1
|
Yamaguchi H, Gomez RA, Sequeira-Lopez MLS. Renin Cells, From Vascular Development to Blood Pressure Sensing. Hypertension 2023; 80:1580-1589. [PMID: 37313725 PMCID: PMC10526986 DOI: 10.1161/hypertensionaha.123.20577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
During embryonic and neonatal life, renin cells contribute to the assembly and branching of the intrarenal arterial tree. During kidney arteriolar development renin cells are widely distributed throughout the renal vasculature. As the arterioles mature, renin cells differentiate into smooth muscle cells, pericytes, and mesangial cells. In adult life, renin cells are confined to the tips of the renal arterioles, thus their name juxtaglomerular cells. Juxtaglomerular cells are sensors that release renin to control blood pressure and fluid-electrolyte homeostasis. Three major mechanisms control renin release: (1) β-adrenergic stimulation, (2) macula densa signaling, and (3) the renin baroreceptor, whereby a decrease in arterial pressure leads to increased renin release whereas an increase in pressure results in decrease renin release. Cells from the renin lineage exhibit plasticity in response to hypotension or hypovolemia, whereas relentless, chronic stimulation induces concentric arterial and arteriolar hypertrophy, leading to focal renal ischemia. The renin cell baroreceptor is a nuclear mechanotransducer within the renin cell that transmits external forces to the chromatin to regulate Ren1 gene expression. In addition to mechanotransduction, the pressure sensor of the renin cell may enlist additional molecules and structures including soluble signals and membrane proteins such as gap junctions and ion channels. How these various components integrate their actions to deliver the exact amounts of renin to meet the organism needs is unknown. This review describes the nature and origins of renin cells, their role in kidney vascular development and arteriolar diseases, and the current understanding of the blood pressure sensing mechanism.
Collapse
Affiliation(s)
- Hiroki Yamaguchi
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - R. Ariel Gomez
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Maria Luisa S. Sequeira-Lopez
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| |
Collapse
|
2
|
Neyra JS, Medrano S, Goes Martini AD, Sequeira-Lopez MLS, Gomez RA. The role of Gata3 in renin cell identity. Am J Physiol Renal Physiol 2023; 325:F188-F198. [PMID: 37345845 PMCID: PMC10396225 DOI: 10.1152/ajprenal.00098.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/09/2023] [Accepted: 06/14/2023] [Indexed: 06/23/2023] Open
Abstract
Renin cells are precursors for other cell types in the kidney and show high plasticity in postnatal life in response to challenges to homeostasis. Our previous single-cell RNA-sequencing studies revealed that the dual zinc-finger transcription factor Gata3, which is important for cell lineage commitment and differentiation, is expressed in mouse renin cells under normal conditions and homeostatic threats. We identified a potential Gata3-binding site upstream of the renin gene leading us to hypothesize that Gata3 is essential for renin cell identity. We studied adult mice with conditional deletion of Gata3 in renin cells: Gata3fl/fl;Ren1dCre/+ (Gata3-cKO) and control Gata3fl/fl;Ren1d+/+ counterparts. Gata3 immunostaining revealed that Gata3-cKO mice had significantly reduced Gata3 expression in juxtaglomerular, mesangial, and smooth muscle cells, indicating a high degree of deletion of Gata3 in renin lineage cells. Gata3-cKO mice exhibited a significant increase in blood urea nitrogen, suggesting hypovolemia and/or compromised renal function. By immunostaining, renin-expressing cells appeared very thin compared with their normal plump shape in control mice. Renin cells were ectopically localized to Bowman's capsule in some glomeruli, and there was aberrant expression of actin-α2 signals in the mesangium, interstitium, and Bowman's capsule in Gata3-cKO mice. Distal tubules showed dilated morphology with visible intraluminal casts. Under physiological threat, Gata3-cKO mice exhibited a lower increase in mRNA levels than controls. Hematoxylin-eosin, periodic acid-Schiff, and Masson's trichrome staining showed increased glomerular fusion, absent cubical epithelial cells in Bowman's capsule, intraglomerular aneurysms, and tubular dilation. In conclusion, our results indicate that Gata3 is crucial to the identity of cells of the renin lineage.NEW & NOTEWORTHY Gata3, a dual zinc-finger transcription factor, is responsible for the identity and localization of renin cells in the kidney. Mice with a conditional deletion of Gata3 in renin lineage cells have abnormal kidneys with juxtaglomerular cells that lose their characteristic location and are misplaced outside and around arterioles and glomeruli. The fundamental role of Gata3 in renin cell development offers a new model to understand how transcription factors control cell location, function, and pathology.
