and CLIC5 (rs321329) genes. CONCLUSIONS These data provide some evidence for a link between biology of the glomerular protein nephrin and antihypertensive action of angiotensin receptor antagonists and encourage additional studies on aldehyde dehydrogenase–mediated reactions in antihypertensive drug action.
Collapse
Affiliation(s)
- Timo P. Hiltunen
- Department of Medicine, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland (T.P.H., K.K.K.)
| | - Kati M. Donner
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland (K.M.D., A.P.S., J.S., S.R.)
| | - Antti‐Pekka Sarin
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland (K.M.D., A.P.S., J.S., S.R.)
- Public Health Genomics Unit, National Institute for Health and Welfare, University of Helsinki, Helsinki, Finland (A.P.S.)
| | - Janna Saarela
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland (K.M.D., A.P.S., J.S., S.R.)
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland (K.M.D., A.P.S., J.S., S.R.)
- Department of Public Health, Hjelt Institute, University of Helsinki, Helsinki, Finland (S.R.)
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom (S.R.)
| | - Arlene B. Chapman
- Mayo Clinic, Rochester, MN (A.B.C.)
- Department of Medicine, Renal Division, Emory University School of Medicine, Atlanta, GA (A.B.C.)
| | - John G. Gums
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, University of Florida, Gainesville, FL (J.G.G., Y.G., R.M.C.D.H., J.A.J.)
- Department of Community Health and Family Medicine, Gainesville, FL (J.G.G.)
| | - Yan Gong
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, University of Florida, Gainesville, FL (J.G.G., Y.G., R.M.C.D.H., J.A.J.)
| | - Rhonda M. Cooper‐DeHoff
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, University of Florida, Gainesville, FL (J.G.G., Y.G., R.M.C.D.H., J.A.J.)
- Department of Medicine, Gainesville, FL (R.M.C.D.H.)
| | - Francesca Frau
- Department of Health Sciences, Genomics and Bioinformatics Unit, University of Milan and Filarete Foundation, Milan, Italy (F.F., E.S.)
| | - Valeria Glorioso
- Hypertension and Related Disease Centre, AOU‐University of Sassari, Sassari, Italy (V.G., R.Z., N.G.)
| | - Roberta Zaninello
- Hypertension and Related Disease Centre, AOU‐University of Sassari, Sassari, Italy (V.G., R.Z., N.G.)
| | - Erika Salvi
- Department of Health Sciences, Genomics and Bioinformatics Unit, University of Milan and Filarete Foundation, Milan, Italy (F.F., E.S.)
| | - Nicola Glorioso
- Hypertension and Related Disease Centre, AOU‐University of Sassari, Sassari, Italy (V.G., R.Z., N.G.)
| | - Eric Boerwinkle
- University of Florida, Gainesville, FL (E.B.)
- Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX (E.B.)
| | - Stephen T. Turner
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, MN (S.T.T.)
| | - Julie A. Johnson
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, University of Florida, Gainesville, FL (J.G.G., Y.G., R.M.C.D.H., J.A.J.)
| | - Kimmo K. Kontula
- Department of Medicine, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland (T.P.H., K.K.K.)
