1
|
Nagalakshmi VK, Smith JP, Matsuoka D, Gomez RA, Lopez MLSS. Piezo channels in JG cells do not regulate renin expression or renin release to the circulation. Clin Sci (Lond) 2024; 138:1527-1536. [PMID: 39564667 PMCID: PMC11843579 DOI: 10.1042/cs20242089] [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: 09/12/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 11/21/2024]
Abstract
Renin-expressing juxtaglomerular (JG) cells possess an intrinsic pressure-sensing mechanism(s) that regulates renin synthesis and release in response to changes in perfusion pressure. Although we recently described the structure of the nuclear mechanotransducer that controls renin transcription, the acute pressure-sensing mechanism that controls the rapid release of renin has not been identified. In JG cells there is an inverse relationship between intracellular calcium and renin release, the 'calcium paradox'. Since the discovery of Piezo2 as the 'touch' receptors, there has been a significant interest in exploring whether they are also involved in other tissues beyond the skin. Given that Piezo receptors are permeable to calcium upon mechanical stimuli, it would be reasonable to hypothesize that Piezo2 controls renin synthesis and/or release in JG cells. To test this hypothesis, we used a variety of novel mouse models and JG cell-specific techniques to define whether Piezo2 controls renin expression and/or release in JG cells. Our in vivo data using constitutive and inducible Cre driver mouse lines and a variety of novel experimental approaches indicate that Piezo2 channels are not necessary for renin synthesis or release in JG cells during normal conditions or when homeostasis is threatened by hypotension, sodium depletion, or inverse changes in blood pressure. Furthermore, Piezo1 channels do not compensate for the lack of Piezo2 in JG cells. Efforts should be devoted to identifying the acute mechanosensory mechanisms controlling renin release.
Collapse
Affiliation(s)
- Vidya K. Nagalakshmi
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA
| | - Jason P. Smith
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA
| | - Daisuke Matsuoka
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA
| | - R. Ariel Gomez
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA
| | | |
Collapse
|
2
|
Nagalakshmi VK, Li M, Liang X, Medrano S, Belyea BC, Gomez RA, Sequeira-Lopez MLS. Cells of the renin lineage promote kidney regeneration post-release of ureteral obstruction in neonatal mice. Acta Physiol (Oxf) 2023; 238:e14014. [PMID: 37309075 PMCID: PMC10527043 DOI: 10.1111/apha.14014] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 06/07/2023] [Accepted: 06/07/2023] [Indexed: 06/14/2023]
Abstract
AIM Ureteral obstruction leads to significant changes in kidney renin expression. It is unclear whether those changes are responsible for the progression of kidney damage, repair, or regeneration. In the current study, we aimed to elucidate the contribution of renin-producing cells (RPCs) and the cells of the renin lineage (CoRL) towards kidney damage and regeneration using a model of partial and reversible unilateral ureteral obstruction (pUUO) in neonatal mice. METHODS Renin cells are progenitors for other renal cell types collectively called CoRL. We labeled the CoRL with green fluorescent protein (GFP) using genetic approaches. We performed lineage tracing to analyze the changes in the distribution of CoRL during and after the release of obstruction. We also ablated the RPCs and CoRL by cell-specific expression of Diphtheria Toxin Sub-unit A (DTA). Finally, we evaluated the kidney damage and regeneration during and after the release of obstruction in the absence of CoRL. RESULTS In the obstructed kidneys, there was a 163% increase in the renin-positive area and a remarkable increase in the distribution of GFP+ CoRL. Relief of obstruction abrogated these changes. In addition, DTA-expressing animals did not respond to pUUO with increased RPCs and CoRL. Moreover, reduction in CoRL significantly compromised the kidney's ability to recover from the damage after the release of obstruction. CONCLUSIONS CoRL play a role in the regeneration of the kidneys post-relief of obstruction.
Collapse
Affiliation(s)
- Vidya K. Nagalakshmi
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA
| | - Minghong Li
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA
| | - Xiuyin Liang
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA
| | - Silvia Medrano
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA
| | - Brian C Belyea
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA
| | - R. Ariel Gomez
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA
| | | |
Collapse
|
3
|
Guessoum O, Zainab M, Sequeira‐Lopez MLS, Gomez RA. Proliferation does not contribute to murine models of renin cell recruitment. Acta Physiol (Oxf) 2020. [PMCID: PMC7583373 DOI: 10.1111/apha.13532] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Aim Renin cells are essential for regulation of blood pressure and fluid‐electrolyte homeostasis. During homeostatic threat, the number of renin cells in the kidney increases, a process termed as recruitment. It has been proposed that recruitment occurs by proliferation, yet no systematic studies have been performed. We sought to determine the extent to which proliferation contributes to the recruitment process. Methods Mice were subjected to recruitment before analysing the renin cells’ cell cycle. For acute threats, we subjected SV129 and C57Bl6 mice to a low sodium diet plus captopril. Tissue sections from treated mice were co‐stained for proliferation markers (Ki67, PCNA, pH3 and BrdU) and renin. Chronic recruitment was studied in deletion models of aldosterone synthase and angiotensinogen through co‐immunostaining and counting mitotic figures in periodic acid‐Schiff‐stained sections. Finally, RNA‐seq of renin cells isolated from recruited mice was performed to study mitotic signature. Results Mice subjected to low salt and captopril displayed increases in renin cell number (312 ± 40 in controls to 692 ± 85 in recruited animals, P<.0001), 10‐fold increases in renin mRNA and fourfold increases in circulating renin. Co‐staining these kidney sections for proliferation markers revealed negligible proliferation of renin cells (<2%), indistinguishable from control animals. Similarly, chronic models of recruitment—aldosterone synthase KO and angiotensinogen KO—had negligible proliferation. Additionally, the transcriptome of recruited renin cells revealed overall downregulation of mitotic pathways when compared to proliferative cell lines. Conclusion Acute and chronic physiological threats to homeostasis produced a distinct increase in renin‐synthesizing cells, but we found no evidence to suggest the involvement of proliferation.
Collapse
Affiliation(s)
- Omar Guessoum
- Department of Biology University of Virginia Charlottesville VA USA
- Department of Pediatrics University of Virginia Charlottesville VA USA
- Child Health Research Center University of Virginia Charlottesville VA USA
| | - Momna Zainab
- Department of Biology University of Virginia Charlottesville VA USA
- Child Health Research Center University of Virginia Charlottesville VA USA
| | - Maria Luisa S. Sequeira‐Lopez
- Department of Biology University of Virginia Charlottesville VA USA
- Department of Pediatrics University of Virginia Charlottesville VA USA
- Child Health Research Center University of Virginia Charlottesville VA USA
| | - R. Ariel Gomez
- Department of Biology University of Virginia Charlottesville VA USA
- Department of Pediatrics University of Virginia Charlottesville VA USA
- Child Health Research Center University of Virginia Charlottesville VA USA
| |
Collapse
|
4
|
Martinez MF, Martini AG, Sequeira-Lopez MLS, Gomez RA. Ctcf is required for renin expression and maintenance of the structural integrity of the kidney. Clin Sci (Lond) 2020; 134:1763-1774. [PMID: 32619009 PMCID: PMC7881370 DOI: 10.1042/cs20200184] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/23/2020] [Accepted: 07/03/2020] [Indexed: 12/17/2022]
Abstract
Renin cells are crucial for the regulation of blood pressure and fluid electrolyte homeostasis. We have recently shown that renin cells possess unique chromatin features at regulatory regions throughout the genome that may determine the identity and memory of the renin phenotype. The 3-D structure of chromatin may be equally important in the determination of cell identity and fate. CCCTC-binding factor (Ctcf) is a highly conserved chromatin organizer that may regulate the renin phenotype by controlling chromatin structure. We found that Ctcf binds at several conserved DNA sites surrounding and within the renin locus, suggesting that Ctcf may regulate the transcriptional activity of renin cells. In fact, deletion of Ctcf in cells of the renin lineage led to decreased endowment of renin-expressing cells accompanied by decreased circulating renin, hypotension, and severe morphological abnormalities of the kidney, including defects in arteriolar branching, and ultimately renal failure. We conclude that control of chromatin architecture by Ctcf is necessary for the appropriate expression of renin, control of renin cell number and structural integrity of the kidney.
