1
|
Zhuo X, Wu Y, Fu X, Liang X, Xiang Y, Li J, Mao C, Jiang Y. The Yin‐Yang roles of protease‐activated receptors in inflammatory signalling and diseases. FEBS J 2022; 289:4000-4020. [DOI: 10.1111/febs.16406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 01/26/2022] [Accepted: 02/15/2022] [Indexed: 12/15/2022]
Affiliation(s)
- Xin Zhuo
- School of Life Science and Engineering Southwest Jiaotong University Chengdu China
| | - Yue Wu
- School of Life Science and Engineering Southwest Jiaotong University Chengdu China
| | - Xiujuan Fu
- School of Life Science and Engineering Southwest Jiaotong University Chengdu China
| | - Xiaoyu Liang
- School of Life Science and Engineering Southwest Jiaotong University Chengdu China
| | - Yuxin Xiang
- School of Life Science and Engineering Southwest Jiaotong University Chengdu China
| | - Jianbin Li
- School of Life Science and Engineering Southwest Jiaotong University Chengdu China
| | - Canquan Mao
- School of Life Science and Engineering Southwest Jiaotong University Chengdu China
| | - Yuhong Jiang
- School of Life Science and Engineering Southwest Jiaotong University Chengdu China
| |
Collapse
|
2
|
Chen H, Smith M, Herz J, Li T, Hasley R, Le Saout C, Zhu Z, Cheng J, Gronda A, Martina JA, Irusta PM, Karpova T, McGavern DB, Catalfamo M. The role of protease-activated receptor 1 signaling in CD8 T cell effector functions. iScience 2021; 24:103387. [PMID: 34841225 PMCID: PMC8605340 DOI: 10.1016/j.isci.2021.103387] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 09/28/2021] [Accepted: 10/28/2021] [Indexed: 12/15/2022] Open
Abstract
CD8 T cells are essential for adaptive immunity against viral infections. Protease activated receptor 1 (PAR1) is expressed by CD8 T cells; however, its role in T cell effector function is not well defined. Here we show that in human CD8 T cells, PAR1 stimulation accelerates calcium mobilization. Furthermore, PAR1 is involved in cytotoxic T cell function by facilitating granule trafficking via actin polymerization and repositioning of the microtubule organizing center (MTOC) toward the immunological synapse. In vivo, PAR1-/- mice have reduced cytokine-producing T cells in response to a lymphocytic choriomeningitis virus (LCMV) infection and fail to efficiently control the virus. Specific deletion of PAR1 in LCMV GP33-specific CD8 T cells results in reduced expansion and diminished effector function. These data demonstrate that PAR1 plays a role in T cell activation and function, and this pathway could represent a new therapeutic strategy to modulate CD8 T cell effector function.
Collapse
Affiliation(s)
- Hui Chen
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, USA
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mindy Smith
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jasmin Herz
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Tong Li
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, USA
| | - Rebecca Hasley
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Cecile Le Saout
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ziang Zhu
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, USA
| | - Jie Cheng
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, USA
| | - Andres Gronda
- Department of Human Science, Georgetown University, Washington, DC, USA
| | - José A. Martina
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Pablo M. Irusta
- Department of Human Science, Georgetown University, Washington, DC, USA
| | - Tatiana Karpova
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dorian B. McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Marta Catalfamo
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, USA
| |
Collapse
|
3
|
Bergeron A, Brezai A, Shukr R, Villeneuve L, Allen BG, Qureshi WMS, Hentges KE, Calderone A. Filamentous nestin and nonmuscle myosin IIB are associated with a migratory phenotype in neonatal rat cardiomyocytes. J Cell Physiol 2020; 236:1281-1294. [PMID: 32654195 DOI: 10.1002/jcp.29934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 06/28/2020] [Accepted: 07/01/2020] [Indexed: 12/13/2022]
Abstract
Cardiomyocyte migration represents a requisite event of cardiogenesis and the regenerative response of the injured adult zebrafish and neonatal rodent heart. The present study tested the hypothesis that the appearance of the intermediate filament protein nestin in neonatal rat ventricular cardiomyocytes (NNVMs) was associated in part with the acquisition of a migratory phenotype. The cotreatment of NNVMs with phorbol 12,13-dibutyrate (PDBu) and the p38α/β mitogen-activated protein kinase inhibitor SB203580 led to the de novo synthesis of nestin. The intermediate filament protein was subsequently reorganized into a filamentous pattern and redistributed to the leading edge of elongated membrane protrusions translating to significant lengthening of NNVMs. PDBu/SB203580 treatment concomitantly promoted the reorganization of nonmuscle myosin IIB (NMIIB) located predominantly at the periphery of the plasma membrane of NNVMs to a filamentous phenotype extending to the leading edge of elongated membrane protrusions. Coimmunoprecipitation assay revealed a physical interaction between NMIIB and nestin after PDBu/SB203580 treatment of NNVMs. In wild-type and heterozygous NMIIB embryonic hearts at E11.5-E14.5 days, nestin immunoreactivity was identified in a subpopulation of cardiomyocytes elongating perpendicular to the compact myocardium, at the leading edge of nascent trabeculae and during thickening of the compact myocardium. In mutant embryonic hearts lacking NMIIB protein expression, trabeculae formation was reduced, the compact myocardium significantly thinner and nestin immunoreactivity undetectable in cardiomyocytes at E14.5 days. These data suggest that NMIIB and nestin may act in a coordinated fashion to facilitate the acquisition of a migratory phenotype in neonatal and embryonic cardiomyocytes.