| |
Collapse
|
40
|
Keaney JF, Loscalzo J. Pharmacogenomics of hypertension: a genome‐wide, placebo‐controlled cross‐over study, using four classes of antihypertensive drugs. J Am Heart Assoc 2015; 4:e001778. [PMID: 25628411 PMCID: PMC4330084 DOI: 10.1161/jaha.115.001778] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Identification of genetic markers of antihypertensive drug responses could assist in individualization of hypertension treatment. METHODS AND RESULTS We conducted a genome-wide association study to identify gene loci influencing the responsiveness of 228 male patients to 4 classes of antihypertensive drugs. The Genetics of Drug Responsiveness in Essential Hypertension (GENRES) study is a double-blind, placebo-controlled cross-over study where each subject received amlodipine, bisoprolol,hydrochlorothiazide, and losartan, each as a monotherapy, in a randomized order. Replication analyses were performed in 4 studies with patients of European ancestry (PEAR Study, N=386; GERA I and II Studies, N=196 and N=198; SOPHIA Study, N=372). We identified 3 single-nucleotide polymorphisms within the ACY3 gene that showed associations with bisoprolol response reaching genome-wide significance (P<5x10(-8))however, this could not be replicated in the PEAR Study using atenolol. In addition, 39 single-nucleotide polymorphisms showed P values of 10(-5) to 10(-7). The 20 top-associated single-nucleotide polymorphisms were different for each antihypertensive drug. None of these top single-nucleotide polymorphisms co-localized with the panel of >40 genes identified in genome-wide association studies of hypertension. Replication analyses of GENRES results provided suggestive evidence for a missense variant (rs3814995) in the NPHS1 (nephrin) gene influencing losartan response, and for 2 variants influencing hydrochlorothiazide response, located within or close to the ALDH1A3 (rs3825926) and CLIC5 (rs321329) genes. CONCLUSIONS These data provide some evidence for a link between biology of the glomerular protein nephrin and antihypertensive action of angiotensin receptor antagonists and encourage additional studies on aldehyde dehydrogenase–mediated reactions in antihypertensive drug action.
Collapse
Affiliation(s)
- John F. Keaney
- UMass Medical School and UMass Memorial Medical Center, Worcester, MA (J.F.K.)
| | - Joseph Loscalzo
- Harvard Medical School and Brigham and Women's Hospital, Boston, MA (J.L.)
| |
Collapse
|
41
|
Shchukina AA, Bobkova IN, Shestakova MV, Vikulova OK, Zuraeva ZT, Mikhaleva OV. [Urinary excretion of markers for podocyte injury in patients with diabetes mellitus]. TERAPEVT ARKH 2015; 87:62-66. [PMID: 26978176 DOI: 10.17116/terarkh2015871062-66] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
AIM To estimate the urinary excretion of markers for podocyte injury, to specify their value for the early diagnosis of diabetic nephropathy (DN), and to access the risk of its progression in patients with diabetes mellitus (DM) with varying degrees of albuminuria/proteinuria. SUBJECT AND METHODS Seventy-four diabetic patients (30 with type 1 DM and 44 with type 2 DM) were examined and divided into 3 groups according to the urinary concentration in one urinary portion: 1) 41 patients with normal albuminuria (NAU) (<20 mg/l); 2) 13 patients with microalbuminuria (MAU) (20-200 mg/l); 3) 20 patients with proteinuria (PU) (>200 mg/l). A control group included 10 healthy individuals. The urinary levels of the podocyte structural proteins nephrin and podocin were determined by enzyme immunoassay. RESULTS Nephrinuria (NU) was detected in 63, 77, and 80% of the patients with NAU, MAU, and PU, respectively. Podocinuria (PDU) was found in 78, 54, and 83% of those with NAU, MAU, and PU, respectively. NU in DN with PU was significantly higher than that in DM with NAU. In the NAU, MAU, and PU subgroups, podocin excretion was equally higher and did not differ between the types of DM. There was a direct correlation of NU with albuminuria, which was stronger in the MAU subgroup. In the patients with DM with varying degrees of albuminuria, the values of NU and PDU correlated directly to serum creatinine levels and inversely with glomerular filtration rate. NU directly correlated with glycated hemoglobin levels in the patients with types 1 and 2 DM of less than 5 years' duration and a direct significant correlation of systolic blood pressure with NU was found in those with type 2 DM. CONCLUSION Determination of urinary nephrin and podocin levels may be used for the early preclinical diagnosis of DN and the monitoring of the glomerular apparatus in DM.
