1
|
Tamiato A, Tombor LS, Fischer A, Muhly-Reinholz M, Vanicek LR, Toğru BN, Neitz J, Glaser SF, Merten M, Rodriguez Morales D, Kwon J, Klatt S, Schumacher B, Günther S, Abplanalp WT, John D, Fleming I, Wettschureck N, Dimmeler S, Luxán G. Age-Dependent RGS5 Loss in Pericytes Induces Cardiac Dysfunction and Fibrosis. Circ Res 2024; 134:1240-1255. [PMID: 38563133 PMCID: PMC11081481 DOI: 10.1161/circresaha.123.324183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/11/2024] [Accepted: 03/20/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Pericytes are capillary-associated mural cells involved in the maintenance and stability of the vascular network. Although aging is one of the main risk factors for cardiovascular disease, the consequences of aging on cardiac pericytes are unknown. METHODS In this study, we have combined single-nucleus RNA sequencing and histological analysis to determine the effects of aging on cardiac pericytes. Furthermore, we have conducted in vivo and in vitro analysis of RGS5 (regulator of G-protein signaling 5) loss of function and finally have performed pericytes-fibroblasts coculture studies to understand the effect of RGS5 deletion in pericytes on the neighboring fibroblasts. RESULTS Aging reduced the pericyte area and capillary coverage in the murine heart. Single-nucleus RNA sequencing analysis further revealed that the expression of Rgs5 was reduced in cardiac pericytes from aged mice. In vivo and in vitro studies showed that the deletion of RGS5 impaired cardiac function, induced fibrosis, and morphological changes in pericytes characterized by a profibrotic gene expression signature and the expression of different ECM (extracellular matrix) components and growth factors, for example, TGFB2 and PDGFB. Indeed, culturing fibroblasts with the supernatant of RGS5-deficient pericytes induced their activation as evidenced by the increased expression of αSMA (alpha smooth muscle actin) in a TGFβ (transforming growth factor beta)2-dependent mechanism. CONCLUSIONS Our results have identified RGS5 as a crucial regulator of pericyte function during cardiac aging. The deletion of RGS5 causes cardiac dysfunction and induces myocardial fibrosis, one of the hallmarks of cardiac aging.
Collapse
Affiliation(s)
- Anita Tamiato
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
- Cardiopulmonary Institute (A.T., L.S.T., S.F.G., M.M., S.K., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.), Goethe University Frankfurt, Germany
- German Center for Cardiovascular Research Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Frankfurt am Main, Germany (A.T., L.S.T., S.F.G., M.M., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.)
| | - Lukas S. Tombor
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
- Cardiopulmonary Institute (A.T., L.S.T., S.F.G., M.M., S.K., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.), Goethe University Frankfurt, Germany
- German Center for Cardiovascular Research Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Frankfurt am Main, Germany (A.T., L.S.T., S.F.G., M.M., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.)
| | - Ariane Fischer
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
| | - Marion Muhly-Reinholz
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
| | - Leah Rebecca Vanicek
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
| | - Büşra Nur Toğru
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
| | - Jessica Neitz
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
| | - Simone Franziska Glaser
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
- German Center for Cardiovascular Research Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Frankfurt am Main, Germany (A.T., L.S.T., S.F.G., M.M., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.)
| | - Maximilian Merten
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
- Cardiopulmonary Institute (A.T., L.S.T., S.F.G., M.M., S.K., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.), Goethe University Frankfurt, Germany
- German Center for Cardiovascular Research Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Frankfurt am Main, Germany (A.T., L.S.T., S.F.G., M.M., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.)
| | - David Rodriguez Morales
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
| | - Jeonghyeon Kwon
- Department of Pharmacology (J.K., N.W.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stephan Klatt
- Cardiopulmonary Institute (A.T., L.S.T., S.F.G., M.M., S.K., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.), Goethe University Frankfurt, Germany
- Institute for Vascular Signalling, Center of Molecular Medicine (S.K., I.F.), Goethe University Frankfurt, Germany
| | - Bianca Schumacher
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
- Cardiopulmonary Institute (A.T., L.S.T., S.F.G., M.M., S.K., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.), Goethe University Frankfurt, Germany
- German Center for Cardiovascular Research Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Frankfurt am Main, Germany (A.T., L.S.T., S.F.G., M.M., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.)
| | - Stefan Günther
- Cardiopulmonary Institute (A.T., L.S.T., S.F.G., M.M., S.K., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.), Goethe University Frankfurt, Germany
- German Center for Cardiovascular Research Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Frankfurt am Main, Germany (A.T., L.S.T., S.F.G., M.M., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.)
- Bioinformatics and Deep Sequencing Platform (S.G.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Wesley T. Abplanalp
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
- Cardiopulmonary Institute (A.T., L.S.T., S.F.G., M.M., S.K., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.), Goethe University Frankfurt, Germany
- German Center for Cardiovascular Research Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Frankfurt am Main, Germany (A.T., L.S.T., S.F.G., M.M., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.)
| | - David John
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
- Cardiopulmonary Institute (A.T., L.S.T., S.F.G., M.M., S.K., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.), Goethe University Frankfurt, Germany
- German Center for Cardiovascular Research Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Frankfurt am Main, Germany (A.T., L.S.T., S.F.G., M.M., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.)
| | - Ingrid Fleming
- Cardiopulmonary Institute (A.T., L.S.T., S.F.G., M.M., S.K., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.), Goethe University Frankfurt, Germany
- Institute for Vascular Signalling, Center of Molecular Medicine (S.K., I.F.), Goethe University Frankfurt, Germany
- German Center for Cardiovascular Research Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Frankfurt am Main, Germany (A.T., L.S.T., S.F.G., M.M., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.)
| | - Nina Wettschureck
- Cardiopulmonary Institute (A.T., L.S.T., S.F.G., M.M., S.K., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.), Goethe University Frankfurt, Germany
- German Center for Cardiovascular Research Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Frankfurt am Main, Germany (A.T., L.S.T., S.F.G., M.M., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.)
- Department of Pharmacology (J.K., N.W.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stefanie Dimmeler
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
- Cardiopulmonary Institute (A.T., L.S.T., S.F.G., M.M., S.K., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.), Goethe University Frankfurt, Germany
- German Center for Cardiovascular Research Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Frankfurt am Main, Germany (A.T., L.S.T., S.F.G., M.M., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.)
| | - Guillermo Luxán
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine (A.T., L.S.T., A.F., M.M.-R., L.R.V., B.N.T., J.N., S.F.G., M.M., D.R.M., B.S., W.T.A., D.J., S.D., G.L.), Goethe University Frankfurt, Germany
- Cardiopulmonary Institute (A.T., L.S.T., S.F.G., M.M., S.K., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.), Goethe University Frankfurt, Germany
- German Center for Cardiovascular Research Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Frankfurt am Main, Germany (A.T., L.S.T., S.F.G., M.M., B.S., S.G., W.T.A., D.J., I.F., N.W., S.D., G.L.)
| |
Collapse
|
2
|
Carlsson R, Enström A, Paul G. Molecular Regulation of the Response of Brain Pericytes to Hypoxia. Int J Mol Sci 2023; 24:5671. [PMID: 36982744 PMCID: PMC10053233 DOI: 10.3390/ijms24065671] [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: 02/19/2023] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
The brain needs sufficient oxygen in order to function normally. This is achieved by a large vascular capillary network ensuring that oxygen supply meets the changing demand of the brain tissue, especially in situations of hypoxia. Brain capillaries are formed by endothelial cells and perivascular pericytes, whereby pericytes in the brain have a particularly high 1:1 ratio to endothelial cells. Pericytes not only have a key location at the blood/brain interface, they also have multiple functions, for example, they maintain blood-brain barrier integrity, play an important role in angiogenesis and have large secretory abilities. This review is specifically focused on both the cellular and the molecular responses of brain pericytes to hypoxia. We discuss the immediate early molecular responses in pericytes, highlighting four transcription factors involved in regulating the majority of transcripts that change between hypoxic and normoxic pericytes and their potential functions. Whilst many hypoxic responses are controlled by hypoxia-inducible factors (HIF), we specifically focus on the role and functional implications of the regulator of G-protein signaling 5 (RGS5) in pericytes, a hypoxia-sensing protein that is regulated independently of HIF. Finally, we describe potential molecular targets of RGS5 in pericytes. These molecular events together contribute to the pericyte response to hypoxia, regulating survival, metabolism, inflammation and induction of angiogenesis.
Collapse
Affiliation(s)
- Robert Carlsson
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Centre and Wallenberg Centre for Molecular Medicine, Lund University, 22184 Lund, Sweden
| | - Andreas Enström
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Centre and Wallenberg Centre for Molecular Medicine, Lund University, 22184 Lund, Sweden
| | - Gesine Paul
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Centre and Wallenberg Centre for Molecular Medicine, Lund University, 22184 Lund, Sweden
- Department of Neurology, Scania University Hospital, 22185 Lund, Sweden
| |
Collapse
|
3
|
Chen T, Song S, Jiang H, Lian H, Hu S. Single Cell Sequencing Reveals Mechanisms of Persistent Truncus Arteriosus Formation after PDGFRα and PDGFRβ Double Knockout in Cardiac Neural Crest Cells. Genes (Basel) 2022; 13:genes13101708. [PMID: 36292593 PMCID: PMC9601305 DOI: 10.3390/genes13101708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/13/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022] Open
Abstract
Persistent truncus arteriosus (PTA) is an uncommon and complex congenital cardiac malformation accounting for about 1.2% of all congenital heart diseases (CHDs), which is caused by a deficiency in the embryonic heart outflow tract’s (OFT) septation and remodeling. PDGFRα and PDGFRβ double knockout (DKO) in cardiac neural crest cells (CNCCs) has been reported to cause PTA, but the underlying mechanisms remain unclear. Here, we constructed a PTA mouse model with PDGFRα and PDGFRβ double knockout in Pax3+ CNCCs and described the condensation failure into OFT septum of CNCC-derived cells due to disturbance of cell polarity in the DKO group. In addition, we further explored the mechanism with single-cell RNA sequencing. We found that two main cell differentiation trajectories into vascular smooth muscle cells (VSMCs) from cardiomyocytes (CMs) and mesenchymal cells (MSs), respectively, were interrupted in the DKO group. The process of CM differentiation into VSMC stagnated in a transitional CM I-like state, which contributed to the failure of OFT remodeling and muscular septum formation. On the other hand, a Penk+ transitional MS II cluster closely related to cell condensation into the OFT septum disappeared, which led to the OFT’s septation absence directly. In conclusion, the disturbance of CNCC-derived cells caused by PDGFRα and PDGFRβ knockout can lead to the OFT septation disorder and the occurrence of PTA.
