51
|
Briet M, Barhoumi T, Mian MOR, Coelho SC, Ouerd S, Rautureau Y, Coffman TM, Paradis P, Schiffrin EL. Aldosterone-Induced Vascular Remodeling and Endothelial Dysfunction Require Functional Angiotensin Type 1a Receptors. Hypertension 2016; 67:897-905. [PMID: 27045029 DOI: 10.1161/hypertensionaha.115.07074] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 01/18/2016] [Indexed: 12/30/2022]
Abstract
We investigated the role of angiotensin type 1a receptors (AGTR1a) in vascular injury induced by aldosterone activation of mineralocorticoid receptors in Agtr1a(-/-) and wild-type (WT) mice infused with aldosterone for 14 days while receiving 1% NaCl in drinking water. Aldosterone increased systolic blood pressure (BP) by ≈30 mm Hg in WT mice and ≈50 mm Hg in Agtr1a(-/-) mice. Aldosterone induced aortic and small artery remodeling, impaired endothelium-dependent relaxation in WT mice, and enhanced fibronectin and collagen deposition and vascular inflammation. None of these vascular effects were observed in Agtr1a(-/-) mice. Aldosterone effects were prevented by the AGTR1 antagonist losartan in WT mice. In contrast to aldosterone, norepinephrine caused similar BP increase and mesenteric artery remodeling in WT and Agtr1a(-/-) mice. Agtr1a(-/-) mice infused with aldosterone did not increase sodium excretion in response to a sodium chloride challenge, suggesting that sodium retention could contribute to the exaggerated BP rise induced by aldosterone. Agtr1a(-/-) mice had decreased mesenteric artery expression of the calcium-activated potassium channel Kcnmb1, which may enhance myogenic tone and together with sodium retention, exacerbate BP responses to aldosterone/salt in Agtr1a(-/-) mice. We conclude that although aldosterone activation of mineralocorticoid receptors raises BP more in Agtr1a(-/-) mice, AGTR1a is required for mineralocorticoid receptor stimulation to induce vascular remodeling and inflammation and endothelial dysfunction.
Collapse
Affiliation(s)
- Marie Briet
- Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, PQ, Canada.,Department of Medicine, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, PQ, Canada.,Division of Nephrology (T.M.C.), Department of Medicine, Duke University, Durham, NC
| | - Tlili Barhoumi
- Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, PQ, Canada
| | - Muhammad Oneeb Rehman Mian
- Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, PQ, Canada
| | - Suellen C Coelho
- Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, PQ, Canada
| | - Sofiane Ouerd
- Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, PQ, Canada
| | - Yohann Rautureau
- Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, PQ, Canada
| | - Thomas M Coffman
- Division of Nephrology (T.M.C.), Department of Medicine, Duke University, Durham, NC
| | - Pierre Paradis
- Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, PQ, Canada
| | - Ernesto L Schiffrin
- Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, PQ, Canada.,Department of Medicine, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, PQ, Canada
| |
Collapse
|
52
|
Abstract
Although angiotensin II subtype-2 receptor (AT2R) was discovered over 2 decades ago, its contribution to physiology and pathophysiology is not fully elucidated. Current knowledge suggests that under normal physiologic conditions, AT2R counterbalances the effects of angiotensin II subtype-1 receptor (AT1R). A major obstacle for AT2R investigations was the lack of specific agonists. Most of the earlier AT2R studies were performed using the peptidic agonist, CG42112A, or the nonpeptidic antagonist PD123319. CGP42112A is nonspecific for AT2R and in higher concentrations can bind to AT1R. Recently, the development of specific nonpeptidic AT2R agonists boosted the efforts in identifying the therapeutic potentials for AT2R stimulation. Unlike AT1R, AT2R is involved in vasodilation by the release of bradykinin and nitric oxide, anti-inflammation, and healing from injury. Interestingly, the vasodilatory effects of AT2R stimulation were not associated with significant reduction in blood pressure. In the kidney, AT2R stimulation produced natriuresis, increased renal blood flow, and reduced tissue inflammation. In animal studies, enhanced AT2R function led to reduction of cardiac inflammation and fibrosis, and reduced the size of the infarcted area. Similarly, AT2R stimulation demonstrated protective effects in vasculature and brain.
Collapse
|
53
|
Lu H, Cassis LA, Kooi CWV, Daugherty A. Structure and functions of angiotensinogen. Hypertens Res 2016; 39:492-500. [PMID: 26888118 PMCID: PMC4935807 DOI: 10.1038/hr.2016.17] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 01/20/2016] [Accepted: 01/21/2016] [Indexed: 12/13/2022]
Abstract
Angiotensinogen (AGT) is the sole precursor of all angiotensin peptides. Although AGT is generally considered as a passive substrate of the renin-angiotensin system, there is accumulating evidence that the regulation and functions of AGT are intricate. Understanding the diversity of AGT properties has been enhanced by protein structural analysis and animal studies. In addition to whole-body genetic deletion, AGT can be regulated in vivo by cell-specific procedures, adeno-associated viral approaches and antisense oligonucleotides. Indeed, the availability of these multiple manipulations of AGT in vivo has provided new insights into the multifaceted roles of AGT. In this review, the combination of structural and functional studies is highlighted to focus on the increasing recognition that AGT exerts effects beyond being a sole provider of angiotensin peptides.
Collapse
Affiliation(s)
- Hong Lu
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA.,Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Lisa A Cassis
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Craig W Vander Kooi
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA
| | - Alan Daugherty
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA.,Department of Physiology, University of Kentucky, Lexington, KY, USA.,Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
54
|
Bernardi S, Michelli A, Zuolo G, Candido R, Fabris B. Update on RAAS Modulation for the Treatment of Diabetic Cardiovascular Disease. J Diabetes Res 2016; 2016:8917578. [PMID: 27652272 PMCID: PMC5019930 DOI: 10.1155/2016/8917578] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 07/27/2016] [Indexed: 02/07/2023] Open
Abstract
Since the advent of insulin, the improvements in diabetes detection and the therapies to treat hyperglycemia have reduced the mortality of acute metabolic emergencies, such that today chronic complications are the major cause of morbidity and mortality among diabetic patients. More than half of the mortality that is seen in the diabetic population can be ascribed to cardiovascular disease (CVD), which includes not only myocardial infarction due to premature atherosclerosis but also diabetic cardiomyopathy. The importance of renin-angiotensin-aldosterone system (RAAS) antagonism in the prevention of diabetic CVD has demonstrated the key role that the RAAS plays in diabetic CVD onset and development. Today, ACE inhibitors and angiotensin II receptor blockers represent the first line therapy for primary and secondary CVD prevention in patients with diabetes. Recent research has uncovered new dimensions of the RAAS and, therefore, new potential therapeutic targets against diabetic CVD. Here we describe the timeline of paradigm shifts in RAAS understanding, how diabetes modifies the RAAS, and what new parts of the RAAS pathway could be targeted in order to achieve RAAS modulation against diabetic CVD.
Collapse
Affiliation(s)
- Stella Bernardi
- Department of Medical Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, 34100 Trieste, Italy
- Division of Medicina Clinica, Azienda Sanitaria Universitaria Integrata di Trieste (ASUITS), Cattinara Teaching Hospital, Strada di Fiume, 34100 Trieste, Italy
- *Stella Bernardi:
| | - Andrea Michelli
- Department of Medical Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, 34100 Trieste, Italy
| | - Giulia Zuolo
- Department of Medical Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, 34100 Trieste, Italy
| | - Riccardo Candido
- Diabetes Centre, Azienda Sanitaria Universitaria Integrata di Trieste (ASUITS), Via Puccini, 34100 Trieste, Italy
| | - Bruno Fabris
- Department of Medical Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, 34100 Trieste, Italy
- Division of Medicina Clinica, Azienda Sanitaria Universitaria Integrata di Trieste (ASUITS), Cattinara Teaching Hospital, Strada di Fiume, 34100 Trieste, Italy
| |
Collapse
|
55
|
Lu H, Wu C, Howatt DA, Balakrishnan A, Moorleghen JJ, Chen X, Zhao M, Graham MJ, Mullick AE, Crooke RM, Feldman DL, Cassis LA, Vander Kooi CW, Daugherty A. Angiotensinogen Exerts Effects Independent of Angiotensin II. Arterioscler Thromb Vasc Biol 2015; 36:256-65. [PMID: 26681751 DOI: 10.1161/atvbaha.115.306740] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 12/03/2015] [Indexed: 01/16/2023]
Abstract
OBJECTIVE This study determined whether angiotensinogen (AGT) has angiotensin II-independent effects using multiple genetic and pharmacological manipulations. APPROACH AND RESULTS All study mice were in low-density lipoprotein receptor -/- background and fed a saturated fat-enriched diet. In mice with floxed alleles and a neomycin cassette in intron 2 of the AGT gene (hypoAGT mice), plasma AGT concentrations were >90% lower compared with their wild-type littermates. HypoAGT mice had lower systolic blood pressure, less atherosclerosis, and diminished body weight gain and liver steatosis. Low plasma AGT concentrations and all phenotypes were recapitulated in mice with hepatocyte-specific deficiency of AGT or pharmacological inhibition of AGT by antisense oligonucleotide administration. In contrast, inhibition of AGT cleavage by a renin inhibitor, aliskiren, failed to alter body weight gain and liver steatosis in low-density lipoprotein receptor -/- mice. In mice with established adiposity, administration of AGT antisense oligonucleotide versus aliskiren led to equivalent reductions of systolic blood pressure and atherosclerosis. AGT antisense oligonucleotide administration ceased body weight gain and further reduced body weight, whereas aliskiren did not affect body weight gain during continuous saturated fat-enriched diet feeding. Structural comparisons of AGT proteins in zebrafish, mouse, rat, and human revealed 4 highly conserved sequences within the des(angiotensin I)AGT domain. des(angiotensin I)AGT, through adeno-associated viral infection in hepatocyte-specific AGT-deficient mice, increased body weight gain and liver steatosis, but did not affect atherosclerosis. CONCLUSIONS AGT contributes to body weight gain and liver steatosis through functions of the des(angiotensin I)AGT domain, which are independent of angiotensin II production.
Collapse
Affiliation(s)
- Hong Lu
- From the Saha Cardiovascular Research Center (H.L., C.W., D.A.H., A.B., J.J.M., X.C., M.Z., A.D.); Departments of Physiology (H.L., A.D.), Pharmacology and Nutritional Sciences (C.W., L.A.C., A.D.), and Molecular and Cellular Biochemistry (C.W.V.K.), University of Kentucky, Lexington; Isis Pharmaceuticals, Inc, Carlsbad, CA (M.J.G., A.E.M., R.M.C.); and Novartis Pharmaceuticals Corporation, East Hanover, NJ (D.L.F.)
| | - Congqing Wu
- From the Saha Cardiovascular Research Center (H.L., C.W., D.A.H., A.B., J.J.M., X.C., M.Z., A.D.); Departments of Physiology (H.L., A.D.), Pharmacology and Nutritional Sciences (C.W., L.A.C., A.D.), and Molecular and Cellular Biochemistry (C.W.V.K.), University of Kentucky, Lexington; Isis Pharmaceuticals, Inc, Carlsbad, CA (M.J.G., A.E.M., R.M.C.); and Novartis Pharmaceuticals Corporation, East Hanover, NJ (D.L.F.)
| | - Deborah A Howatt
- From the Saha Cardiovascular Research Center (H.L., C.W., D.A.H., A.B., J.J.M., X.C., M.Z., A.D.); Departments of Physiology (H.L., A.D.), Pharmacology and Nutritional Sciences (C.W., L.A.C., A.D.), and Molecular and Cellular Biochemistry (C.W.V.K.), University of Kentucky, Lexington; Isis Pharmaceuticals, Inc, Carlsbad, CA (M.J.G., A.E.M., R.M.C.); and Novartis Pharmaceuticals Corporation, East Hanover, NJ (D.L.F.)
| | - Anju Balakrishnan
- From the Saha Cardiovascular Research Center (H.L., C.W., D.A.H., A.B., J.J.M., X.C., M.Z., A.D.); Departments of Physiology (H.L., A.D.), Pharmacology and Nutritional Sciences (C.W., L.A.C., A.D.), and Molecular and Cellular Biochemistry (C.W.V.K.), University of Kentucky, Lexington; Isis Pharmaceuticals, Inc, Carlsbad, CA (M.J.G., A.E.M., R.M.C.); and Novartis Pharmaceuticals Corporation, East Hanover, NJ (D.L.F.)
| | - Jessica J Moorleghen
- From the Saha Cardiovascular Research Center (H.L., C.W., D.A.H., A.B., J.J.M., X.C., M.Z., A.D.); Departments of Physiology (H.L., A.D.), Pharmacology and Nutritional Sciences (C.W., L.A.C., A.D.), and Molecular and Cellular Biochemistry (C.W.V.K.), University of Kentucky, Lexington; Isis Pharmaceuticals, Inc, Carlsbad, CA (M.J.G., A.E.M., R.M.C.); and Novartis Pharmaceuticals Corporation, East Hanover, NJ (D.L.F.)