Collapse
Affiliation(s)
- A A Shchukina
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia, Moscow, Russia
| | - I N Bobkova
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia, Moscow, Russia
| | - M V Shestakova
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia, Moscow, Russia; Institute of Diabetes, Endocrinology Research Center, Ministry of Health of Russia, Moscow, Russia
| | - O K Vikulova
- Institute of Diabetes, Endocrinology Research Center, Ministry of Health of Russia, Moscow, Russia
| | - Z T Zuraeva
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia, Moscow, Russia
| | - O V Mikhaleva
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia, Moscow, Russia
| |
Collapse
|
42
|
Pourghasem M, Shafi H, Babazadeh Z. Histological changes of kidney in diabetic nephropathy. CASPIAN JOURNAL OF INTERNAL MEDICINE 2015; 6:120-7. [PMID: 26644877 PMCID: PMC4650785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 05/15/2015] [Accepted: 05/20/2015] [Indexed: 11/23/2022]
Abstract
Diabetes mellitus is the most common cause of chronic renal disorders and end-stage kidney disease in developed countries. It is the major cause of dialysis and transplantation. Failure in renal function causes wide disorders in the body. Diabetes results in wide range of alterations in the renal tissue. It is believed that early histological changes in diabetic nephropathy are detectable 2 years after diabetes is diagnosed. The glomerular alterations are the most important lesions in the diabetic nephropathy (DN). The Renal Pathology Society provides a new pathological classification for the detection of histopathology of DN. It divides diabetic nephropathy into four hierarchical glomerular lesions. Alloxan or streptozotocin induced diabetic rat is the one most widely used specie to study DN. Histological changes in the rat DN closely resemble the human disease and the most information of this review was obtained through the study of rat DN. All cell types of the kidney such as mesangial cells, podocytes and tubulointerstitial cells are liable to be affected in the event of DN. Severity of renal lesions is associated to the clinical aspect of renal outcome, but the aim of this article was only to review the histological changes of kidney in diabetes mellitus.
Collapse
Affiliation(s)
- Mohsen Pourghasem
- Cellular and Molecular Biology Research Center (CMBRC), Babol University of Medical Sciences, Babol, Iran
| | - Hamid Shafi
- Fatemeh Zahra Infertility and Reproductive Health Research Center, Babol University of Medical Sciences, Babol, Iran
| | - Zahra Babazadeh
- Cellular and Molecular Biology Research Center (CMBRC), Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
43
|
Márquez E, Riera M, Pascual J, Soler MJ. Renin-angiotensin system within the diabetic podocyte. Am J Physiol Renal Physiol 2014; 308:F1-10. [PMID: 25339703 DOI: 10.1152/ajprenal.00531.2013] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Diabetic kidney disease is the leading cause of end-stage renal disease. Podocytes are differentiated cells necessary for the development and maintenance of the glomerular basement membrane and the capillary tufts, as well as the function of the glomerular filtration barrier. The epithelial glomerular cells express a local renin-angiotensin system (RAS) that varies in different pathological situations such as hyperglycemia or mechanical stress. RAS components have been shown to be altered in diabetic podocytopathy, and their modulation may modify diabetic nephropathy progression. Podocytes are a direct target for angiotensin II-mediated injury by altered expression and distribution of podocyte proteins. Furthermore, angiotensin II promotes podocyte injury indirectly by inducing cellular hypertrophy, increased apoptosis, and changes in the anionic charge of the glomerular basement membrane, among other effects. RAS blockade has been shown to decrease the level of proteinuria and delay the progression of chronic kidney disease. This review summarizes the local intraglomerular RAS and its imbalance in diabetic podocytopathy. A better understanding of the intrapodocyte RAS might provide a new approach for diabetic kidney disease treatment.