Collapse
Affiliation(s)
- Alexandre Bergeron
- Research Center, Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Andra Brezai
- Research Center, Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Rami Shukr
- Research Center, Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Louis Villeneuve
- Research Center, Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Bruce G Allen
- Research Center, Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Wasay M S Qureshi
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Kathryn E Hentges
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Angelino Calderone
- Research Center, Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada.,Department of Pharmacology and Physiology, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
4
|
Mohamed R, Cao Y, Afroz R, Xu S, Ta HT, Barras M, Zheng W, Little PJ, Kamato D. ROS directly activates transforming growth factor β type 1 receptor signalling in human vascular smooth muscle cells. Biochim Biophys Acta Gen Subj 2020; 1864:129463. [DOI: 10.1016/j.bbagen.2019.129463] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/25/2019] [Accepted: 10/07/2019] [Indexed: 12/11/2022]
|
5
|
Merkus D, van Beusekom HMM, Boomars KA. Protease-activated receptor 1 as potential therapeutic target in pulmonary arterial hypertension. Cardiovasc Res 2019; 115:1260-1261. [PMID: 30903137 DOI: 10.1093/cvr/cvz071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Daphne Merkus
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, CA Rotterdam, The Netherlands
| | - Heleen M M van Beusekom
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, CA Rotterdam, The Netherlands
| | - Karin A Boomars
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
6
|
Hertig V, Brezai A, Bergeron A, Villeneuve L, Gillis MA, Calderone A. p38α MAPK inhibition translates to cell cycle re-entry of neonatal rat ventricular cardiomyocytes and de novo nestin expression in response to thrombin and after apex resection. Sci Rep 2019; 9:8203. [PMID: 31160695 PMCID: PMC6547723 DOI: 10.1038/s41598-019-44712-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 05/20/2019] [Indexed: 12/18/2022] Open
Abstract
The present study tested the hypothesis that p38α MAPK inhibition leads to cell cycle re-entry of neonatal ventricular cardiomyocytes (NNVMs) and de novo nestin expression in response to thrombin and after apex resection of the neonatal rat heart. Thrombin (1 U/ml) treatment of 1-day old NNVMs did not induce cell cycle re-entry or nestin expression. Acute exposure of NNVMs to thrombin increased p38α MAPK and HSP27 phosphorylation and p38α/β MAPK inhibitor SB203580 abrogated HSP27 phosphorylation. Thrombin and SB203580 co-treatment of NNVMs led to bromodeoxyuridine incorporation and nestin expression. SB203580 (5 mg/kg) administration immediately after apex resection of 1-day old neonatal rat hearts and continued for two additional days shortened the fibrin clot length sealing the exposed left ventricular chamber. SB203580-treatment increased the density of troponin-T(+)-NNVMs that incorporated bromodeoxyuridine and expressed nuclear phosphohistone-3. Nestin(+)-NNVMs were selectively detected at the border of the fibrin clot and SB203580 potentiated the density that re-entered the cell cycle. These data suggest that the greater density of ventricular cardiomyocytes and nestin(+)-ventricular cardiomyocytes that re-entered the cell cycle after SB203580 treatment of the apex-resected neonatal rat heart during the acute phase of fibrin clot formation may be attributed in part to inhibition of thrombin-mediated p38α MAPK signalling.
Collapse
Affiliation(s)
- Vanessa Hertig
- Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Andra Brezai
- Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Alexandre Bergeron
- Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Louis Villeneuve
- Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | | | - Angelino Calderone
- Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada.
- Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
7
|
Wu J, Zhang C, Liu C, Zhang A, Li A, Zhang J, Zhang Y. Aortic constriction induces hypertension and cardiac hypertrophy via (pro)renin receptor activation and the PLC‑β3 signaling pathway. Mol Med Rep 2018; 19:573-580. [PMID: 30431106 DOI: 10.3892/mmr.2018.9653] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 10/04/2018] [Indexed: 11/06/2022] Open
Abstract
The (pro)renin receptor [(P)RR] serves an important role in cardiovascular complications. However, the precise mechanisms of (P)RR in the heart remain obscure. The authors hypothesized that overexpression of (P)RR would be associated with activation of the relevant signal pathway which could lead to organ injury. The aim of the present study was to test the role of cardiac (P)RR and its potential signaling pathway components including phospholipase C (PLC), protein kinase C (PKC), extracellular signal‑regulated kinase (ERK)1/2 and Raf‑1 proto‑oncogene, serine/threonine kinase (Raf‑1). Hypertension and cardiac hypertrophy were induced by partial abdominal aortic ligation in Sprague‑Dawley rats. The expression levels of cardiac (P)RR, PLC‑β3, PKC, ERK1/2 and Raf‑1 were measured following administration of the handle region peptide (HRP) and PLC‑β3 inhibitor U73122. The expression of (P)RR and PLC‑β3 significantly increased in the left ventricle (P<0.05). Levels of PKC‑α, ERK1/2 and Raf‑1 in the heart rose significantly (P<0.05). HRP and U73122 significantly decreased the levels of cardiac (P)RR and PLC‑β3. Furthermore, levels of PKC‑α, ERK1/2 and Raf‑1 were also decreased (P<0.05). Cardiac parameters, blood pressure and plasma Angiotensin (Ang) I and Ang II levels were altered significantly (P<0.05). The results demonstrated that hypertension induced by aortic restriction activated the (P)RR in the heart. This action led to hypertension and cardiac hypertrophy via the (P)RR‑PLC‑β3‑PKC‑ERK1/2‑Raf‑1 signaling pathway. These results provide a mechanism by which elevated (P)RR levels in hypertension may contribute to the development of cardiac remodeling.
Collapse
Affiliation(s)
- Junyan Wu
- Institute of Cardiovascular Diseases, Taian Maternal and Child Health Hospital, Taian, Shandong 271000, P.R. China
| | - Cong Zhang
- Department of Endocrinology, Peking Union Medical College Hospital, Beijing 100730, P.R. China
| | - Chuanjun Liu
- Institute of Cardiovascular Diseases, Taian Maternal and Child Health Hospital, Taian, Shandong 271000, P.R. China
| | - Aihua Zhang
- Institute of Cardiovascular Diseases, Taian Maternal and Child Health Hospital, Taian, Shandong 271000, P.R. China
| | - An Li
- Institute of Cardiovascular Diseases, Taian Maternal and Child Health Hospital, Taian, Shandong 271000, P.R. China
| | - Jingjun Zhang
- Department of Neurology, Taishan Medical University, Taian, Shandong 271000, P.R. China
| | - Yanling Zhang
- Institute of Cardiovascular Diseases, Taian Maternal and Child Health Hospital, Taian, Shandong 271000, P.R. China
| |
Collapse
|
8
|
Yao ZH, Xie HJ, Yuan YL, Huo YT, Cao J, Lai WY, Cai RJ, Cheng YX. Contraction-dependent TGF-β1 activation is required for thrombin-induced remodeling in human airway smooth muscle cells. Life Sci 2018; 197:130-139. [PMID: 29428600 DOI: 10.1016/j.lfs.2018.02.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 02/06/2018] [Accepted: 02/07/2018] [Indexed: 01/08/2023]
Abstract
AIMS Thrombin is a serine proteinase that is not only involved in coagulation cascade, but also mediates a number of biological responses relevant to tissues repair, and induces bronchoconstriction. TGF-β plays a pivotal role in airway remodeling due to its effects on airway smooth muscle proliferation and extracellular matrix (ECM) deposition. Recently, bronchoconstriction itself is found to constitute a form of strain and is highly relevant to asthmatic airway remodeling. However, the underlying mechanisms remain unknown. Here, we investigated the role of contraction- dependent TGF-β activation in thrombin-induced remodeling in human airway smooth muscle (HASM) cells. MATERIALS AND METHODS Primary HASM cells were treated with or without thrombin in the absence or presence of anti-TGF-β antibody, cytochalasin D and formoterol. CFSE labeling index or CCK-8 assay were performed to test cell proliferation. RT-PCR and Western blotting were used to examined ECM mRNA level and collagen Iα1, α-actin protein expression, respectively. Immunofluorescence was also used to confirm contraction induced by thrombin in HASM cells. KEY FINDING Thrombin stimulation enhanced HASM cells proliferation and activated TGF-β signaling. Thrombin induced ECM mRNA and collagen Iα1 protein expression, and these effects are mediated by TGF-β. Abrogation of TGF-β activation by contraction inhibitors cytochalasin D and formoterol prevents the thrombin-induced effects. SIGNIFICANCE These findings suggest that contraction-dependent TGF-β activation could be a mechanism by which thrombin leads to the development of asthmatic airway remodeling. Blocking physical forces with bronchodilator would be an intriguing way in reducing airway remodeling in asthma.