Collapse
Affiliation(s)
- Tianyun Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100006, China
| | - Shen Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100006, China
| | - Haobin Jiang
- Division of Thoracic Surgery, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou 310027, China
| | - Hong Lian
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100006, China
| | - Shengshou Hu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100006, China
- Correspondence:
| |
Collapse
|
4
|
A regulator of G protein signaling 5 marked subpopulation of vascular smooth muscle cells is lost during vascular disease. PLoS One 2022; 17:e0265132. [PMID: 35320283 PMCID: PMC8942229 DOI: 10.1371/journal.pone.0265132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/23/2022] [Indexed: 11/19/2022] Open
Abstract
Vascular smooth muscle cell (VSMC) subpopulations relevant to vascular disease and injury repair have been depicted in healthy vessels and atherosclerosis profiles. However, whether VSMC subpopulation associated with vascular homeostasis exists in the healthy artery and how are their nature and fate in vascular remodeling remains elusive. Here, using single-cell RNA-sequencing (scRNA-seq) to detect VSMC functional heterogeneity in an unbiased manner, we showed that VSMC subpopulations in healthy artery presented transcriptome diversity and that there was significant heterogeneity in differentiation state and development within each subpopulation. Notably, we detected an independent subpopulation of VSMCs that highly expressed regulator of G protein signaling 5 (RGS5), upregulated the genes associated with inhibition of cell proliferation and construction of cytoskeleton compared with the general subpopulation, and mainly enriched in descending aorta. Additionally, the proportion of RGS5high VSMCs was markedly decreased or almost disappeared in the vascular tissues of neointimal formation, abdominal aortic aneurysm and atherosclerosis. Specific spatiotemporal characterization of RGS5high VSMC subpopulation suggested that this subpopulation was implicated in vascular homeostasis. Together, our analyses identify homeostasis-relevant transcriptional signatures of VSMC subpopulations in healthy artery, which may explain the regional vascular resistance to atherosclerosis at some extent.
Collapse
|
5
|
Guan X, Xin H, Xu M, Ji J, Li J. The Role and Mechanism of SIRT6 in Regulating Phenotype Transformation of Vascular Smooth Muscle Cells in Abdominal Aortic Aneurysm. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:3200798. [PMID: 35035519 PMCID: PMC8758316 DOI: 10.1155/2022/3200798] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND Data mining of current gene expression databases has not been previously performed to determine whether sirtuin 6 (SIRT6) expression participates in the pathological process of abdominal aortic aneurysm (AAA). The present study was aimed at investigating the role and mechanism of SIRT6 in regulating phenotype transformation of vascular smooth muscle cells (VSMC) in AAA. METHODS Three gene expression microarray datasets of AAA patients in the Gene Expression Omnibus (GEO) database and one dataset of SIRT6-knockout (KO) mice were selected, and the differentially expressed genes (DEGs) were identified using GEO2R. Furthermore, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses of both the AAA-related DEGs and the SIRT6-related DEGs were conducted. RESULTS GEO2R analysis showed that the expression of SIRT6 was downregulated for three groups and upregulated for one group in the three datasets, and none of them satisfied statistical significance. There were top 5 DEGs (KYNU, NPTX2, SCRG1, GRK5, and RGS5) in both of the human AAA group and SIRT6-KO mouse group. Top 25 ontology of the SIRT6-KO-related DEGs showed that several pathways including tryptophan catabolic process to kynurenine and negative regulation of cell growth were enriched in the tissues of thickness aortic wall biopsies of AAA patients. CONCLUSIONS Although SIRT6 mRNA level itself did not change among AAA patients, SIRT6 may play an important role in regulating several signaling pathways with significant association with AAA, suggesting that SIRT6 mRNA upregulation is a protective factor for VSMC against AAA.
Collapse
Affiliation(s)
- Xiaomei Guan
- Department of Vascular Surgery, Affiliated Hospital of Qingdao University, Qingdao 266700, China
| | - Hai Xin
- Department of Vascular Surgery, Affiliated Hospital of Qingdao University, Qingdao 266700, China
| | - Meiling Xu
- Department of Interventional Operating Room, Affiliated Hospital of Qingdao University, Qingdao 266700, China
| | - Jianlei Ji
- Department of Kidney Transplantation, Affiliated Hospital of Qingdao University, Qingdao 266700, China
| | - Jun Li
- Department of Vascular Surgery, Affiliated Hospital of Qingdao University, Qingdao 266700, China
| |
Collapse
|
6
|
Demirel E, Arnold C, Garg J, Jäger MA, Sticht C, Li R, Kuk H, Wettschureck N, Hecker M, Korff T. RGS5 Attenuates Baseline Activity of ERK1/2 and Promotes Growth Arrest of Vascular Smooth Muscle Cells. Cells 2021; 10:1748. [PMID: 34359918 PMCID: PMC8306326 DOI: 10.3390/cells10071748] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/25/2021] [Accepted: 07/07/2021] [Indexed: 01/14/2023] Open
Abstract
The regulator of G-protein signaling 5 (RGS5) acts as an inhibitor of Gαq/11 and Gαi/o activity in vascular smooth muscle cells (VSMCs), which regulate arterial tone and blood pressure. While RGS5 has been described as a crucial determinant regulating the VSMC responses during various vascular remodeling processes, its regulatory features in resting VSMCs and its impact on their phenotype are still under debate and were subject of this study. While Rgs5 shows a variable expression in mouse arteries, neither global nor SMC-specific genetic ablation of Rgs5 affected the baseline blood pressure yet elevated the phosphorylation level of the MAP kinase ERK1/2. Comparable results were obtained with 3D cultured resting VSMCs. In contrast, overexpression of RGS5 in 2D-cultured proliferating VSMCs promoted their resting state as evidenced by microarray-based expression profiling and attenuated the activity of Akt- and MAP kinase-related signaling cascades. Moreover, RGS5 overexpression attenuated ERK1/2 phosphorylation, VSMC proliferation, and migration, which was mimicked by selectively inhibiting Gαi/o but not Gαq/11 activity. Collectively, the heterogeneous expression of Rgs5 suggests arterial blood vessel type-specific functions in mouse VSMCs. This comprises inhibition of acute agonist-induced Gαq/11/calcium release as well as the support of a resting VSMC phenotype with low ERK1/2 activity by suppressing the activity of Gαi/o.
Collapse
Affiliation(s)
- Eda Demirel
- Department of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, 69120 Heidelberg, Germany
| | - Caroline Arnold
- Department of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, 69120 Heidelberg, Germany
| | - Jaspal Garg
- Department of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, 69120 Heidelberg, Germany
| | - Marius Andreas Jäger
- Department of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, 69120 Heidelberg, Germany
| | - Carsten Sticht
- NGS Core Facility, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Rui Li
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Hanna Kuk
- The Ottawa Department of Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Markus Hecker
- Department of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, 69120 Heidelberg, Germany
| | - Thomas Korff
- Department of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, 69120 Heidelberg, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| |
Collapse
|
7
|
Hariharan A, Weir N, Robertson C, He L, Betsholtz C, Longden TA. The Ion Channel and GPCR Toolkit of Brain Capillary Pericytes. Front Cell Neurosci 2020; 14:601324. [PMID: 33390906 PMCID: PMC7775489 DOI: 10.3389/fncel.2020.601324] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/13/2020] [Indexed: 12/14/2022] Open
Abstract
Brain pericytes reside on the abluminal surface of capillaries, and their processes cover ~90% of the length of the capillary bed. These cells were first described almost 150 years ago (Eberth, 1871; Rouget, 1873) and have been the subject of intense experimental scrutiny in recent years, but their physiological roles remain uncertain and little is known of the complement of signaling elements that they employ to carry out their functions. In this review, we synthesize functional data with single-cell RNAseq screens to explore the ion channel and G protein-coupled receptor (GPCR) toolkit of mesh and thin-strand pericytes of the brain, with the aim of providing a framework for deeper explorations of the molecular mechanisms that govern pericyte physiology. We argue that their complement of channels and receptors ideally positions capillary pericytes to play a central role in adapting blood flow to meet the challenge of satisfying neuronal energy requirements from deep within the capillary bed, by enabling dynamic regulation of their membrane potential to influence the electrical output of the cell. In particular, we outline how genetic and functional evidence suggest an important role for Gs-coupled GPCRs and ATP-sensitive potassium (KATP) channels in this context. We put forth a predictive model for long-range hyperpolarizing electrical signaling from pericytes to upstream arterioles, and detail the TRP and Ca2+ channels and Gq, Gi/o, and G12/13 signaling processes that counterbalance this. We underscore critical questions that need to be addressed to further advance our understanding of the signaling topology of capillary pericytes, and how this contributes to their physiological roles and their dysfunction in disease.
Collapse
Affiliation(s)
- Ashwini Hariharan
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Nick Weir
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Colin Robertson
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Liqun He
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Christer Betsholtz
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Department of Medicine Huddinge (MedH), Karolinska Institutet & Integrated Cardio Metabolic Centre, Huddinge, Sweden
| | - Thomas A Longden
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
8
|
Stern C, Schreier B, Nolze A, Rabe S, Mildenberger S, Gekle M. Knockout of vascular smooth muscle EGF receptor in a mouse model prevents obesity-induced vascular dysfunction and renal damage in vivo. Diabetologia 2020; 63:2218-2234. [PMID: 32548701 PMCID: PMC7476975 DOI: 10.1007/s00125-020-05187-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 04/06/2020] [Indexed: 12/17/2022]
Abstract
AIMS/HYPOTHESIS Obesity causes type 2 diabetes leading to vascular dysfunction and finally renal end-organ damage. Vascular smooth muscle (VSM) EGF receptor (EGFR) modulates vascular wall homeostasis in part via serum response factor (SRF), a major regulator of VSM differentiation and a sensor for glucose. We investigated the role of VSM-EGFR during obesity-induced renovascular dysfunction, as well as EGFR-hyperglycaemia crosstalk. METHODS The role of VSM-EGFR during high-fat diet (HFD)-induced type 2 diabetes was investigated in a mouse model with inducible, VSM-specific EGFR-knockout (KO). Various structural and functional variables as well as transcriptome changes, in vivo and ex vivo, were assessed. The impact of hyperglycaemia on EGFR-induced signalling and SRF transcriptional activity and the underlying mechanisms were investigated at the cellular level. RESULTS We show that VSM-EGFR mediates obesity/type 2 diabetes-induced vascular dysfunction, remodelling and transcriptome dysregulation preceding renal damage and identify an EGFR-glucose synergism in terms of SRF activation, matrix dysregulation and mitochondrial function. EGFR deletion protects the animals from HFD-induced endothelial dysfunction, creatininaemia and albuminuria. Furthermore, we show that HFD leads to marked changes of the aortic transcriptome in wild-type but not in KO animals, indicative of EGFR-dependent SRF activation, matrix dysregulation and mitochondrial dysfunction, the latter confirmed at the cellular level. Studies at the cellular level revealed that high glucose potentiated EGFR/EGF receptor 2 (ErbB2)-induced stimulation of SRF activity, enhancing the graded signalling responses to EGF, via the EGFR/ErbB2-ROCK-actin-MRTF pathway and promoted mitochondrial dysfunction. CONCLUSIONS/INTERPRETATION VSM-EGFR contributes to HFD-induced vascular and subsequent renal alterations. We propose that a potentiated EGFR/ErbB2-ROCK-MRTF-SRF signalling axis and mitochondrial dysfunction underlie the role of EGFR. This advanced working hypothesis will be investigated in mechanistic depth in future studies. VSM-EGFR may be a therapeutic target in cases of type 2 diabetes-induced renovascular disease. DATA AVAILABILITY The datasets generated during and/or analysed during the current study are available in: (1) share_it, the data repository of the academic libraries of Saxony-Anhalt ( https://doi.org/10.25673/32049.2 ); and (2) in the gene expression omnibus database with the study identity GSE144838 ( https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE144838 ). Graphical abstract.