Collapse
Affiliation(s)
- Jesus S Neyra
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Silvia Medrano
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Alexandre De Goes Martini
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Maria Luisa S Sequeira-Lopez
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - R Ariel Gomez
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| |
Collapse
|
3
|
Márquez M, Muñoz M, Córdova A, Puebla M, Figueroa XF. Connexin 40-Mediated Regulation of Systemic Circulation and Arterial Blood Pressure. J Vasc Res 2023; 60:87-100. [PMID: 37331352 DOI: 10.1159/000531035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 05/05/2023] [Indexed: 06/20/2023] Open
Abstract
Vascular system is a complex network in which different cell types and vascular segments must work in concert to regulate blood flow distribution and arterial blood pressure. Although paracrine/autocrine signaling is involved in the regulation of vasomotor tone, direct intercellular communication via gap junctions plays a central role in the control and coordination of vascular function in the microvascular network. Gap junctions are made up by connexin (Cx) proteins, and among the four Cxs expressed in the cardiovascular system (Cx37, Cx40, Cx43, and Cx45), Cx40 has emerged as a critical signaling pathway in the vessel wall. This Cx is predominantly found in the endothelium, but it is involved in the development of the cardiovascular system and in the coordination of endothelial and smooth muscle cell function along the length of the vessels. In addition, Cx40 participates in the control of vasomotor tone through the transmission of electrical signals from the endothelium to the underlying smooth muscle and in the regulation of arterial blood pressure by renin-angiotensin system in afferent arterioles. In this review, we discuss the participation of Cx40-formed channels in the development of cardiovascular system, control and coordination of vascular function, and regulation of arterial blood pressure.
Collapse
Affiliation(s)
- Mónica Márquez
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Matías Muñoz
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexandra Córdova
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mariela Puebla
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Xavier F Figueroa
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
4
|
Jindal S, Chockalingam S, Ghosh SS, Packirisamy G. Connexin and gap junctions: perspectives from biology to nanotechnology based therapeutics. Transl Res 2021; 235:144-167. [PMID: 33582245 DOI: 10.1016/j.trsl.2021.02.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 01/10/2021] [Accepted: 02/09/2021] [Indexed: 12/11/2022]
Abstract
The concept of gap junctions and their role in intercellular communication has been known for around 50 years. Considerable progress has been made in understanding the fundamental biology of connexins in mediating gap junction intercellular communication (GJIC) and their role in various cellular processes including pathological conditions. However, this understanding has not led to development of advanced therapeutics utilizing GJIC. Inadequacies in strategies that target specific connexin protein in the affected tissue, with minimal or no collateral damage, are the primary reason for the lack of development of efficient therapeutic models. Herein, nanotechnology has a role to play, giving plenty of scope to circumvent these problems and develop more efficient connexin based therapeutics. AsODN, antisense oligodeoxynucleotides; BMPs, bone morphogenetic proteins; BMSCs, bone marrow stem cells; BG, bioglass; Cx, Connexin; CxRE, connexin-responsive elements; CoCr NPs, cobalt-chromium nanoparticles; cGAMP, cyclic guanosine monophosphate-adenosine monophosphate; cAMP, cyclic adenosine monophosphate; ERK1/2, extracellular signal-regulated kinase 1/2; EMT, epithelial-mesenchymal transition; EPA, eicosapentaenoic acids; FGFR1, fibroblast growth factor receptor 1; FRAP, fluorescence recovery after photobleaching; 5-FU, 5-fluorouracil; GJ, gap junction; GJIC, gap junctional intercellular communication; HGPRTase, hypoxanthine phosphoribosyltransferase; HSV-TK, herpes virus thymidine kinase; HSA, human serum albumin; HA, hyaluronic acid; HDAC, histone deacetylase; IRI, ischemia reperfusion injury; IL-6, interleukin-6; IL-8, interleukin-8; IONPs, iron-oxide nanoparticles; JNK, c-Jun N-terminal kinase; LAMP, local activation of molecular fluorescent probe; MSCs, mesenchymal stem cells; MMP, matrix metalloproteinase; MI, myocardial infarction; MAPK, mitogen-activated protein kinase; NF-κB, nuclear factor kappa B; NO, nitric oxide; PKC, protein kinase C; QDs, quantum dots; ROI, region of interest; RGO, reduced graphene oxide; siRNA, small interfering RNA; TGF-β1, transforming growth factor-β1; TNF-α, tumor necrosis factor-α; UCN, upconversion nanoparticles; VEGF, vascular endothelial growth factor. In this review, we discuss briefly the role of connexins and gap junctions in various physiological and pathological processes, with special emphasis on cancer. We further discuss the application of nanotechnology and tissue engineering in developing treatments for various connexin based disorders.