Collapse
Affiliation(s)
- Zhi-Hui Yao
- Department of Respiratory Disease, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China; Department of Respiratory Disease, Hengyang NO.1 Peoples Hospital, Hengyang, Hunan, China
| | - Hao-Jun Xie
- Department of Respiratory Disease, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Ya-Lu Yuan
- Department of Respiratory Disease, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China; Department of Critical Care Medicine, Affiliated Foshan Hospital of Southern Medical University, Foshan, Guangdong, China
| | - Ya-Ting Huo
- Department of Respiratory Disease, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Jing Cao
- Department of Respiratory Disease, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Wen-Yan Lai
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Rui-Jun Cai
- Department of Thoracic Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuan-Xiong Cheng
- Department of Respiratory Disease, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.
| |
Collapse
|
9
|
Castaldi A, Chesini GP, Taylor AE, Sussman MA, Brown JH, Purcell NH. Sphingosine 1-phosphate elicits RhoA-dependent proliferation and MRTF-A mediated gene induction in CPCs. Cell Signal 2016; 28:871-9. [PMID: 27094722 PMCID: PMC5004781 DOI: 10.1016/j.cellsig.2016.04.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 04/01/2016] [Accepted: 04/10/2016] [Indexed: 12/16/2022]
Abstract
Although c-kit(+) cardiac progenitor cells (CPCs) are currently used in clinical trials there remain considerable gaps in our understanding of the molecular mechanisms underlying their proliferation and differentiation. G-protein coupled receptors (GPCRs) play an important role in regulating these processes in mammalian cell types thus we assessed GPCR mRNA expression in c-kit(+) cells isolated from adult mouse hearts. Our data provide the first comprehensive overview of the distribution of this fundamental class of cardiac receptors in CPCs and reveal notable distinctions from that of adult cardiomyocytes. We focused on GPCRs that couple to RhoA activation in particular those for sphingosine-1-phosphate (S1P). The S1P2 and S1P3 receptors are the most abundant S1P receptor subtypes in mouse and human CPCs while cardiomyocytes express predominantly S1P1 receptors. Treatment of CPCs with S1P, as with thrombin and serum, increased proliferation through a pathway requiring RhoA signaling, as evidenced by significant attenuation when Rho was inhibited by treatment with C3 toxin. Further analysis demonstrated that both S1P- and serum-induced proliferation are regulated through the S1P2 and S1P3 receptor subtypes which couple to Gα12/13 to elicit RhoA activation. The transcriptional co-activator MRTF-A was activated by S1P as assessed by its nuclear accumulation and induction of a RhoA/MRTF-A luciferase reporter. In addition S1P treatment increased expression of cardiac lineage markers Mef2C and GATA4 and the smooth muscle marker GATA6 through activation of MRTF-A. In conclusion, we delineate an S1P-regulated signaling pathway in CPCs that introduces the possibility of targeting S1P2/3 receptors, Gα12/13 or RhoA to influence the proliferation and commitment of c-kit(+) CPCs and improve the response of the myocardium following injury.
Collapse
Affiliation(s)
- Alessandra Castaldi
- Department of Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0636, USA
| | - Gino P Chesini
- Department of Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0636, USA
| | - Amy E Taylor
- Department of Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0636, USA
| | - Mark A Sussman
- San Diego State Heart Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Joan Heller Brown
- Department of Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0636, USA.
| | - Nicole H Purcell
- Department of Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0636, USA
| |
Collapse
|
10
|
Jumeau C, Rupin A, Chieng-Yane P, Mougenot N, Zahr N, David-Dufilho M, Hatem SN. Direct Thrombin Inhibitors Prevent Left Atrial Remodeling Associated With Heart Failure in Rats. JACC Basic Transl Sci 2016; 1:328-339. [PMID: 27642643 PMCID: PMC5012373 DOI: 10.1016/j.jacbts.2016.05.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 05/02/2016] [Accepted: 05/02/2016] [Indexed: 01/31/2023]
Abstract
The present study tested the hypothesis that thrombin participates in formation of left atrial remodeling and that direct oral anticoagulants, such as direct thrombin inhibitors (DTIs), can prevent its progression. In a rat model of heart failure associated with left atrial dilation, we found that chronic treatment with DTIs reduces the atrial remodeling and the duration of atrial fibrillation (AF) episodes induced by burst pacing by inhibiting myocardial hypertrophy and fibrosis. In addition to the prevention of thromboembolism complicating AF, DTIs may be of interest to slow down the progression of the arrhythmogenic substrate.