Collapse
Affiliation(s)
- Christian Stern
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Strasse 6, 06112, Halle, Germany
| | - Barbara Schreier
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Strasse 6, 06112, Halle, Germany
| | - Alexander Nolze
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Strasse 6, 06112, Halle, Germany
| | - Sindy Rabe
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Strasse 6, 06112, Halle, Germany
| | - Sigrid Mildenberger
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Strasse 6, 06112, Halle, Germany
| | - Michael Gekle
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Strasse 6, 06112, Halle, Germany.
| |
Collapse
|
9
|
Liu X, Chen W, Li W, Li Y, Priest JR, Zhou B, Wang J, Zhou Z. Single-Cell RNA-Seq of the Developing Cardiac Outflow Tract Reveals Convergent Development of the Vascular Smooth Muscle Cells. Cell Rep 2020; 28:1346-1361.e4. [PMID: 31365875 DOI: 10.1016/j.celrep.2019.06.092] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 04/17/2019] [Accepted: 06/25/2019] [Indexed: 02/07/2023] Open
Abstract
Cardiac outflow tract (OFT) is a major hotspot for congenital heart diseases. A thorough understanding of the cellular diversity, transitions, and regulatory networks of normal OFT development is essential to decipher the etiology of OFT malformations. We performed single-cell transcriptomic sequencing of 55,611 mouse OFT cells from three developmental stages that generally correspond to the early, middle, and late stages of OFT remodeling and septation. Known cellular transitions, such as endothelial-to-mesenchymal transition, have been recapitulated. In particular, we identified convergent development of the vascular smooth muscle cell (VSMC) lineage where intermediate cell subpopulations were found to be involved in either myocardial-to-VSMC trans-differentiation or mesenchymal-to-VSMC transition. Finally, we uncovered transcriptional regulators potentially governing cellular transitions. Our study provides a single-cell reference map of cell states for normal OFT development and paves the way for further studies of the etiology of OFT malformations at the single-cell level.
Collapse
Affiliation(s)
- Xuanyu Liu
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Center of Laboratory Medicine, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Wen Chen
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Center of Laboratory Medicine, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Wenke Li
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Center of Laboratory Medicine, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Yan Li
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - James R Priest
- Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Bin Zhou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University. School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| | - Jikui Wang
- Henan Key Laboratory for Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University. Xinxiang 453003, China.
| | - Zhou Zhou
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Center of Laboratory Medicine, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China.
| |
Collapse
|
10
|
Figueiredo AM, Villacampa P, Diéguez-Hurtado R, José Lozano J, Kobialka P, Cortazar AR, Martinez-Romero A, Angulo-Urarte A, Franco CA, Claret M, Aransay AM, Adams RH, Carracedo A, Graupera M. Phosphoinositide 3-Kinase-Regulated Pericyte Maturation Governs Vascular Remodeling. Circulation 2020; 142:688-704. [PMID: 32466671 DOI: 10.1161/circulationaha.119.042354] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Pericytes regulate vessel stabilization and function, and their loss is associated with diseases such as diabetic retinopathy or cancer. Despite their physiological importance, pericyte function and molecular regulation during angiogenesis remain poorly understood. METHODS To decipher the transcriptomic programs of pericytes during angiogenesis, we crossed Pdgfrb(BAC)-CreERT2 mice into RiboTagflox/flox mice. Pericyte morphological changes were assessed in mural cell-specific R26-mTmG reporter mice, in which low doses of tamoxifen allowed labeling of single-cell pericytes at high resolution. To study the role of phosphoinositide 3-kinase (PI3K) signaling in pericyte biology during angiogenesis, we used genetic mouse models that allow selective inactivation of PI3Kα and PI3Kβ isoforms and their negative regulator phosphate and tensin homolog deleted on chromosome 10 (PTEN) in mural cells. RESULTS At the onset of angiogenesis, pericytes exhibit molecular traits of cell proliferation and activated PI3K signaling, whereas during vascular remodeling, pericytes upregulate genes involved in mature pericyte cell function, together with a remarkable decrease in PI3K signaling. Immature pericytes showed stellate shape and high proliferation, and mature pericytes were quiescent and elongated. Unexpectedly, we demonstrate that PI3Kβ, but not PI3Kα, regulates pericyte proliferation and maturation during vessel formation. Genetic PI3Kβ inactivation in pericytes triggered early pericyte maturation. Conversely, unleashing PI3K signaling by means of PTEN deletion delayed pericyte maturation. Pericyte maturation was necessary to undergo vessel remodeling during angiogenesis. CONCLUSIONS Our results identify new molecular and morphological traits associated with pericyte maturation and uncover PI3Kβ activity as a checkpoint to ensure appropriate vessel formation. In turn, our results may open new therapeutic opportunities to regulate angiogenesis in pathological processes through the manipulation of pericyte PI3Kβ activity.
Collapse
Affiliation(s)
- Ana M Figueiredo
- Vascular Biology and Signalling Group, ProCURE, Oncobell Program, Institut d´Investigació Biomèdica de Bellvitge (IDIBELL), Gran Via de l'Hospitalet 199, 08908 L´Hospitalet de Llobregat, Barcelona, Spain (A.M.F., P.V., P.K., A.M.-R., A.A.-U., M.G.)
| | - Pilar Villacampa
- Vascular Biology and Signalling Group, ProCURE, Oncobell Program, Institut d´Investigació Biomèdica de Bellvitge (IDIBELL), Gran Via de l'Hospitalet 199, 08908 L´Hospitalet de Llobregat, Barcelona, Spain (A.M.F., P.V., P.K., A.M.-R., A.A.-U., M.G.)
| | - Rodrigo Diéguez-Hurtado
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, and Faculty of Medicine, University of Münster, Germany (R.D.-H., R.H.A.)
| | - Juan José Lozano
- Bioinformatics Platform, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain (J.J.L.)
| | - Piotr Kobialka
- Vascular Biology and Signalling Group, ProCURE, Oncobell Program, Institut d´Investigació Biomèdica de Bellvitge (IDIBELL), Gran Via de l'Hospitalet 199, 08908 L´Hospitalet de Llobregat, Barcelona, Spain (A.M.F., P.V., P.K., A.M.-R., A.A.-U., M.G.)
| | - Ana Rosa Cortazar
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain (A.R.C., A.M.A., A.C.)
| | - Anabel Martinez-Romero
- Vascular Biology and Signalling Group, ProCURE, Oncobell Program, Institut d´Investigació Biomèdica de Bellvitge (IDIBELL), Gran Via de l'Hospitalet 199, 08908 L´Hospitalet de Llobregat, Barcelona, Spain (A.M.F., P.V., P.K., A.M.-R., A.A.-U., M.G.)
| | - Ana Angulo-Urarte
- Vascular Biology and Signalling Group, ProCURE, Oncobell Program, Institut d´Investigació Biomèdica de Bellvitge (IDIBELL), Gran Via de l'Hospitalet 199, 08908 L´Hospitalet de Llobregat, Barcelona, Spain (A.M.F., P.V., P.K., A.M.-R., A.A.-U., M.G.)
| | - Claudio A Franco
- CIBERONC (A.R.C., A.M.A., A.C., M.G.) and CIBERehd (A.M.A.), Instituto de Salud Carlos III, Madrid, Spain. Instituto de Medicina Molecular-João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal (C.A.F.)
| | - Marc Claret
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (M.C.)
| | - Ana María Aransay
- CIBERONC (A.R.C., A.M.A., A.C., M.G.) and CIBERehd (A.M.A.), Instituto de Salud Carlos III, Madrid, Spain. Instituto de Medicina Molecular-João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal (C.A.F.)
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, and Faculty of Medicine, University of Münster, Germany (R.D.-H., R.H.A.)
| | - Arkaitz Carracedo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain (A.R.C., A.M.A., A.C.)
| | - Mariona Graupera
- Vascular Biology and Signalling Group, ProCURE, Oncobell Program, Institut d´Investigació Biomèdica de Bellvitge (IDIBELL), Gran Via de l'Hospitalet 199, 08908 L´Hospitalet de Llobregat, Barcelona, Spain (A.M.F., P.V., P.K., A.M.-R., A.A.-U., M.G.)
| |
Collapse
|
11
|
Röhl S, Suur BE, Lengquist M, Seime T, Caidahl K, Hedin U, Arner A, Matic L, Razuvaev A. Lack of PCSK6 Increases Flow-Mediated Outward Arterial Remodeling in Mice. Cells 2020; 9:cells9041009. [PMID: 32325687 PMCID: PMC7225991 DOI: 10.3390/cells9041009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 12/17/2022] Open
Abstract
Proprotein convertases (PCSKs) process matrix metalloproteases and cytokines, but their function in the vasculature is largely unknown. Previously, we demonstrated upregulation of PCSK6 in atherosclerotic plaques from symptomatic patients, localization to smooth muscle cells (SMCs) in the fibrous cap and positive correlations with inflammation, extracellular matrix remodeling and cytokines. Here, we hypothesize that PCSK6 could be involved in flow-mediated vascular remodeling and aim to evaluate its role in the physiology of this process using knockout mice. Pcsk6−/− and wild type mice were randomized into control and increased blood flow groups and induced in the right common carotid artery (CCA) by ligation of the left CCA. The animals underwent repeated ultrasound biomicroscopy (UBM) examinations followed by euthanization with subsequent evaluation using wire myography, transmission electron microscopy or histology. The Pcsk6−/− mice displayed a flow-mediated increase in lumen circumference over time, assessed with UBM. Wire myography revealed differences in the flow-mediated remodeling response detected as an increase in lumen circumference at optimal stretch with concomitant reduction in active tension. Furthermore, a flow-mediated reduction in expression of SMC contractile markers SMA, MYH11 and LMOD1 was seen in the Pcsk6−/− media. Absence of PCSK6 increases outward remodeling and reduces medial contractility in response to increased blood flow.