Collapse
Affiliation(s)
- Shlok Jindal
- Nanobiotechnology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - S Chockalingam
- Department of Biotechnology, National Institute of Technology Warangal, Warangal, Telangana, India
| | - Siddhartha Sankar Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Gopinath Packirisamy
- Nanobiotechnology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India; Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India.
| |
Collapse
|
5
|
Abstract
Renin cells are essential for survival perfected throughout evolution to ensure normal development and defend the organism against a variety of homeostatic threats. During embryonic and early postnatal life, they are progenitors that participate in the morphogenesis of the renal arterial tree. In adult life, they are capable of regenerating injured glomeruli, control blood pressure, fluid-electrolyte balance, tissue perfusion, and in turn, the delivery of oxygen and nutrients to cells. Throughout life, renin cell descendants retain the plasticity or memory to regain the renin phenotype when homeostasis is threatened. To perform all of these functions and maintain well-being, renin cells must regulate their identity and fate. Here, we review the major mechanisms that control the differentiation and fate of renin cells, the chromatin events that control the memory of the renin phenotype, and the major pathways that determine their plasticity. We also examine how chronic stimulation of renin cells alters their fate leading to the development of a severe and concentric hypertrophy of the intrarenal arteries and arterioles. Lastly, we provide examples of additional changes in renin cell fate that contribute to equally severe kidney disorders.
Collapse
Affiliation(s)
- Maria Luisa S. Sequeira-Lopez
- Departments of Pediatrics an Biology, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - R. Ariel Gomez
- Departments of Pediatrics an Biology, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| |
Collapse
|
6
|
Lozić M, Filipović N, Jurić M, Kosović I, Benzon B, Šolić I, Kelam N, Racetin A, Watanabe K, Katsuyama Y, Ogata M, Saraga-Babić M, Vukojević K. Alteration of Cx37, Cx40, Cx43, Cx45, Panx1, and Renin Expression Patterns in Postnatal Kidneys of Dab1-/- ( yotari) Mice. Int J Mol Sci 2021; 22:1284. [PMID: 33525532 PMCID: PMC7865779 DOI: 10.3390/ijms22031284] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/24/2021] [Accepted: 01/25/2021] [Indexed: 12/23/2022] Open
Abstract
Numerous evidence corroborates roles of gap junctions/hemichannels in proper kidney development. We analyzed how Dab1 gene functional silencing influences expression and localization of Cx37, Cx40, Cx43, Cx45, Panx1 and renin in postnatal kidneys of yotari mice, by using immunohistochemistry and electron microscopy. Dab1 Δ102/221 might lead to the activation of c-Src tyrosine kinase, causing the upregulation of Cx43 in the medulla of yotari mice. The expression of renin was more prominent in yotari mice (p < 0.001). Renin granules were unusually present inside the vascular walls of glomeruli capillaries, in proximal and distal convoluted tubules and in the medulla. Disfunction of Cx40 is likely responsible for increased atypically positioned renin cells which release renin in an uncontrolled fashion, but this doesn't rule out simultaneous involvement of other Cxs, such as Cx45 which was significantly increased in the yotari cortex. The decreased Cx37 expression in yotari medulla might contribute to hypertension reduction provoked by high renin expression. These findings imply the relevance of Cxs/Panx1 as markers of impaired kidney function (high renin) in yotari mice and that they have a role in the preservation of intercellular signaling and implicate connexopathies as the cause of premature death of yotari mice.
Collapse
Affiliation(s)
- Mirela Lozić
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia; (M.L.); (N.F.); (M.J.); (I.K.); (B.B.); (I.Š.); (N.K.); (A.R.); (M.S.-B.)
| | - Natalija Filipović
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia; (M.L.); (N.F.); (M.J.); (I.K.); (B.B.); (I.Š.); (N.K.); (A.R.); (M.S.-B.)
| | - Marija Jurić
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia; (M.L.); (N.F.); (M.J.); (I.K.); (B.B.); (I.Š.); (N.K.); (A.R.); (M.S.-B.)
| | - Ivona Kosović
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia; (M.L.); (N.F.); (M.J.); (I.K.); (B.B.); (I.Š.); (N.K.); (A.R.); (M.S.-B.)
| | - Benjamin Benzon
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia; (M.L.); (N.F.); (M.J.); (I.K.); (B.B.); (I.Š.); (N.K.); (A.R.); (M.S.-B.)
| | - Ivana Šolić
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia; (M.L.); (N.F.); (M.J.); (I.K.); (B.B.); (I.Š.); (N.K.); (A.R.); (M.S.-B.)
| | - Nela Kelam
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia; (M.L.); (N.F.); (M.J.); (I.K.); (B.B.); (I.Š.); (N.K.); (A.R.); (M.S.-B.)
| | - Anita Racetin
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia; (M.L.); (N.F.); (M.J.); (I.K.); (B.B.); (I.Š.); (N.K.); (A.R.); (M.S.-B.)