Collapse
Affiliation(s)
- Eva Márquez
- Department of Nephrology, Hospital del Mar, Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; and
| | - Marta Riera
- Department of Nephrology, Hospital del Mar, Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; and Red de Investigación Renal (REDINREN), Instituto Carlos III, Madrid, Spain
| | - Julio Pascual
- Department of Nephrology, Hospital del Mar, Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; and Red de Investigación Renal (REDINREN), Instituto Carlos III, Madrid, Spain
| | - María José Soler
- Department of Nephrology, Hospital del Mar, Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; and Red de Investigación Renal (REDINREN), Instituto Carlos III, Madrid, Spain
| |
Collapse
|
44
|
Gao N, Wang H, Zhang X, Yang Z. The inhibitory effect of angiotensin II on BKCa channels in podocytes via oxidative stress. Mol Cell Biochem 2014; 398:217-22. [PMID: 25234195 DOI: 10.1007/s11010-014-2221-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 09/13/2014] [Indexed: 01/11/2023]
Abstract
Angiotensin II (Ang II) is an important active substance of the renin-angiotensin system (RAS). The present study has confirmed that abnormalities of Ang II may be related with cerebrovascular diseases, endocrine diseases, cardiovascular diseases, liver diseases, such as: cerebral hypoxia, diabetes, obesity, atrial fibrillation, and liver cirrhosis. However, understanding effects of Ang II on podocytes is not enough. This study was to investigate the effects of oxidative stress on the large conductance, Ca(2+)-activated K(+) channels (BKCa). Results from the 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay showed that Ang II induced podocyte death in a concentration-dependent manner. The measurement of superoxide dismutase (SOD) generation demonstrated that Ang II decreased the total SOD of cellular levels. Meaningfully, pretreatment of a type of ROS scavenger formulations named N-(mercaptopropionyl)-glycine (N-MPG) could inhibit podocyte apoptosis induced by Ang II. Meanwhile, patch-clamp technique was used in this study to detect the effects of Ang II on currents of BKCa channel in podocytes. The results indicated that Ang II inhibited the current amplitude of BKCa channel and decreased the slope of I-V curve. Ang II also made the activation curves of BKCa channel shift to the left. These results may provide a theoretical basis for potential treatment of chronic glomerular disease in the future.
Collapse
Affiliation(s)
- Na Gao
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Nankai University, Tianjin, 300071, China
| | | | | | | |
Collapse
|
45
|
Maezawa Y, Takemoto M, Yokote K. Cell biology of diabetic nephropathy: Roles of endothelial cells, tubulointerstitial cells and podocytes. J Diabetes Investig 2014; 6:3-15. [PMID: 25621126 PMCID: PMC4296695 DOI: 10.1111/jdi.12255] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 05/15/2014] [Indexed: 02/06/2023] Open
Abstract
Diabetic nephropathy is the major cause of end-stage renal failure throughout the world in both developed and developing countries. Diabetes affects all cell types of the kidney, including endothelial cells, tubulointerstitial cells, podocytes and mesangial cells. During the past decade, the importance of podocyte injury in the formation and progression of diabetic nephropathy has been established and emphasized. However, recent findings provide additional perspectives on pathogenesis of diabetic nephropathy. Glomerular endothelial damage is already present in the normoalbuminuric stage of the disease when podocyte injury starts. Genetic targeting of mice that cause endothelial injury leads to accelerated diabetic nephropathy. Tubulointerstitial damage, previously considered to be a secondary effect of glomerular protein leakage, was shown to have a primary significance in the progression of diabetic nephropathy. Emerging evidence suggests that the glomerular filtration barrier and tubulointerstitial compartment is a composite, dynamic entity where any injury of one cell type spreads to other cell types, and leads to the dysfunction of the whole apparatus. Accumulation of novel knowledge would provide a better understanding of the pathogenesis of diabetic nephropathy, and might lead to a development of a new therapeutic strategy for the disease.