Collapse
Affiliation(s)
- Samuel Röhl
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 76 Stockholm, Sweden; (S.R.); (B.E.S.); (M.L.); (T.S.); (K.C.); (U.H.)
| | - Bianca E. Suur
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 76 Stockholm, Sweden; (S.R.); (B.E.S.); (M.L.); (T.S.); (K.C.); (U.H.)
| | - Mariette Lengquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 76 Stockholm, Sweden; (S.R.); (B.E.S.); (M.L.); (T.S.); (K.C.); (U.H.)
| | - Till Seime
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 76 Stockholm, Sweden; (S.R.); (B.E.S.); (M.L.); (T.S.); (K.C.); (U.H.)
| | - Kenneth Caidahl
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 76 Stockholm, Sweden; (S.R.); (B.E.S.); (M.L.); (T.S.); (K.C.); (U.H.)
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 76 Stockholm, Sweden; (S.R.); (B.E.S.); (M.L.); (T.S.); (K.C.); (U.H.)
| | - Anders Arner
- Department of Clinical Sciences Lund, Thoracic Surgery, Lund University, 221 84 Lund, Sweden;
| | - Ljubica Matic
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 76 Stockholm, Sweden; (S.R.); (B.E.S.); (M.L.); (T.S.); (K.C.); (U.H.)
- Correspondence: (L.M.); (A.R.); Tel.: +46-(0)-73-962-42-79 (L.M.); +46-(0)-76-238-44-75 (A.R.)
| | - Anton Razuvaev
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 76 Stockholm, Sweden; (S.R.); (B.E.S.); (M.L.); (T.S.); (K.C.); (U.H.)
- Correspondence: (L.M.); (A.R.); Tel.: +46-(0)-73-962-42-79 (L.M.); +46-(0)-76-238-44-75 (A.R.)
| |
Collapse
|
12
|
Özen I, Roth M, Barbariga M, Gaceb A, Deierborg T, Genové G, Paul G. Loss of Regulator of G-Protein Signaling 5 Leads to Neurovascular Protection in Stroke. Stroke 2019; 49:2182-2190. [PMID: 30354999 PMCID: PMC6116795 DOI: 10.1161/strokeaha.118.020124] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background and Purpose- In ischemic stroke, breakdown of the blood-brain barrier (BBB) aggravates brain damage. Pericyte detachment contributes to BBB disruption and neurovascular dysfunction, but little is known about its regulation in stroke. Here, we investigated how loss of RGS5 (regulator of G protein signaling 5) in pericytes affects BBB breakdown in stroke and its consequences. Method- We used RGS5 knockout and control mice and applied a permanent middle cerebral occlusion model. We analyzed pericyte numbers, phenotype, and vessel morphology using immunohistochemistry and confocal microscopy. We investigated BBB breakdown by measuring endothelial coverage, tight junctions, and AQP4 (aquaporin 4) in addition to BBB permeability (fluorescent-conjugated dextran extravasation). Tissue hypoxia was assessed with pimonidazole hydrochloride and neuronal death quantified with the terminal deoxynucleotidyl transferase dUTP nick end labeling assay. Results- We demonstrate that loss of RGS5 increases pericyte numbers and their endothelial coverage, which is associated with higher capillary density and length, and significantly less BBB damage after stroke. Loss of RGS5 in pericytes results in reduced vascular leakage and preserved tight junctions and AQP4, decreased cerebral hypoxia, and partial neuronal protection in the infarct area. Conclusions- Our findings show that loss of RGS5 affects pericyte-related BBB preservation in stroke and identifies RGS5 as an important target for neurovascular protection.
Collapse
Affiliation(s)
- Ilknur Özen
- From the Translational Neurology Group, Department of Clinical Science (I.O., M.R., M.B., A.G., G.P.)
| | - Michaela Roth
- From the Translational Neurology Group, Department of Clinical Science (I.O., M.R., M.B., A.G., G.P.)
| | - Marco Barbariga
- From the Translational Neurology Group, Department of Clinical Science (I.O., M.R., M.B., A.G., G.P.)
| | - Abderahim Gaceb
- From the Translational Neurology Group, Department of Clinical Science (I.O., M.R., M.B., A.G., G.P.)
| | - Tomas Deierborg
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Sciences (T.D.)
| | - Guillem Genové
- Lund University, Sweden; Department of Medicine, Integrated Cardio Metabolic Center, Karolinska Institute, Huddinge, Sweden (G.G.)
| | - Gesine Paul
- From the Translational Neurology Group, Department of Clinical Science (I.O., M.R., M.B., A.G., G.P.).,Wallenberg Centre for Molecular Medicine (G.P.).,Department of Neurology, Scania University Hospital, Lund, Sweden (G.P.)
| |
Collapse
|
13
|
Tung CH, Kuo LW, Huang MF, Wu YY, Tsai YT, Wu JE, Hsu KF, Chen YL, Hong TM. MicroRNA-150-5p promotes cell motility by inhibiting c-Myb-mediated Slug suppression and is a prognostic biomarker for recurrent ovarian cancer. Oncogene 2019; 39:862-876. [PMID: 31570789 DOI: 10.1038/s41388-019-1025-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 09/05/2019] [Accepted: 09/12/2019] [Indexed: 02/06/2023]
Abstract
Treatment of ovarian cancer (OvCa) remains challenging owing to its high recurrence rates. Detachment of cancer cells into the peritoneal fluid plays a key role in OvCa relapse, but how this occurs remains incompletely understood. Here we examined global miRNA expression profiles of paired primary/recurrent OvCa specimens and identified a novel biomarker, microRNA-150-5p (miR-150-5p), that was significantly upregulated in 16 recurrent OvCa tissues compared with their matched primary specimens. Analyses of cohorts from two other groups confirmed that expression of miR-150-5p was associated with early relapse and poor survival of OvCa patients. Inhibition of miR-150-5p significantly inhibited the migration and invasion of OvCa cells and induced a mesenchymal-epithelial transition (MET) phenotype. We demonstrated that the proto-oncogene, MYB, is an miR-150-5p target in OvCa cells and that the miR-150-5p/c-Myb/Slug axis plays important roles in regulating epithelial-mesenchymal transition (EMT) in OvCa cells. Expression of MYB was significantly correlated with good clinical outcome in OvCa and was negatively correlated with Slug expression in late-stage clinical specimens. These results suggest that miR-150-5p upregulation mediates the progression of recurrent OvCa by targeting the c-Myb/Slug pathway. Inhibition of miR-150-5p may serve as a new therapeutic strategy for preventing recurrence of OvCa.
Collapse
Affiliation(s)
- Chia-Hao Tung
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Li-Wei Kuo
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Meng-Fan Huang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Ying Wu
- Clinical Medicine Research Center, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Yao-Tsung Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jia-En Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Keng-Fu Hsu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yuh-Ling Chen
- Institute of Oral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Tse-Ming Hong
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan. .,Clinical Medicine Research Center, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
14
|
Disease-relevant transcriptional signatures identified in individual smooth muscle cells from healthy mouse vessels. Nat Commun 2018; 9:4567. [PMID: 30385745 PMCID: PMC6212435 DOI: 10.1038/s41467-018-06891-x] [Citation(s) in RCA: 196] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 09/19/2018] [Indexed: 02/07/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) show pronounced heterogeneity across and within vascular beds, with direct implications for their function in injury response and atherosclerosis. Here we combine single-cell transcriptomics with lineage tracing to examine VSMC heterogeneity in healthy mouse vessels. The transcriptional profiles of single VSMCs consistently reflect their region-specific developmental history and show heterogeneous expression of vascular disease-associated genes involved in inflammation, adhesion and migration. We detect a rare population of VSMC-lineage cells that express the multipotent progenitor marker Sca1, progressively downregulate contractile VSMC genes and upregulate genes associated with VSMC response to inflammation and growth factors. We find that Sca1 upregulation is a hallmark of VSMCs undergoing phenotypic switching in vitro and in vivo, and reveal an equivalent population of Sca1-positive VSMC-lineage cells in atherosclerotic plaques. Together, our analyses identify disease-relevant transcriptional signatures in VSMC-lineage cells in healthy blood vessels, with implications for disease susceptibility, diagnosis and prevention.
Collapse
|
15
|
Nam Y, Min YS, Sohn UD. Recent advances in pharmacological research on the management of irritable bowel syndrome. Arch Pharm Res 2018; 41:955-966. [PMID: 30132170 DOI: 10.1007/s12272-018-1068-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 08/16/2018] [Indexed: 12/17/2022]
Abstract
Irritable bowel syndrome (IBS), a common gastrointestinal (GI) disorder, is associated with various factors, including lifestyle, infection, stress, intestinal flora, and related diseases. The pharmacotherapeutic stimulation of receptors and downstream signaling pathways is effective in reducing IBS symptoms; however, it is still associated with adverse effects. Various receptors related to GI motility and visceral hypersensitivity should be considered to enhance the benefit/risk ratio of IBS treatments. This review discusses recent pharmacological advances in IBS management. Several receptors related to GI motility and abdominal pain are investigated in various angles. 5-Hydroxytryptamine (5-HT) is an important neurotransmitter that activates the colonic mucosal 5-HT4 receptor without causing severe cardiovascular adverse effects. The clinical potential of ramosetron for diarrhea-predominant IBS has been suggested because of a lower risk of ischemic colitis than conventional 5-HT3 receptor antagonists. Toll-like receptors (TLRs), especially TLR2 and TLR4, show a significant effect on the post-infection symptoms and lipopolysaccharide-mediated regulation of GI motility. Histamine is a well-known nitrogenous compound that regulates inflammatory responses and visceral hypersensitivity. Histamine 1 receptor-mediated sensitization of the transient receptor potential vanilloid 1 is associated with IBS. Pharmacological approaches based on these signaling pathways could be useful in the development of novel IBS treatments.
Collapse
Affiliation(s)
- Yoonjin Nam
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, 84 Heukseok-RO, Dongjak-Gu, Seoul, 06974, Republic of Korea
| | - Young Sil Min
- Department of Medical Plant Science, Jung Won University, 85 Munmu-ro, Goesan-eup, Goesan-gun, Chungbuk, 28024, Republic of Korea
| | - Uy Dong Sohn
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, 84 Heukseok-RO, Dongjak-Gu, Seoul, 06974, Republic of Korea.
| |
Collapse
|
16
|
Liu C, Hu Q, Jing J, Zhang Y, Jin J, Zhang L, Mu L, Liu Y, Sun B, Zhang T, Kong Q, Wang G, Wang D, Zhang Y, Liu X, Zhao W, Wang J, Feng T, Li H. Regulator of G protein signaling 5 (RGS5) inhibits sonic hedgehog function in mouse cortical neurons. Mol Cell Neurosci 2017; 83:65-73. [PMID: 28684360 DOI: 10.1016/j.mcn.2017.06.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Revised: 02/21/2017] [Accepted: 06/20/2017] [Indexed: 12/12/2022] Open
Abstract
Regulator of G protein signaling 5 (RGS5) acts as a GTPase-activating protein (GAP) for the Gαi subunit and negatively regulates G protein-coupled receptor signaling. However, its presence and function in postmitotic differentiated primary neurons remains largely uncharacterized. During neural development, sonic hedgehog (Shh) signaling is involved in cell signaling pathways via Gαi activity. In particular, Shh signaling is essential for embryonic neural tube patterning, which has been implicated in neuronal polarization involving neurite outgrowth. Here, we examined whether RGS5 regulates Shh signaling in neurons. RGS5 transcripts were found to be expressed in cortical neurons and their expression gradually declined in a time-dependent manner in culture system. When an adenovirus expressing RGS5 was introduced into an in vitro cell culture model of cortical neurons, RGS5 overexpression significantly reduced neurite outgrowth and FM4-64 uptake, while cAMP-PKA signaling was also affected. These findings suggest that RGS5 inhibits Shh function during neurite outgrowth and the presynaptic terminals of primary cortical neurons mature via modulation of cAMP.