- Department of Medical Genetics, School of Medicine, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
| | - Koichiro Watanabe
- Department of Anatomy, Shiga University of Medical Science, Ötsu 520-2192, Japan; (K.W.); (Y.K.)
| | - Yu Katsuyama
- Department of Anatomy, Shiga University of Medical Science, Ötsu 520-2192, Japan; (K.W.); (Y.K.)
| | - Masaki Ogata
- Division of Anatomy, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, 981-Miyagi 8558, Japan;
| | - Mirna Saraga-Babić
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia; (M.L.); (N.F.); (M.J.); (I.K.); (B.B.); (I.Š.); (N.K.); (A.R.); (M.S.-B.)
| | - Katarina Vukojević
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia; (M.L.); (N.F.); (M.J.); (I.K.); (B.B.); (I.Š.); (N.K.); (A.R.); (M.S.-B.)
- Department of Medical Genetics, School of Medicine, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
| |
Collapse
|
7
|
Steglich A, Hickmann L, Linkermann A, Bornstein S, Hugo C, Todorov VT. Beyond the Paradigm: Novel Functions of Renin-Producing Cells. Rev Physiol Biochem Pharmacol 2020; 177:53-81. [PMID: 32691160 DOI: 10.1007/112_2020_27] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The juxtaglomerular renin-producing cells (RPC) of the kidney are referred to as the major source of circulating renin. Renin is the limiting factor in renin-angiotensin system (RAS), which represents a proteolytic cascade in blood plasma that plays a central role in the regulation of blood pressure. Further cells disseminated in the entire organism express renin at a low level as part of tissue RASs, which are thought to locally modulate the effects of systemic RAS. In recent years, it became increasingly clear that the renal RPC are involved in developmental, physiological, and pathophysiological processes outside RAS. Based on recent experimental evidence, a novel concept emerges postulating that next to their traditional role, the RPC have non-canonical RAS-independent progenitor and renoprotective functions. Moreover, the RPC are part of a widespread renin lineage population, which may act as a global stem cell pool coordinating homeostatic, stress, and regenerative responses throughout the organism. This review focuses on the RAS-unrelated functions of RPC - a dynamic research area that increasingly attracts attention.
Collapse
Affiliation(s)
- Anne Steglich
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Linda Hickmann
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Andreas Linkermann
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Stefan Bornstein
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Christian Hugo
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Vladimir T Todorov
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.
| |
Collapse
|
8
|
Le Gal L, Pellegrin M, Santoro T, Mazzolai L, Kurtz A, Meda P, Wagner C, Haefliger J. Connexin37-Dependent Mechanisms Selectively Contribute to Modulate Angiotensin II -Mediated Hypertension. J Am Heart Assoc 2019; 8:e010823. [PMID: 30943815 PMCID: PMC6507190 DOI: 10.1161/jaha.118.010823] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 01/30/2019] [Indexed: 12/23/2022]
Abstract
Background Gap junction channels made of Connexin37 (Cx37) are expressed by aortic endothelial and smooth muscle cells of hypertensive mice, as well as by the renin-secreting cells of kidneys. Methods and Results To decipher whether Cx37 has any role in hypertension, angiotensin II (Ang II ) was infused in normotensive wild-type and Cx37-deficient mice (Cx37-/-). After 2 to 4 weeks, the resulting increase in blood pressure was lower in Cx37-/- than in wild-type mice, suggesting an alteration in the Ang II response. To investigate this possibility, mice were submitted to a 2-kidney, 1-clip procedure, a renin-dependent model of hypertension. Two weeks after this clipping, Cx37-/- mice were less hypertensive than wild-type mice and, 2 weeks later, their blood pressure had returned to control values, in spite of abnormally high plasma renin levels. In contrast, Cx37-/- and wild-type mice that received N-nitro-l-arginine-methyl-ester, a renin-independent model of hypertension, featured a similar and sustained increase in blood pressure. The data indicate that loss of Cx37 selectively altered the Ang II -dependent pathways. Consistent with this conclusion, aortas of Cx37-/- mice featured an increased basal expression of the Ang II type 2 receptors ( AT 2R), and increased transcripts levels of downstream signaling proteins, such as Cnksr1 and Ptpn6 ( SHP -1). Accordingly, the response of Cx37-/- mice aortas to an ex vivo Ang II exposure was altered, since phosphorylation levels of several proteins of the Ang II pathway ( MLC 2, ERK , and AKT ) remained unchanged. Conclusions These findings provide evidence that Cx37 selectively influences Ang II signaling, mostly via a modulation of the expression of the Ang II type 2 receptor.