| | - Xiaofeng Chen
- From the Saha Cardiovascular Research Center (H.L., C.W., D.A.H., A.B., J.J.M., X.C., M.Z., A.D.); Departments of Physiology (H.L., A.D.), Pharmacology and Nutritional Sciences (C.W., L.A.C., A.D.), and Molecular and Cellular Biochemistry (C.W.V.K.), University of Kentucky, Lexington; Isis Pharmaceuticals, Inc, Carlsbad, CA (M.J.G., A.E.M., R.M.C.); and Novartis Pharmaceuticals Corporation, East Hanover, NJ (D.L.F.)
| | - Mingming Zhao
- From the Saha Cardiovascular Research Center (H.L., C.W., D.A.H., A.B., J.J.M., X.C., M.Z., A.D.); Departments of Physiology (H.L., A.D.), Pharmacology and Nutritional Sciences (C.W., L.A.C., A.D.), and Molecular and Cellular Biochemistry (C.W.V.K.), University of Kentucky, Lexington; Isis Pharmaceuticals, Inc, Carlsbad, CA (M.J.G., A.E.M., R.M.C.); and Novartis Pharmaceuticals Corporation, East Hanover, NJ (D.L.F.)
| | - Mark J Graham
- From the Saha Cardiovascular Research Center (H.L., C.W., D.A.H., A.B., J.J.M., X.C., M.Z., A.D.); Departments of Physiology (H.L., A.D.), Pharmacology and Nutritional Sciences (C.W., L.A.C., A.D.), and Molecular and Cellular Biochemistry (C.W.V.K.), University of Kentucky, Lexington; Isis Pharmaceuticals, Inc, Carlsbad, CA (M.J.G., A.E.M., R.M.C.); and Novartis Pharmaceuticals Corporation, East Hanover, NJ (D.L.F.)
| | - Adam E Mullick
- From the Saha Cardiovascular Research Center (H.L., C.W., D.A.H., A.B., J.J.M., X.C., M.Z., A.D.); Departments of Physiology (H.L., A.D.), Pharmacology and Nutritional Sciences (C.W., L.A.C., A.D.), and Molecular and Cellular Biochemistry (C.W.V.K.), University of Kentucky, Lexington; Isis Pharmaceuticals, Inc, Carlsbad, CA (M.J.G., A.E.M., R.M.C.); and Novartis Pharmaceuticals Corporation, East Hanover, NJ (D.L.F.)
| | - Rosanne M Crooke
- From the Saha Cardiovascular Research Center (H.L., C.W., D.A.H., A.B., J.J.M., X.C., M.Z., A.D.); Departments of Physiology (H.L., A.D.), Pharmacology and Nutritional Sciences (C.W., L.A.C., A.D.), and Molecular and Cellular Biochemistry (C.W.V.K.), University of Kentucky, Lexington; Isis Pharmaceuticals, Inc, Carlsbad, CA (M.J.G., A.E.M., R.M.C.); and Novartis Pharmaceuticals Corporation, East Hanover, NJ (D.L.F.)
| | - David L Feldman
- From the Saha Cardiovascular Research Center (H.L., C.W., D.A.H., A.B., J.J.M., X.C., M.Z., A.D.); Departments of Physiology (H.L., A.D.), Pharmacology and Nutritional Sciences (C.W., L.A.C., A.D.), and Molecular and Cellular Biochemistry (C.W.V.K.), University of Kentucky, Lexington; Isis Pharmaceuticals, Inc, Carlsbad, CA (M.J.G., A.E.M., R.M.C.); and Novartis Pharmaceuticals Corporation, East Hanover, NJ (D.L.F.)
| | - Lisa A Cassis
- From the Saha Cardiovascular Research Center (H.L., C.W., D.A.H., A.B., J.J.M., X.C., M.Z., A.D.); Departments of Physiology (H.L., A.D.), Pharmacology and Nutritional Sciences (C.W., L.A.C., A.D.), and Molecular and Cellular Biochemistry (C.W.V.K.), University of Kentucky, Lexington; Isis Pharmaceuticals, Inc, Carlsbad, CA (M.J.G., A.E.M., R.M.C.); and Novartis Pharmaceuticals Corporation, East Hanover, NJ (D.L.F.)
| | - Craig W Vander Kooi
- From the Saha Cardiovascular Research Center (H.L., C.W., D.A.H., A.B., J.J.M., X.C., M.Z., A.D.); Departments of Physiology (H.L., A.D.), Pharmacology and Nutritional Sciences (C.W., L.A.C., A.D.), and Molecular and Cellular Biochemistry (C.W.V.K.), University of Kentucky, Lexington; Isis Pharmaceuticals, Inc, Carlsbad, CA (M.J.G., A.E.M., R.M.C.); and Novartis Pharmaceuticals Corporation, East Hanover, NJ (D.L.F.)
| | - Alan Daugherty
- From the Saha Cardiovascular Research Center (H.L., C.W., D.A.H., A.B., J.J.M., X.C., M.Z., A.D.); Departments of Physiology (H.L., A.D.), Pharmacology and Nutritional Sciences (C.W., L.A.C., A.D.), and Molecular and Cellular Biochemistry (C.W.V.K.), University of Kentucky, Lexington; Isis Pharmaceuticals, Inc, Carlsbad, CA (M.J.G., A.E.M., R.M.C.); and Novartis Pharmaceuticals Corporation, East Hanover, NJ (D.L.F.).
| |
Collapse
|
56
|
|
57
|
Karnik SS, Unal H, Kemp JR, Tirupula KC, Eguchi S, Vanderheyden PML, Thomas WG. International Union of Basic and Clinical Pharmacology. XCIX. Angiotensin Receptors: Interpreters of Pathophysiological Angiotensinergic Stimuli [corrected]. Pharmacol Rev 2015; 67:754-819. [PMID: 26315714 PMCID: PMC4630565 DOI: 10.1124/pr.114.010454] [Citation(s) in RCA: 225] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The renin angiotensin system (RAS) produced hormone peptides regulate many vital body functions. Dysfunctional signaling by receptors for RAS peptides leads to pathologic states. Nearly half of humanity today would likely benefit from modern drugs targeting these receptors. The receptors for RAS peptides consist of three G-protein-coupled receptors—the angiotensin II type 1 receptor (AT1 receptor), the angiotensin II type 2 receptor (AT2 receptor), the MAS receptor—and a type II trans-membrane zinc protein—the candidate angiotensin IV receptor (AngIV binding site). The prorenin receptor is a relatively new contender for consideration, but is not included here because the role of prorenin receptor as an independent endocrine mediator is presently unclear. The full spectrum of biologic characteristics of these receptors is still evolving, but there is evidence establishing unique roles of each receptor in cardiovascular, hemodynamic, neurologic, renal, and endothelial functions, as well as in cell proliferation, survival, matrix-cell interaction, and inflammation. Therapeutic agents targeted to these receptors are either in active use in clinical intervention of major common diseases or under evaluation for repurposing in many other disorders. Broad-spectrum influence these receptors produce in complex pathophysiological context in our body highlights their role as precise interpreters of distinctive angiotensinergic peptide cues. This review article summarizes findings published in the last 15 years on the structure, pharmacology, signaling, physiology, and disease states related to angiotensin receptors. We also discuss the challenges the pharmacologist presently faces in formally accepting newer members as established angiotensin receptors and emphasize necessary future developments.
Collapse
Affiliation(s)
- Sadashiva S Karnik
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Hamiyet Unal
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Jacqueline R Kemp
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Kalyan C Tirupula
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Satoru Eguchi
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Patrick M L Vanderheyden
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Walter G Thomas
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| |
Collapse
|
58
|
Angiotensin II receptor blockade promotes repair of skeletal muscle through down-regulation of aging-promoting C1q expression. Sci Rep 2015; 5:14453. [PMID: 26571361 PMCID: PMC4585890 DOI: 10.1038/srep14453] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 08/24/2015] [Indexed: 01/31/2023] Open
Abstract
Disruption of angiotensin II type 1 (AT1) receptor prolonged life span in mice. Since aging-related decline in skeletal muscle function was retarded in Atgr1a−/− mice, we examined the role of AT1 receptor in muscle regeneration after injury. Administration of AT1 receptor blocker irbesartan increased the size of regenerating myofibers, decreased fibrosis, and enhanced functional muscle recovery after cryoinjury. We recently reported that complement C1q, secreted by macrophages, activated Wnt/β-catenin signaling and promoted aging-related decline in regenerative capacity of skeletal muscle. Notably, irbesartan induced M2 polarization of macrophages, but reduced C1q expression in cryoinjured muscles and in cultured macrophage cells. Irbesartan inhibited up-regulation of Axin2, a downstream gene of Wnt/β-catenin pathway, in cryoinjured muscles. In addition, topical administration of C1q reversed beneficial effects of irbesartan on skeletal muscle regeneration after injury. These results suggest that AT1 receptor blockade improves muscle repair and regeneration through down-regulation of the aging-promoting C1q-Wnt/β-catenin signaling pathway.
Collapse
|
59
|
Ramkumar N, Stuart D, Calquin M, Quadri S, Wang S, Van Hoek AN, Siragy HM, Ichihara A, Kohan DE. Nephron-specific deletion of the prorenin receptor causes a urine concentration defect. Am J Physiol Renal Physiol 2015; 309:F48-56. [PMID: 25995108 DOI: 10.1152/ajprenal.00126.2015] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 05/11/2015] [Indexed: 01/27/2023] Open
Abstract
The prorenin receptor (PRR), a recently discovered component of the renin-angiotensin system, is expressed in the nephron in general and the collecting duct in particular. However, the physiological significance of nephron PRR remains unclear, partly due to developmental abnormalities associated with global or renal-specific PRR gene knockout (KO). Therefore, we developed mice with inducible nephron-wide PRR deletion using Pax8-reverse tetracycline transactivator and LC-1 transgenes and loxP flanked PRR alleles such that ablation of PRR occurs in adulthood, after induction with doxycycline. Nephron-specific PRR KO mice have normal survival to ∼1 yr of age and no renal histological defects. Compared with control mice, PRR KO mice had 65% lower medullary PRR mRNA and protein levels and markedly diminished renal PRR immunofluorescence. During both normal water intake and mild water restriction, PRR KO mice had significantly lower urine osmolality, higher water intake, and higher urine volume compared with control mice. No differences were seen in urine vasopressin excretion, urine Na(+) and K(+) excretion, plasma Na(+), or plasma osmolality between the two groups. However, PRR KO mice had reduced medullary aquaporin-2 levels and arginine vasopressin-stimulated cAMP accumulation in the isolated renal medulla compared with control mice. Taken together, these results suggest nephron PRR can potentially modulate renal water excretion.
Collapse
Affiliation(s)
- Nirupama Ramkumar
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center, Salt Lake City, Utah;
| | - Deborah Stuart
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Matias Calquin
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Syed Quadri
- Department of Medicine, University of Virginia Health System, Charlottesville, Virginia; and
| | - Shuping Wang
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Alfred N Van Hoek
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Helmy M Siragy
- Department of Medicine, University of Virginia Health System, Charlottesville, Virginia; and
| | - Atsuhiro Ichihara
- Department of Medicine II, Endocrinology and Hypertension, Tokyo Women's Medical University, Tokyo, Japan
| | - Donald E Kohan
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center, Salt Lake City, Utah
| |
Collapse
|
60
|
Kamo T, Akazawa H, Komuro I. Pleiotropic Effects of Angiotensin II Receptor Signaling in Cardiovascular Homeostasis and Aging. Int Heart J 2015; 56:249-54. [PMID: 25912907 DOI: 10.1536/ihj.14-429] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Most of the pathophysiological actions of angiotensin II (Ang II) are mediated through the Ang II type 1 (AT1) receptor, a member of the seven-transmembrane G protein-coupled receptor family. Essentially, AT1 receptor signaling is beneficial for organismal survival and procreation, because it is crucial for normal organ development, and blood pressure and electrolyte homeostasis. On the other hand, AT1 receptor signaling has detrimental effects, such as promoting various aging-related diseases that include cardiovascular diseases, diabetes, chronic kidney disease, dementia, osteoporosis, and cancer. Pharmacological or genetic blockade of AT1 receptor signaling in rodents has been shown to prevent the progression of aging-related phenotypes and promote longevity. In this way, AT1 receptor signaling exerts antagonistic and pleiotropic effects according to the ages and pathophysiological conditions. Here we review the pleiotropic effects of AT1 receptor signaling in cardiovascular homeostasis and aging.