Collapse
Affiliation(s)
- Chuanliang Liu
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China; Vocational College Daxing'an Mountains, Jiagedaqi District, Heilongjiang 165000, China
| | - Qiongqiong Hu
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Jia Jing
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Yun Zhang
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Jing Jin
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Liulei Zhang
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Lili Mu
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Yumei Liu
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Bo Sun
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Tongshuai Zhang
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Qingfei Kong
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Guangyou Wang
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Dandan Wang
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Yao Zhang
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Xijun Liu
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Wei Zhao
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Jinghua Wang
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China.
| | - Tao Feng
- Department of Neurology, The Nangang Branch of Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150001, China.
| | - Hulun Li
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China; Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, Heilongjiang 150086, China
| |
Collapse
|
17
|
Maihöfer NA, Suleiman S, Dreymüller D, Manley PW, Rossaint R, Uhlig S, Martin C, Rieg AD. Imatinib relaxes the pulmonary venous bed of guinea pigs. Respir Res 2017; 18:32. [PMID: 28178968 PMCID: PMC5299687 DOI: 10.1186/s12931-017-0514-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 01/19/2017] [Indexed: 12/15/2022] Open
Abstract
Background Recently, the IMPRES study revealed that systemic imatinib improves exercise capacity in patients with advanced pulmonary arterial hypertension. Imatinib blocks the tyrosine kinase activity of the platelet-derived growth factor (PDGF)-receptor (PDGFR), acts antiproliferative and relaxes pulmonary arteries. However so far, the relaxant effects of imatinib on pulmonary veins (PVs) and on the postcapillary resistance are unknown, although pulmonary hypertension (PH) due to left heart disease (LHD) is most common and primarily affects PVs. Next, it is unknown whether activation of PDGFR alters the pulmonary venous tone. Due to the reported adverse effects of systemic imatinib, we evaluated the effects of nebulized imatinib on the postcapillary resistance. Methods Precision-cut lung slices (PCLS) were prepared from guinea pigs. PVs were pre-constricted with Endothelin-1 (ET-1) and the imatinib-induced relaxation was studied by videomicroscopy; PDGF-BB-related vascular properties were evaluated as well. The effects of perfused/nebulized imatinib on the postcapillary resistance were studied in cavine isolated perfused lungs (IPL). Intracellular cAMP/cGMP was measured by ELISA in PVs. Results In PCLS, imatinib (100 μM) relaxed pre-constricted PVs (126%). In PVs, imatinib increased cAMP, but not cGMP and inhibition of adenyl cyclase or protein kinase A reduced the imatinib-induced relaxation. Further, inhibition of KATP-channels, \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$ {\mathrm{BK}}_{\mathrm{Ca}}^{2+} $$\end{document}BKCa2+-channels or Kv-channels diminished the imatinib-induced relaxation, whereas inhibition of NO-signaling was without effect. In the IPL, perfusion or nebulization of imatinib reduced the ET-1-induced increase of the postcapillary resistance. In PCLS, PDGF-BB contracted PVs, which was blocked by imatinib and by the PDGFR-β kinase inhibitor SU6668, whereas inhibition of PDGFR-α (ponatinib) had no significant effect. Conversely, PDGFR-β kinase inhibitors (SU6668/DMPQ) relaxed PVs pre-constricted with ET-1 comparable to imatinib, whereas the PDGFR-α kinase inhibitor ponatinib did not. Conclusions Imatinib-induced relaxation depends on cAMP and on the activation of K+-channels. Perfused or nebulized imatinib significantly reduces the postcapillary resistance in the pre-constricted (ET-1) pulmonary venous bed. Hence, nebulization of imatinib is feasible and might reduce systemic side effects. Conversely, PDGF-BB contracts PVs by activation of PDGFR-β suggesting that imatinib-induced relaxation depends on PDGFR-β-antagonism. Imatinib combines short-term relaxant and long-term antiproliferative effects. Thus, imatinib might be a promising therapy for PH due to LHD.
Collapse
Affiliation(s)
- Nina A Maihöfer
- Institute of Pharmacology and Toxicology, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany
| | - Said Suleiman
- Institute of Pharmacology and Toxicology, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany
| | - Daniela Dreymüller
- Institute of Pharmacology and Toxicology, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany
| | | | - Rolf Rossaint
- Department of Anesthesiology, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany
| | - Stefan Uhlig
- Institute of Pharmacology and Toxicology, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany
| | - Christian Martin
- Institute of Pharmacology and Toxicology, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany
| | - Annette D Rieg
- Institute of Pharmacology and Toxicology, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany. .,Department of Anesthesiology, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany.
| |
Collapse
|
18
|
Padel T, Özen I, Boix J, Barbariga M, Gaceb A, Roth M, Paul G. Platelet-derived growth factor-BB has neurorestorative effects and modulates the pericyte response in a partial 6-hydroxydopamine lesion mouse model of Parkinson's disease. Neurobiol Dis 2016; 94:95-105. [PMID: 27288154 DOI: 10.1016/j.nbd.2016.06.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 05/24/2016] [Accepted: 06/05/2016] [Indexed: 12/28/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease where the degeneration of the nigrostriatal pathway leads to specific motor deficits. There is an unmet medical need for regenerative treatments that stop or reverse disease progression. Several growth factors have been investigated in clinical trials to restore the dopaminergic nigrostriatal pathway damaged in PD. Platelet-derived growth factor-BB (PDGF-BB), a molecule that recruits pericytes to stabilize microvessels, was recently investigated in a phase-1 clinical trial, showing a dose-dependent increase in dopamine transporter binding in the putamen of PD patients. Interestingly, evidence is accumulating that PD is paralleled by microvascular changes, however, whether PDGF-BB modifies pericytes in PD is not known. Using a pericyte reporter mouse strain, we investigate the functional and restorative effect of PDGF-BB in a partial 6-hydroxydopamine medial forebrain bundle lesion mouse model of PD, and whether this restorative effect is accompanied by changes in pericyte features. We demonstrate that a 2-week treatment with PDGF-BB leads to behavioural recovery using several behavioural tests, and partially restores the nigrostriatal pathway. Interestingly, we find that pericytes are activated in the striatum of PD lesioned mice and that these changes are reversed by PDGF-BB treatment. The modulation of brain pericytes may contribute to the PDGF-BB-induced neurorestorative effects, PDGF-BB allowing for vascular stabilization in PD. Pericytes might be a new cell target of interest for future regenerative therapies.
Collapse
Affiliation(s)
- Thomas Padel
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden
| | - Ilknur Özen
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden
| | - Jordi Boix
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden
| | - Marco Barbariga
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden
| | - Abderahim Gaceb
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden
| | - Michaela Roth
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden
| | - Gesine Paul
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden; Department of Neurology, Scania University Hospital, 22185 Lund, Sweden.
| |
Collapse
|
19
|
Nishio M, Nakahara M, Sato C, Saeki K, Akutsu H, Umezawa A, Tobe K, Yasuda K, Yuo A, Saeki K. New categorization of human vascular endothelial cells by pro- vs anti-proliferative phenotypes. World J Transl Med 2015; 4:88-100. [DOI: 10.5528/wjtm.v4.i3.88] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 08/28/2015] [Accepted: 10/13/2015] [Indexed: 02/05/2023] Open
Abstract
AIM: To integrally understand the effects of human vascular endothelial cells (VECs) on the proliferation of vascular smooth muscle cells (VSMCs).
METHODS: Various kinds of human VECs of different origins were co-cultured with human aortic smooth muscle cells, a representative of human VSMCs. To exclude the irrelevant effects due to growth competition between VECs and VSMCs, the proliferation of VECs had previously been arrested via a low-dose gamma ray irradiation. To discriminately analyze the proliferation of VSMCs from that of VECs, the former cells were labeled with red fluorescent dye while the latter cells were labeled with green fluorescent dye before performing co-culture experiments. After 4 d, total cells were harvested and subjected to flow cytometric analyses. Decrements in red fluorescence intensities due to proliferation-mediated dilutions were measured and mathematically processed using a specific software to quantitatively evaluate the proliferation of VSMCs. The findings obtained from the flow cytometry-based analyses were further validated by microscopic observations.
RESULTS: Commercially available primary cultured human VECs exclusively promoted VSMC proliferation regardless of their tissue origins and we termed these pro-proliferative VECs as “type-I”. By contrast, VECs freshly generated from human bone marrow-derived endothelial progenitors cells or human pluripotent stem cells including embryonic stem cells and induced pluripotent stem cells suppressed VSMC proliferation and we termed these anti-proliferative VECs as “type-II”. Repetitive subcultures as well as oxidative stress induced “type-II VECs to type-I” conversion along with an induction of Regulator of G-protein signaling 5 (RGS5). Compatibly, anti-oxidant treatments suppressed both the subculture-dependent “type-II to type-I” conversion and an induction of RGS5 gene. Immunostaining studies of clinical specimens indicated that RGS5 protein expressions in endothelial layers were low in normal arteries but they were up-regulated in pathological arteries including hypertension, atherosclerosis and autoimmune vasculitis in a dose-dependent manner. Overexpression and knockdown of RGS5 caused that “type-II to type-I” and “type-I to type-II” phenotype conversions of VECs, respectively.
CONCLUSION: Human VECs are categorized into two types: pro-proliferative RGS5high VECs (type-I) and anti-proliferative RGS5low VECs (type-II).