Collapse
Affiliation(s)
- Loïc Le Gal
- Department of MedicineUniversity of LausanneSwitzerland
| | - Maxime Pellegrin
- Division of AngiologyHeart and Vessel DepartmentCentre Hospitalier Universitaire VaudoisUniversity of LausanneSwitzerland
| | - Tania Santoro
- Department of MedicineUniversity of LausanneSwitzerland
| | - Lucia Mazzolai
- Division of AngiologyHeart and Vessel DepartmentCentre Hospitalier Universitaire VaudoisUniversity of LausanneSwitzerland
| | - Armin Kurtz
- Department of PhysiologyUniversity of RegensburgGermany
| | - Paolo Meda
- Department of Cell Physiology and MetabolismSchool of MedicineCMUUniversity of GenevaSwitzerland
| | | | | |
Collapse
|
9
|
Oliveira M, Lira R, Freire T, Luna C, Martins M, Almeida A, Carvalho S, Cortez E, Stumbo AC, Thole A, Carvalho L. Bone marrow mononuclear cell transplantation rescues the glomerular filtration barrier and epithelial cellular junctions in a renovascular hypertension model. Exp Physiol 2019; 104:740-754. [DOI: 10.1113/ep087330] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 02/19/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Mariana Oliveira
- Laboratory of Stem Cell ResearchHistology and Embryology DepartmentBiology InstituteState University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Rafaelle Lira
- Laboratory of Stem Cell ResearchHistology and Embryology DepartmentBiology InstituteState University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Thiago Freire
- Laboratory of Stem Cell ResearchHistology and Embryology DepartmentBiology InstituteState University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Camila Luna
- Laboratory of Stem Cell ResearchHistology and Embryology DepartmentBiology InstituteState University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Marcela Martins
- Laboratory of Stem Cell ResearchHistology and Embryology DepartmentBiology InstituteState University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Aline Almeida
- Laboratory of Stem Cell ResearchHistology and Embryology DepartmentBiology InstituteState University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Simone Carvalho
- Laboratory of Stem Cell ResearchHistology and Embryology DepartmentBiology InstituteState University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Erika Cortez
- Laboratory of Stem Cell ResearchHistology and Embryology DepartmentBiology InstituteState University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Ana Carolina Stumbo
- Laboratory of Stem Cell ResearchHistology and Embryology DepartmentBiology InstituteState University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Alessandra Thole
- Laboratory of Stem Cell ResearchHistology and Embryology DepartmentBiology InstituteState University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Lais Carvalho
- Laboratory of Stem Cell ResearchHistology and Embryology DepartmentBiology InstituteState University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
10
|
Novielli-Kuntz NM, Jelen M, Barr K, DeLalio LJ, Feng Q, Isakson BE, Gros R, Laird DW. Ablation of both Cx40 and Panx1 results in similar cardiovascular phenotypes exhibited in Cx40 knockout mice. Biosci Rep 2019; 39:BSR20182350. [PMID: 30745457 PMCID: PMC6393227 DOI: 10.1042/bsr20182350] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/10/2019] [Accepted: 02/05/2019] [Indexed: 11/30/2022] Open
Abstract
Connexins (Cxs) and pannexins (Panxs) are highly regulated large-pore channel-forming proteins that participate in cellular communication via small molecular exchange with the extracellular microenvironment, or in the case of connexins, directly between cells. Given the putative functional overlap between single membrane-spanning connexin hemichannels and Panx channels, and cardiovascular system prevalence, we generated the first Cx40-/-Panx1-/- mouse with the anticipation that this genetic modification would lead to a severe cardiovascular phenotype. Mice null for both Cx40 and Panx1 produced litter sizes and adult growth progression similar to wild-type (WT), Cx40-/- and Panx1-/- mice. Akin to Cx40-/- mice, Cx40-/-Panx1-/- mice exhibited cardiac hypertrophy and elevated systolic, diastolic, and mean arterial blood pressure compared with WT and Panx1-/- mice; however assessment of left ventricular ejection fraction and fractional shortening revealed no evidence of cardiac dysfunction between groups. Furthermore, Cx40-/-, Panx1-/-, and Cx40-/-Panx1-/- mice demonstrated impaired endothelial-mediated vasodilation of aortic segments to increasing concentrations of methacholine (MCh) compared with WT, highlighting roles for both Cx40 and Panx1 in vascular endothelial cell (EC) function. Surprisingly, elevated kidney renin mRNA expression, plasma renin activity, and extraglomerular renin-producing cell populations found in Cx40-/- mice was further exaggerated in double knockout mice. Thus, while gestation and gross development were conserved in Cx40-/-Panx1-/- mice, they exhibit cardiac hypertrophy, hypertension, and impaired endothelial-mediated vasodilation that phenocopies Cx40-/- mice. Nevertheless, the augmented renin homeostasis observed in the double knockout mice suggests that both Cx40 and Panx1 may play an integrative role.