Collapse
Affiliation(s)
- Yoshiro Maezawa
- Department of Clinical Cell Biology and Medicine, Chiba University Graduate School of Medicine Chiba, Japan ; Division of Diabetes, Metabolism and Endocrinology, Chiba University Hospital Chiba, Japan
| | - Minoru Takemoto
- Department of Clinical Cell Biology and Medicine, Chiba University Graduate School of Medicine Chiba, Japan ; Division of Diabetes, Metabolism and Endocrinology, Chiba University Hospital Chiba, Japan
| | - Koutaro Yokote
- Department of Clinical Cell Biology and Medicine, Chiba University Graduate School of Medicine Chiba, Japan ; Division of Diabetes, Metabolism and Endocrinology, Chiba University Hospital Chiba, Japan
| |
Collapse
|
46
|
Lu W, Liu S, Zhao Z, Liu Y, Li T. The effect of connective tissue growth factor on renal fibrosis and podocyte injury in hypertensive rats. Ren Fail 2014; 36:1420-7. [DOI: 10.3109/0886022x.2014.934692] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
47
|
Abstract
The high prevalence of vitamin D deficiency in patients with chronic kidney disease is believed to be an important risk factor for the cardiorenal syndrome commonly seen in this patient population. African Americans suffer a disproportionally high incidence of renal and cardiovascular disease with poor disease outcome, which may be partly attributed to their low vitamin D status in part owing to low subcutaneous photoproduction of vitamin D. Mounting evidence from animal and clinical studies has shown beneficial effects of vitamin D therapy on the renal and cardiovascular systems, and the underlying renoprotective and cardioprotective mechanisms of vitamin D receptor (VDR)-mediated signaling are under intense investigation. In this article, our most recent understanding of the renal protective mechanism of the podocyte VDR signaling against diabetic nephropathy and the anti-atherosclerotic role of macrophage VDR signaling in the regulation of atherosclerosis is reviewed.
Collapse
Affiliation(s)
- Yan Chun Li
- Department of Medicine, The University of Chicago, Chicago, IL.
| |
Collapse
|
48
|
Plagov A, Lan X, Rai P, Kumar D, Lederman R, Rehman S, Malhotra A, Ding G, Chander PN, Singhal PC. Modulation of renin angiotensin system predominantly alters sclerotic phenotype of glomeruli in HIVAN. Histol Histopathol 2014; 29:1575-81. [PMID: 24892944 DOI: 10.14670/hh-29.1575] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
HIV-associated nephropathy (HIVAN) is a common complication of HIV-1 infection in patients with African ancestry in general and with APOL1 gene risk variants in particular. Although collapsing glomerulopathy is considered a hallmark of HIVAN, significant numbers of glomeruli in patients with HIVAN also display other variants of focal segmental glomerulosclerosis (FSGS). We propose that collapsed glomeruli as well as glomeruli with other variants of FSGS are manifestations of HIVAN and their prevalence depends on associated host factors. We explored the role of the renin-angiotensin system (RAS) in the manifestation of any specific glomerular phenotype in HIVAN. To evaluate the role of the RAS we have used a genetically engineered mouse model of HIVAN (Tg26) with two and four copies of angiotensinogen (Agt) gene (Tg26/Agt2 and Tg26/Agt4). In Tg26/Agt2, 1 out of 6 glomeruli exhibited sclerosed phenotype, whereas 1 out of 25 glomeruli displayed collapsed phenotype; on the other hand, in Tg26/Agt4, 1 out of 3 glomeruli exhibited sclerotic phenotype and only 1 out of 7 glomeruli showed collapsed phenotype. To inhibit the effect of RAS, Tg26/Agt2 were administered captopril, aliskiren, aliskiren plus captopril or aliskiren plus telmisartan by miniosmotic pumps for 4 weeks. In all experimental groups there was a significant reduction in percentage of sclerosed glomeruli and only minimal reduction in collapsed glomeruli compared to normal saline receiving Tg26/Agt2. These findings suggest that the manifestation of the sclerosed phenotype in HIVAN is predominantly dependent on activation of the RAS.