Collapse
|
20
|
Daniel JM, Prock A, Dutzmann J, Sonnenschein K, Thum T, Bauersachs J, Sedding DG. Regulator of G-Protein Signaling 5 Prevents Smooth Muscle Cell Proliferation and Attenuates Neointima Formation. Arterioscler Thromb Vasc Biol 2015; 36:317-27. [PMID: 26663397 DOI: 10.1161/atvbaha.115.305974] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 11/24/2015] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Regulator of G-protein signaling 5 (RGS5) is abundantly expressed in vascular smooth muscle cells (SMCs) and inhibits G-protein signaling by enhancing the guanosine triphosphate-hydrolyzing activity of Gα-subunits. In the present study, we investigated the effects of RGS5 on vascular SMC function in vitro and neointima formation after wire-induced injury in mice and determined the underlying mechanisms. APPROACH AND RESULTS We found a robust expression of RGS5 in native arteries of C57BL/6 mice and a highly significant downregulation within neointimal lesions 10 and 21 days after vascular injury as assessed by quantitative polymerase chain reaction, immunoblotting, and immunohistochemistry. In vitro, RGS5 was found significantly downregulated after mitogenic stimulation of human coronary artery SMCs. To restore RGS5 levels, SMCs were transduced with adenoviral vectors encoding wild-type RGS5 or a nondegradable mutant. RGS5-WT and, even more prominently, the C2A-RGS5 mutant prevented SMC proliferation and migration. In contrast, the siRNA-mediated knockdown of RGS5 significantly augmented SMC proliferation. Following overexpression of RGS5, fluorescence-activated cell sorting analysis of propidium iodide-stained cells indicated cell cycle arrest in G0/G1 phase. Mechanistically, inhibition of the phosphorylation of the extracellular signal-regulated kinase 1/2 and mitogen-activated protein kinase downstream signaling was shown to be responsible for the anti-proliferative effect of RGS5. Following wire-induced injury of the femoral artery in C57BL/6 mice, adenoviral-mediated overexpression of RGS5-WT or C2A-RGS5 significantly reduced SMC proliferation and neointima formation in vivo. CONCLUSIONS Downregulation of RGS5 is an important prerequisite for SMC proliferation in vitro and in vivo. Therefore, reconstitution of RGS5 levels represents a promising therapeutic option to prevent vascular remodeling processes.
Collapse
Affiliation(s)
- Jan-Marcus Daniel
- From the Department of Cardiology and Angiology (J.-M.D., J.D., K.S., J.B., D.G.S.), REBIRTH Excellence Cluster (J.-M.D., T.T., J.B., D.G.S.), and Institute of Molecular and Translational Therapeutic Strategies (IMTTS) (K.S., T.T.), Hannover Medical School, Hannover, Germany; Department of General, Visceral, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany (A.P.); and National Heart and Lung Institute, Imperial College London, London, UK (T.T.)
| | - André Prock
- From the Department of Cardiology and Angiology (J.-M.D., J.D., K.S., J.B., D.G.S.), REBIRTH Excellence Cluster (J.-M.D., T.T., J.B., D.G.S.), and Institute of Molecular and Translational Therapeutic Strategies (IMTTS) (K.S., T.T.), Hannover Medical School, Hannover, Germany; Department of General, Visceral, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany (A.P.); and National Heart and Lung Institute, Imperial College London, London, UK (T.T.)
| | - Jochen Dutzmann
- From the Department of Cardiology and Angiology (J.-M.D., J.D., K.S., J.B., D.G.S.), REBIRTH Excellence Cluster (J.-M.D., T.T., J.B., D.G.S.), and Institute of Molecular and Translational Therapeutic Strategies (IMTTS) (K.S., T.T.), Hannover Medical School, Hannover, Germany; Department of General, Visceral, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany (A.P.); and National Heart and Lung Institute, Imperial College London, London, UK (T.T.)
| | - Kristina Sonnenschein
- From the Department of Cardiology and Angiology (J.-M.D., J.D., K.S., J.B., D.G.S.), REBIRTH Excellence Cluster (J.-M.D., T.T., J.B., D.G.S.), and Institute of Molecular and Translational Therapeutic Strategies (IMTTS) (K.S., T.T.), Hannover Medical School, Hannover, Germany; Department of General, Visceral, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany (A.P.); and National Heart and Lung Institute, Imperial College London, London, UK (T.T.)
| | - Thomas Thum
- From the Department of Cardiology and Angiology (J.-M.D., J.D., K.S., J.B., D.G.S.), REBIRTH Excellence Cluster (J.-M.D., T.T., J.B., D.G.S.), and Institute of Molecular and Translational Therapeutic Strategies (IMTTS) (K.S., T.T.), Hannover Medical School, Hannover, Germany; Department of General, Visceral, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany (A.P.); and National Heart and Lung Institute, Imperial College London, London, UK (T.T.)
| | - Johann Bauersachs
- From the Department of Cardiology and Angiology (J.-M.D., J.D., K.S., J.B., D.G.S.), REBIRTH Excellence Cluster (J.-M.D., T.T., J.B., D.G.S.), and Institute of Molecular and Translational Therapeutic Strategies (IMTTS) (K.S., T.T.), Hannover Medical School, Hannover, Germany; Department of General, Visceral, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany (A.P.); and National Heart and Lung Institute, Imperial College London, London, UK (T.T.)
| | - Daniel G Sedding
- From the Department of Cardiology and Angiology (J.-M.D., J.D., K.S., J.B., D.G.S.), REBIRTH Excellence Cluster (J.-M.D., T.T., J.B., D.G.S.), and Institute of Molecular and Translational Therapeutic Strategies (IMTTS) (K.S., T.T.), Hannover Medical School, Hannover, Germany; Department of General, Visceral, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany (A.P.); and National Heart and Lung Institute, Imperial College London, London, UK (T.T.).
| |
Collapse
|
21
|
Stembridge M, Ainslie PN, Shave R. Mechanisms underlying reductions in stroke volume at rest and during exercise at high altitude. Eur J Sport Sci 2015; 16:577-84. [DOI: 10.1080/17461391.2015.1071876] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
22
|
Althoff TF, Offermanns S. G-protein-mediated signaling in vascular smooth muscle cells — implications for vascular disease. J Mol Med (Berl) 2015; 93:973-81. [DOI: 10.1007/s00109-015-1305-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 05/14/2015] [Accepted: 06/02/2015] [Indexed: 10/24/2022]
|
23
|
Holobotovskyy V, Chong YS, Burchell J, He B, Phillips M, Leader L, Murphy TV, Sandow SL, McKitrick DJ, Charles AK, Tare M, Arnolda LF, Ganss R. Regulator of G protein signaling 5 is a determinant of gestational hypertension and preeclampsia. Sci Transl Med 2015; 7:290ra88. [DOI: 10.1126/scitranslmed.aaa5038] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Preeclampsia is a systemic vascular disorder of pregnancy and is associated with increased sensitivity to angiotensin II (AngII) and hypertension. The cause of preeclampsia remains unknown. We identified the role of regulator of G protein (heterotrimeric guanine nucleotide–binding protein) signaling 5 (RGS5) in blood pressure regulation during pregnancy and preeclampsia. RGS5 expression in human myometrial vessels is markedly suppressed in gestational hypertension and/or preeclampsia. In pregnant RGS5-deficient mice, reduced vascular RGS5 expression causes gestational hypertension by enhancing vascular sensitivity to AngII. Further challenge by increasing AngII results in preeclampsia-like symptoms, namely, more severe hypertension, proteinuria, placental pathology, and reduced birth weight. In pregnant heterozygote null mice, treatment with peroxisome proliferator–activated receptor (PPAR) agonists normalizes vascular function and blood pressure through effects on RGS5. These findings highlight a key role of RGS5 at the interface between AngII and PPAR signaling. Because preeclampsia is refractory to current standard therapies, our study opens an unrecognized and urgently needed opportunity for treatment of gestational hypertension and preeclampsia.
Collapse
Affiliation(s)
- Vasyl Holobotovskyy
- Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Yee Seng Chong
- Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Jennifer Burchell
- Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Bo He
- Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Michael Phillips
- Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia 6009, Australia
- Royal Perth Hospital, Perth, Western Australia 6009, Australia
| | - Leo Leader
- School of Women’s and Children’s Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2031, Australia
| | - Timothy V. Murphy
- Physiology, School of Medical Sciences, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Shaun L. Sandow
- Physiology, School of Medical Sciences, University of New South Wales, Sydney, New South Wales 2052, Australia
- Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Maroochydore, Queensland 4558, Australia
| | - Douglas J. McKitrick
- School of Medicine and Pharmacology, The University of Western Australia & Cardiology Department, Royal Perth Hospital, Perth, Western Australia 6000, Australia
| | - Adrian K. Charles
- Princess Margaret Hospital and School of Paediatrics and Child Health, The University of Western Australia, Perth, Western Australia 6008, Australia
| | - Marianne Tare
- Department of Physiology, Monash University, Clayton, Victoria 3800, Australia
- School of Rural Health, Monash University, Churchill, Victoria 3842, Australia
| | - Leonard F. Arnolda
- Medical School, Australian National University, and Cardiology Department, Canberra Hospital, Canberra, Australian Capital Territory 2606, Australia
| | - Ruth Ganss
- Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia 6009, Australia
| |
Collapse
|
24
|
Mao Y, Su J, Lei L, Meng L, Qi Y, Huo Y, Tang C. Impaired regulator of G protein signaling 2 transcription facilitates vascular remodeling in injured rat aorta. J Cardiovasc Med (Hagerstown) 2015; 15:572-8. [PMID: 24914635 DOI: 10.2459/jcm.0b013e3283629c30] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND The activation of Gq-protein-coupled receptors induces proliferation of vascular smooth muscle cells (VSMCs) proliferation and is involved in vascular remodeling. The regulator of G protein signaling 2 (RGS2), which accelerates the termination of Gq protein signaling, may play a role in vascular remodeling. However, this role remains unclear. METHODS Aortic balloon injury or sham operation was produced in male Wistar rats. Histological examination and gene expression analysis were performed after surgery. In cultured VSMCs after modulation of RGS2 expression, cell proliferation was also evaluated. RESULTS At day 3 after injury, RGS2 transcription was reduced by 52.8% (P <0.05 vs. sham group) with vascular remodeling. In cultured VSMCs stimulated by endothelin-1, phenylephrine or angiotensin II, the proliferation of RGS2 overexpressed cells was significantly inhibited; the proliferation of RGS2 downregulated cells was significantly promoted, compared with that of RGS2 normal cells. Moreover, after incubation with angiotensin II of high concentration (>10 μmol/l) or long term (>8 h), the RGS2 expression was clearly downregulated in cultured VSMCs. Administration of an angiotensin receptor blocker, valsartan (20 mg/kg per day) starting from 1 week preballoon injury to 3 days after injury, restored aortic RGS2 transcription and improved vascular remodeling. CONCLUSION These results suggested that the inhibiting effect of RGS2 on VSMC proliferation is downregulated in vascular remodeling of injured rat aorta, and this effect is likely to be mediated by angiotensin II signaling.