Collapse
Affiliation(s)
| | - Meghan Jelen
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Canada
| | - Kevin Barr
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Canada
| | - Leon J DeLalio
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, U.S.A
| | - Qingping Feng
- Department of Physiology and Pharmacology London, ON, Canada
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, U.S.A
| | - Robert Gros
- Department of Physiology and Pharmacology London, ON, Canada
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Dale W Laird
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Canada
- Department of Physiology and Pharmacology London, ON, Canada
| |
Collapse
|
11
|
Hassan GS, Jacques D, D'Orléans-Juste P, Magder S, Bkaily G. Physical contact between human vascular endothelial and smooth muscle cells modulates cytosolic and nuclear calcium homeostasis. Can J Physiol Pharmacol 2018; 96:655-661. [PMID: 29756482 DOI: 10.1139/cjpp-2018-0093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The interaction between vascular endothelial cells (VECs) and vascular smooth muscle cells (VSMCs) plays an important role in the modulation of vascular tone. There is, however, no information on whether direct physical communication regulates the intracellular calcium levels of human VECs (hVECs) and (or) human VSMCs (hVSMCs). Thus, the objective of the study is to verify whether co-culture of hVECs and hVSMCs modulates cytosolic ([Ca2+]c) and nuclear calcium ([Ca2+]n) levels via physical contact and (or) factors released by both cell types. Quantitative 3D confocal microscopy for [Ca2+]c and [Ca2+]n measurement was performed in cultured hVECs or hVSMCs or in co-culture of hVECs-hVSMCs. Our results show that: (1) physical contact between hVECs-hVECs or hVSMCs-hVSMCs does not affect [Ca2+]c and [Ca2+]n in these 2 cell types; (2) physical contact between hVECs and hVSMCs induces a significant increase only of [Ca2+]n of hVECs without affecting the level of [Ca2+]c and [Ca2+]n of hVSMCs; and (3) preconditioned culture medium of hVECs or hVSMCs does not affect [Ca2+]c and [Ca2+]n of both types of cells. We concluded that physical contact between hVECs and hVSMCs only modulates [Ca2+]n in hVECs. The increase of [Ca2+]n in hVECs may modulate nuclear functions that are calcium dependent.
Collapse
Affiliation(s)
- Ghada S Hassan
- a Department of Anatomy and Cell Biology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Danielle Jacques
- a Department of Anatomy and Cell Biology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Pedro D'Orléans-Juste
- b Department of Pharmacology and Physiology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Sheldon Magder
- c McGill University Health Centre, Montreal, QC H3A 1A1, Canada
| | - Ghassan Bkaily
- a Department of Anatomy and Cell Biology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| |
Collapse
|
12
|
Affiliation(s)
- Michael Bader
- From the Max Delbrück Center of Molecular Medicine, Berlin, Germany; Charite-University Medicine, Berlin, Germany; Berlin Institute of Health (BIH), Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Germany; and Institute for Biology, University of Lübeck, Germany.
| |
Collapse
|
13
|
Meda P. Gap junction proteins are key drivers of endocrine function. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:124-140. [PMID: 28284720 DOI: 10.1016/j.bbamem.2017.03.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/03/2017] [Accepted: 03/06/2017] [Indexed: 01/07/2023]
Abstract
It has long been known that the main secretory cells of exocrine and endocrine glands are connected by gap junctions, made by a variety of connexin species that ensure their electrical and metabolic coupling. Experiments in culture systems and animal models have since provided increasing evidence that connexin signaling contributes to control the biosynthesis and release of secretory products, as well as to the life and death of secretory cells. More recently, genetic studies have further provided the first lines of evidence that connexins also control the function of human glands, which are central to the pathogenesis of major endocrine diseases. Here, we summarize the recent information gathered on connexin signaling in these systems, since the last reviews on the topic, with particular regard to the pancreatic beta cells which produce insulin, and the renal cells which produce renin. These cells are keys to the development of various forms of diabetes and hypertension, respectively, and combine to account for the exploding, worldwide prevalence of the metabolic syndrome. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- Paolo Meda
- Department of Cell Physiology and Metabolism, University of Geneva Medical School, Switzerland.