Collapse
Affiliation(s)
- Takehiro Kamo
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| | | | | |
Collapse
|
61
|
Sparks MA, Stegbauer J, Chen D, Gomez JA, Griffiths RC, Azad HA, Herrera M, Gurley SB, Coffman TM. Vascular Type 1A Angiotensin II Receptors Control BP by Regulating Renal Blood Flow and Urinary Sodium Excretion. J Am Soc Nephrol 2015; 26:2953-62. [PMID: 25855778 DOI: 10.1681/asn.2014080816] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 01/27/2015] [Indexed: 01/10/2023] Open
Abstract
Inappropriate activation of the type 1A angiotensin (AT1A) receptor contributes to the pathogenesis of hypertension and its associated complications. To define the role for actions of vascular AT1A receptors in BP regulation and hypertension pathogenesis, we generated mice with cell-specific deletion of AT1A receptors in smooth muscle cells (SMKO mice) using Loxp technology and Cre transgenes with robust expression in both conductance and resistance arteries. We found that elimination of AT1A receptors from vascular smooth muscle cells (VSMCs) caused a modest (approximately 7 mmHg) yet significant reduction in baseline BP and exaggerated sodium sensitivity in mice. Additionally, the severity of angiotensin II (Ang II)-dependent hypertension was dramatically attenuated in SMKO mice, and this protection against hypertension was associated with enhanced urinary excretion of sodium. Despite the lower BP, acute vasoconstrictor responses to Ang II in the systemic vasculature were largely preserved (approximately 80% of control levels) in SMKO mice because of exaggerated activity of the sympathetic nervous system rather than residual actions of AT1B receptors. In contrast, Ang II-dependent responses in the renal circulation were almost completely eliminated in SMKO mice (approximately 5%-10% of control levels). These findings suggest that direct actions of AT1A receptors in VSMCs are essential for regulation of renal blood flow by Ang II and highlight the capacity of Ang II-dependent vascular responses in the kidney to effect natriuresis and BP control.
Collapse
Affiliation(s)
- Matthew A Sparks
- Division of Nephrology, Department of Medicine, Durham VA and Duke University Medical Centers, Durham, North Carolina
| | - Johannes Stegbauer
- Division of Nephrology, Department of Medicine, Durham VA and Duke University Medical Centers, Durham, North Carolina; Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Daian Chen
- Division of Nephrology, Department of Medicine, Durham VA and Duke University Medical Centers, Durham, North Carolina
| | - Jose A Gomez
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, North Carolina; and
| | - Robert C Griffiths
- Division of Nephrology, Department of Medicine, Durham VA and Duke University Medical Centers, Durham, North Carolina
| | - Hooman A Azad
- Division of Nephrology, Department of Medicine, Durham VA and Duke University Medical Centers, Durham, North Carolina
| | - Marcela Herrera
- Division of Nephrology, Department of Medicine, Durham VA and Duke University Medical Centers, Durham, North Carolina
| | - Susan B Gurley
- Division of Nephrology, Department of Medicine, Durham VA and Duke University Medical Centers, Durham, North Carolina
| | - Thomas M Coffman
- Division of Nephrology, Department of Medicine, Durham VA and Duke University Medical Centers, Durham, North Carolina; Cardiovascular and Metabolic Disorders Research Program, Duke-National University of Singapore, Graduate Medical School, Singapore
| |
Collapse
|
62
|
Nagalakshmi VK, Yu J. The ureteric bud epithelium: morphogenesis and roles in metanephric kidney patterning. Mol Reprod Dev 2015; 82:151-66. [PMID: 25783232 DOI: 10.1002/mrd.22462] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 01/12/2015] [Indexed: 01/03/2023]
Abstract
The mammalian metanephric kidney is composed of two epithelial components, the collecting duct system and the nephron epithelium, that differentiate from two different tissues -the ureteric bud epithelium and the nephron progenitors, respectively-of intermediate mesoderm origin. The collecting duct system is generated through reiterative ureteric bud branching morphogenesis, whereas the nephron epithelium is formed in a process termed nephrogenesis, which is initiated with the mesenchymal-epithelial transition of the nephron progenitors. Ureteric bud branching morphogenesis is regulated by nephron progenitors, and in return, the ureteric bud epithelium regulates nephrogenesis. The metanephric kidney is physiologically divided along the corticomedullary axis into subcompartments that are enriched with specific segments of these two epithelial structures. Here, we provide an overview of the major molecular and cellular processes underlying the morphogenesis and patterning of the ureteric bud epithelium and its roles in the cortico-medullary patterning of the metanephric kidney.
Collapse
Affiliation(s)
- Vidya K Nagalakshmi
- Department of Cell Biology and Division of Center of Immunity, Inflammation and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | | |
Collapse
|
63
|
Sparks MA, Crowley SD, Gurley SB, Mirotsou M, Coffman TM. Classical Renin-Angiotensin system in kidney physiology. Compr Physiol 2015; 4:1201-28. [PMID: 24944035 DOI: 10.1002/cphy.c130040] [Citation(s) in RCA: 374] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The renin-angiotensin system has powerful effects in control of the blood pressure and sodium homeostasis. These actions are coordinated through integrated actions in the kidney, cardiovascular system and the central nervous system. Along with its impact on blood pressure, the renin-angiotensin system also influences a range of processes from inflammation and immune responses to longevity. Here, we review the actions of the "classical" renin-angiotensin system, whereby the substrate protein angiotensinogen is processed in a two-step reaction by renin and angiotensin converting enzyme, resulting in the sequential generation of angiotensin I and angiotensin II, the major biologically active renin-angiotensin system peptide, which exerts its actions via type 1 and type 2 angiotensin receptors. In recent years, several new enzymes, peptides, and receptors related to the renin-angiotensin system have been identified, manifesting a complexity that was previously unappreciated. While the functions of these alternative pathways will be reviewed elsewhere in this journal, our focus here is on the physiological role of components of the "classical" renin-angiotensin system, with an emphasis on new developments and modern concepts.
Collapse
Affiliation(s)
- Matthew A Sparks
- Division of Nephrology, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | | | | | | | | |
Collapse
|
64
|
Abstract
Stem cells are endowed with the potential for self-renewal and multipotency. Pluripotent embryonic stem cells have an early role in the formation of the three germ layers (ectoderm, mesoderm and endoderm), whereas adult tissue stem cells and progenitor cells are critical mediators of organ homeostasis. The adrenal cortex is an exceptionally dynamic endocrine organ that is homeostatically maintained by paracrine and endocrine signals throughout postnatal life. In the past decade, much has been learned about the stem and progenitor cells of the adrenal cortex and the multiple roles that these cell populations have in normal development and homeostasis of the adrenal gland and in adrenal diseases. In this Review, we discuss the evidence for the presence of adrenocortical stem cells, as well as the various signalling molecules and transcriptional networks that are critical for the embryological establishment and postnatal maintenance of this vital population of cells. The implications of these pathways and cells in the pathophysiology of disease are also addressed.
Collapse
Affiliation(s)
- Elisabeth M Walczak
- Division of Nephrology, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Gary D Hammer
- Center for Organogenesis, Alfred Taubman Biomedical Sciences Research Building, Room 1528, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA
| |
Collapse
|
65
|
Storch U, Blodow S, Gudermann T, Mederos Y Schnitzler M. Cysteinyl leukotriene 1 receptors as novel mechanosensors mediating myogenic tone together with angiotensin II type 1 receptors-brief report. Arterioscler Thromb Vasc Biol 2014; 35:121-6. [PMID: 25395620 DOI: 10.1161/atvbaha.114.304844] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Myogenic vasoconstriction is mediated by vascular smooth muscle cells of resistance arteries sensing mechanical stretch. Angiotensin II AT1 receptors and in particular AT1BRs in murine vascular smooth muscle cells have been characterized as mechanosensors that cannot fully account for myogenic vasoconstriction observed. Therefore, we aimed at uncovering novel vascular mechanosensors by expression profiling and functional characterization of candidate proteins. APPROACH AND RESULTS Analyzing myogenic tone of isolated murine mesenteric arteries of AT1A and AT1B receptor double gene-deficient (AT1A/1B (-/-)) mice ex vivo, we observed a decreased myogenic tone at high intraluminal pressures and an unexpected hyper-reactivity at low intraluminal pressures because of upregulation of cysteinyl leukotriene 1 receptors (CysLT1Rs). Pharmacological blockade of CysLT1Rs with pranlukast significantly reduced myogenic tone not only in AT1A/1B (-/-) but also in wild-type arteries. In wild-type arteries, additional blockade of angiotensin II AT1 receptors with candesartan resulted in an additive reduction of myogenic tone. Furthermore, calcium imaging experiments were performed with fura-2-loaded human embryonic kidney 293 cells overexpressing CysLT1Rs and with isolated mesenteric vascular smooth muscle cells. Hypo-osmotically induced membrane stretch provoked calcium transients that were significantly reduced by pranlukast. Incubations of isolated mesenteric vascular smooth muscle cells with the 5-lipoxygenase inhibitor zileuton had no effect. Furthermore, the Gq/11-protein inhibitor YM 254890 profoundly reduced myogenic tone to the same extent as induced by the application of pranlukast plus candesartan. CONCLUSIONS Here, we identify a novel, hitherto unappreciated role of CysLT1Rs in vascular regulation. We identified CysLT1Rs as novel mechanosensors in the vasculature involved in myogenic vasoconstriction. Moreover, our findings suggest that myogenic tone is determined by AT1 and CysLT1 receptors acting together as mechanosensors via Gq/11-protein activation.
Collapse
Affiliation(s)
- Ursula Storch
- From the Walther Straub Institute of Pharmacology and Toxicology (U.S., S.B., T.G., M.MyS.), Ludwig Maximilians University of Munich, Munich, Germany; and DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance (T.G., M.MyS.), Munich, Germany
| | - Stephanie Blodow
- From the Walther Straub Institute of Pharmacology and Toxicology (U.S., S.B., T.G., M.MyS.), Ludwig Maximilians University of Munich, Munich, Germany; and DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance (T.G., M.MyS.), Munich, Germany
| | - Thomas Gudermann
- From the Walther Straub Institute of Pharmacology and Toxicology (U.S., S.B., T.G., M.MyS.), Ludwig Maximilians University of Munich, Munich, Germany; and DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance (T.G., M.MyS.), Munich, Germany.
| | - Michael Mederos Y Schnitzler
- From the Walther Straub Institute of Pharmacology and Toxicology (U.S., S.B., T.G., M.MyS.), Ludwig Maximilians University of Munich, Munich, Germany; and DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance (T.G., M.MyS.), Munich, Germany.
| |
Collapse
|
66
|
Li W, Hartwig S, Rosenblum ND. Developmental origins and functions of stromal cells in the normal and diseased mammalian kidney. Dev Dyn 2014; 243:853-63. [DOI: 10.1002/dvdy.24134] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Revised: 03/20/2014] [Accepted: 03/24/2014] [Indexed: 01/05/2023] Open
Affiliation(s)
- Winny Li
- Institute of Medical Science, University of Toronto; Toronto Canada
- Program in Developmental and Stem Cell Biology; The Hospital for Sick Children; Toronto Ontario Canada
| | - Sunny Hartwig
- Department of Biomedical Science; Atlantic Veterinary College, University of Prince Edward Island; Prince Edward Island Canada
| | - Norman D. Rosenblum
- Institute of Medical Science, University of Toronto; Toronto Canada
- Program in Developmental and Stem Cell Biology; The Hospital for Sick Children; Toronto Ontario Canada
- Division of Nephrology; Hospital for Sick Children; Toronto Ontario Canada
- Department of Physiology; University of Toronto; Toronto Ontario Canada
- Department of Pediatrics; University of Toronto; Toronto Ontario Canada
| |
Collapse
|
67
|
Ohsawa M, Tamura K, Wakui H, Maeda A, Dejima T, Kanaoka T, Azushima K, Uneda K, Tsurumi-Ikeya Y, Kobayashi R, Matsuda M, Uchida S, Toya Y, Kobori H, Nishiyama A, Yamashita A, Ishikawa Y, Umemura S. Deletion of the angiotensin II type 1 receptor-associated protein enhances renal sodium reabsorption and exacerbates angiotensin II-mediated hypertension. Kidney Int 2014; 86:570-81. [PMID: 24694992 PMCID: PMC4149871 DOI: 10.1038/ki.2014.95] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 02/07/2014] [Accepted: 02/13/2014] [Indexed: 12/13/2022]
Abstract
Angiotensin II type 1 receptor (AT1R)–associated protein (ATRAP) promotes AT1R internalization along with suppression of pathological activation of tissue AT1R signaling. However, the functional significance of ATRAP in renal sodium handling and blood pressure regulation under pathological stimuli is not fully resolved. Here we show the blood pressure of mice with a gene-targeted disruption of ATRAP was comparable to that of wild-type mice at baseline. However, in ATRAP-knockout mice, angiotensin II–induced hypertension was exacerbated and the extent of positive sodium balance was increased by angiotensin II. Renal expression of the sodium-proton antiporter 3, a major sodium transporter in the proximal tubules, urinary pH, renal angiotensinogen production, and angiotensin II content was unaffected. Stimulation of the renal expression and activity of the epithelial sodium channel (ENaC), a major sodium transporter in the distal tubules, was significantly enhanced by chronic angiotensin II infusion. The circulating and urinary aldosterone levels were comparable. The blood pressure response and renal ENaC expression by aldosterone were not affected. Thus, ATRAP deficiency exacerbated angiotensin II–mediated hypertension by pathological activation of renal tubular AT1R by angiotensin II. This directly stimulates ENaC in the distal tubules and enhances sodium retention in an aldosterone-independent manner.