Collapse
Affiliation(s)
- Maria Florencia Martinez
- Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, 22908. United States
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, 22908. United States
| | - Alexandre G. Martini
- Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, 22908. United States
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, 22908. United States
| | - Maria Luisa S. Sequeira-Lopez
- Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, 22908. United States
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, 22908. United States
- Department of Biology, University of Virginia School of Medicine, Charlottesville, Virginia, 22908. United States
| | - R. Ariel Gomez
- Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, 22908. United States
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, 22908. United States
- Department of Biology, University of Virginia School of Medicine, Charlottesville, Virginia, 22908. United States
| |
Collapse
|
5
|
Mohamed TH, Watanabe H, Kaur R, Belyea BC, Walker PD, Gomez RA, Sequeira-Lopez MLS. Renin-Expressing Cells Require β1-Integrin for Survival and for Development and Maintenance of the Renal Vasculature. Hypertension 2020; 76:458-467. [PMID: 32594804 DOI: 10.1161/hypertensionaha.120.14959] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Juxtaglomerular cells are crucial for blood pressure and fluid-electrolyte homeostasis. The factors that maintain the life of renin cells are unknown. In vivo, renin cells receive constant cell-to-cell, mechanical, and neurohumoral stimulation that maintain their identity and function. Whether the presence of this niche is crucial for the vitality of the juxtaglomerular cells is unknown. Integrins are the largest family of cell adhesion molecules that mediate cell-to-cell and cell-to-matrix interactions. Of those, β1-integrin is the most abundant in juxtaglomerular cells. However, its role in renin cell identity and function has not been ascertained. To test the hypothesis that cell-matrix interactions are fundamental not only to maintain the identity and function of juxtaglomerular cells but also to keep them alive, we deleted β1-integrin in vivo in cells of the renin lineage. In mutant mice, renin cells died by apoptosis, resulting in decreased circulating renin, hypotension, severe renal-vascular abnormalities, and renal failure. Results indicate that cell-to-cell and cell-to-matrix interactions via β1-integrin is essential for juxtaglomerular cells survival, suggesting that the juxtaglomerular niche is crucial not only for the tight regulation of renin release but also for juxtaglomerular cell survival-a sine qua non condition to maintain homeostasis.
Collapse
Affiliation(s)
- Tahagod H Mohamed
- From the Child Health Research Center, Department of Pediatrics (T.H.M., H.W., R.K., B.C.B., R.A.G., M.L.S.S.-L.), University of Virginia School of Medicine, Charlottesville
| | - Hirofumi Watanabe
- From the Child Health Research Center, Department of Pediatrics (T.H.M., H.W., R.K., B.C.B., R.A.G., M.L.S.S.-L.), University of Virginia School of Medicine, Charlottesville
| | - Rajwinderjit Kaur
- From the Child Health Research Center, Department of Pediatrics (T.H.M., H.W., R.K., B.C.B., R.A.G., M.L.S.S.-L.), University of Virginia School of Medicine, Charlottesville
| | - Brian C Belyea
- From the Child Health Research Center, Department of Pediatrics (T.H.M., H.W., R.K., B.C.B., R.A.G., M.L.S.S.-L.), University of Virginia School of Medicine, Charlottesville
| | - Patrick D Walker
- Renal Pathology Division, Arkana Laboratories, Little Rock, AR (P.D.W.)
| | - R Ariel Gomez
- From the Child Health Research Center, Department of Pediatrics (T.H.M., H.W., R.K., B.C.B., R.A.G., M.L.S.S.-L.), University of Virginia School of Medicine, Charlottesville.,Department of Biology (R.A.G., M.L.S.S.-L.), University of Virginia School of Medicine, Charlottesville
| | - Maria Luisa S Sequeira-Lopez
- From the Child Health Research Center, Department of Pediatrics (T.H.M., H.W., R.K., B.C.B., R.A.G., M.L.S.S.-L.), University of Virginia School of Medicine, Charlottesville.,Department of Biology (R.A.G., M.L.S.S.-L.), University of Virginia School of Medicine, Charlottesville
| |
Collapse
|
6
|
Schramm A, Schweda F, Sequeira-Lopez MLS, Hofmann F, Sandner P, Schlossmann J. Protein Kinase G Is Involved in Acute but Not in Long-Term Regulation of Renin Secretion. Front Pharmacol 2019; 10:800. [PMID: 31379575 PMCID: PMC6657341 DOI: 10.3389/fphar.2019.00800] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 06/21/2019] [Indexed: 12/27/2022] Open
Abstract
Pharmacological inhibition of the renin–angiotensin–aldosterone system (RAAS) is, in combination with diuretics, the first-choice treatment for hypertension, although 10–20% of patients do not respond adequately. Next to the RAAS, the nitric oxide/cGMP/protein kinase G (PKG) system is the second fundamental blood pressure regulator. Whether both systems influence each other is not well-studied. It has been shown that nitric oxide (NO) supports renin recruitment via activation of soluble guanylate cyclase (sGC) and subsequent generation of cGMP. Whether this leads to an ensuing activation of PKGs in this context is not known. PKGIα, as well as PKGII, is expressed in renin-producing cells. Hence, we analyzed whether these enzymes play a role regarding renin synthesis, secretion, or recruitment. We generated renin-cell-specific PKGI-knockout mice and either stimulated or inhibited the renin system in these mice by salt diets. To exclude the possibility that one kinase isoform can compensate the lack of the other, we also studied double-knockout animals with a conditional knockout of PKGI in juxtaglomerular cells (JG cells) and a ubiquitous knockout of PKGII. We analyzed blood pressure, renin mRNA and renal renin protein content as well as plasma renin concentration. Furthermore, we stimulated the cGMP system in these mice using BAY 41-8543, an sGC stimulator, and examined renin regulation either after acute administration or after 7 days (application once daily). We did not reveal any striking differences regarding long-term renin regulation in the studied mouse models. Yet, when we studied the acute effect of BAY 41-8543 on renin secretion in isolated perfused kidneys as well as in living animals, we found that the administration of the substance led to a significant increase in plasma renin concentration in control animals. This effect was completely abolished in double-knockout animals. However, after 7 days of once daily application, we did not detect a persistent increase in renin mRNA or protein in any studied genotype. Therefore, we conclude that in mice, cGMP and PKG are involved in the acute regulation of renin release but have no influence on long-term renin adjustment.