Collapse
Key Words
- AF, atrial fibrillation
- ANP, atrial natriuretic peptide
- BNP, brain natriuretic peptide
- CTGF, connective tissue growth factor
- DTI, direct thrombin inhibitor
- MHC, myosin heavy chain
- MI, myocardial infarction
- NFATc3, nuclear factor of activated T cells 3
- PAI, plasminogen activator inhibitor
- PAR, protease-activated receptor
- anticoagulant
- atrial arrhythmia
- direct thrombin inhibitor
- heart failure
- remodeling
Collapse
Affiliation(s)
- Céline Jumeau
- Sorbonne University, Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche 1166, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
- Servier Research Institute, Suresnes, France
| | - Alain Rupin
- Servier Research Institute, Suresnes, France
| | | | - Nathalie Mougenot
- Inserm-Sorbonnes-Universités, Unité Mixte de Service 28 Université Pierre et Marie Curie, Paris, France
| | - Noël Zahr
- INSERM Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Pitié-Salpêtrière Department of Pharmacology and Centre d'Investigation Clinique 1421, Paris, France
| | - Monique David-Dufilho
- Sorbonne University, Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche 1166, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Stéphane N. Hatem
- Sorbonne University, Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche 1166, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
- Cardiology Department, ICAN, Paris, France
| |
Collapse
|
11
|
Gu Y, Wang C, Li G, Huang LYM. EXPRESS: F-actin links Epac-PKC signaling to purinergic P2X3 receptors sensitization in dorsal root ganglia following inflammation. Mol Pain 2016; 12:12/0/1744806916660557. [PMID: 27385722 PMCID: PMC4955968 DOI: 10.1177/1744806916660557] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Sensitization of purinergic P2X3 receptors (P2X3Rs) contributes to the production of exaggerated nociceptive responses following inflammatory injury. We showed previously that prostaglandin E2 (PGE2) potentiates P2X3R-mediated ATP currents in dorsal root ganglion neurons isolated from both control and complete Freund’s adjuvant-induced inflamed rats. PGE2 potentiation of ATP currents depends only on PKA signaling in control neurons, but it depends on both PKA and PKC signaling in inflamed neurons. We further found that inflammation evokes an increase in exchange proteins directly activated by cAMP (Epacs) in dorsal root ganglions. This increase promotes the activation of PKC to produce a much enhanced PGE2 effect on ATP currents and to elicit Epac-dependent flinch nocifensive behavioral responses in complete Freund’s adjuvant rats. The link between Epac-PKC signaling and P2X3R sensitization remains unexplored. Here, we show that the activation of Epacs promotes the expression of phosphorylated PKC and leads to an increase in the cytoskeleton, F-actin, expression at the cell perimeter. Depolymerization of F-actin blocks PGE2-enhanced ATP currents and inhibits P2X3R-mediated nocifensive responses after inflammation. Thus, F-actin is dynamically involved in the Epac-PKC-dependent P2X3R sensitization. Furthermore, Epacs induce a PKC-dependent increase in the membrane expression of P2X3Rs. This increase is abolished by F-actin depolymerization, suggesting that F-actin mediates Epac-PKC signaling of P2X3R membrane expression. Thus, after inflammation, an Epac-PKC dependent increase in F-actin in dorsal root ganglion neurons enhances the membrane expression of P2X3Rs to bring about sensitization of P2X3Rs and abnormal pain behaviors.
Collapse
Affiliation(s)
- Yanping Gu
- University of Texas Medical Branch at Galveston
| | - Congying Wang
- University of Texas Medical Branch at GalvestonUniversity of Texas Medical Branch at Galveston
| | - Guangwen Li
- University of Texas Medical Branch at Galveston
| | - Li-Yen Mae Huang
- University of Texas Medical Branch at GalvestonUniversity of Texas Medical Branch at Galveston
| |
Collapse
|
12
|
Chien PTY, Lin CC, Hsiao LD, Yang CM. c-Src/Pyk2/EGFR/PI3K/Akt/CREB-activated pathway contributes to human cardiomyocyte hypertrophy: Role of COX-2 induction. Mol Cell Endocrinol 2015; 409:59-72. [PMID: 25869400 DOI: 10.1016/j.mce.2015.04.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 03/23/2015] [Accepted: 04/07/2015] [Indexed: 12/27/2022]
Abstract
Thrombin and COX-2 regulating cardiac hypertrophy are via various signaling cascades. Several transcriptional factors including CREB involve in COX-2 expression. However, the interplay among thrombin, CREB, and COX-2 in primary human neonatal ventricular cardiomyocytes remains unclear. In this study, thrombin-induced COX-2 promoter activity, mRNA and protein expression, and PGE2 synthesis were attenuated by pretreatment with the inhibitors of c-Src (PP1), Pyk2 (PF431396), EGFR (AG1478), PI3K/Akt (LY294002/SH-5), and p300 (GR343), or transfection with siRNAs of c-Src, Pyk2, EGFR, p110, Akt, CREB, and p300. Moreover, thrombin-stimulated phosphorylation of c-Src, Pyk2, EGFR, Akt, CREB and p300 was attenuated by their respective inhibitors. These results indicate that thrombin-induced COX-2 expression is mediated through PAR-1/c-Src/Pyk2/EGFR/PI3K/Akt linking to CREB and p300 cascades. Functionally, thrombin-induced hypertrophy and ANF/BNP release were, at least in part, mediated through a PAR-1/COX-2-dependent pathway. We uncover the importance of COX-2 regarding human cardiomyocyte hypertrophy that will provide a therapeutic intervention in cardiovascular diseases.