Collapse
Affiliation(s)
- Masato Ohsawa
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kouichi Tamura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiromichi Wakui
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Akinobu Maeda
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Toru Dejima
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tomohiko Kanaoka
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kengo Azushima
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kazushi Uneda
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yuko Tsurumi-Ikeya
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Ryu Kobayashi
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Miyuki Matsuda
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Shinichi Uchida
- Department of Nephrology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshiyuki Toya
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiroyuki Kobori
- Department of Pharmacology, Kagawa University School of Medicine, Kagawa, Japan
| | - Akira Nishiyama
- Department of Pharmacology, Kagawa University School of Medicine, Kagawa, Japan
| | - Akio Yamashita
- Department of Molecular Biology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yoshihiro Ishikawa
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Satoshi Umemura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
68
|
Gomez RA, Belyea B, Medrano S, Pentz ES, Sequeira-Lopez MLS. Fate and plasticity of renin precursors in development and disease. Pediatr Nephrol 2014; 29:721-6. [PMID: 24337407 PMCID: PMC3999616 DOI: 10.1007/s00467-013-2688-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 10/04/2013] [Accepted: 10/28/2013] [Indexed: 01/04/2023]
Abstract
Renin-expressing cells appear early in the embryo and are distributed broadly throughout the body as organogenesis ensues. Their appearance in the metanephric kidney is a relatively late event in comparison with other organs such as the fetal adrenal gland. The functions of renin cells in extra renal tissues remain to be investigated. In the kidney, they participate locally in the assembly and branching of the renal arterial tree and later in the endocrine control of blood pressure and fluid-electrolyte homeostasis. Interestingly, this endocrine function is accomplished by the remarkable plasticity of renin cell descendants along the kidney arterioles and glomeruli which are capable of reacquiring the renin phenotype in response to physiological demands, increasing circulating renin and maintaining homeostasis. Given that renin cells are sensors of the status of the extracellular fluid and perfusion pressure, several signaling mechanisms (β-adrenergic receptors, Notch pathway, gap junctions and the renal baroreceptor) must be coordinated to ensure the maintenance of renin phenotype--and ultimately the availability of renin--during basal conditions and in response to homeostatic threats. Notably, key transcriptional (Creb/CBP/p300, RBP-J) and posttranscriptional (miR-330, miR125b-5p) effectors of those signaling pathways are prominent in the regulation of renin cell identity. The next challenge, it seems, would be to understand how those factors coordinate their efforts to control the endocrine and contractile phenotypes of the myoepithelioid granulated renin-expressing cell.
Collapse
Affiliation(s)
- R Ariel Gomez
- Department of Pediatrics, University of Virginia School of Medicine, 409 Lane Road, Room 2001, Charlottesville, VA, 22908, USA,
| | | | | | | | | |
Collapse
|
69
|
Yosypiv IV. Renin-angiotensin system in ureteric bud branching morphogenesis: implications for kidney disease. Pediatr Nephrol 2014; 29:609-20. [PMID: 24061643 DOI: 10.1007/s00467-013-2616-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 08/20/2013] [Accepted: 08/21/2013] [Indexed: 12/26/2022]
Abstract
Failure of normal branching morphogenesis of the ureteric bud (UB), a key ontogenic process that controls organogenesis of the metanephric kidney, leads to congenital anomalies of the kidney and urinary tract (CAKUT), the leading cause of end-stage kidney disease in children. Recent studies have revealed a central role of the renin-angiotensin system (RAS), the cardinal regulator of blood pressure and fluid/electrolyte homeostasis, in the control of normal kidney development. Mice or humans with mutations in the RAS genes exhibit a spectrum of CAKUT which includes renal medullary hypoplasia, hydronephrosis, renal hypodysplasia, duplicated renal collecting system and renal tubular dysgenesis. Emerging evidence indicates that severe hypoplasia of the inner medulla and papilla observed in angiotensinogen (Agt)- or angiotensin (Ang) II AT 1 receptor (AT 1 R)-deficient mice is due to aberrant UB branching morphogenesis resulting from disrupted RAS signaling. Lack of the prorenin receptor (PRR) in the UB in mice causes reduced UB branching, resulting in decreased nephron endowment, marked kidney hypoplasia, urinary concentrating and acidification defects. This review provides a mechanistic rational supporting the hypothesis that aberrant signaling of the intrarenal RAS during distinct stages of metanephric kidney development contributes to the pathogenesis of the broad phenotypic spectrum of CAKUT. As aberrant RAS signaling impairs normal renal development, these findings advocate caution for the use of RAS inhibitors in early infancy and further underscore a need to avoid their use during pregnancy and to identify the types of molecular processes that can be targeted for clinical intervention.
Collapse
Affiliation(s)
- Ihor V Yosypiv
- Section of Pediatric Nephrology, Department of Pediatrics, Hypertension and Renal Center of Excellence, Tulane University Health Sciences Center, 1430 Tulane Avenue, New Orleans, LA, 70112, USA,
| |
Collapse
|
70
|
Xie L, Sparks MA, Li W, Qi Y, Liu C, Coffman TM, Johnson GA. Quantitative susceptibility mapping of kidney inflammation and fibrosis in type 1 angiotensin receptor-deficient mice. NMR IN BIOMEDICINE 2013; 26:1853-63. [PMID: 24154952 PMCID: PMC3956055 DOI: 10.1002/nbm.3039] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 08/06/2013] [Accepted: 08/23/2013] [Indexed: 05/18/2023]
Abstract
Disruption of the regulatory role of the kidneys leads to diverse renal pathologies; one major hallmark is inflammation and fibrosis. Conventional magnitude MRI has been used to study renal pathologies; however, the quantification or even detection of focal lesions caused by inflammation and fibrosis is challenging. We propose that quantitative susceptibility mapping (QSM) may be particularly sensitive for the identification of inflammation and fibrosis. In this study, we applied QSM in a mouse model deficient for angiotensin receptor type 1 (AT1). This model is known for graded pathologies, including focal interstitial fibrosis, cortical inflammation, glomerulocysts and inner medullary hypoplasia. We acquired high-resolution MRI on kidneys from AT1-deficient mice that were perfusion fixed with contrast agent. Two MR sequences were used (three-dimensional spin echo and gradient echo) to produce three image contrasts: T1, T2* (magnitude) and QSM. T1 and T2* (magnitude) images were acquired to segment major renal structures and to provide landmarks for the focal lesions of inflammation and fibrosis in the three-dimensional space. The volumes of major renal structures were measured to determine the relationship of the volumes to the degree of renal abnormalities and magnetic susceptibility values. Focal lesions were segmented from QSM images and were found to be closely associated with the major vessels. Susceptibilities were relatively more paramagnetic in wild-type mice: 1.46 ± 0.36 in the cortex, 2.14 ± 0.94 in the outer medulla and 2.10 ± 2.80 in the inner medulla (10(-2) ppm). Susceptibilities were more diamagnetic in knockout mice: -7.68 ± 4.22 in the cortex, -11.46 ± 2.13 in the outer medulla and -7.57 ± 5.58 in the inner medulla (10(-2) ppm). This result was consistent with the increase in diamagnetic content, e.g. proteins and lipids, associated with inflammation and fibrosis. Focal lesions were validated with conventional histology. QSM was very sensitive in detecting pathology caused by small focal inflammation and fibrosis. QSM offers a new MR contrast mechanism to study this common disease marker in the kidney.
Collapse
Affiliation(s)
- Luke Xie
- Center for In Vivo Microscopy, Department of Radiology, Duke University Medical Center, Durham, NC, USA
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Matthew A. Sparks
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, NC, USA
| | - Wei Li
- Brain Imaging and Analysis Center, Duke University Medical Center, Durham, NC, USA
| | - Yi Qi
- Center for In Vivo Microscopy, Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | - Chunlei Liu
- Center for In Vivo Microscopy, Department of Radiology, Duke University Medical Center, Durham, NC, USA
- Brain Imaging and Analysis Center, Duke University Medical Center, Durham, NC, USA
| | - Thomas M. Coffman
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, NC, USA
| | - G. Allan Johnson
- Center for In Vivo Microscopy, Department of Radiology, Duke University Medical Center, Durham, NC, USA
- Brain Imaging and Analysis Center, Duke University Medical Center, Durham, NC, USA
- Correspondence to: G. A. Johnson, Center for In Vivo Microscopy, Department of Radiology, Duke University Medical Center, Box 3302, Durham, NC 27710, USA.
| |
Collapse
|
71
|
Blodow S, Schneider H, Storch U, Wizemann R, Forst AL, Gudermann T, Mederos y Schnitzler M. Novel role of mechanosensitive AT1B receptors in myogenic vasoconstriction. Pflugers Arch 2013; 466:1343-53. [PMID: 24101294 DOI: 10.1007/s00424-013-1372-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 09/26/2013] [Accepted: 09/27/2013] [Indexed: 01/15/2023]
Abstract
Myogenic vasoconstriction is an inherent property of vascular smooth muscle cells (VSMCs) of resistance arteries harboring ill-defined mechanosensing and mechanotransducing elements. G protein-coupled receptors (GPCRs) are discussed as mechanosensors in VSMCs. In this study, we sought to identify and characterize the role and impact of GPCRs on myogenic vasoconstriction. Thus, we analyzed mRNA expression levels of GPCRs in resistance versus preceding conduit arteries revealing a significant enrichment of several GPCRs in resistance vessels. Selective pharmacological blockade of the highly expressed GPCRs in isolated murine mesenteric arteries ex vivo decreased myogenic vasoconstriction. In particular, candesartan and losartan most prominently suppressed myogenic tone, suggesting that AT1 receptors play a central role in myogenic vasoconstriction. Analyzing angiotensinogen(-/-) mice, a relevant contribution of locally produced angiotensin II to myogenic tone could be excluded. Investigation of AT1A (-/-) and AT1B (-/-) murine mesenteric arteries revealed that AT1B, but not AT1A, receptors are key components of myogenic regulation. This notion was supported by examining fura-2-loaded isolated AT1A (-/-) and AT1B (-/-) VSMCs. Our results indicate that in VSMCs, AT1B receptors are more mechanosensitive than AT1A receptors even at comparable receptor expression levels. Furthermore, we demonstrate that the mechanosensitivity of GPCRs is agonist-independent and positively correlates with receptor expression levels.
Collapse
Affiliation(s)
- Stephanie Blodow
- Walther-Straub-Institute of Pharmacology and Toxicology, Ludwig Maximilians University of Munich, Goethestr. 33, 80336, Munich, Germany
| | | | | | | | | | | | | |
Collapse
|
72
|
Mederle K, Schweda F, Kattler V, Doblinger E, Miyata K, Höcherl K, Oike Y, Castrop H. The angiotensin II AT1 receptor-associated protein Arap1 is involved in sepsis-induced hypotension. Crit Care 2013; 17:R130. [PMID: 23844607 PMCID: PMC4056110 DOI: 10.1186/cc12809] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 07/11/2013] [Indexed: 11/22/2022] Open
Abstract
Introduction Hypotension in septic patients results from hypovolemia, vasodilatation and hyporeactivity to vasoconstrictors, such as angiotensin II. The AT1 receptor-associated protein 1 (Arap1) is expressed in vascular smooth muscle cells and increases the surface expression of the AT1-receptor in vitro. We hypothesized that dysregulation of Arap1 may contribute to vascular hyporeactivity to angiotensin II during endotoxemia. Methods Arap1-deficient mice were used to assess the role of Arap1 in sepsis-induced hypotension. The isolated perfused kidney was used as an in vitro model to determine the relevance of Arap1 for vascular resistance and sensitivity to angiotensin II. Results During endotoxemia, mean arterial blood pressure (MAP) decreased in both genotypes, with the time course of sepsis-induced hypotension being markedly accelerated in Arap1-/- compared to +/+ mice. However, baseline MAP was similar in Arap1-/- and wildtype mice (102 ± 2 vs.103 ± 2 mmHg; telemetry measurements; n = 10; P = 0.66). Following lipopolysaccharide (LPS) injections (3 mg/kg), Arap1 expression was successively down-regulated in the wildtype mice, reaching levels below 10% of baseline expression. The endotoxemia-related decline in Arap1 expression could be recapitulated in cultured mesangial cells by incubation with pro-inflammatory cytokines, such as tumor necrosis factor α and interferon γ. Plasma renin concentration was increased in Arap1-/- mice compared to wildtype mice (66 ± 6 vs. 41 ± 4 ng AngI/ml/h; n = 23; P = 0.001), presumably contributing to preserved MAP under baseline conditions. The sensitivity of the vasculature to angiotensin II was reduced in Arap1-/- compared to +/+ mice, as determined in the isolated perfused kidney. Conclusions Our data suggest that down-regulation of Arap1 expression during sepsis contributes to the development of hypotension by causing reduced vascular sensitivity to angiotensin II.