Collapse
Affiliation(s)
- Andrea Schramm
- Institute of Pharmacy, Department of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany
| | - Frank Schweda
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | | | - Franz Hofmann
- Institute of Pharmacology and Toxicology, Technical University of Munich, Munich, Germany
| | - Peter Sandner
- Bayer AG, Drug Discovery-Cardiology, Wuppertal, Germany
| | - Jens Schlossmann
- Institute of Pharmacy, Department of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
7
|
Martinez MF, Medrano S, Brown RI, Tufan T, Shang S, Bertoncello N, Guessoum O, Adli M, Belyea BC, Sequeira-Lopez MLS, Gomez RA. Super-enhancers maintain renin-expressing cell identity and memory to preserve multi-system homeostasis. J Clin Invest 2018; 128:4787-4803. [PMID: 30130256 PMCID: PMC6205391 DOI: 10.1172/jci121361] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 08/14/2018] [Indexed: 02/06/2023] Open
Abstract
Renin cells are crucial for survival - they control fluid-electrolyte and blood pressure homeostasis, vascular development, regeneration, and oxygen delivery to tissues. During embryonic development, renin cells are progenitors for multiple cell types that retain the memory of the renin phenotype. When there is a threat to survival, those descendants are transformed and reenact the renin phenotype to restore homeostasis. We tested the hypothesis that the molecular memory of the renin phenotype resides in unique regions and states of these cells' chromatin. Using renin cells at various stages of stimulation, we identified regions in the genome where the chromatin is open for transcription, mapped histone modifications characteristic of active enhancers such as H3K27ac, and tracked deposition of transcriptional activators such as Med1, whose deletion results in ablation of renin expression and low blood pressure. Using the rank ordering of super-enhancers, epigenetic rewriting, and enhancer deletion analysis, we found that renin cells harbor a unique set of super-enhancers that determine their identity. The most prominent renin super-enhancer may act as a chromatin sensor of signals that convey the physiologic status of the organism, and is responsible for the transformation of renin cell descendants to the renin phenotype, a fundamental process to ensure homeostasis.
Collapse
Affiliation(s)
| | | | | | - Turan Tufan
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Stephen Shang
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | | | - Omar Guessoum
- Child Health Research Center
- Department of Pediatrics
- Department of Biology, and
| | - Mazhar Adli
- Child Health Research Center
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | | | | | - R. Ariel Gomez
- Child Health Research Center
- Department of Pediatrics
- Department of Biology, and
| |
Collapse
|
8
|
Gomez RA, Sequeira-Lopez MLS. Novel Functions of Renin Precursors in Homeostasis and Disease. Physiology (Bethesda) 2017; 31:25-33. [PMID: 26661526 DOI: 10.1152/physiol.00039.2015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Renin progenitors appear early and are found in multiple tissues throughout the embryo. Besides their well known role in blood pressure and fluid homeostasis, renin progenitors participate in tissue morphogenesis, repair, and regeneration, and may integrate immune and endocrine responses. In the bone marrow, renin cells offer clues to understand normal and neoplastic hematopoiesis.
Collapse
Affiliation(s)
- R Ariel Gomez
- University of Virginia School of Medicine, Child Health Research Center, Charlottesville, Virginia
| | | |
Collapse
|
9
|
Uwaezuoke SN, Okafor HU, Muoneke VN, Odetunde OI, Odimegwu CL. Chronic kidney disease in children and the role of epigenetics: Future therapeutic trajectories. Biomed Rep 2016; 5:660-664. [PMID: 28105334 PMCID: PMC5228463 DOI: 10.3892/br.2016.781] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 10/04/2016] [Indexed: 01/08/2023] Open
Abstract
Global differences in the observed causes of chronic kidney disease (CKD) in children are well documented and are attributed to dissimilarities in clime, race, hereditary, and ancestry. Thus, familial clustering and disparities in CKD prevalence rates across ethnic and racial groups indicate that the progression of renal disease has a strong genetic component. Mammalian studies have demonstrated a feasible nexus between nutrition and non-genetic exposure (around the time of conception and in epigenetic changes) in the expression of major genes identified in renal organogenesis. The major consequence is a reduction in the number of nephrons, with subsequent predisposition to hypertension and CKD. Identifying these epigenetic changes is crucial (due to their potentially reversible nature), as they may serve as future therapeutic targets to prevent kidney fibrosis and CKD. Despite progress in the field of epigenetics in oncology, research in other subspecialties of medicine is largely experimental with few existing studies regarding the clinical implication of epigenetics in renal disease. Therapeutic trajectories for CKD in children based on the influence of epigenetics may eventually revolutionize the management of this disease. The aim of the current narrative review is to appraise the role of epigenetics in CKD, and highlight the potential future therapeutic pathways.
Collapse
Affiliation(s)
- Samuel N. Uwaezuoke
- Department of Pediatrics, College of Medicine, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu 400001, Nigeria
| | - Henrietta U. Okafor
- Department of Pediatrics, College of Medicine, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu 400001, Nigeria
| | - Vivian N. Muoneke
- Department of Pediatrics, College of Medicine, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu 400001, Nigeria
| | - Odutola I. Odetunde
- Department of Pediatrics, College of Medicine, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu 400001, Nigeria
| | - Chioma L. Odimegwu
- Department of Pediatrics, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu 400001, Nigeria
| |
Collapse
|
10
|
Lichtnekert J, Kaverina NV, Eng DG, Gross KW, Kutz JN, Pippin JW, Shankland SJ. Renin-Angiotensin-Aldosterone System Inhibition Increases Podocyte Derivation from Cells of Renin Lineage. J Am Soc Nephrol 2016; 27:3611-3627. [PMID: 27080979 DOI: 10.1681/asn.2015080877] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 02/20/2016] [Indexed: 12/17/2022] Open
Abstract
Because adult podocytes cannot proliferate and are therefore unable to self-renew, replacement of these cells depends on stem/progenitor cells. Although podocyte number is higher after renin-angiotensin-aldosterone system (RAAS) inhibition in glomerular diseases, the events explaining this increase are unclear. Cells of renin lineage (CoRL) have marked plasticity, including the ability to acquire a podocyte phenotype. To test the hypothesis that RAAS inhibition partially replenishes adult podocytes by increasing CoRL number, migration, and/or transdifferentiation, we administered tamoxifen to Ren1cCreERxRs-tdTomato-R CoRL reporter mice to induce permanent labeling of CoRL with red fluorescent protein variant tdTomato. We then induced experimental FSGS, typified by abrupt podocyte depletion, with a cytopathic antipodocyte antibody. RAAS inhibition by enalapril (angiotensin-converting enzyme inhibitor) or losartan (angiotensin-receptor blocker) in FSGS mice stimulated the proliferation of CoRL, increasing the reservoir of these cells in the juxtaglomerular compartment (JGC). Compared with water or hydralazine, RAAS inhibition significantly increased the migration of CoRL from the JGC to the intraglomerular compartment (IGC), with more glomeruli containing RFP+CoRL and, within these glomeruli, more RFP+CoRL. Moreover, RAAS inhibition in FSGS mice increased RFP+CoRL transdifferentiation in the IGC to phenotypes, consistent with those of podocytes (coexpression of synaptopodin and Wilms tumor protein), parietal epithelial cells (PAX 8), and mesangial cells (α8 integrin). These results show that in the context of podocyte depletion in FSGS, RAAS inhibition augments CoRL proliferation and plasticity toward three different glomerular cell lineages.
Collapse
Affiliation(s)
| | | | | | - Kenneth W Gross
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, New York
| | - J Nathan Kutz
- Department of Applied Mathematics, University of Washington, Seattle, Washington; and
| | | | | |
Collapse
|
11
|
Sequeira-Lopez MLS, Lin EE, Li M, Hu Y, Sigmund CD, Gomez RA. The earliest metanephric arteriolar progenitors and their role in kidney vascular development. Am J Physiol Regul Integr Comp Physiol 2014; 308:R138-49. [PMID: 25427768 DOI: 10.1152/ajpregu.00428.2014] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The development of the kidney arterioles is poorly understood. Mature arterioles contain several functionally and morphologically distinct cell types, including smooth muscle, endothelial, and juxtaglomerular cells, and they are surrounded by interconnected pericytes, fibroblasts, and other interstitial cells. We have shown that the embryonic kidney possesses all of the necessary precursors for the development of the renal arterial tree, and those precursors assemble in situ to form the kidney arterioles. However, the identity of those precursors was unclear. Within the embryonic kidney, several putative progenitors marked by the expression of either the winged-forkhead transcription factor 1 (Foxd1+ progenitor), the aspartyl-protease renin (Ren+ progenitor), and/or hemangioblasts (Scl+ progenitor) are likely to differentiate and endow most of the cells of the renal arterial tree. However, the lineage relationships and the role of these distinct progenitors in renal vascular morphogenesis have not been delineated. We, therefore, designed a series of experiments to ascertain the hierarchical lineage relationships between Foxd1+ and Ren+ progenitors and the role of these two precursors in the morphogenesis and patterning of the renal arterial tree. Results show that 1) Foxd1+ cells are the precursors for all the mural cells (renin cells, smooth muscle cells, perivascular fibroblasts, and pericytes) of the renal arterial tree and glomerular mesangium, and 2) Foxd1 per se directs the origin, number, orientation, and cellular composition of the renal vessels.