Collapse
Affiliation(s)
- Andrei Plagov
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Xiqian Lan
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Partab Rai
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Dileep Kumar
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Rivka Lederman
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Shabina Rehman
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Ashwani Malhotra
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Guohua Ding
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Praveen N Chander
- Department of Pathology, New York Medical College, Valhalla, NY, USA
| | - Pravin C Singhal
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Manhasset, NY, USA.
| |
Collapse
|
49
|
Kittikulsuth W, Looney SW, Pollock DM. Endothelin ET(B) receptors contribute to sex differences in blood pressure elevation in angiotensin II hypertensive rats on a high-salt diet. Clin Exp Pharmacol Physiol 2014; 40:362-70. [PMID: 23713708 DOI: 10.1111/1440-1681.12084] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 02/23/2013] [Accepted: 03/05/2013] [Indexed: 11/27/2022]
Abstract
Female rats are more resistant to blood pressure increases induced by high salt (HS) intake or angiotensin (Ang) II infusion. Because endothelin ET(B) receptors on endothelial and epithelial cells mediate tonic vasodilation and sodium excretion, we hypothesized that ET(B) receptors limit the hypertensive response and renal injury induced by HS diet alone or with chronic AngII infusion (AngII/HS) in female compared with male rats. A 4 week HS diet (4% NaCl) did not significantly change blood pressure (measured by telemetry) in either male or female Sprague-Dawley rats. Administration of the ET(B) receptor antagonist A-192621 (1, 3 and 10 mg/kg per day in food) during HS feeding caused a dose-dependent increase in blood pressure in both sexes. In AngII/HS rats, males had a larger increase in blood pressure than females. The increase in blood pressure produced by ET(B) receptor blockade in male AngII/HS rats was not significant. However, A-192621 treatment resulted in a significant further increase in blood pressure in female AngII/HS rats. Male rats had consistently higher protein excretion rates before and during AngII/HS, but this was not significantly affected by ET(B) receptor blockade in either sex. In conclusion, ET(B) receptors play a significantly greater beneficial role in protecting female compared with male rats against AngII-induced hypertension and may contribute to the sex differences in AngII-induced hypertension.
Collapse
Affiliation(s)
- Wararat Kittikulsuth
- Section of Experimental Medicine, Department of Medicine, Georgia Regents University, Augusta, GA, USA
| | | | | |
Collapse
|
50
|
Liu Y, Hitomi H, Diah S, Deguchi K, Mori H, Masaki T, Nakano D, Kobori H, Nishiyama A. Roles of Na⁺/H⁺ exchanger type 1 and intracellular pH in angiotensin II-induced reactive oxygen species generation and podocyte apoptosis. J Pharmacol Sci 2014; 122:176-83. [PMID: 23800993 DOI: 10.1254/jphs.12291fp] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
A growing body of evidence suggests that podocyte apoptosis is a major cause of decreased podocyte number, which leads to albuminuria and glomerular injury. The aim of this study was to clarify the molecular mechanisms of angiotensin II (Ang II)-induced apoptosis in cultured mouse podocytes. We examined the effects of Ang II (100 nmol/L) on apoptosis, superoxide anions, and cytosol pH in podocytes. For intracellular pH measurements, image analysis was conducted using confocal laser microscopy after incubation with carboxy-seminaphthorhodafluor-1. Superoxide anions and intracellular pH were elevated with Ang II treatment. Apoptotic cell numbers, as measured by TUNEL staining and caspase 3 activity, were also augmented in the Ang II-treated group. Pre-treatment with olmesartan (100 nmol/L, an Ang II type 1-receptor blocker), apocynin (50 μmol/L, NADPH oxidase inhibitor), or 5-N,N hexamethylene amiloride [30 μmol/L, Na⁺/H⁺ exchanger type 1 (NHE-1) inhibitor] abolished Ang II-induced podocyte apoptosis, whereas NHE-1 mRNA and protein expression was not affected by Ang II treatment. Moreover, Ang II increased NHE-1 phosphorylation. These results suggest that superoxide production, NHE-1 activation, and intracellular alkalization were early features prior to apoptosis in Ang II-treated mouse podocytes, and may offer new insights into the mechanisms responsible for Ang II-induced podocyte injury.
Collapse
Affiliation(s)
- Ya Liu
- Department of Pharmacology, Faculty of Medicine, Kagawa University, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|