Collapse
|
73
|
Arsenic upregulates the expression of angiotensin II Type I receptor in mouse aortic endothelial cells. Toxicol Lett 2013; 220:70-5. [DOI: 10.1016/j.toxlet.2013.04.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 04/08/2013] [Accepted: 04/09/2013] [Indexed: 11/22/2022]
|
74
|
Benigni A, Orisio S, Noris M, Iatropoulos P, Castaldi D, Kamide K, Rakugi H, Arai Y, Todeschini M, Ogliari G, Imai E, Gondo Y, Hirose N, Mari D, Remuzzi G. Variations of the angiotensin II type 1 receptor gene are associated with extreme human longevity. AGE (DORDRECHT, NETHERLANDS) 2013; 35:993-1005. [PMID: 22569962 PMCID: PMC3636412 DOI: 10.1007/s11357-012-9408-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Accepted: 04/04/2012] [Indexed: 05/31/2023]
Abstract
Longevity phenotype in humans results from the influence of environmental and genetic factors. Few gene polymorphisms have been identified so far with a modest effect on lifespan leaving room for the search of other players in the longevity game. It has been recently demonstrated that targeted disruption of the mouse homolog of the human angiotensin II type 1 receptor (AT1R) gene (AGTR1) translates into marked prolongation of animal lifespan (Benigni et al., J Clin Invest 119(3):524-530, 2009). Based on the above study in mice, here we sought to search for AGTR1 variations associated to reduced AT1 receptor protein levels and to prolonged lifespan in humans. AGTR1 was sequenced in 173 Italian centenarians and 376 younger controls. A novel non-synonymous mutation was detected in a centenarian. Two polymorphisms in AGTR1 promoter, rs422858 and rs275653, in complete linkage disequilibrium, were significantly associated with the ability to attain extreme old age. We then replicated the study of rs275653 in a large independent cohort of Japanese origin (598 centenarians and semi-supercentenarians, 422 younger controls) and indeed confirmed its association with exceptional old age. In combined analyses, rs275653 was associated to extreme longevity either at recessive model (P = 0.007, odds ratio (OR) 3.57) or at genotype level (P = 0.015). Significance was maintained after correcting for confounding factors. Fluorescence activated cell sorting analysis revealed that subjects homozygous for the minor allele of rs275653 had less AT1R-positive peripheral blood polymorphonuclear cells. Moreover, rs275653 was associated to lower blood pressure in centenarians. These findings highlight the role of AGTR1 as a possible candidate among longevity-enabling genes.
Collapse
Affiliation(s)
- Ariela Benigni
- />Department of Molecular Medicine, Mario Negri Institute for Pharmacological Research, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87, 24126 Bergamo, Italy
| | - Silvia Orisio
- />Department of Molecular Medicine, Mario Negri Institute for Pharmacological Research, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87, 24126 Bergamo, Italy
| | - Marina Noris
- />Transplant Research Center, “Chiara Cucchi De Alessandri & Gilberto Crespi”, Mario Negri Institute for Pharmacological Research, 24020 Ranica, Italy
| | - Paraskevas Iatropoulos
- />Clinical Research Center for Rare Diseases “Aldo e Cele Daccò”, Mario Negri Institute for Pharmacological Research, 24020 Ranica, Italy
| | - Davide Castaldi
- />Ph.D. School of Informatics, DISCo, University of Milan—Bicocca, 20126 Milan, Italy
| | - Kei Kamide
- />Department of Geriatric Medicine and Nephrology, Osaka University Graduate School of Medicine, 565-0871 Osaka, Japan
| | - Hiromi Rakugi
- />Department of Geriatric Medicine and Nephrology, Osaka University Graduate School of Medicine, 565-0871 Osaka, Japan
| | - Yasumichi Arai
- />Division of Geriatric Medicine, Department of Internal Medicine, Keio University School of Medicine, 160-8582 Tokyo, Japan
| | - Marta Todeschini
- />Transplant Research Center, “Chiara Cucchi De Alessandri & Gilberto Crespi”, Mario Negri Institute for Pharmacological Research, 24020 Ranica, Italy
| | - Giulia Ogliari
- />Department of Medical Sciences, Geriatric Unit, IRCCS Ca’ Granda Foundation Maggiore Policlinico Hospital, University of Milan, 20122 Milan, Italy
| | - Enyu Imai
- />Department of Nephrology, Nagoya University Graduate School of Medicine, 466-8550 Nagoya, Japan
| | - Yasuyuki Gondo
- />Department of Clinical Thanatology and Geriatric Behavioral Science, Osaka University Graduate School of Medicine, 565-0871 Osaka, Japan
| | - Nobuyoshi Hirose
- />Division of Geriatric Medicine, Department of Internal Medicine, Keio University School of Medicine, 160-8582 Tokyo, Japan
| | - Daniela Mari
- />Department of Medical Sciences, Geriatric Unit, IRCCS Ca’ Granda Foundation Maggiore Policlinico Hospital, University of Milan, 20122 Milan, Italy
| | - Giuseppe Remuzzi
- />Mario Negri Institute for Pharmacological Research, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87, 24126 Bergamo, Italy
- />Unit of Nephrology and Dialysis, Azienda Ospedaliera Ospedali Riuniti di Bergamo, 24128 Bergamo, Italy
| |
Collapse
|
75
|
Madsen K, Tinning AR, Marcussen N, Jensen BL. Postnatal development of the renal medulla; role of the renin-angiotensin system. Acta Physiol (Oxf) 2013; 208:41-9. [PMID: 23432903 DOI: 10.1111/apha.12088] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 12/15/2012] [Accepted: 02/13/2013] [Indexed: 01/04/2023]
Abstract
Adverse events during foetal development can predispose the individual for cardiovascular disease later in life, a correlation known as foetal programming of adult hypertension. The 'programming' events have been associated with the kidneys due to the significant role in extracellular volume control and long-term blood pressure regulation. Previously, nephron endowment and functional consequences of a low nephron number have been extensively investigated without achieving a full explanation of the underlying pathophysiological mechanisms. In this review, we will focus on mechanisms of postnatal development in the renal medulla with regard to the programming effects. The renin-angiotensin system is critically involved in mammalian kidney development and impaired signalling gives rise to developmental renal lesions that have been associated with hypertension later in life. A consistent finding in both experimental animal models and in human case reports is atrophy of the renal medulla with developmental lesions to both medullary nephron segments and vascular development with concomitant functional disturbances reaching into adulthood. A review of current knowledge of the role of the renin-angiotensin system for renal medullary development will be given.
Collapse
Affiliation(s)
| | - A. R. Tinning
- Department of Cardiovascular and Renal Research; Institute of Molecular Medicine; University of Southern Denmark; Odense; Denmark
| | - N. Marcussen
- Department of Pathology; Odense University Hospital; Odense; Denmark
| | - B. L. Jensen
- Department of Cardiovascular and Renal Research; Institute of Molecular Medicine; University of Southern Denmark; Odense; Denmark
| |
Collapse
|
76
|
Rasouly HM, Lu W. Lower urinary tract development and disease. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2013; 5:307-42. [PMID: 23408557 PMCID: PMC3627353 DOI: 10.1002/wsbm.1212] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Congenital anomalies of the lower urinary tract (CALUT) are a family of birth defects of the ureter, the bladder, and the urethra. CALUT includes ureteral anomaliesc such as congenital abnormalities of the ureteropelvic junction (UPJ) and ureterovesical junction (UVJ), and birth defects of the bladder and the urethra such as bladder-exstrophy-epispadias complex (BEEC), prune belly syndrome (PBS), and posterior urethral valves (PUVs). CALUT is one of the most common birth defects and is often associated with antenatal hydronephrosis, vesicoureteral reflux (VUR), urinary tract obstruction, urinary tract infections (UTI), chronic kidney disease, and renal failure in children. Here, we discuss the current genetic and molecular knowledge about lower urinary tract development and genetic basis of CALUT in both human and mouse models. We provide an overview of the developmental processes leading to the formation of the ureter, the bladder, and the urethra, and different genes and signaling pathways controlling these developmental processes. Human genetic disorders that affect the ureter, the bladder and the urethra and associated gene mutations are also presented. As we are entering the postgenomic era of personalized medicine, information in this article may provide useful interpretation for the genetic and genomic test results collected from patients with lower urinary tract birth defects. With evidence-based interpretations, clinicians may provide more effective personalized therapies to patients and genetic counseling for their families.
Collapse
Affiliation(s)
- Hila Milo Rasouly
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, MA 02118, USA
| | - Weining Lu
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, MA 02118, USA
| |
Collapse
|
77
|
Wakui H, Tamura K, Masuda SI, Tsurumi-Ikeya Y, Fujita M, Maeda A, Ohsawa M, Azushima K, Uneda K, Matsuda M, Kitamura K, Uchida S, Toya Y, Kobori H, Nagahama K, Yamashita A, Umemura S. Enhanced angiotensin receptor-associated protein in renal tubule suppresses angiotensin-dependent hypertension. Hypertension 2013; 61:1203-10. [PMID: 23529167 DOI: 10.1161/hypertensionaha.111.00572] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We have previously shown that angiotensin II type 1 receptor-associated protein (ATRAP/Agtrap) interacts with the angiotensin II type 1 receptor and promotes constitutive internalization of the receptor so as to inhibit the pathological activation of its downstream signaling but preserve baseline physiological signaling activity. The present study was designed to investigate the role of renal ATRAP in angiotensin II-dependent hypertension. We generated transgenic mice dominantly expressing ATRAP in the renal tubules, including renal distal tubules. The renal ATRAP transgenic mice exhibited no significant change in blood pressure at baseline on normal salt diet. However, in the renal ATRAP transgenic mice compared with wild-type mice, the following took place: (1) the development of high blood pressure in response to angiotensin II infusion was significantly suppressed based on radiotelemetry, (2) the extent of daily positive sodium balance was significantly reduced during angiotensin II infusion in metabolic cage analysis, and (3) the renal Na+ -Cl- cotransporter activation and α-subunit of the epithelial sodium channel induction by angiotensin II infusion were inhibited. Furthermore, adenoviral overexpression of ATRAP suppressed the angiotensin II-mediated increase in the expression of α-subunit of the epithelial sodium channel in mouse distal convoluted tubule cells. These results indicate that renal tubule-dominant ATRAP activation provokes no evident effects on blood pressure at baseline but exerts an inhibitory effect on the pathological elevation of blood pressure in response to angiotensin II stimulation, thereby suggesting that ATRAP is a potential target of interest in blood pressure modulation under pathological conditions.
Collapse
Affiliation(s)
- Hiromichi Wakui
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Gonzalez-Rodriguez P, Tong W, Xue Q, Li Y, Hu S, Zhang L. Fetal hypoxia results in programming of aberrant angiotensin ii receptor expression patterns and kidney development. Int J Med Sci 2013; 10:532-8. [PMID: 23532764 PMCID: PMC3607238 DOI: 10.7150/ijms.5566] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 02/26/2013] [Indexed: 01/30/2023] Open
Abstract
AIMS The present study tested the hypothesis that fetal hypoxia adversely affects kidney development in fetal and offspring rats and alter the expression patterns of angiotensin II type 1 (AT1R) and type 2 (AT2R) receptors. METHODS Time-dated pregnant rats were divided between normoxic and hypoxic (10.5% O2 last period of gestation) groups. Protein expression, in the offspring, was determined using western blot. RESULTS Hypoxic treatment significantly decreased body and kidney weight in 21-day fetuses (E21) and 7-day neonates (P7). In 3-month-old offspring there were no significant differences in body and kidney weight between hypoxic and control animals. Fetal hypoxia had no effect on kidney AT1R density in E21 or P7, but significantly decreased kidney AT1R protein and mRNA abundance in both male and female adults. In contrast, kidney AT2R density was not affected by fetal hypoxia throughout the developmental stages studied. The hypoxia-mediated reduction of nephron numbers was progressively from P7 worsened into the adulthood with females affected more than males. CONCLUSION The results suggest that fetal hypoxia causes programming of aberrant kidney development and accelerates the aging process of the kidney during the postnatal development, which may contribute to an increased risk of cardiovascular disease.
Collapse
Affiliation(s)
- Pablo Gonzalez-Rodriguez
- Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | | | | | | | | | | |
Collapse
|
79
|
Herrera M, Sparks MA, Alfonso-Pecchio AR, Harrison-Bernard LM, Coffman TM. Lack of specificity of commercial antibodies leads to misidentification of angiotensin type 1 receptor protein. Hypertension 2012; 61:253-8. [PMID: 23150519 DOI: 10.1161/hypertensionaha.112.203679] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The angiotensin II type 1 receptor (AT(1)R) mediates most hypertensive actions of angiotensin II. To understand the molecular regulation of the AT(1)R in normal physiology and pathophysiology, methods for sensitive and specific detection of AT(1)R protein are required. Here, we examined the specificity of a panel of putative AT(1)R antibodies that are commonly used by investigators in the field. For these studies, we carried out Western blotting and immunohistochemistry with kidney tissue from wild-type mice and genetically modified mice lacking the major murine AT(1)R isoform, AT(1A) (AT(1A)KO), or with combined deficiency of both the AT(1A) and AT(1B) isoforms (AT(1AB)KO). For the 3 antibodies tested, Western blots of protein homogenates from wild-type kidneys yielded distinct bands with the expected size range for AT(1)R. In addition, these bands appeared identical in samples from mice lacking 1 or both murine AT(1)R isoforms. Additionally, the pattern of immunohistochemical staining in kidneys, liver, and adrenal glands of wild-type mice was very similar to that of AT(1AB)KO mice completely lacking all AT(1)R. We verified the absence of AT(1)R subtypes in each mouse line by the following: (1) quantitative polymerase chain reaction documenting the absence of mRNA species, and (2) functionally by assessing angiotensin II-dependent vasoconstriction, which was substantially blunted in both AT(1A)KOs and AT(1AB)KOs. Finally, these antibodies failed to detect epitope-tagged AT(1A)R protein overexpressed in human embryonic kidney cells. We conclude that anti-AT(1)R antibodies available from commercial sources and commonly used in published studies exhibit nonspecific binding in mouse tissue that may lead to erroneous results.