Collapse
Affiliation(s)
| | - Eugene E Lin
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia; and
| | - Minghong Li
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia; and
| | - Yan Hu
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia; and
| | - Curt D Sigmund
- Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - R Ariel Gomez
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia; and
| |
Collapse
|
12
|
Schmid J, Oelbe M, Saftig P, Schwake M, Schweda F. Parallel regulation of renin and lysosomal integral membrane protein 2 in renin-producing cells: further evidence for a lysosomal nature of renin secretory vesicles. Pflugers Arch 2013; 465:895-905. [PMID: 23229015 DOI: 10.1007/s00424-012-1192-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 11/23/2012] [Accepted: 11/23/2012] [Indexed: 01/21/2023]
Abstract
The protease renin is the key enzyme in the renin-angiotensin system (RAS) that regulates extracellular volume and blood pressure. Renin is synthesized in renal juxtaglomerular cells (JG cells) as the inactive precursor prorenin. Activation of prorenin by cleavage of the prosegment occurs in renin storage vesicles that have lysosomal properties. To characterize the renin storage vesicles more precisely, the expression and functional relevance of the major lysosomal membrane proteins lysosomal-associated membrane protein 1 (LAMP-1), LAMP-2, and lysosomal integral membrane protein 2 (LIMP-2) were determined in JG cells. Immunostaining experiments revealed strong coexpression of renin with the LIMP-2 (SCARB2), while faint staining of LAMP-1 and LAMP-2 was detected in some JG cells only. Stimulation of the renin system (ACE inhibitor, renal hypoperfusion) resulted in the recruitment of renin-producing cells in the afferent arterioles and parallel upregulation of LIMP-2, but not LAMP-1 or LAMP-2. Despite the coregulation of renin and LIMP-2, LIMP-2-deficient mice had normal renal renin mRNA levels, renal renin and prorenin contents, and plasma renin and prorenin concentrations under control conditions and in response to stimulation with a low salt diet (with or without angiotensin-converting enzyme (ACE) inhibition). No differences in the size or number of renin vesicles were detected using electron microscopy. Acute stimulation of renin release by isoproterenol exerted similar responses in both genotypes in vivo and in isolated perfused kidneys. Renin and the major lysosomal protein LIMP-2 are colocalized and coregulated in renal JG cells, further corroborating the lysosomal nature of renin storage vesicles. LIMP-2 does not appear to play an obvious role in the regulation of renin synthesis or release.
Collapse
Affiliation(s)
- Johannes Schmid
- Institut für Physiologie, Universität Regensburg, 93040, Regensburg, Germany
| | | | | | | | | |
Collapse
|
13
|
Buys ES, Raher MJ, Kirby A, Shahid M, Mohd S, Baron DM, Hayton SR, Tainsh LT, Sips PY, Rauwerdink KM, Yan Q, Tainsh RET, Shakartzi HR, Stevens C, Decaluwé K, Rodrigues-Machado MDG, Malhotra R, Van de Voorde J, Wang T, Brouckaert P, Daly MJ, Bloch KD. Genetic modifiers of hypertension in soluble guanylate cyclase α1-deficient mice. J Clin Invest 2012; 122:2316-25. [PMID: 22565307 DOI: 10.1172/jci60119] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Accepted: 03/21/2012] [Indexed: 01/09/2023] Open
Abstract
Nitric oxide (NO) plays an essential role in regulating hypertension and blood flow by inducing relaxation of vascular smooth muscle. Male mice deficient in a NO receptor component, the α1 subunit of soluble guanylate cyclase (sGCα1), are prone to hypertension in some, but not all, mouse strains, suggesting that additional genetic factors contribute to the onset of hypertension. Using linkage analyses, we discovered a quantitative trait locus (QTL) on chromosome 1 that was linked to mean arterial pressure (MAP) in the context of sGCα1 deficiency. This region is syntenic with previously identified blood pressure-related QTLs in the human and rat genome and contains the genes coding for renin. Hypertension was associated with increased activity of the renin-angiotensin-aldosterone system (RAAS). Further, we found that RAAS inhibition normalized MAP and improved endothelium-dependent vasorelaxation in sGCα1-deficient mice. These data identify the RAAS as a blood pressure-modifying mechanism in a setting of impaired NO/cGMP signaling.
Collapse
Affiliation(s)
- Emmanuel S Buys
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Pentz ES, Cordaillat M, Carretero OA, Tucker AE, Sequeira Lopez MLS, Gomez RA. Histone acetyl transferases CBP and p300 are necessary for maintenance of renin cell identity and transformation of smooth muscle cells to the renin phenotype. Am J Physiol Heart Circ Physiol 2012; 302:H2545-52. [PMID: 22523253 DOI: 10.1152/ajpheart.00782.2011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In response to a homeostatic threat circulating renin increases by increasing the number of cells expressing renin by dedifferentiation and re-expression of renin in arteriolar smooth muscle cells (aSMCs) that descended from cells that expressed renin in early life. However, the mechanisms that govern the maintenance and reacquisition of the renin phenotype are not well understood. The cAMP pathway is important for renin synthesis and release: the transcriptional effects are mediated by binding of cAMP responsive element binding protein with its co-activators, CBP and p300, to the cAMP response element in the renin promoter. We have shown previously that mice with conditional deletion of CBP and p300 (cKO) in renin cells had severely reduced renin expression in adult life. In this study we investigated when the loss of renin-expressing cells in the cKO occurred and found that the loss of renin expression becomes evident after differentiation of the kidney is completed during postnatal life. To determine whether CBP/p300 is necessary for re-expression of renin we subjected cKO mice to low sodium diet + captopril to induce retransformation of aSMCs to the renin phenotype. The cKO mice did not increase circulating renin, their renin mRNA and protein expression were greatly diminished compared with controls, and only a few aSMCs re-expressed renin. These studies underline the crucial importance of the CREB/CBP/p300 complex for the ability of renin cells to retain their cellular memory and regain renin expression, a fundamental survival mechanism, in response to a threat to homeostasis.
Collapse
Affiliation(s)
- Ellen Steward Pentz
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA
| | | | | | | | | | | |
Collapse
|
15
|
Schnermann J, Briggs JP. Synthesis and secretion of renin in mice with induced genetic mutations. Kidney Int 2012; 81:529-38. [PMID: 22258323 DOI: 10.1038/ki.2011.451] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The juxtaglomerular (JG) cell product renin is rate limiting in the generation of the bioactive octapeptide angiotensin II. Rates of synthesis and secretion of the aspartyl protease renin by JG cells are controlled by multiple afferent and efferent pathways originating in the CNS, cardiovascular system, and kidneys, and making critical contributions to the maintenance of extracellular fluid volume and arterial blood pressure. Since both excesses and deficits of angiotensin II have deleterious effects, it is not surprising that control of renin is secured by a complex system of feedforward and feedback relationships. Mice with genetic alterations have contributed to a better understanding of the networks controlling renin synthesis and secretion. Essential input for the setting of basal renin generation rates is provided by β-adrenergic receptors acting through cyclic adenosine monophosphate, the primary intracellular activation mechanism for renin mRNA generation. Other major control mechanisms include COX-2 and nNOS affecting renin through PGE2, PGI2, and nitric oxide. Angiotensin II provides strong negative feedback inhibition of renin synthesis, largely an indirect effect mediated by baroreceptor and macula densa inputs. Adenosine appears to be a dominant factor in the inhibitory arms of the baroreceptor and macula densa mechanisms. Targeted gene mutations have also shed light on a number of novel aspects related to renin processing and the regulation of renin synthesis and secretion.