| |
Collapse
|
14
|
Šeda O, Křenová D, Oliyarnyk O, Šedová L, Krupková M, Liška F, Chylíková B, Kazdová L, Křen V. Heterozygous connexin 50 mutation affects metabolic syndrome attributes in spontaneously hypertensive rat. Lipids Health Dis 2016; 15:199. [PMID: 27871290 PMCID: PMC5117636 DOI: 10.1186/s12944-016-0376-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 11/14/2016] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Several members of connexin family of transmembrane proteins were previously implicated in distinct metabolic conditions. In this study we aimed to determine the effects of complete and heterozygous form of connexin50 gene (Gja8) mutation L7Q on metabolic profile and oxidative stress parameters in spontaneously hypertensive inbred rat strain (SHR). METHODS Adult, standard chow-fed male rats of SHR, heterozygous SHR-Dca+/- and SHR-Dca-/- coisogenic strains were used. At the age of 4 months, dexamethasone (2.6 μg/ml) was administered in the drinking water for three days. The lipidemic profile (cholesterol and triacylglycerol concentration in 20 lipoprotein fractions, chylomicron, VLDL, LDL and HDL particle sizes) together with 33 cytokines and hormones in serum and several oxidative stress parameters in plasma, liver, kidney and heart were assessed. RESULTS SHR and SHR-Dca-/- rats had similar concentrations of triacylglycerols and cholesterol in all major lipoprotein fractions. The heterozygotes reached significantly highest levels of total (SHR-Dca+/-: 51.3 ± 7.2 vs. SHR: 34.5 ± 2.4 and SHR-Dca-/-: 34.4 ± 2.5 mg/dl, p = 0.026), chylomicron and VLDL triacylglycerols. The heterozygotes showed significantly lowest values of HDL cholesterol (40.9 ± 2.3 mg/dl) compared both to SHR (51.8 ± 2.2 mg/dl) and SHR-Dca-/- (48.6 ± 2.7 mg/dl). Total and LDL cholesterol in SHR-Dca+/- was lower compared to SHR. Glucose tolerance was improved and insulin concentrations were lowest in SHR-Dca-/- (1.11 ± 0.20 pg/ml) in comparison with both SHR (2.32 ± 0.49 pg/ml) and SHR-Dca+/- (3.04 ± 0.21 pg/ml). The heterozygous rats showed profile suggestive of increased oxidative stress as well as highest serum concentrations of several pro-inflammatory cytokines including interleukins 6, 12, 17, 18 and tumor necrosis factor alpha. CONCLUSIONS Our results demonstrate that connexin50 mutation in heterozygous state affects significantly the lipid profile and the oxidative stress parameters in the spontaneously hypertensive rat strain.
Collapse
Affiliation(s)
- Ondřej Šeda
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, 128 00, Prague 2, Czech Republic. .,Division BIOCEV, Institute of Molecular Genetics of the Academy of Sciences of the Czech Republic, Laboratory of Rat Models of Metabolic Disorders, Vídeňská 1083, 142 20, Prague 4, Czech Republic.