Collapse
Affiliation(s)
- Marcela Herrera
- Division of Nephrology, Department of Medicine, Duke University Medical Center, Durham, NC, USA.
| | | | | | | | | |
Collapse
|
80
|
Regional variation in aortic AT1b receptor mRNA abundance is associated with contractility but unrelated to atherosclerosis and aortic aneurysms. PLoS One 2012; 7:e48462. [PMID: 23119030 PMCID: PMC3485205 DOI: 10.1371/journal.pone.0048462] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 09/27/2012] [Indexed: 11/19/2022] Open
Abstract
Background Angiotensin II (AngII), the main bioactive peptide of the renin angiotensin system, exerts most of its biological actions through stimulation of AngII type 1 (AT1) receptors. This receptor is expressed as 2 structurally similar subtypes in rodents, termed AT1a and AT1b. Although AT1a receptors have been studied comprehensively, roles of AT1b receptors in the aorta have not been defined. Methodology/Results We initially compared the regional distribution of AT1b receptor mRNA with AT1a receptor mRNA in the aorta. mRNA abundance of both subtypes increased from the proximal to the distal aorta, with the greatest abundance in the infra-renal region. Corresponding to the high mRNA abundance for both receptors, only aortic rings from the infra-renal aorta contracted in response to AngII stimulation. Despite the presence of both receptor transcripts, deletion of AT1b receptors, but not AT1a receptors, diminished AngII-induced contractility. To determine whether absence of AT1b receptors influenced aortic pathologies, we bred AT1b receptor deficient mice into an LDL receptor deficient background. Mice were fed a diet enriched in saturated fat and infused with AngII (1,000 ng/kg/min). Parameters that could influence development of aortic pathologies, including systolic blood pressure and plasma cholesterol concentrations, were not impacted by AT1b receptor deficiency. Absence of AT1b receptors also had no effect on size of aortic atherosclerotic lesions and aortic aneurysms in both the ascending and abdominal regions. Conclusions/Significance Regional abundance of AT1b receptor mRNA coincided with AngII-induced regional contractility, but it was not associated with AngII-induced aortic pathologies.
Collapse
|
81
|
Chen F. Plumbing the depths of urinary tract obstruction by using murine models. Organogenesis 2012; 5:297-305. [PMID: 19568351 DOI: 10.4161/org.8055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2009] [Accepted: 02/02/2009] [Indexed: 11/19/2022] Open
Abstract
Urinary tract obstruction leads to obstructive nephropathy, which in turn, frequently results in renal failure. Congenital urinary tract obstruction can be traced back to errors during the organogenesis of the urinary system. A fundamental understanding of the causes of urinary tract obstruction and the developmental processes involved are critical for improving the diagnostic and therapeutic strategies for this disease. A number of laboratories, including ours, have been using genetically engineered and spontaneously occurring mouse models to study the primary causes and the pathogenesis of urinary tract obstruction. These studies have shown that urinary tract obstruction is a very heterogeneous disease that can be caused by a diverse set of factors targeting multiple levels of the urinary system. Accumulating evidence also indicates that the development of the urinary tract requires the integration of progenitor cells of diverse embryonic origins, leading to the formation of multiple junctions prone to developmental errors. In addition, the high sensitivity of the pyeloureteral peristaltic machinery to disturbance affecting the structural or functional integrity of its components also contributes to the high incidence rate of urinary tract obstruction.
Collapse
Affiliation(s)
- Feng Chen
- Assistant Professor of Medicine and Cell Biology and Physiology; Washington University School of Medicine; St. Louis, Missouri USA
| |
Collapse
|
82
|
Yosypiv IV. Hypothesis: a new role for the Renin-Angiotensin system in ureteric bud branching. Organogenesis 2012; 1:26-32. [PMID: 19521557 DOI: 10.4161/org.1.1.1071] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2004] [Accepted: 04/12/2004] [Indexed: 11/19/2022] Open
Abstract
Branching morphogenesis in the developing mammalian kidney involves growth and branching of the ureteric bud (UB), leading to formation of its daughter collecting ducts, calyces, pelvis and ureters. Even subtle defects in the efficiency and/or accuracy of this process have profound effects on the ultimate development of the kidney and result in congenital abnormalities of the kidney and urinary tract. This review summarizes current knowledge regarding a number of genes known to regulate UB development and emphasizes an emerging role for the renin-angiotensin system (RAS) in renal branching morphogenesis. Mutations in the genes encoding components of the RAS in mice cause renal papillary hypoplasia, hydronephrosis, and urinary concentrating defect. These findings imply that UB-derived epithelia are targets for angiotensin (ANG) II actions during metanephric kidney development. Here, it is proposed that papillary hypoplasia in RAS-deficient mice is secondary to an intrinsic defect in the development of the renal medulla. This hypothesis is based on the following observations: (a) UB and surrounding stroma express angiotensinogen (AGT) and ANG II AT(1) receptors in vivo; (b) ANG II stimulates UB cell process extension, branching and cord formation in collagen gel cultures in vitro; and (c) AT(1) blockade inhibits ANG II-induced UB cell branching. It is further postulated that ANG II is a novel stroma-derived factor involved in stroma/UB cross-talk which regulates UB branching morphogenesis.
Collapse
|
83
|
Six commercially available angiotensin II AT1 receptor antibodies are non-specific. Cell Mol Neurobiol 2012; 32:1353-65. [PMID: 22843099 DOI: 10.1007/s10571-012-9862-y] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 07/05/2012] [Indexed: 12/31/2022]
Abstract
Commercially available Angiotensin II AT1 receptor antibodies are widely employed for receptor localization and quantification, but they have not been adequately validated. In this study, six commercially available AT1 receptor antibodies were characterized by established criteria: sc-1173 and sc-579 from Santa Cruz Biotechnology, Inc., AAR-011 from Alomone Labs, Ltd., AB15552 from Millipore, and ab18801 and ab9391 from Abcam. The immunostaining patterns observed were different for every antibody tested, and were unrelated to the presence or absence of AT1 receptors. The antibodies detected a 43 kDa band in western blots, corresponding to the predicted size of the native AT1 receptor. However, identical bands were observed in wild-type mice and in AT1A knock-out mice not expressing the target protein. Moreover, immunoreactivity detected in rat hypothalamic 4B cells not expressing AT1 receptors or transfected with AT1A receptor construct was identical, as revealed by western blotting and immunocytochemistry in cultured 4B cells. Additional prominent immunoreactive bands above and below 43 kDa were observed by western blotting in extracts from tissues of AT1A knock-out and wild-type mice and in 4B cells with or without AT1 receptor expression. In all cases, the patterns of immunoreactivity were independent of the AT1 receptor expression and different for each antibody studied. We conclude that, in our experimental setup, none of the commercially available AT1 receptor antibodies tested met the criteria for specificity and that competitive radioligand binding remains the only reliable approach to study AT1 receptor physiology in the absence of full antibody characterization.
Collapse
|
84
|
Li XC, Shao Y, Zhuo JL. AT1a receptor signaling is required for basal and water deprivation-induced urine concentration in AT1a receptor-deficient mice. Am J Physiol Renal Physiol 2012; 303:F746-56. [PMID: 22739536 DOI: 10.1152/ajprenal.00644.2011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
It is well recognized that ANG II interacts with arginine vasopressin (AVP) to regulate water reabsorption and urine concentration in the kidney. The present study used ANG II type 1a (AT(1a)) receptor-deficient (Agtr1a(-/-)) mice to test the hypothesis that AT(1a) receptor signaling is required for basal and water deprivation-induced urine concentration in the renal medulla. Eight groups of wild-type (WT) and Agtr1a(-/-) mice were treated with or without 24-h water deprivation and 1-desamino-8-d-AVP (DDAVP; 100 ng/h ip) for 2 wk or with losartan (10 mg/kg ip) during water deprivation. Under basal conditions, Agtr1a(-/-) mice had lower systolic blood pressure (P < 0.01), greater than threefold higher 24-h urine excretion (WT mice: 1.3 ± 0.1 ml vs. Agtr1a(-/-) mice: 5.9 ± 0.7 ml, P < 0.01), and markedly decreased urine osmolality (WT mice: 1,834 ± 86 mosM/kg vs. Agtr1a(-/-) mice: 843 ± 170 mosM/kg, P < 0.01), without significant changes in 24-h urinary Na(+) excretion. These responses in Agtr1a(-/-) mice were associated with lower basal plasma AVP (WT mice: 105 ± 8 pg/ml vs. Agtr1a(-/-) mice: 67 ± 6 pg/ml, P < 0.01) and decreases in total lysate and membrane aquaporin-2 (AQP2; 48.6 ± 7% of WT mice, P < 0.001) and adenylyl cyclase isoform III (55.6 ± 8% of WT mice, P < 0.01) proteins. Although 24-h water deprivation increased plasma AVP to the same levels in both strains, 24-h urine excretion was still higher, whereas urine osmolality remained lower, in Agtr1a(-/-) mice (P < 0.01). Water deprivation increased total lysate AQP2 proteins in the inner medulla but had no effect on adenylyl cyclase III, phosphorylated MAPK ERK1/2, and membrane AQP2 proteins in Agtr1a(-/-) mice. Furthermore, infusion of DDAVP for 2 wk was unable to correct the urine-concentrating defects in Agtr1a(-/-) mice. These results demonstrate that AT(1a) receptor-mediated ANG II signaling is required to maintain tonic AVP release and regulate V(2) receptor-mediated responses to water deprivation in the inner medulla.
Collapse
Affiliation(s)
- Xiao C Li
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology, University of MississippiMedical Center, 2500 N. State St., Jackson, MS 39216-4505, USA
| | | | | |
Collapse
|
85
|
Song R, Preston G, Khalili A, El-Dahr SS, Yosypiv IV. Angiotensin II regulates growth of the developing papillas ex vivo. Am J Physiol Renal Physiol 2012; 302:F1112-20. [PMID: 22301625 DOI: 10.1152/ajprenal.00435.2011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
We tested the hypothesis that lack of angiotensin (ANG) II production in angiotensinogen (AGT)-deficient mice or pharmacologic antagonism of ANG II AT(1) receptor (AT(1)R) impairs growth of the developing papillas ex vivo, thus contributing to the hypoplastic renal medulla phenotype observed in AGT- or AT(1)R-null mice. Papillas were dissected from Hoxb7(GFP+) or AGT(+/+), (+/-), (-/-) mouse metanephroi on postnatal day P3 and grown in three-dimentional collagen matrix gels in the presence of media (control), ANG II (10(-5) M), or the specific AT(1)R antagonist candesartan (10(-6) M) for 24 h. Percent reduction in papillary length was attenuated in AGT(+/+) and in AGT(+/-) compared with AGT(-/-) (-18.4 ± 1.3 vs. -32.2 ± 1.6%, P < 0.05, -22.8 ± 1.3 vs. -32.2 ± 1.6%, P < 0.05, respectively). ANG II blunted the decrease in papilla length observed in respective media-treated controls in Hoxb7(GFP+) (-1.5 ± 0.3 vs. -10.0 ± 1.4%, P < 0.05) or AGT(+/+), (+/-), and (-/-) papillas (-12.8 ± 0.7 vs. -18.4 ± 1.3%, P < 0.05, -16.8 ± 1.1 vs. -23 ± 1.2%, P < 0.05; -26.2 ± 1.6 vs. -32.2 ± 1.6%, P < 0.05, respectively). In contrast, percent decrease in the length of Hoxb7(GFP+) papillas in the presence of the AT(1)R antagonist candesartan was higher compared with control (-24.3 ± 2.1 vs. -10.5 ± 1.8%, P < 0.05). The number of proliferating phospho-histone H3 (pH3)-positive collecting duct cells was lower, whereas the number of caspase 3-positive cells undergoing apoptosis was higher in candesartan- vs. media-treated papillas (pH3: 12 ± 1.4 vs. 21 ± 2.1, P < 0.01; caspase 3: 3.8 ± 0.5 vs. 1.7 ± 0.2, P < 0.01). Using quantitative RT-PCR, we demonstrate that AT(1)R signaling regulates the expression of genes implicated in morphogenesis of the renal medulla. We conclude that AT(1)R prevents shrinkage of the developing papillas observed ex vivo via control of Wnt7b, FGF7, β-catenin, calcineurin B1, and α3 integrin gene expression, collecting duct cell proliferation, and survival.