Collapse
Affiliation(s)
- Jurgen Schnermann
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | |
Collapse
|
16
|
Brunskill EW, Sequeira-Lopez MLS, Pentz ES, Lin E, Yu J, Aronow BJ, Potter SS, Gomez RA. Genes that confer the identity of the renin cell. J Am Soc Nephrol 2011; 22:2213-25. [PMID: 22034642 DOI: 10.1681/asn.2011040401] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Renin-expressing cells modulate BP, fluid-electrolyte homeostasis, and kidney development, but remarkably little is known regarding the genetic regulatory network that governs the identity of these cells. Here we compared the gene expression profiles of renin cells with most cells in the kidney at various stages of development as well as after a physiologic challenge known to induce the transformation of arteriolar smooth muscle cells into renin-expressing cells. At all stages, renin cells expressed a distinct set of genes characteristic of the renin phenotype, which was vastly different from other cell types in the kidney. For example, cells programmed to exhibit the renin phenotype expressed Akr1b7, and maturing cells expressed angiogenic factors necessary for the development of the kidney vasculature and RGS (regulator of G-protein signaling) genes, suggesting a potential relationship between renin cells and pericytes. Contrary to the plasticity of arteriolar smooth muscle cells upstream from the glomerulus, which can transiently acquire the embryonic phenotype in the adult under physiologic stress, the adult juxtaglomerular cell always possessed characteristics of both smooth muscle and renin cells. Taken together, these results identify the gene expression profile of renin-expressing cells at various stages of maturity, and suggest that juxtaglomerular cells maintain properties of both smooth muscle and renin-expressing cells, likely to allow the rapid control of body fluids and BP through both contractile and endocrine functions.
Collapse
Affiliation(s)
- Eric W Brunskill
- Harrison Distinguished Professor of Pediatrics and Biology, University of Virginia, 409 Lane Road, MR4 Building, Room 2001, Charlottesville, VA 22908, USA
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Castellanos Rivera RM, Monteagudo MC, Pentz ES, Glenn ST, Gross KW, Carretero O, Sequeira-Lopez MLS, Gomez RA. Transcriptional regulator RBP-J regulates the number and plasticity of renin cells. Physiol Genomics 2011; 43:1021-8. [PMID: 21750232 DOI: 10.1152/physiolgenomics.00061.2011] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Renin-expressing cells are crucial in the control of blood pressure and fluid-electrolyte homeostasis. Notch receptors convey cell-cell signals that may regulate the renin cell phenotype. Because the common downstream effector for all Notch receptors is the transcription factor RBP-J, we used a conditional knockout approach to delete RBP-J in cells of the renin lineage. The resultant RBP-J conditional knockout (cKO) mice displayed a severe reduction in the number of renin-positive juxtaglomerular apparatuses (JGA) and a reduction in the total number of renin positive cells per JGA and along the afferent arterioles. This reduction in renin protein was accompanied by a decrease in renin mRNA expression, decreased circulating renin, and low blood pressure. To investigate whether deletion of RBP-J altered the ability of mice to increase the number of renin cells normally elicited by a physiological threat, we treated RBP-J cKO mice with captopril and sodium depletion for 10 days. The resultant treated RBP-J cKO mice had a 65% reduction in renin mRNA levels (compared with treated controls) and were unable to increase circulating renin. Although these mice attempted to increase the number of renin cells, the cells were unusually thin and had few granules and barely detectable amounts of immunoreactive renin. As a consequence, the cells were incapable of fully adopting the endocrine phenotype of a renin cell. We conclude that RBP-J is required to maintain basal renin expression and the ability of smooth muscle cells along the kidney vasculature to regain the renin phenotype, a fundamental mechanism to preserve homeostasis.
Collapse
Affiliation(s)
- Ruth M Castellanos Rivera
- Department of Pediatrics, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Woroniecki R, Gaikwad AB, Susztak K. Fetal environment, epigenetics, and pediatric renal disease. Pediatr Nephrol 2011; 26:705-11. [PMID: 21174217 PMCID: PMC3063864 DOI: 10.1007/s00467-010-1714-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Revised: 10/26/2010] [Accepted: 10/27/2010] [Indexed: 12/31/2022]
Abstract
The notion that some adult diseases may have their origins in utero has recently captured scientists' attention. Some of these effects persist across generations and may involve epigenetic mechanisms. Epigenetic modifications, DNA methylation together with covalent modifications of histones, alter chromatin density and accessibility of DNA to cellular machinery, modulating the transcriptional potential of the underlying DNA sequence. Here, we will discuss the different epigenetic modifications and their potential role in and contribution to renal disease development.
Collapse
Affiliation(s)
- Robert Woroniecki
- Department of Pediatrics, Nephrology, Albert Einstein College of Medicine, 1300 Morris Park Ave., Ullmann Bldg 619, Bronx, NY 10461, USA
| | - Anil Bhanudas Gaikwad
- Department of Medicine, Division of Nephrology, Albert Einstein College of Medicine, 1300 Morris Park Ave., Ullmann Bldg 619, Bronx, NY 10461, USA
| | - Katalin Susztak
- Department of Medicine, Division of Nephrology, Albert Einstein College of Medicine, 1300 Morris Park Ave., Ullmann Bldg 619, Bronx, NY 10461, USA, Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Ave., Ullmann Bldg 619, Bronx, NY 10461, USA
| |
Collapse
|
19
|
Moreno C, Hoffman M, Stodola TJ, Didier DN, Lazar J, Geurts AM, North PE, Jacob HJ, Greene AS. Creation and characterization of a renin knockout rat. Hypertension 2011; 57:614-9. [PMID: 21242461 PMCID: PMC3513323 DOI: 10.1161/hypertensionaha.110.163840] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Accepted: 12/15/2010] [Indexed: 12/14/2022]
Abstract
The renin-angiotensin system plays an important role in the control of blood pressure (BP) and renal function. To illuminate the importance of renin in the context of a disease background in vivo, we used zinc-finger nucleases (ZFNs) designed to target the renin gene and create a renin knockout in the SS/JrHsdMcwi (SS) rat. ZFN against renin caused a 10-bp deletion in exon 5, resulting in a frameshift mutation. Plasma renin activity was undetectable in the Ren-/- rat, and renin protein was absent from the juxtaglomerular cells in the kidney. Body weight was lower in the Ren-/- rats (than in the Ren+/- or wild-type littermates), and conscious BP on low-salt diet (0.4% NaCl) was 58 ± 2 mm Hg in the Ren-/- male rats versus 117 mm Hg in the Ren+/- littermates, a reduction of almost 50 mm Hg. Blood urea nitrogen (BUN) and plasma creatinine levels were elevated in the Ren-/- strain (BUN 112 ± 7 versus 23 ± 2 mg/dL and creatinine 0.53 ± 0.02 versus 0.26 ± 0.02 mg/dL), and kidney morphology was abnormal with a rudimentary inner renal medulla, cortical interstitial fibrosis, thickening of arterial walls, and abnormally shaped glomeruli. The development of the first rat knockout in the renin-angiotensin system demonstrates the efficacy of the ZFN technology for creating knockout rats for cardiovascular disease on any genetic background and emphasizes the role of renin in BP regulation and kidney function even in the low-renin SS rat.