Collapse
Affiliation(s)
- Yuanjie Mao
- aDepartment of Cardiology, Peking University First Hospital, Beijing, China bDepartment of Biochemistry, National Cerebral and Cardiovascular Center, Osaka, Japan cCardiology Division, Cardiovascular Research Center, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island, USA dDepartment of Endocrinology, Beijing Aerospace Center Hospital eDepartment of Physiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
25
|
Ganss R. Keeping the Balance Right. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 133:93-121. [DOI: 10.1016/bs.pmbts.2015.02.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
26
|
Transcription factor cAMP response element modulator (Crem) restrains Pdgf-dependent proliferation of vascular smooth muscle cells in mice. Pflugers Arch 2014; 467:2165-77. [PMID: 25425331 PMCID: PMC4564437 DOI: 10.1007/s00424-014-1652-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 11/10/2014] [Accepted: 11/10/2014] [Indexed: 12/26/2022]
Abstract
Transcription factors of the cAMP response element-binding protein (Creb)/cAMP response element modulator (Crem) family were linked to the switch from a contractile to a proliferating phenotype in vascular smooth muscle cells (VSMCs). Here, we analyzed the vascular function of Crem in mice with a global inactivation of Crem (Crem(-/-)). CRE-mediated transcriptional activity was enhanced in primary Crem(-/-) VSMCs under nonstimulated conditions and under stimulation with Forskolin and platelet-derived growth factor (Pdgf) whereas stimulation with nitric oxide or cGMP showed no effect. This elevated CRE-mediated transcriptional activity as a result of Crem inactivation did not alter aortic contractility or fractions of proliferating or apoptotic aortic VSMCs in situ, and no impact of Crem inactivation on the development of atherosclerotic plaques was observed. Crem(-/-) mice exhibited an increased neointima formation after carotid ligation associated with an increased proliferation of VSMCs in the carotid media. Pdgf-stimulated proliferation of primary aortic Crem(-/-) VSMCs was increased along with an upregulation of messenger RNA (mRNA) levels of Pdgf receptor, alpha polypeptide (Pdgfra), cyclophilin A (Ppia), the regulator of G-protein signaling 5 (Rgs5), and Rho GTPase-activating protein 12 (Arhgap12). Taken together, our data reveal the inhibition of Pdgf-stimulated proliferation of VSMCs by repressing the Pdgf-stimulated CRE-mediated transcriptional activation as the predominant function of Crem in mouse vasculature suggesting an important role of Crem in vasculoproliferative diseases.
Collapse
|
27
|
Regulator of G-protein signaling-5 is a marker of hepatic stellate cells and expression mediates response to liver injury. PLoS One 2014; 9:e108505. [PMID: 25290689 PMCID: PMC4188519 DOI: 10.1371/journal.pone.0108505] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 08/22/2014] [Indexed: 12/11/2022] Open
Abstract
Liver fibrosis is mediated by hepatic stellate cells (HSCs), which respond to a variety of cytokine and growth factors to moderate the response to injury and create extracellular matrix at the site of injury. G-protein coupled receptor (GPCR)-mediated signaling, via endothelin-1 (ET-1) and angiotensin II (AngII), increases HSC contraction, migration and fibrogenesis. Regulator of G-protein signaling-5 (RGS5), an inhibitor of vasoactive GPCR agonists, functions to control GPCR-mediated contraction and hypertrophy in pericytes and smooth muscle cells (SMCs). Therefore we hypothesized that RGS5 controls GPCR signaling in activated HSCs in the context of liver injury. In this study, we localize RGS5 to the HSCs and demonstrate that Rgs5 expression is regulated during carbon tetrachloride (CCl4)-induced acute and chronic liver injury in Rgs5LacZ/LacZ reporter mice. Furthermore, CCl4 treated RGS5-null mice develop increased hepatocyte damage and fibrosis in response to CCl4 and have increased expression of markers of HSC activation. Knockdown of Rgs5 enhances ET-1-mediated signaling in HSCs in vitro. Taken together, we demonstrate that RGS5 is a critical regulator of GPCR signaling in HSCs and regulates HSC activation and fibrogenesis in liver injury.
Collapse
|
28
|
Bökenkamp R, van Brempt R, van Munsteren JC, van den Wijngaert I, de Hoogt R, Finos L, Goeman J, Groot ACGD, Poelmann RE, Blom NA, DeRuiter MC. Dlx1 and Rgs5 in the ductus arteriosus: vessel-specific genes identified by transcriptional profiling of laser-capture microdissected endothelial and smooth muscle cells. PLoS One 2014; 9:e86892. [PMID: 24489801 PMCID: PMC3904938 DOI: 10.1371/journal.pone.0086892] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 12/17/2013] [Indexed: 12/31/2022] Open
Abstract
Closure of the ductus arteriosus (DA) is a crucial step in the transition from fetal to postnatal life. Patent DA is one of the most common cardiovascular anomalies in children with significant clinical consequences especially in premature infants. We aimed to identify genes that specify the DA in the fetus and differentiate it from the aorta. Comparative microarray analysis of laser-captured microdissected endothelial (ECs) and vascular smooth muscle cells (SMCs) from the DA and aorta of fetal rats (embryonic day 18 and 21) identified vessel-specific transcriptional profiles. We found a strong age-dependency of gene expression. Among the genes that were upregulated in the DA the regulator of the G-protein coupled receptor 5 (Rgs5) and the transcription factor distal-less homeobox 1 (Dlx1) exhibited the highest and most significant level of differential expression. The aorta showed a significant preferential expression of the Purkinje cell protein 4 (Pcp4) gene. The results of the microarray analysis were validated by real-time quantitative PCR and immunohistochemistry. Our study confirms vessel-specific transcriptional profiles in ECs and SMCs of rat DA and aorta. Rgs5 and Dlx1 represent novel molecular targets for the regulation of DA maturation and closure.
Collapse
Affiliation(s)
- Regina Bökenkamp
- Department of Pediatric Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Intensive Care, Leiden University Medical Center, Leiden, The Netherlands
- * E-mail:
| | - Ronald van Brempt
- Department of Intensive Care, Leiden University Medical Center, Leiden, The Netherlands
- Johnson and Johnson Pharmaceutical Research and Development, Beerse, Belgium
| | | | | | - Ronald de Hoogt
- Johnson and Johnson Pharmaceutical Research and Development, Beerse, Belgium
| | - Livio Finos
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands
| | - Jelle Goeman
- Biostatistics, Department for Health Evidence, Radboud University Medical Center, Nimegen, The Netherlands
| | - Adriana Cornelia Gittenberger-de Groot
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Robert Eugen Poelmann
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Nicolaas Andreas Blom
- Department of Pediatric Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
29
|
Mahoney WM, Gunaje J, Daum G, Dong XR, Majesky MW. Regulator of G-protein signaling - 5 (RGS5) is a novel repressor of hedgehog signaling. PLoS One 2013; 8:e61421. [PMID: 23637832 PMCID: PMC3630190 DOI: 10.1371/journal.pone.0061421] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 03/10/2013] [Indexed: 01/11/2023] Open
Abstract
Hedgehog (Hh) signaling plays fundamental roles in morphogenesis, tissue repair, and human disease. Initiation of Hh signaling is controlled by the interaction of two multipass membrane proteins, patched (Ptc) and smoothened (Smo). Recent studies identify Smo as a G-protein coupled receptor (GPCR)-like protein that signals through large G-protein complexes which contain the Gαi subunit. We hypothesize Regulator of G-Protein Signaling (RGS) proteins, and specifically RGS5, are endogenous repressors of Hh signaling via their ability to act as GTPase activating proteins (GAPs) for GTP-bound Gαi, downstream of Smo. In support of this hypothesis, we demonstrate that RGS5 over-expression inhibits sonic hedgehog (Shh)-mediated signaling and osteogenesis in C3H10T1/2 cells. Conversely, signaling is potentiated by siRNA-mediated knock-down of RGS5 expression, but not RGS4 expression. Furthermore, using immuohistochemical analysis and co-immunoprecipitation (Co-IP), we demonstrate that RGS5 is present with Smo in primary cilia. This organelle is required for canonical Hh signaling in mammalian cells, and RGS5 is found in a physical complex with Smo in these cells. We therefore conclude that RGS5 is an endogenous regulator of Hh-mediated signaling and that RGS proteins are potential targets for novel therapeutics in Hh-mediated diseases.
Collapse
Affiliation(s)
- William M. Mahoney
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
- Center for Cardiovascular Biology, University of Washington, Seattle, Washington, United States of America
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, United States of America
- * E-mail: (WMM); (MWM)
| | - Jagadambika Gunaje
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
- Center for Cardiovascular Biology, University of Washington, Seattle, Washington, United States of America
| | - Guenter Daum
- Department of Surgery, University of Washington, Seattle, Washington, United States of America
- Center for Cardiovascular Biology, University of Washington, Seattle, Washington, United States of America
| | - Xiu Rong Dong
- Seattle Children’s Research Institute, University of Washington, Seattle, Washington, United States of America
| | - Mark W. Majesky
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
- Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
- Center for Cardiovascular Biology, University of Washington, Seattle, Washington, United States of America
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, United States of America
- Seattle Children’s Research Institute, University of Washington, Seattle, Washington, United States of America
- * E-mail: (WMM); (MWM)
| |
Collapse
|
30
|
Holobotovskyy V, Manzur M, Tare M, Burchell J, Bolitho E, Viola H, Hool LC, Arnolda LF, McKitrick DJ, Ganss R. Regulator of G-protein signaling 5 controls blood pressure homeostasis and vessel wall remodeling. Circ Res 2013; 112:781-91. [PMID: 23303165 DOI: 10.1161/circresaha.111.300142] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Regulator of G-protein signaling 5 (RGS5) modulates G-protein-coupled receptor signaling and is prominently expressed in arterial smooth muscle cells. Our group first reported that RGS5 is important in vascular remodeling during tumor angiogenesis. We hypothesized that RGS5 may play an important role in vessel wall remodeling and blood pressure regulation. OBJECTIVE To demonstrate that RGS5 has a unique and nonredundant role in the pathogenesis of hypertension and to identify crucial RGS5-regulated signaling pathways. METHODS AND RESULTS We observed that arterial RGS5 expression is downregulated with chronically elevated blood pressure after angiotensin II infusion. Using a knockout mouse model, radiotelemetry, and pharmacological inhibition, we subsequently showed that loss of RGS5 results in profound hypertension. RGS5 signaling is linked to the renin-angiotensin system and directly controls vascular resistance, vessel contractility, and remodeling. RGS5 deficiency aggravates pathophysiological features of hypertension, such as medial hypertrophy and fibrosis. Moreover, we demonstrate that protein kinase C, mitogen-activated protein kinase/extracellular signal-regulated kinase, and Rho kinase signaling pathways are major effectors of RGS5-mediated hypertension. CONCLUSIONS Loss of RGS5 results in hypertension. Loss of RGS5 signaling also correlates with hyper-responsiveness to vasoconstrictors and vascular stiffening. This establishes a significant, distinct, and causal role of RGS5 in vascular homeostasis. RGS5 modulates signaling through the angiotensin II receptor 1 and major Gαq-coupled downstream pathways, including Rho kinase. So far, activation of RhoA/Rho kinase has not been associated with RGS molecules. Thus, RGS5 is a crucial regulator of blood pressure homeostasis with significant clinical implications for vascular pathologies, such as hypertension.