Collapse
Affiliation(s)
- Renfang Song
- Division of Pediatric Nephrology, Department of Pediatrics, Hypertension, and Renal Center of Excellence, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| | | | | | | | | |
Collapse
|
86
|
Nahmod KA, Walther T, Cambados N, Fernandez N, Meiss R, Tappenbeck N, Wang Y, Raffo D, Simian M, Schwiebs A, Pozner RG, Fuxman Bass JI, Pozzi AG, Geffner JR, Kordon EC, Schere-Levy C. AT1 receptor blockade delays postlactational mammary gland involution: a novel role for the renin angiotensin system. FASEB J 2012; 26:1982-94. [PMID: 22286690 DOI: 10.1096/fj.11-191932] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Angiotensin II (AngII), the main effector peptide of the renin-angiotensin system (RAS), participates in multiple biological processes, including cell growth, apoptosis, and tissue remodeling. Since AngII activates, in different cell types, signal transducing pathways that are critical for mammary gland postlactational regression, we investigated the role of the RAS during this process. We found that exogenous administration of AngII in mammary glands of lactating Balb/c mice induced epithelium apoptosis [2.9±0.5% (control) vs. 9.6±1.1% (AngII); P < 0.001] and activation of the proapoptotic factor STAT3, an effect inhibited by irbesartan, an AT(1) receptor blocker. Subsequently, we studied the expression kinetics of RAS components during involution. We found that angiotensin-converting enzyme (ACE) mRNA expression peaked 6 h after weaning (5.7-fold; P<0.01), while induction of angiotensinogen and AT(1) and AT(2) receptors expression was detected 96 h after weaning (6.2-, 10-, and 6.2-fold increase, respectively; P<0.01). To assess the role of endogenously generated AngII, mice were treated with losartan, an AT(1) receptor blocker, during mammary involution. Mammary glands from losartan-treated mice showed activation of the survival factors AKT and BCL-(XL), significantly lower LIF and TNF-α mRNA expression (P<0.05), reduced apoptosis [12.1±2.1% (control) vs. 4.8±0.7% (losartan); P<0.001] and shedding of epithelial cells, inhibition of MMP-9 activity in a dose-dependent manner (80%; P<0.05; with losartan IC(50) value of 6.9 mg/kg/d] and lower collagen deposition and adipocyte invasion causing a delayed involution compared to vehicle-treated mice. Furthermore, mammary glands of forced weaned AT(1A)- and/or AT(1B)-deficient mice exhibited retarded apoptosis of epithelial cells [6.3±0.95% (WT) vs. 3.3±0.56% (AT(1A)/AT(1B) DKO); P<0.05] with remarkable delayed postlactational regression compared to wild-type animals. Taken together, these results strongly suggest that AngII, via the AT(1) receptor, plays a major role in mouse mammary gland involution identifying a novel role for the RAS. angiotensin system.
Collapse
Affiliation(s)
- Karen A Nahmod
- IFIBYNE-CONICET, Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Abstract
The mature renal medulla, the inner part of the kidney, consists of the medullary collecting ducts, loops of Henle, vasa recta and the interstitium. The unique spatial arrangement of these components is essential for the regulation of urine concentration and other specialized kidney functions. Thus, the proper and timely assembly of medulla constituents is a crucial morphogenetic event leading to the formation of a functioning metanephric kidney. Mechanisms that direct renal medulla formation are poorly understood. This review describes the current understanding of the key molecular and cellular mechanisms underlying morphological aspects of medulla formation. Given that hypoplasia of the renal medulla is a common manifestation of congenital obstructive nephropathy and other types of congenital anomalies of the kidney and urinary tract (CAKUT), better understanding of how disruptions in medulla formation are linked to CAKUT will enable improved diagnosis, treatment and prevention of CAKUT and their associated morbidity.
Collapse
Affiliation(s)
- Renfang Song
- Section of Pediatric Nephrology, Department of Pediatrics, Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, USA
| | | |
Collapse
|
88
|
Gribouval O, Morinière V, Pawtowski A, Arrondel C, Sallinen SL, Saloranta C, Clericuzio C, Viot G, Tantau J, Blesson S, Cloarec S, Machet MC, Chitayat D, Thauvin C, Laurent N, Sampson JR, Bernstein JA, Clemenson A, Prieur F, Daniel L, Levy-Mozziconacci A, Lachlan K, Alessandri JL, Cartault F, Rivière JP, Picard N, Baumann C, Delezoide AL, Belar Ortega M, Chassaing N, Labrune P, Yu S, Firth H, Wellesley D, Bitzan M, Alfares A, Braverman N, Krogh L, Tolmie J, Gaspar H, Doray B, Majore S, Bonneau D, Triau S, Loirat C, David A, Bartholdi D, Peleg A, Brackman D, Stone R, DeBerardinis R, Corvol P, Michaud A, Antignac C, Gubler MC. Spectrum of mutations in the renin-angiotensin system genes in autosomal recessive renal tubular dysgenesis. Hum Mutat 2011; 33:316-26. [PMID: 22095942 DOI: 10.1002/humu.21661] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Accepted: 11/08/2011] [Indexed: 11/11/2022]
Abstract
Autosomal recessive renal tubular dysgenesis (RTD) is a severe disorder of renal tubular development characterized by early onset and persistent fetal anuria leading to oligohydramnios and the Potter sequence, associated with skull ossification defects. Early death occurs in most cases from anuria, pulmonary hypoplasia, and refractory arterial hypotension. The disease is linked to mutations in the genes encoding several components of the renin-angiotensin system (RAS): AGT (angiotensinogen), REN (renin), ACE (angiotensin-converting enzyme), and AGTR1 (angiotensin II receptor type 1). Here, we review the series of 54 distinct mutations identified in 48 unrelated families. Most of them are novel and ACE mutations are the most frequent, observed in two-thirds of families (64.6%). The severity of the clinical course was similar whatever the mutated gene, which underlines the importance of a functional RAS in the maintenance of blood pressure and renal blood flow during the life of a human fetus. Renal hypoperfusion, whether genetic or secondary to a variety of diseases, precludes the normal development/ differentiation of proximal tubules. The identification of the disease on the basis of precise clinical and histological analyses and the characterization of the genetic defects allow genetic counseling and early prenatal diagnosis.
Collapse
Affiliation(s)
- Olivier Gribouval
- Inserm U983, Faculté de Médecine Paris Descartes, Université Paris Descartes, Paris, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Stegbauer J, Gurley SB, Sparks MA, Woznowski M, Kohan DE, Yan M, Lehrich RW, Coffman TM. AT1 receptors in the collecting duct directly modulate the concentration of urine. J Am Soc Nephrol 2011; 22:2237-46. [PMID: 22052052 DOI: 10.1681/asn.2010101095] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Mice lacking AT(1) angiotensin receptors have an impaired capacity to concentrate the urine, but the underlying mechanism is unknown. To determine whether direct actions of AT(1) receptors in epithelial cells of the collecting duct regulate water reabsorption, we used Cre-Loxp technology to specifically eliminate AT(1A) receptors from the collecting duct in mice (CD-KOs). Although levels of AT(1A) receptor mRNA in the inner medulla of CD-KO mice were significantly reduced, their kidneys appeared structurally normal. Under basal conditions, plasma and urine osmolalities and urine volumes were similar between CD-KO mice and controls. The increase in urine osmolality in response to water deprivation or vasopressin administration, however, was consistently attenuated in CD-KO mice. Similarly, levels of aquaporin-2 protein in inner and outer medulla after water deprivation were significantly lower in CD-KO mice compared with controls, despite its normal localization to the apical membrane. In summary, these results demonstrate that AT(1A) receptors in epithelial cells of the collecting duct directly modulate aquaporin-2 levels and contribute to the concentration of urine.
Collapse
Affiliation(s)
- Johannes Stegbauer
- Department of Medicine, Division of Nephrology, Duke University Medical Center, MSRBII Room 2018, 106 Research Drive, Durham, NC 27710, USA
| | | | | | | | | | | | | | | |
Collapse
|
90
|
Abstract
The renin-angiotensin system (RAS) plays an important role in regulating blood pressure, water-salt balance and the pathogenesis of cardiovascular diseases. Angiotensin II (Ang II) is the physiologically active mediator and mediates the main pathophysiological actions in RAS. Ang II exerts the effects by activating its receptors, primarily type 1 (AT1R) and type 2 (AT2R). Most of the known pathophysiological effects of Ang II are mediated by AT1R activation. The precise physiological function of AT2R is still not clear. Generally, AT2R is considered to oppose the effects of AT1R. Lectin-like oxidized low-density lipoprotein scavenger receptor-1 (LOX-1) is one of the major receptors responsible for binding, internalizing and degrading ox-LDL. The activation of LOX-1 has been known to be related to many pathophysiological events, including endothelial dysfunction and injury, fibroblast growth, and vascular smooth muscle cell hypertrophy. Many of these alterations are present in atherosclerosis, hypertension, and myocardial ischemia and remodeling. A growing body of evidence suggests the existence of a cross-talk between LOX-1 and Ang II receptors. Their interplays are embodied in the reciprocal regulation of their expression and activity. Their interplays are involved in a series of signals. Recent studies suggests that reactive oxygen species (ROS), nitric oxide (NO), protein kinase C (PKC) and mitogen activated protein kinases (MAPKs) are important signals responsible for their cross-talk. This paper reviews these aspects of dyslipidemia and RAS activation.
Collapse
Affiliation(s)
- Xianwei Wang
- Division of Cardiology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | | | | |
Collapse
|
91
|
Yosypiv IV. Renin-angiotensin system in ureteric bud branching morphogenesis: insights into the mechanisms. Pediatr Nephrol 2011; 26:1499-512. [PMID: 21359618 DOI: 10.1007/s00467-011-1820-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Revised: 01/24/2011] [Accepted: 02/01/2011] [Indexed: 12/31/2022]
Abstract
Branching morphogenesis of the ureteric bud (UB) is a key developmental process that controls organogenesis of the entire metanephros. Notably, aberrant UB branching may result in a spectrum of congenital anomalies of the kidney and urinary tract (CAKUT). Genetic, biochemical and physiological studies have demonstrated that the renin-angiotensin system (RAS), a key regulator of the blood pressure and fluid/electrolyte homeostasis, also plays a critical role in kidney development. All the components of the RAS are expressed in the metanephros. Moreover, mutations in the genes encoding components of the RAS in mice or humans cause diverse types of CAKUT which include renal papillary hypoplasia, hydronephrosis, duplicated collecting system, renal tubular dysgenesis, renal vascular abnormalities, abnormal glomerulogenesis and urinary concentrating defect. Despite widely accepted role of the RAS in metanephric kidney and renal collecting system (ureter, pelvis, calyces and collecting ducts) development, the mechanisms by which an intact RAS exerts its morphogenetic actions are incompletely defined. Emerging evidence indicates that defects in UB branching morphogenesis may be causally linked to the pathogenesis of renal collecting system anomalies observed under conditions of aberrant RAS signaling. This review describes the role of the RAS in UB branching morphogenesis and highlights emerging insights into the cellular and molecular mechanisms whereby RAS regulates this critical morphogenetic process.
Collapse
Affiliation(s)
- Ihor V Yosypiv
- Section of Pediatric Nephrology, Department of Pediatrics, SL-37 Hypertension and Renal Center of Excellence, Tulane University Health Sciences Center, 1430 Tulane Avenue, New Orleans, LA 70112, USA.
| |
Collapse
|
92
|
Yu J. Wnt signaling and renal medulla formation. Pediatr Nephrol 2011; 26:1553-7. [PMID: 21533626 DOI: 10.1007/s00467-011-1888-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Revised: 03/07/2011] [Accepted: 03/23/2011] [Indexed: 11/29/2022]
Abstract
The renal medulla, the inner compartment of the metanephric kidney, plays vital roles in the regulation of body water, electrolyte homeostasis, and systemic blood pressure. It is composed of the loops-of-Henle, the medullary collecting ducts, the vasa recta, and the medullary interstitium. Its epithelial and endothelial components display ordered spatial organization. This organization serves as the structural basis for its function in urine concentration. The urine concentration ability of a renal medulla is also related to its length among species. In this review, the current understanding of the molecular and cellular mechanisms underlying renal medulla formation (elongation) is summarized, with a focus on the role of Wnt signaling in this developmental process. Renal medulla blunting and effacement is a common symptom of many renal and urological destructions. The knowledge in renal medulla formation should assist efforts in repair and regeneration of a damaged renal medulla, so to improve renal physiology in diseased situations.