Collapse
Affiliation(s)
- Carol Moreno
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Gross KW, Gomez RA, Sigmund CD. Twists and turns in the search for the elusive renin processing enzyme: focus on "Cathepsin B is not the processing enzyme for mouse prorenin". Am J Physiol Regul Integr Comp Physiol 2010; 298:R1209-11. [PMID: 20237305 DOI: 10.1152/ajpregu.00188.2010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
21
|
Sequeira-Lopez MLS, Weatherford ET, Borges GR, Monteagudo MC, Pentz ES, Harfe BD, Carretero O, Sigmund CD, Gomez RA. The microRNA-processing enzyme dicer maintains juxtaglomerular cells. J Am Soc Nephrol 2010; 21:460-7. [PMID: 20056748 PMCID: PMC2831866 DOI: 10.1681/asn.2009090964] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2009] [Indexed: 11/03/2022] Open
Abstract
Juxtaglomerular cells are highly specialized myoepithelioid granulated cells located in the glomerular afferent arterioles. These cells synthesize and release renin, which distinguishes them from other cells. How these cells maintain their identity, restricted localization, and fate is unknown and is fundamental to the control of BP and homeostasis of fluid and electrolytes. Because microRNAs may control cell fate via temporal and spatial gene regulation, we generated mice with a conditional deletion of Dicer, the RNase III endonuclease that produces mature microRNAs in cells of the renin lineage. Deletion of Dicer severely reduced the number of juxtaglomerular cells, decreased expression of the renin genes (Ren1 and Ren2), lowered plasma renin concentration, and decreased BP. As a consequence of the disappearance of renin-producing cells, the kidneys developed striking vascular abnormalities and prominent striped fibrosis. We conclude that microRNAs maintain the renin-producing juxtaglomerular cells and the morphologic integrity and function of the kidney.
Collapse
Affiliation(s)
| | - Eric T. Weatherford
- Departments of Internal Medicine and Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
| | - Giulianna R. Borges
- Departments of Internal Medicine and Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
| | - Maria C. Monteagudo
- *Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Ellen S. Pentz
- *Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Brian D. Harfe
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, Florida; and
| | - Oscar Carretero
- Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Curt D. Sigmund
- Departments of Internal Medicine and Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
| | - R. Ariel Gomez
- *Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia
| |
Collapse
|
22
|
Wang CH, Li F, Takahashi N. The renin angiotensin system and the metabolic syndrome. THE OPEN HYPERTENSION JOURNAL 2010; 3:1-13. [PMID: 21132096 PMCID: PMC2995894 DOI: 10.2174/1876526203010001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The renin angiotensin system (RAS) is important for fluid and blood pressure regulation. Recent studies suggest that an overactive RAS is involved in the metabolic syndrome. This article discusses recent advances on how genetic alteration of the RAS affects cardiovascular and metabolic phenotypes, with a special emphasis on the potential role of angiotensin-independent effects of renin.
Collapse
Affiliation(s)
- Chih-Hong Wang
- Department of Pathology and Laboratory Medicine The University of North Carolina at Chapel Hill Chapel Hill, NC 27599-7525
| | - Feng Li
- Department of Pathology and Laboratory Medicine The University of North Carolina at Chapel Hill Chapel Hill, NC 27599-7525
| | - Nobuyuki Takahashi
- Department of Pathology and Laboratory Medicine The University of North Carolina at Chapel Hill Chapel Hill, NC 27599-7525
- Tohoku University, Graduate Schools of Pharmaceutical Sciences and Medicine, Sendai, 980-8578, Japan
| |
Collapse
|
23
|
Casellas D. Methods for imaging Renin-synthesizing, -storing, and -secreting cells. Int J Hypertens 2009; 2010:298747. [PMID: 20948562 PMCID: PMC2949082 DOI: 10.4061/2010/298747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Revised: 07/07/2009] [Accepted: 09/08/2009] [Indexed: 12/04/2022] Open
Abstract
Renin-producing cells have been the object of intense research efforts for the past fifty years within the field of hypertension. Two decades ago, research focused on the concept and characterization of the intrarenal renin-angiotensin system. Early morphological studies led to the concept of the juxtaglomerular apparatus, a minute organ that links tubulovascular structures and function at the single nephron level. The kidney, thus, appears as a highly "topological organ" in which anatomy and function are intimately linked. This point is reflected by a concurrent and constant development of functional and structural approaches. After summarizing our current knowledge about renin cells and their distribution along the renal vascular tree, particularly along glomerular afferent arterioles, we reviewed a variety of imaging techniques that permit a fine characterization of renin synthesis, storage, and release at the single-arteriolar, -cell, or -granule level. Powerful tools such as multiphoton microscopy and transgenesis bear the promises of future developments of the field.
Collapse
Affiliation(s)
- Daniel Casellas
- Groupe Rein et Hypertension (EA3127), Institut Universitaire de Recherche Clinique, 641 Avenue du Doyen Giraud, 34093 Montpellier Cédex 5, France
| |
Collapse
|
24
|
Gomez RA, Pentz ES, Jin X, Cordaillat M, Sequeira Lopez MLS. CBP and p300 are essential for renin cell identity and morphological integrity of the kidney. Am J Physiol Heart Circ Physiol 2009; 296:H1255-62. [PMID: 19252086 PMCID: PMC2685333 DOI: 10.1152/ajpheart.01266.2008] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2008] [Accepted: 02/24/2009] [Indexed: 01/05/2023]
Abstract
The mechanisms that govern the identity of renin cells are not well understood. We and others have identified cAMP as an important pathway in the regulation of renin synthesis and release. Recently, experiments in cells from the renin lineage led us to propose that acquisition and maintenance of renin cell identity are mediated by cAMP and histone acetylation at the cAMP responsive element (CRE) of the renin gene. Ultimately, the transcriptional effects of cAMP depend on binding of the appropriate transcription factors to CRE. It has been suggested that access of transcription factors to this region of the promoter is facilitated by the coactivators CREB-binding protein (CBP) and p300, which possess histone acetyltransferase activity and may be, in turn, responsible for the remodeling of chromatin underlying expression of the renin gene. We hypothesized that CBP and p300 are therefore required for expression of the renin gene and maintenance of the renin cell. Because mice homozygous for the deletion of CBP or p300 die before kidney organogenesis begins, no data on kidney or juxtaglomerular cell development in these mice are available. Therefore, to define the role of these histone acetyltransferases in renin cell identity in vivo, we used a conditional deletion approach, in which floxed CBP and p300 mice were crossed with mice expressing cre recombinase in renin cells. Results show that the histone acetyltransferases CBP and p300 are necessary for maintenance of renin cell identity and structural integrity of the kidney.
Collapse
Affiliation(s)
- R Ariel Gomez
- Dept. of Pediatrics, Univ. of Virginia School of Medicine, 409 Lane Rd., MR4 2001, Charlottesville, VA 22908, USA.
| | | | | | | | | |
Collapse
|
25
|
Pentz ES, Lopez MLSS, Cordaillat M, Gomez RA. Identity of the renin cell is mediated by cAMP and chromatin remodeling: an in vitro model for studying cell recruitment and plasticity. Am J Physiol Heart Circ Physiol 2007; 294:H699-707. [PMID: 18055510 DOI: 10.1152/ajpheart.01152.2007] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The renin-angiotensin system (RAS) regulates blood pressure and fluid-electrolyte homeostasis. A key step in the RAS cascade is the regulation of renin synthesis and release by the kidney. We and others have shown that a major mechanism to control renin availability is the regulation of the number of cells capable of making renin. The kidney possesses a pool of cells, mainly in its vasculature but also in the glomeruli, capable of switching from smooth muscle to endocrine renin-producing cells when homeostasis is threatened. The molecular mechanisms governing the ability of these cells to turn the renin phenotype on and off have been very difficult to study in vivo. We, therefore, developed an in vitro model in which cells of the renin lineage are labeled with cyan fluorescent protein and cells actively making renin mRNA are labeled with yellow fluorescent protein. The model allowed us to determine that it is possible to culture cells of the renin lineage for numerous passages and that the memory to express the renin gene is maintained in culture and can be reenacted by cAMP and chromatin remodeling (histone H4 acetylation) at the cAMP-responsive element in the renin gene.