| | - Drahomíra Křenová
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, 128 00, Prague 2, Czech Republic
| | - Olena Oliyarnyk
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Vídeňská 1958/9, 140 21, Prague 4, Czech Republic
| | - Lucie Šedová
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, 128 00, Prague 2, Czech Republic.,Division BIOCEV, Institute of Molecular Genetics of the Academy of Sciences of the Czech Republic, Laboratory of Rat Models of Metabolic Disorders, Vídeňská 1083, 142 20, Prague 4, Czech Republic
| | - Michaela Krupková
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, 128 00, Prague 2, Czech Republic
| | - František Liška
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, 128 00, Prague 2, Czech Republic
| | - Blanka Chylíková
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, 128 00, Prague 2, Czech Republic
| | - Ludmila Kazdová
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Vídeňská 1958/9, 140 21, Prague 4, Czech Republic
| | - Vladimír Křen
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, 128 00, Prague 2, Czech Republic
| |
Collapse
|
15
|
Wang LJ, Liu WD, Zhang L, Ma KT, Zhao L, Shi WY, Zhang WW, Wang YZ, Li L, Si JQ. Enhanced expression of Cx43 and gap junction communication in vascular smooth muscle cells of spontaneously hypertensive rats. Mol Med Rep 2016; 14:4083-4090. [PMID: 27748857 PMCID: PMC5101886 DOI: 10.3892/mmr.2016.5783] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 08/30/2016] [Indexed: 11/24/2022] Open
Abstract
Niflumic acid (NFA) is a novel gap junction (GJ) inhibitor. The aim of the present study was to investigate the effect of NFA on GJ communication and the expression of connexin (Cx) in vascular smooth muscle cells (VSMCs) of mesenteric arterioles of spontaneously hypertensive rats (SHR). Whole-cell patch clamp recording demonstrated that NFA at 1×10–4 M significantly inhibited the inward current and its effect was reversible. The time for charging and discharging of cell membrane capacitance (Cinput) reduced from 9.73 to 0.48 ms (P<0.05; n=6). Pressure myograph measurement showed that NFA at 3×10-4 M fully neutralized the contraction caused by phenylephrine. The relaxation responses of normotensive control Wistar Kyoto (WKY) rats were significantly higher, compared with those of the SHRs (P<0.05; n=6). Western blot and reverse transcription-quantitative polymerase chain reaction analyses showed that the mRNA and protein expression levels of Cx43 of the third-level branch of mesenteric arterioles of the SHRs and WKY rats were higher, compared with those of the first-level branch. The mRNA and protein expression levels of Cx43 of the primary and third-level branches of the mesenteric arterioles in the SHRs were higher, compared with those in the WKY rats (P<0.05; n=6). The mRNA levels of Cx43 in the mesenteric arterioles were significantly downregulated by NFA in a concentration-dependent manner (P<0.01; n=6). The protein levels of Cx43 in primary cultured VSMCs isolated from the mesenteric arterioles were also significantly downregulated by NFA in a concentration-dependent manner (P<0.01; n=6). These results showed that the vasorelaxatory effects of GJ inhibitors were reduced in the SHRs, which was associated with a higher protein expression level of Cx43 in the mesenteric arterioles of the SHRs. NFA also relaxed the mesenteric arterioles by reducing the expression of Cx43, which decreased blood pressure. Therefore, regulation of the expression of GJs may be a therapeutic target for the treatment of hypertension.
Collapse
Affiliation(s)
- Li-Jie Wang
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Wei-Dong Liu
- Department of Gastroenterology, The People's Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, Xinjiang 830001, P.R. China
| | - Liang Zhang
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Ke-Tao Ma
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Lei Zhao
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Wen-Yan Shi
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Wen-Wen Zhang
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Ying-Zi Wang
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Li Li
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Jun-Qiang Si
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| |
Collapse
|
16
|
Yang R, Hu ZL, Kong QT, Li WW, Zhang LL, Du X, Huang SY, Xia XY, Sang H. A known mutation in GJB6 in a large Chinese family with hidrotic ectodermal dysplasia. J Eur Acad Dermatol Venereol 2016; 30:1362-5. [PMID: 27137747 DOI: 10.1111/jdv.13600] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Accepted: 12/16/2015] [Indexed: 12/18/2022]
Affiliation(s)
- R. Yang
- Department of Dermatology; Jinling Hospital; School of Medicine; Nanjing University; Nanjing China
| | - ZL. Hu
- Department of Dermatology; Jinling Hospital; School of Medicine; Nanjing University; Nanjing China
| | - QT. Kong
- Department of Dermatology; Jinling Hospital; School of Medicine; Nanjing University; Nanjing China
| | - WW. Li
- Department of Reproduction and Genetics; Institute of Laboratory Medicine; Jinling Hospital; School of Medicine; Nanjing University; Nanjing China
| | - LL. Zhang
- Department of Dermatology; Jinling Hospital; School of Medicine; Nanjing University; Nanjing China
| | - X. Du
- Department of Dermatology; Jinling Hospital; School of Medicine; Nanjing University; Nanjing China
| | - SY. Huang
- Department of Dermatology; Jinling Hospital; School of Medicine; Nanjing University; Nanjing China
| | - XY. Xia
- Department of Reproduction and Genetics; Institute of Laboratory Medicine; Jinling Hospital; School of Medicine; Nanjing University; Nanjing China
| | - H. Sang
- Department of Dermatology; Jinling Hospital; School of Medicine; Nanjing University; Nanjing China
| |
Collapse
|