Collapse
Affiliation(s)
- Jing Yu
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
| |
Collapse
|
93
|
Song R, Preston G, Yosypiv IV. Angiotensin II stimulates in vitro branching morphogenesis of the isolated ureteric bud. Mech Dev 2011; 128:359-67. [PMID: 21820050 DOI: 10.1016/j.mod.2011.07.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 07/14/2011] [Accepted: 07/15/2011] [Indexed: 12/19/2022]
Abstract
Mutations in the renin-angiotensin system (RAS) genes are associated with congenital anomalies of the kidney and urinary tract (CAKUT). As angiotensin (Ang) II, the principal effector peptide growth factor of the RAS, stimulates ureteric bud (UB) branching in whole intact embryonic (E) metanephroi, defects in UB morphogenesis may be causally linked to CAKUT observed under conditions of disrupted RAS. In the present study, using the isolated intact UB (iUB) assay, we tested the hypothesis that Ang II stimulates UB morphogenesis by directly acting on the UB, identified Ang II target genes in the iUB by microarray and examined the effect of Ang II on UB cell migration in vitro. We show that isolated E11.5 mouse iUBs express Ang II AT(1) and AT(2) receptor mRNA. Treatment of E11.5 iUBs grown in collagen matrix gels with Ang II (10(-5)M) increases the number of iUB tips after 48h of culture compared to control (4.8±0.4 vs. 2.4±0.2, p<0.01). A number of genes required for UB branching as well as novel genes whose role in UB development is currently unknown are targets of Ang II signaling in the iUB. In addition, Ang II increases UB cell migration (346±5.1 vs. 275±4.4, p<0.01) in vitro. In summary, Ang II stimulates UB cell migration and directly induces morphogenetic response in the iUB. We conclude that Ang II-regulated genes in the iUB may be important mediators of Ang II-induced UB branching. We hypothesize that Ang II-dependent cell movements play an important role in UB branching morphogenesis.
Collapse
Affiliation(s)
- Renfang Song
- Division of Pediatric Nephrology, Department of Pediatrics, Hypertension and Renal Center of Excellence, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| | | | | |
Collapse
|
94
|
Song R, Yosypiv IV. (Pro)renin Receptor in Kidney Development and Disease. Int J Nephrol 2011; 2011:247048. [PMID: 21755055 PMCID: PMC3132641 DOI: 10.4061/2011/247048] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2011] [Revised: 03/20/2011] [Accepted: 04/20/2011] [Indexed: 12/24/2022] Open
Abstract
The renin-angiotensin system (RAS), a key regulator of the blood pressure and fluid/electrolyte homeostasis, also plays a critical role in kidney development. All the components of the RAS are expressed in the developing metanephros. Moreover, mutations in the genes encoding components of the RAS in mice or humans are associated with a broad spectrum of congenital anomalies of the kidney and urinary tract (CAKUT). These forms of CAKUT include renal papillary hypoplasia, hydronephrosis, duplicated collecting system, renal tubular dysgenesis, renal vascular abnormalities, and aberrant glomerulogenesis. Emerging evidence indicates that (pro)renin receptor (PRR), a novel component of the RAS, is essential for proper kidney development and that aberrant PRR signaling is causally linked to cardiovascular and renal disease. This paper describes the role of the RAS in kidney development and highlights emerging insights into the cellular and molecular mechanisms by which the PRR may regulate this critical morphogenetic process.
Collapse
Affiliation(s)
- Renfang Song
- Section of Pediatric Nephrology, Department of Pediatrics, Hypertension and Renal Center of Excellence, Tulane University Health Sciences Center, 1430 Tulane Avenue, New Orleans, LA 70112, USA
| | | |
Collapse
|
95
|
Yamamoto R, Akazawa H, Fujihara H, Ozasa Y, Yasuda N, Ito K, Kudo Y, Qin Y, Ueta Y, Komuro I. Angiotensin II type 1 receptor signaling regulates feeding behavior through anorexigenic corticotropin-releasing hormone in hypothalamus. J Biol Chem 2011; 286:21458-65. [PMID: 21525005 DOI: 10.1074/jbc.m110.192260] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The activation of renin-angiotensin system contributes to the development of metabolic syndrome and diabetes as well as hypertension. However, it remains undetermined how renin-angiotensin system is implicated in feeding behavior. Here, we show that angiotensin II type 1 (AT(1)) receptor signaling regulates the hypothalamic neurocircuit that is involved in the control of food intake. Compared with wild-type Agtr1a(+/+) mice, AT(1) receptor knock-out (Agtr1a(-/-)) mice were hyperphagic and obese with increased adiposity on an ad libitum diet, whereas Agtr1a(-/-) mice were lean with decreased adiposity on a pair-fed diet. In the hypothalamus, mRNA levels of anorexigenic neuropeptide corticotropin-releasing hormone (Crh) were lower in Agtr1a(-/-) mice than in Agtr1a(+/+) mice both on an ad libitum and pair-fed diet. Furthermore, intracerebroventricular administration of CRH suppressed food intake both in Agtr1a(+/+) and Agtr1a(-/-) mice. In addition, the Crh gene promoter was significantly transactivated via the cAMP-responsive element by angiotensin II stimulation. These results thus demonstrate that central AT(1) receptor signaling plays a homeostatic role in the regulation of food intake by maintaining gene expression of Crh in hypothalamus and suggest a therapeutic potential of central AT(1) receptor blockade in feeding disorders.
Collapse
Affiliation(s)
- Rie Yamamoto
- Department of Cardiovascular Science and Medicine, Chiba University Graduate School of Medicine, Chiba, Chiba 260-8670, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Scalia R, Gong Y, Berzins B, Freund B, Feather D, Landesberg G, Mishra G. A novel role for calpain in the endothelial dysfunction induced by activation of angiotensin II type 1 receptor signaling. Circ Res 2011; 108:1102-11. [PMID: 21415394 DOI: 10.1161/circresaha.110.229393] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE The cytosolic protease calpain has been recently implicated in the vascular remodeling of angiotensin II (Ang II) type 1 receptor (AT(1)R) signaling. The role of Ang II/AT(1)R/calpain signaling on endothelial function, an important and early determinant of vascular pathology, remains though totally unknown. Accordingly, we investigated the role of calpain in the endothelial dysfunction of Ang II. OBJECTIVE To demonstrate a mechanistic role for calpain in the endothelial dysfunction induced by Ang II/AT(1)R signaling. To establish endothelial-expressed calpains as an important target of AT(1)R signaling. METHODS AND RESULTS Subchronic administration of nonpressor doses of Ang II to rats and mice significantly increased vascular calpain activity via AT(1)R signaling. Intravital microscopy studies revealed that activation of vascular expressed calpains causes endothelial dysfunction with increased leukocyte-endothelium interactions and albumin permeability in the microcirculation. Western blot and immunohistochemistry studies confirmed that Ang II/AT(1)R signaling preferentially activates the constitutively expressed μ-calpain isoform and demonstrated a calpain-dependent degradation of IκBα, along with upregulation of nuclear factor κB-regulated endothelial cell adhesion molecules. These physiological and biochemical parameters were nearly normalized following inhibition of AT(1)R or calpain in vivo. RNA silencing studies in microvascular endothelial cells, along with knockout and transgenic mouse studies, further confirmed the role of μ-calpain in the endothelial adhesiveness induced by Ang II. CONCLUSIONS This study uncovers a novel role for calpain in the endothelial dysfunction of Ang II/AT(1)R signaling and establishes the calpain system as a novel molecular target of the vascular protective action of renin-angiotensin system inhibition. Our results may have significant clinical implications in vascular disease.
Collapse
Affiliation(s)
- Rosario Scalia
- Department of Physiology, and The Cardiovascular Research Center, Temple University, Philadelphia, PA 19140, USA.
| | | | | | | | | | | | | |
Collapse
|
97
|
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] [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
|
98
|
Wu C, Lu H, Cassis LA, Daugherty A. Molecular and Pathophysiological Features of Angiotensinogen: A Mini Review. ACTA ACUST UNITED AC 2011; 4:183-190. [PMID: 22389749 DOI: 10.7156/v4i4p183] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The renin-angiotensin system is an essential regulatory system for blood pressure and fluid homeostasis. Angiotensinogen is the only known precursor of all the peptides generated in this system. While many of the basic understandings of angiotensinogen have come from research efforts to define its role in blood pressure regulation, novel pathophysiological functions of angiotensinogen have been discovered in the last two decades including kidney developmental abnormalities, atherosclerosis, and obesity. Despite the impressive advance in the understanding of angiotensinogen gene structure and protein functions, some fundamental questions remain unanswered. In this short review, we provide contemporary insights into the molecular characteristics of angiotensinogen and its pathophysiological features. In light of the recent progress, we emphasize some newly recognized functional features of angiotensinogen other than its regulation on blood pressure.
Collapse
Affiliation(s)
- Congqing Wu
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, USA
| | | | | | | |
Collapse
|
99
|
Nahmod K, Gentilini C, Vermeulen M, Uharek L, Wang Y, Zhang J, Schultheiss HP, Geffner J, Walther T. Impaired function of dendritic cells deficient in angiotensin II type 1 receptors. J Pharmacol Exp Ther 2010; 334:854-62. [PMID: 20516139 DOI: 10.1124/jpet.109.161760] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2025] Open
Abstract
Dendritic cells (DC) are highly specialized antigen-presenting cells with a unique ability to activate resting T lymphocytes and initiate primary immune responses. Angiotensin II (AII) is involved in key events of the inflammatory response. Because our previous work implicated an effect of AII on differentiation and function of murine and human DC, we investigated the impact of AII type 1 receptor (AT(1)) deficiency on the phenotypical and functional properties of mouse DC in vitro and in vivo. Bone marrow (BM) cells isolated from mice lacking AII subtype 1a receptor (AT(1a)), AII subtype 1b receptor (AT(1b)), or both receptor isoforms and control littermates [wild type (WT)] were cultured for 7 days in the presence of recombinant mouse granulocyte/macrophage colony-stimulating factor to generate myeloid DC in vitro. Generation of CD11c(+) cells was less efficient in both AT(1a)- and AT(1b)-deficient BM cells than in WT BM cell cultures. Moreover, DC generated from AT(1)-deficient progenitors showed lower levels of expression of major histocompatibility complex II (MHC-II) and CD11c (p < 0.01) and a marked reduction in their allostimulatory activity (p < 0.01 or 0.001). Although AT(1)-deficient DC released comparable levels of interleukin (IL)-10 and IL-12p70 to WT DC, they produced significantly lower levels of tumor necrosis factor alpha (TNF-alpha) (p < 0.05). Remarkably, CD11c(+) cells isolated from the spleen of AT(1) knockout mice challenged with lipopolysaccharide in vivo up-regulated MHC-II, CD40, and CD80 as did WT, but released significantly lower levels of TNF-alpha (p < 0.01). These data provide clear evidence that AT(1) controls differentiation and functionality of DC and thus may have a crucial impact on inflammatory processes where local angiotensinergic systems are known to be activated.
Collapse
Affiliation(s)
- Karen Nahmod
- Laboratory of Immunology, Institute of Hematologic Research, National Academy of Medicine, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Ulmasov B, Xu Z, Talkad V, Oshima K, Neuschwander-Tetri BA. Angiotensin II signaling through the AT1a and AT1b receptors does not have a role in the development of cerulein-induced chronic pancreatitis in the mouse. Am J Physiol Gastrointest Liver Physiol 2010; 299:G70-80. [PMID: 20413721 PMCID: PMC7199229 DOI: 10.1152/ajpgi.00006.2010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The intraorgan renin-angiotensin system (RAS) plays an important role in the pathophysiology of a variety of diseases and has been implicated in fibrogenesis. The role of RAS in the development of chronic pancreatitis is not well established. The blockade of RAS in rat models with angiotensin-converting enzyme inhibitors (ACEi) or angiotensin receptor 1 (AT1) blockers (ARBs) mostly have reduced pancreatic inflammation and fibrosis with a few exceptions. At the same time, the use of ACEi and ARBs in humans is associated with a modest risk of acute pancreatitis. The aim of this study was to elucidate the effect of the AT1 signaling pathway in the development of pancreatitis using AT1a- and AT1b-deficient mice as well as the ARB losartan. Chronic pancreatitis was induced by repetitive cerulein administration in C57BL/6J wild-type (WT) and AT1a- and AT1b-deficient mice (AT1a-/- and AT1b-/-), and pancreatic injury was assessed at day 10. Pancreatic weight of cerulein treated groups was significantly reduced. There was severe parenchymal atrophy and fibrosis assessed by histological examination. Fibrosis was accompanied by activation of pancreatic stellate cells (PSC) evaluated by Western blot analysis for alpha-smooth muscle actin. No differences were seen between cerulein-treated WT, AT1a-/- , AT1b-/- mice, or losartan treated-WT mice with regards to morphological or molecular alterations induced by cerulein. Our results demonstrate that AT1a and AT1b receptor pathways do not seem to be essential for the development of pancreatitis in the mouse model of pancreatitis induced by repetitive cerulein injury.
Collapse
Affiliation(s)
| | - Zekuan Xu
- 3Pathology, Saint Louis University School of Medicine, St. Louis, Missouri;
| | | | - Kiyoko Oshima
- 2Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | | |
Collapse
|