Collapse
Affiliation(s)
- Ellen Steward Pentz
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | | | | | | |
Collapse
|
26
|
Chen L, Kim SM, Oppermann M, Faulhaber-Walter R, Huang Y, Mizel D, Chen M, Lopez MLS, Weinstein LS, Gomez RA, Briggs JP, Schnermann J. Regulation of renin in mice with Cre recombinase-mediated deletion of G protein Gsα in juxtaglomerular cells. Am J Physiol Renal Physiol 2007; 292:F27-37. [PMID: 16822937 DOI: 10.1152/ajprenal.00193.2006] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
By crossing mice with expression of Cre recombinase under control of the endogenous renin promoter (Sequeira Lopez ML, Pentz ES, Nomasa T, Smithies O, Gomez RA. Dev Cell 6: 719–728, 2004) with mice in which exon 1 of the Gnas gene was flanked by loxP sites (Chen M, Gavrilova O, Liu J, Xie T, Deng C, Nguyen AT, Nackers LM, Lorenzo J, Shen L, Weinstein LS. Proc Natl Acad Sci USA), we generated animals with preferential and nearly complete excision of Gsα in juxtaglomerular granular (JG) cells. Compared with wild-type animals, mice with conditional Gsα deficiency had markedly reduced basal levels of renin expression and very low plasma renin concentrations. Furthermore, the acute release responses to furosemide, hydralazine, and isoproterenol were virtually abolished. Consistent with a state of primary renin depletion, Gsα-deficient mice had reduced arterial blood pressure, reduced levels of aldosterone, and a low glomerular filtration rate. Renin content and renin secretion of JG cells in primary culture were drastically reduced, and the stimulatory response to the addition of PGE2or isoproterenol was eliminated. Unexpectedly, Gsα recombination was also observed in the renal medulla, and this was associated with a vasopressin-resistant concentrating defect. Our study shows that Cre recombinase under control of the renin promoter can be used for the excision of floxed targets from JG cells. We conclude that Gsα-mediated signal transduction is essential and nonredundant in the control of renin synthesis and release.
Collapse
Affiliation(s)
- Limeng Chen
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892-1370, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Castrop H, Oppermann M, Weiss Y, Huang Y, Mizel D, Lu H, Germain S, Schweda F, Theilig F, Bachmann S, Briggs J, Kurtz A, Schnermann J. Reporter gene recombination in juxtaglomerular granular and collecting duct cells by human renin promoter-Cre recombinase transgene. Physiol Genomics 2006; 25:277-85. [PMID: 16418317 DOI: 10.1152/physiolgenomics.00302.2005] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
To assess the feasibility of using the renin promoter for expressing Cre recombinase in juxtaglomerular (JG) cells only, we generated five independent transgenic mouse lines (designated hRen-Cre) expressing Cre recombinase under control of a 12.2-kb human renin promoter. In the kidneys of adult mice Cre mRNA (RT-PCR) was found in the renal cortex, with Cre protein (immunohistochemistry) being localized in afferent arterioles and to a lower degree in interlobular arteries. Cre mRNA levels were regulated in a renin-typical fashion by changes in oral salt intake, water restriction, or isoproterenol infusion, indicating the presence of key regulatory elements within 12.2 kb of the 5′-flanking region of the human renin gene. hRen-Cre mice were interbred with both the ROSA26-EGFP and ROSA26-lacZ reporter strains to assess renin promoter activity from Cre-mediated excision of a floxed stop cassette and subsequent enhanced green fluorescent protein (EGFP) and β-galactosidase (β-gal) detection. In adult mice, β-gal staining and EGFP were observed in afferent arterioles and interlobular arteries, overlapping with Cre protein expression. In addition, intense β-gal staining was found in cortical and medullary collecting ducts where Cre expression was minimal. In embryonic kidneys, β-gal staining was detected in the developing collecting duct system beginning at embryonic day 12, showing substantial activity of the human renin promoter in the branching ureteric bud. Our data indicate that besides its well-known activity in JG cells and renal vessels the human renin promoter is transiently active in the collecting duct system during kidney development, complicating the use of this approach for JG cell-specific excision of floxed targets.
Collapse
Affiliation(s)
- H Castrop
- Institute of Physiology, University of Regensburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Prieto-Carrasquero MC, Kobori H, Ozawa Y, Gutiérrez A, Seth D, Navar LG. AT1 receptor-mediated enhancement of collecting duct renin in angiotensin II-dependent hypertensive rats. Am J Physiol Renal Physiol 2005; 289:F632-7. [PMID: 15870381 PMCID: PMC2040262 DOI: 10.1152/ajprenal.00462.2004] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Angiotensin II (ANG II)-infused rats exhibit increases in distal nephron renin expressed in principal cells of connecting tubules and collecting ducts. This study was performed to determine whether the augmentation of distal nephron renin involves ANG II type 1 (AT1) receptor activation. Male Sprague-Dawley rats (200-220 g) were divided into three groups: 1) sham operated (n = 8); 2) ANG II infused (80 ng/min, 13 days, n = 8); and 3) ANG II infused plus AT1 receptor blocker (ARB), olmesartan (5 mg/days, n = 8). ANG II infusion increased systolic blood pressure (BP; 178 +/- 4 vs. 122 +/- 1 mmHg; P < 0.001) and suppressed plasma renin activity (PRA; 0.08 +/- 0.1 vs. 5.3 +/- 0.8 ng ANG I x ml(-1) x h(-1)). ARB treatment prevented the increase in BP (113 +/- 6 mmHg) and led to increases in PRA (15.8 +/- 1.5 ng ANG I x ml(-1) x h(-1)). Renin protein levels measured in the kidney medulla, to avoid contribution from juxtaglomerular apparatus cells, were higher in ANG II-infused rats [1.64 +/- 0.3 vs. 1.00 +/- 0.1 densitometric units (DU) compared with sham-operated rats; P < 0.05], and ARB treatment prevented this increase (1.01 +/- 0.1). Similarly, renin immunoreactivity increased in medullary collecting ducts of ANG II-infused compared with sham-operated rats (2.5 +/- 0.3 vs. 1.0 +/- 0.2 DU; P < 0.001), which was also prevented by ARB (1.01 +/- 0.06). Renin qRTPCR in ANG II-infused rats showed higher mRNA levels in the kidney medulla compared with sham-operated rats (5.5 +/- 2.3 vs. 0.04 +/- 0.02 ratio to GAPDH mRNA levels; P < 0.001); however, renin transcript levels were normalized in the ARB-treated rats. These data demonstrate that the augmentation of distal nephron renin in ANG II-infused hypertensive rats is AT1 receptor mediated. The augmented distal tubular renin may contribute to increased intratubular ANG II levels and distal nephron sodium reabsorption in ANG II-dependent hypertension.