Collapse
Affiliation(s)
- Peter Tzu-Yu Chien
- Graduate Institute of Biomedical Sciences, Health Ageing Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan; Department of Physiology and Pharmacology, Health Ageing Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Chih-Chung Lin
- Department of Anesthetics, Chang Gung Memorial Hospital at Lin-Kou and College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Li-Der Hsiao
- Department of Anesthetics, Chang Gung Memorial Hospital at Lin-Kou and College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Chuen-Mao Yang
- Graduate Institute of Biomedical Sciences, Health Ageing Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan; Department of Physiology and Pharmacology, Health Ageing Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan.
| |
Collapse
|
13
|
Magi S, Nasti AA, Gratteri S, Castaldo P, Bompadre S, Amoroso S, Lariccia V. Gram-negative endotoxin lipopolysaccharide induces cardiac hypertrophy: Detrimental role of Na+–Ca2+ exchanger. Eur J Pharmacol 2015; 746:31-40. [PMID: 25445045 DOI: 10.1016/j.ejphar.2014.10.054] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 10/22/2014] [Accepted: 10/25/2014] [Indexed: 01/18/2023]
|
14
|
Burch ML, Getachew R, Osman N, Febbraio MA, Little PJ. Thrombin-mediated proteoglycan synthesis utilizes both protein-tyrosine kinase and serine/threonine kinase receptor transactivation in vascular smooth muscle cells. J Biol Chem 2013; 288:7410-9. [PMID: 23335513 DOI: 10.1074/jbc.m112.400259] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
G protein-coupled receptor signaling is mediated by three main mechanisms of action; these are the classical pathway, β-arrestin scaffold signaling, and the transactivation of protein-tyrosine kinase receptors such as those for EGF and PDGF. Recently, it has been demonstrated that G protein-coupled receptors can also mediate signals via transactivation of serine/threonine kinase receptors, most notably the transforming growth factor-β receptor family. Atherosclerosis is characterized by the development of lipid-laden plaques in blood vessel walls. Initiation of plaque development occurs via low density lipoprotein retention in the neointima of vessels due to binding with modified proteoglycans secreted by vascular smooth muscle cells. Here we show that transactivation of protein-tyrosine kinase receptors is mediated by matrix metalloproteinase triple membrane bypass signaling. In contrast, serine/threonine kinase receptor transactivation is mediated by a cytoskeletal rearrangement-Rho kinase-integrin system, and both protein-tyrosine kinase and serine/threonine kinase receptor transactivation concomitantly account for the total proteoglycan synthesis stimulated by thrombin in vascular smooth muscle. This work provides evidence of thrombin-mediated proteoglycan synthesis and paves the way for a potential therapeutic target for plaque development and atherosclerosis.
Collapse
Affiliation(s)
- Micah L Burch
- Diabetes Complications Laboratory, Royal Melbourne Institute of Technology University, Bundoora, Victoria 3004, Australia
| | | | | | | | | |
Collapse
|
15
|
Ho H, Soto Hopkin A, Kapadia R, Vasudeva P, Schilling J, Ganesan AK. RhoJ modulates melanoma invasion by altering actin cytoskeletal dynamics. Pigment Cell Melanoma Res 2013; 26:218-25. [PMID: 23253891 DOI: 10.1111/pcmr.12058] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 12/04/2012] [Indexed: 12/25/2022]
Abstract
Rho family GTPases regulate diverse processes in human melanoma ranging from tumor formation to metastasis and chemoresistance. In this study, a combination of in vitro and in vivo approaches was utilized to determine whether RHOJ, a CDC42 homologue that regulates melanoma chemoresistance, also controls melanoma migration. Depletion or overexpression of RHOJ altered cellular morphology, implicating a role for RHOJ in modulating actin cytoskeletal dynamics. RHOJ depletion inhibited melanoma cell migration and invasion in vitro and melanoma tumor growth and lymphatic spread in mice. Molecular studies revealed that RHOJ alters actin cytoskeletal dynamics by inducing the phosphorylation of LIMK, cofilin, and p41-ARC (ARP2/3 complex subunit) in a PAK1-dependent manner in vitro and in tumor xenografts. Taken together, these observations identify RHOJ as a melanoma linchpin determinant that regulates both actin cytoskeletal dynamics and chemoresistance by activating PAK1.
Collapse
Affiliation(s)
- Hsiang Ho
- Department of Medicine, University of California, Irvine, Irvine, CA, USA
| | | | | | | | | | | |
Collapse
|
16
|
Curtis MW, Budyn E, Desai TA, Samarel AM, Russell B. Microdomain heterogeneity in 3D affects the mechanics of neonatal cardiac myocyte contraction. Biomech Model Mechanobiol 2013; 12:95-109. [PMID: 22407215 PMCID: PMC3407350 DOI: 10.1007/s10237-012-0384-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 02/23/2012] [Indexed: 01/26/2023]
Abstract
Cardiac muscle cells are known to adapt to their physical surroundings, optimizing intracellular organization and contractile function for a given culture environment. A previously developed in vitro model system has shown that the inclusion of discrete microscale domains (or microrods) in three dimensions (3D) can alter long-term growth responses of neonatal ventricular myocytes. The aim of this work was to understand how cellular contact with such a domain affects various mechanical changes involved in cardiac muscle cell remodeling. Myocytes were maintained in 3D gels over 5 days in the presence or absence of 100-μm-long microrods, and the effect of this local heterogeneity on cell behavior was analyzed via several imaging techniques. Microrod abutment resulted in approximately twofold increases in the maximum displacement of spontaneously beating myocytes, as based on confocal microscopy scans of the gel xy-plane or the myocyte long axis. In addition, microrods caused significant increases in the proportion of aligned myofibrils (≤20° deviation from long axis) in fixed myocytes. Microrod-related differences in axial contraction could be abrogated by long-term interruption of certain signals of the RhoA-/Rho-associated kinase (ROCK) or protein kinase C (PKC) pathway. Furthermore, microrod-induced increases in myocyte size and protein content were prevented by ROCK inhibition. In all, the data suggest that microdomain heterogeneity in 3D appears to promote the development of axially aligned contractile machinery in muscle cells, an observation that may have relevance to a number of cardiac tissue engineering interventions.