Collapse
Affiliation(s)
- Vasyl Holobotovskyy
- Western Australian Institute for Medical Research, Rear, 50 Murray St, Perth, WA 6010, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Garaffo G, Provero P, Molineris I, Pinciroli P, Peano C, Battaglia C, Tomaiuolo D, Etzion T, Gothilf Y, Santoro M, Merlo GR. Profiling, Bioinformatic, and Functional Data on the Developing Olfactory/GnRH System Reveal Cellular and Molecular Pathways Essential for This Process and Potentially Relevant for the Kallmann Syndrome. Front Endocrinol (Lausanne) 2013; 4:203. [PMID: 24427155 PMCID: PMC3876029 DOI: 10.3389/fendo.2013.00203] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 12/18/2013] [Indexed: 11/28/2022] Open
Abstract
During embryonic development, immature neurons in the olfactory epithelium (OE) extend axons through the nasal mesenchyme, to contact projection neurons in the olfactory bulb. Axon navigation is accompanied by migration of the GnRH+ neurons, which enter the anterior forebrain and home in the septo-hypothalamic area. This process can be interrupted at various points and lead to the onset of the Kallmann syndrome (KS), a disorder characterized by anosmia and central hypogonadotropic hypogonadism. Several genes has been identified in human and mice that cause KS or a KS-like phenotype. In mice a set of transcription factors appears to be required for olfactory connectivity and GnRH neuron migration; thus we explored the transcriptional network underlying this developmental process by profiling the OE and the adjacent mesenchyme at three embryonic ages. We also profiled the OE from embryos null for Dlx5, a homeogene that causes a KS-like phenotype when deleted. We identified 20 interesting genes belonging to the following categories: (1) transmembrane adhesion/receptor, (2) axon-glia interaction, (3) scaffold/adapter for signaling, (4) synaptic proteins. We tested some of them in zebrafish embryos: the depletion of five (of six) Dlx5 targets affected axonal extension and targeting, while three (of three) affected GnRH neuron position and neurite organization. Thus, we confirmed the importance of cell-cell and cell-matrix interactions and identified new molecules needed for olfactory connection and GnRH neuron migration. Using available and newly generated data, we predicted/prioritized putative KS-disease genes, by building conserved co-expression networks with all known disease genes in human and mouse. The results show the overall validity of approaches based on high-throughput data and predictive bioinformatics to identify genes potentially relevant for the molecular pathogenesis of KS. A number of candidate will be discussed, that should be tested in future mutation screens.
Collapse
Affiliation(s)
- Giulia Garaffo
- Department of Molecular Biotechnology and Health Science, University of Torino, Torino, Italy
| | - Paolo Provero
- Department of Molecular Biotechnology and Health Science, University of Torino, Torino, Italy
| | - Ivan Molineris
- Department of Molecular Biotechnology and Health Science, University of Torino, Torino, Italy
| | - Patrizia Pinciroli
- Department of Medical Biotechnology Translational Medicine (BIOMETRA), University of Milano, Milano, Italy
| | - Clelia Peano
- Institute of Biomedical Technology, National Research Council, ITB-CNR, Segrate, Italy
| | - Cristina Battaglia
- Department of Medical Biotechnology Translational Medicine (BIOMETRA), University of Milano, Milano, Italy
- Institute of Biomedical Technology, National Research Council, ITB-CNR, Segrate, Italy
| | - Daniela Tomaiuolo
- Department of Molecular Biotechnology and Health Science, University of Torino, Torino, Italy
| | - Talya Etzion
- The George S. Wise Faculty of Life Sciences, Department of Neurobiology, Tel-Aviv University, Tel-Aviv, Israel
| | - Yoav Gothilf
- The George S. Wise Faculty of Life Sciences, Department of Neurobiology, Tel-Aviv University, Tel-Aviv, Israel
| | - Massimo Santoro
- Department of Molecular Biotechnology and Health Science, University of Torino, Torino, Italy
| | - Giorgio R. Merlo
- Department of Molecular Biotechnology and Health Science, University of Torino, Torino, Italy
- *Correspondence: Giorgio R. Merlo, Department of Molecular Biotechnology and Health Science, University of Torino, Via Nizza 52, Torino 10126, Italy e-mail:
| |
Collapse
|
32
|
Althoff TF, Albarrán Juárez J, Troidl K, Tang C, Wang S, Wirth A, Takefuji M, Wettschureck N, Offermanns S. Procontractile G protein-mediated signaling pathways antagonistically regulate smooth muscle differentiation in vascular remodeling. ACTA ACUST UNITED AC 2012; 209:2277-90. [PMID: 23129751 PMCID: PMC3501360 DOI: 10.1084/jem.20120350] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Vascular smooth muscle (Sm) cells (VSMCs) are highly plastic. Their differentiation state can be regulated by serum response factor (SRF), which activates genes involved in Sm differentiation and proliferation by recruiting cofactors, such as members of the myocardin family and ternary complex factors (TCFs), respectively. However, the extracellular cues and upstream signaling mechanisms regulating SRF-dependent VSMC differentiation under in vivo conditions are poorly understood. In this study, we show that the procontractile signaling pathways mediated by the G proteins G(12)/G(13) and G(q)/G(11) antagonistically regulate VSMC plasticity in different models of vascular remodeling. In mice lacking Gα(12)/Gα(13) or their effector, the RhoGEF protein LARG, RhoA-dependent SRF-regulation was blocked and down-regulation of VSMC differentiation marker genes was enhanced. This was accompanied by an excessive vascular remodeling and exacerbation of atherosclerosis. In contrast, Sm-specific Gα(q)/Gα(11) deficiency blocked activation of extracellular signal-regulated kinase 1/2 and the TCF Elk-1, resulting in a reduced VSMC dedifferentiation in response to flow cessation or vascular injury. These data show that the balanced activity of both G protein-mediated pathways in VSMCs is required for an appropriate vessel remodeling response in vascular diseases and suggest new approaches to modulate Sm differentiation in vascular pathologies.
Collapse
Affiliation(s)
- Till F Althoff
- Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Pett K, Hauton D. The effects of asymmetric ventricular filling on left-right ventricular interaction in the normal rat heart. Pflugers Arch 2012; 464:523-34. [PMID: 23001120 DOI: 10.1007/s00424-012-1154-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 08/31/2012] [Accepted: 09/04/2012] [Indexed: 11/28/2022]
Abstract
Heart failure is characterised by ventricular dysfunction and with the potential for changes to ventricular volumes constraining the mechanical performance of the heart. The contribution of this interaction from geometric changes rather than fibrosis or metabolic changes is unclear. Using the constant pressure Langendorff-perfused rat heart, the volume interaction between left ventricle (LV) and right ventricle (RV) was investigated. RV diastolic stiffness (P < 0.001) and developed pressure (P < 0.001) were significantly lower than LV. When the RV was fixed at the end-diastolic volume (EDV) or EDV + 50 %, both LV systolic and diastolic performance were unaffected with increasing LV balloon volume. However, at fixed LV volume, RV systolic performance was significantly decreased when LV volume increased to EDV + 50 % when RV volume was increased incrementally between 50 and 300 μl (P < 0.001). Systolic interaction in RV was noted as declining RV peak systolic load with increasing LV systolic pressure (P < 0.05) and diastolic interaction was noted for RV when LV volume was increased from EDV to EDV + 50 % (P < 0.05). RV diastolic wall stress was increased with increasing LV balloon volume (P < 0.05), but LV wall stress was unaltered at fixed RV balloon volume. Taken together, increasing LV volume above EDV decreased systolic performance and triggered ventricular constraint in the RV but the RV itself had no effect on the performance of the LV. These results are consistent with overload of the LV impairing pulmonary perfusion by direct ventricular interaction with potential alteration to ventilation-perfusion characteristics within the lung.
Collapse
Affiliation(s)
- Kimberley Pett
- School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | | |
Collapse
|
34
|
Kach J, Sethakorn N, Dulin NO. A finer tuning of G-protein signaling through regulated control of RGS proteins. Am J Physiol Heart Circ Physiol 2012; 303:H19-35. [PMID: 22542620 DOI: 10.1152/ajpheart.00764.2011] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Regulators of G-protein signaling (RGS) proteins are GTPase-activating proteins (GAP) for various Gα subunits of heterotrimeric G proteins. Through this mechanism, RGS proteins regulate the magnitude and duration of G-protein-coupled receptor signaling and are often referred to as fine tuners of G-protein signaling. Increasing evidence suggests that RGS proteins themselves are regulated through multiple mechanisms, which may provide an even finer tuning of G-protein signaling and crosstalk between G-protein-coupled receptors and other signaling pathways. This review summarizes the current data on the control of RGS function through regulated expression, intracellular localization, and covalent modification of RGS proteins, as related to cell function and the pathogenesis of diseases.
Collapse
Affiliation(s)
- Jacob Kach
- Department of Medicine, University of Chicago, Illinois, 60637, USA
| | | | | |
Collapse
|
35
|
Loss of regulator of G protein signaling 5 exacerbates obesity, hepatic steatosis, inflammation and insulin resistance. PLoS One 2012; 7:e30256. [PMID: 22272317 PMCID: PMC3260252 DOI: 10.1371/journal.pone.0030256] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 12/12/2011] [Indexed: 01/22/2023] Open
Abstract
Background The effect of regulator of G protein signaling 5 (RGS5) on cardiac hypertrophy, atherosclerosis and angiogenesis has been well demonstrated, but the role in the development of obesity and insulin resistance remains completely unknown. We determined the effect of RGS5 deficiency on obesity, hepatic steatosis, inflammation and insulin resistance in mice fed either a normal-chow diet (NC) or a high-fat diet (HF). Methodology/Principal Findings Male, 8-week-old RGS5 knockout (KO) and littermate control mice were fed an NC or an HF for 24 weeks and were phenotyped accordingly. RGS5 KO mice exhibited increased obesity, fat mass and ectopic lipid deposition in the liver compared with littermate control mice, regardless of diet. When fed an HF, RGS5 KO mice had a markedly exacerbated metabolic dysfunction and inflammatory state in the blood serum. Meanwhile, macrophage recruitment and inflammation were increased and these increases were associated with the significant activation of JNK, IκBα and NF-κBp65 in the adipose tissue, liver and skeletal muscle of RGS5 KO mice fed an HF relative to control mice. These exacerbated metabolic dysfunction and inflammation are accompanied with decreased systemic insulin sensitivity in the adipose tissue, liver and skeletal muscle of RGS5 KO mice, reflected by weakened Akt/GSK3β phosphorylation. Conclusions/Significance Our data suggest that loss of RGS5 exacerbates HF-induced obesity, hepatic steatosis, inflammation and insulin resistance.
Collapse
|