Collapse
Affiliation(s)
- Minolfa C Prieto-Carrasquero
- Dept. of Physiology and Hypertension, Tulane Univ. Health Sciences Center, 1430 Tulane Ave., New Orleans, LA 70112-2699, USA.
| | | | | | | | | | | |
Collapse
|
29
|
Takahashi N, Lopez MLSS, Cowhig JE, Taylor MA, Hatada T, Riggs E, Lee G, Gomez RA, Kim HS, Smithies O. Ren1cHomozygous Null Mice Are Hypotensive and Polyuric, but Heterozygotes Are Indistinguishable from Wild-Type. J Am Soc Nephrol 2004; 16:125-32. [PMID: 15563565 DOI: 10.1681/asn.2004060490] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Mice lacking Ren1c were generated using C57BL/6-derived embryonic stem cells. Mice homozygous for Ren1c disruption (Ren1c-/-) are born at the expected ratio, but approximately 80% die of dehydration within a few days. The surviving Ren1c-/- mice have no renin mRNA expression in the kidney, hydronephrosis, thickening of renal arterial walls, and fibrosis in the kidney. Plasma renin and angiotensins I and II are undetectable. Urinary aldosterone is 6% wild-type. They have low tail-cuff BP (84 +/- 4 versus 116 +/- 5 mmHg in +/+) and excrete large amounts of urine (5.2 +/- 0.8 ml/d, 725 +/- 34 mOsm versus 1.1 +/- 0.1 ml/d, 2460 +/- 170 mOsm in +/+). After 5 d of drinking 5% dextrose, desmopressin does not increase the osmolality of the urine in -/- mice (624 +/- 19 to 656 +/- 25 mOsm), whereas in +/+, it increases severalfold (583 +/- 44 to 2630 +/- 174 mOsm). Minipump infusion of angiotensin II to Ren1c-/- mice restores BP to wild-type level, but preexisting damage to the medulla prevents complete restoration of the ability of the kidney to concentrate urine. Heterozygous Ren1c+/- mice, in contrast, are indistinguishable from +/+ in BP, urine volume, and osmolality. Kidney renin mRNA, the number of kidney cells producing renin, and plasma renin concentration in the Ren1c+/- mice are also indistinguishable from +/+. These results demonstrate that renin is the only enzyme capable of maintaining plasma angiotensins and that renin expression in the kidney is very tightly regulated at the mRNA level.
Collapse
Affiliation(s)
- Nobuyuki Takahashi
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 701 Brinkhous-Bullitt Building, Chapel Hill, NC 27599-7525, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Hansen PB, Yang T, Huang Y, Mizel D, Briggs J, Schnermann J. Plasma renin in mice with one or two renin genes. ACTA ACUST UNITED AC 2004; 181:431-7. [PMID: 15283755 DOI: 10.1111/j.1365-201x.2004.01315.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
AIM In the present study we have investigated whether the presence of a second renin gene exerts an overriding influence on plasma renin such that mice with two renin genes have consistently higher renin levels than mice with only one renin gene. METHODS Plasma renin was determined as the rate of angiotensin I generation using a radioimmunoassay (RIA) kit with (plasma renin concentration, PRC) or without (plasma renin activity, PRA) the addition of purified rat angiotensinogen as substrate. RESULTS In male 129SvJ, DBA/2 and Swiss Webster mice, strains possessing both Ren-1 and Ren-2, PRC (ng Ang I mL(-1) h(-1)) averaged 178 +/- 36, 563 +/- 57 and 550 +/- 43 while PRA was 2.9 +/- 0.5, 3.6 +/- 0.8 and 7.8 +/- 1.2. In male C57BL/6, C3H and BALB/c mice that express only Ren-1, PRC averaged 426 +/- 133, 917 +/- 105 and 315 +/- 72, and PRA was 3.4 +/- 1.0, 6.9 +/- 1.7 and 4.5 +/- 1.2. In the two renin gene A1AR-/- mice compared with the one renin gene A1AR+/+, PRC averaged 538 +/- 321 and 415 +/- 159 while PRA averaged 3.2 +/- 1.1 and 4.4 +/- 1.4 ng Ang I mL(-1) h(-1). Aldosterone levels showed no significant differences between one renin (C57BL/6, C3H and BALB/c) and two renin (129SvJ, DBA/2 and Swiss Webster) gene mice. Furthermore, by quantitative real-time polymerase chain reaction (RT-PCR) we found no correlation between the number of renin genes and whole kidney renin mRNA levels from one and two renin gene mice. CONCLUSION Our data show that baseline plasma renin is not systematically higher in mice with two renin genes than in one renin gene mice. Thus, the presence of a second renin gene does not seem to be a major determinant of differences in PRC between different mouse strains.
Collapse
Affiliation(s)
- P B Hansen
- National Institute of Diabetes, and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | | | | | | | | | | |
Collapse
|
31
|
Sequeira López MLS, Pentz ES, Nomasa T, Smithies O, Gomez RA. Renin cells are precursors for multiple cell types that switch to the renin phenotype when homeostasis is threatened. Dev Cell 2004; 6:719-28. [PMID: 15130496 DOI: 10.1016/s1534-5807(04)00134-0] [Citation(s) in RCA: 219] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2003] [Revised: 03/08/2004] [Accepted: 03/11/2004] [Indexed: 01/05/2023]
Abstract
Renin-synthesizing cells are crucial in the regulation of blood pressure and fluid-electrolyte homeostasis. Adult mammals subjected to manipulations that threaten homeostasis increase circulating renin by increasing the number of renin-expressing/-releasing cells. We hypothesize that the ability of adult cells to synthesize renin does not occur randomly in any cell type, depending instead on the cell's lineage. To determine the fate of renin-expressing cells, we generated knockin mice expressing cre recombinase in renin-expressing cells and crossed them with reporter mice. Results show that renin-expressing cells are precursors for a variety of cells that differentiate into non-renin-expressing cells such as smooth-muscle, epithelial, mesangial, and extrarenal cells. In the kidney, these cells retain the capability to synthesize renin when additional hormone is required to reestablish homeostasis: specific subpopulations of apparently differentiated cells are "held in reserve" to respond (repeatedly) by de-differentiating and expressing renin in response to stress, and re-differentiating when the crisis passes.
Collapse
|
32
|
Pentz ES, Moyano MA, Thornhill BA, Sequeira Lopez MLS, Gomez RA. Ablation of renin-expressing juxtaglomerular cells results in a distinct kidney phenotype. Am J Physiol Regul Integr Comp Physiol 2003; 286:R474-83. [PMID: 14563659 DOI: 10.1152/ajpregu.00426.2003] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Renin-expressing cells are peculiar in that they act as differentiated cells, producing the hormone renin, while they also seem to act as progenitors for other renal cell types. As such, they may have functions independent of their ability to generate renin/angiotensin. To test this hypothesis, we ablated renin-expressing cells during development by placing diphtheria toxin A chain (DTA) under control of the Ren1d mouse renin promoter by homologous recombination in a two-renin gene strain (Ren2 and Ren1d). Renin-expressing cells are essentially absent from kidneys in homozygotes (DTA/DTA) which, unlike wild-type mice, are unable to recruit renin-expressing cells when homeostasis is threatened. In contrast, renin staining in the submandibular gland (SMG), which expresses mainly Ren2, is normal. Homozygous mice survive normally, but the kidneys are small and have morphological abnormalities: 25% of the glomeruli are hyperplastic or atrophic, tubules are dilated and atrophic, and areas of undifferentiated cells exist near the atrophic glomeruli and tubules. However, in contrast to the very abnormal renal vessels found when renin-angiotensin system genes are deleted, the kidney vessels in homozygotes have normal wall thickness and no decrease in lumen size. Homozygotes have severely reduced kidney and plasma renin concentrations and females have reduced blood pressure. Homozygotes have elevated blood urea nitrogen and potassium levels, which are suggestive of altered renal function. We conclude that renin cells per se are necessary for the morphological integrity of the kidney and may have a role in maintenance of normal kidney function.
Collapse
Affiliation(s)
- Ellen Steward Pentz
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA
| | | | | | | | | |
Collapse
|