Collapse
Affiliation(s)
- Matthew W. Curtis
- Department of Physiology and Biophysics, University of Illinois at Chicago, 835 South Wolcott Avenue, Chicago, IL 60612, USA
| | - Elisa Budyn
- Department of Mechanical and Industrial Engineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Tejal A. Desai
- Department of Physiology and Division of Bioengineering, University of California at San Francisco, San Francisco, CA, USA
| | - Allen M. Samarel
- The Cardiovascular Institute, Loyola University Medical Center, Maywood, IL, USA
| | - Brenda Russell
- Department of Physiology and Biophysics, University of Illinois at Chicago, 835 South Wolcott Avenue, Chicago, IL 60612, USA,
| |
Collapse
|
17
|
Tudpor K, Laínez S, Kwakernaak AJ, Kovalevskaya NV, Verkaart S, van Genesen S, van der Kemp A, Navis G, Bindels RJM, Hoenderop JGJ. Urinary plasmin inhibits TRPV5 in nephrotic-range proteinuria. J Am Soc Nephrol 2012; 23:1824-34. [PMID: 23024298 DOI: 10.1681/asn.2011111126] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Urinary proteins that leak through the abnormal glomerulus in nephrotic syndrome may affect tubular transport by interacting with membrane transporters on the luminal side of tubular epithelial cells. Patients with nephrotic syndrome can develop nephrocalcinosis, which animal models suggest may develop from impaired transcellular Ca(2+) reabsorption via TRPV5 in the distal convoluted tubule (DCT). In nephrotic-range proteinuria, filtered plasminogen reaches the luminal side of DCT, where it is cleaved into active plasmin by urokinase. In this study, we found that plasmin purified from the urine of patients with nephrotic-range proteinuria inhibits Ca(2+) uptake in TRPV5-expressing human embryonic kidney 293 cells through the activation of protease-activated receptor-1 (PAR-1). Preincubation with a plasmin inhibitor, a PAR-1 antagonist, or a protein kinase C (PKC) inhibitor abolished the effect of plasmin on TRPV5. In addition, ablation of the PKC phosphorylation site S144 rendered TRPV5 resistant to the action of plasmin. Patch-clamp experiments showed that a decreased TRPV5 pore size and a reduced open probability accompany the plasmin-mediated reduction in Ca(2+) uptake. Furthermore, high-resolution nuclear magnetic resonance spectroscopy demonstrated specific interactions between calmodulin and residues 133-154 of the N-terminus of TRPV5 for both wild-type and phosphorylated (S144pS) peptides. In summary, PAR-1 activation by plasmin induces PKC-mediated phosphorylation of TRPV5, thereby altering calmodulin-TRPV5 binding, resulting in decreased channel activity. These results indicate that urinary plasmin could contribute to the downstream effects of proteinuria on the tubulointerstitium by negatively modulating TRPV5.
Collapse
Affiliation(s)
- Kukiat Tudpor
- Department of Physiology, Nijmegen Centre for Molecular Life Sciences, Radboud University, Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Gilling CE, Mittal AK, Chaturvedi NK, Iqbal J, Aoun P, Bierman PJ, Bociek RG, Weisenburger DD, Joshi SS. Lymph node-induced immune tolerance in chronic lymphocytic leukaemia: a role for caveolin-1. Br J Haematol 2012; 158:216-231. [PMID: 22571278 DOI: 10.1111/j.1365-2141.2012.09148.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 03/23/2012] [Indexed: 11/28/2022]
Abstract
Emerging evidence indicates that the tumour microenvironment (TME) regulates the behaviour of chronic lymphocytic leukaemia (CLL). However, the precise mechanism and molecules involved in this process remain unknown. Gene expression profiles of CLL cells from lymph node (LN), bone marrow (BM) and peripheral blood (PB) indicate overexpression of a tolerogenic signature in CLL cells in lymph nodes (LN-CLL). Based on their role in B cell biology, the progression of CLL, or immune regulation, a few genes of this 83-gene signature were selected for further analyses. We observed a significant correlation between the clinical outcomes and the expression of CAV1 (P = 0·041), FGFR1 isoform 8 (P = 0·032), PTPN6 (P = 0·031) and ZWINT (P < 0·001). CAV1, a molecule involved in the regulation of tumour progression in other cancers, was seven-fold higher in LN-CLL cells compared to BM- and PB-CLL cells. Knockdown of CAV1 expression in CLL cells resulted in significantly decreased migration (P = 0·016) and proliferation (P = 0·04). When CAV1 was knocked down in B and T cell lines, we observed an inability to form immune synapses. Furthermore, CAV1 knockdown in CLL cells impaired their ability to form immune synapses with autologous T lymphocytes and allogeneic, healthy T cells. Subsequent analyses of microarray data showed differential expression of cytoskeletal genes, specifically those involved in actin polymerization. Therefore, we report a novel role for CAV1 in tumour-induced immunosuppression during the progression of CLL.
Collapse
Affiliation(s)
- Christine E Gilling
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Amit K Mittal
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Nagendra K Chaturvedi
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Javeed Iqbal
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Patricia Aoun
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Philip J Bierman
- Internal Medicine-Oncology/Hematology Section, University of Nebraska Medical Center, Omaha, NE, USA
| | - Robert G Bociek
- Internal Medicine-Oncology/Hematology Section, University of Nebraska Medical Center, Omaha, NE, USA
| | - Dennis D Weisenburger
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Shantaram S Joshi
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
19
|
Ruiz-Loredo AY, López E, López-Colomé AM. Thrombin stimulates stress fiber assembly in RPE cells by PKC/CPI-17-mediated MLCP inactivation. Exp Eye Res 2012; 96:13-23. [DOI: 10.1016/j.exer.2012.01.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Revised: 01/11/2012] [Accepted: 01/17/2012] [Indexed: 12/29/2022]
|
20
|
PPARα activation inhibits endothelin-1-induced cardiomyocyte hypertrophy by prevention of NFATc4 binding to GATA-4. Arch Biochem Biophys 2011; 518:71-8. [PMID: 22198280 DOI: 10.1016/j.abb.2011.11.024] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2011] [Revised: 11/13/2011] [Accepted: 11/14/2011] [Indexed: 11/21/2022]
Abstract
Peroxisome proliferator-activated receptor alpha (PPARα) has been implicated in the pathogenesis of cardiac hypertrophy, although its mechanism of action remains largely unknown. To determine the effect of PPARα activation on endothelin-1 (ET-1)-induced cardiomyocyte hypertrophy and explore its molecular mechanisms, we evaluated the interaction of PPARα with nuclear factor of activated T-cells c4 (NFATc4) in nuclei of cardiomyocytes from neonatal rats in primary culture. In ET-1-stimulated cardiomyocytes, data from electrophoretic mobility-shift assays (EMSA) and co-immunoprecipitation (co-IP) revealed that fenofibrate (Fen), a PPARα activator, in a concentration-dependent manner, enhanced the association of NFATc4 with PPARα and decreased its interaction with GATA-4, in promoter complexes involved in activation of the rat brain natriuretic peptide (rBNP) gene. Effects of PPARα overexpression were similar to those of its activation by Fen. PPARα depletion by small interfering RNA abolished inhibitory effects of Fen on NFATc4 binding to GATA-4 and the rBNP DNA. Quantitative RT-PCR and confocal microscopy confirmed inhibitory effects of PPARα activation on elevation of rBNP mRNA levels and ET-1-induced cardiomyocyte hypertrophy. Our results suggest that activated PPARα can compete with GATA-4 binding to NFATc4, thereby decreasing transactivation of NFATc4, and interfering with ET-1 induced cardiomyocyte hypertrophy.
Collapse
|
21
|
Cornely R, Rentero C, Enrich C, Grewal T, Gaus K. Annexin A6 is an organizer of membrane microdomains to regulate receptor localization and signalling. IUBMB Life 2011; 63:1009-17. [PMID: 21990038 DOI: 10.1002/iub.540] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2011] [Accepted: 06/16/2011] [Indexed: 12/13/2022]
Abstract
Annexin A6 (AnxA6) belongs to the conserved annexin protein family--a group of Ca(2+) -dependent membrane binding proteins. It is the largest of all annexin proteins and upon activation, binds to negatively charged phospholipids in the plasma membrane and endosomes. In addition, AnxA6 associates with cholesterol-rich membrane microdomains termed lipid rafts. Membrane cholesterol triggers Ca(2+) -independent translocation of AnxA6 to membranes and AnxA6 levels determine the number of caveolae, a form of specialized rafts at the cell surface. AnxA6 also has an F-actin binding domain and interacts with cytoskeleton components. Taken together, this suggests that AnxA6 has a scaffold function to link membrane microdomains with the organization of the cytoskeleton. Such a link facilitates AnxA6 to participate in plasma membrane repair and it would also impact on receptor signalling at the cell surface, growth factor, and lipoprotein receptor trafficking, Ca(2+) -channel activity and T cell activation. Hence, the regulation of cell surface receptors by AnxA6 may be facilitated by its unique structure that allows recruitment of interaction partners and simultaneously bridging specialized membrane domains with cortical actin surrounding activated receptors.
Collapse
Affiliation(s)
- Rhea Cornely
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
| | | | | | | | | |
Collapse
|