1
|
Campos I, Richter B, Thomas SM, Czaya B, Yanucil C, Kentrup D, Fajol A, Li Q, Secor SM, Faul C. FGFR4 Is Required for Concentric Growth of Cardiac Myocytes during Physiologic Cardiac Hypertrophy. J Cardiovasc Dev Dis 2024; 11:320. [PMID: 39452290 PMCID: PMC11508992 DOI: 10.3390/jcdd11100320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/04/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024] Open
Abstract
Fibroblast growth factor (FGF) 23 is a bone-derived hormone that promotes renal phosphate excretion. Serum FGF23 is increased in chronic kidney disease (CKD) and contributes to pathologic cardiac hypertrophy by activating FGF receptor (FGFR) 4 on cardiac myocytes, which might lead to the high cardiovascular mortality in CKD patients. Increases in serum FGF23 levels have also been observed following endurance exercise and in pregnancy, which are scenarios of physiologic cardiac hypertrophy as an adaptive response of the heart to increased demand. To determine whether FGF23/FGFR4 contributes to physiologic cardiac hypertrophy, we studied FGFR4 knockout mice (FGFR4-/-) during late pregnancy. In comparison to virgin littermates, pregnant wild-type and FGFR4-/- mice showed increases in serum FGF23 levels and heart weight; however, the elevation in myocyte area observed in pregnant wild-type mice was abrogated in pregnant FGFR4-/- mice. This outcome was supported by treatments of cultured cardiac myocytes with serum from fed Burmese pythons, another model of physiologic hypertrophy, where the co-treatment with an FGFR4-specific inhibitor abrogated the serum-induced increase in cell area. Interestingly, we found that in pregnant mice, the heart, and not the bone, shows elevated FGF23 expression, and that increases in serum FGF23 are not accompanied by changes in phosphate metabolism. Our study suggests that in physiologic cardiac hypertrophy, the heart produces FGF23 that contributes to hypertrophic growth of cardiac myocytes in a paracrine and FGFR4-dependent manner, and that the kidney does not respond to heart-derived FGF23.
Collapse
Affiliation(s)
- Isaac Campos
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Beatrice Richter
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Sarah Madison Thomas
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Brian Czaya
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Christopher Yanucil
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Dominik Kentrup
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Abul Fajol
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Qing Li
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Stephen M. Secor
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35487, USA;
| | - Christian Faul
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| |
Collapse
|
2
|
Støle TP, Lunde M, Gehmlich K, Christensen G, Louch WE, Carlson CR. Exploring Syndecan-4 and MLP and Their Interaction in Primary Cardiomyocytes and H9c2 Cells. Cells 2024; 13:947. [PMID: 38891079 PMCID: PMC11172336 DOI: 10.3390/cells13110947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
The transmembrane proteoglycan syndecan-4 is known to be involved in the hypertrophic response to pressure overload. Although multiple downstream signaling pathways have been found to be involved in this response in a syndecan-4-dependent manner, there are likely more signaling components involved. As part of a larger syndecan-4 interactome screening, we have previously identified MLP as a binding partner to the cytoplasmic tail of syndecan-4. Interestingly, many human MLP mutations have been found in patients with hypertrophic (HCM) and dilated cardiomyopathy (DCM). To gain deeper insight into the role of the syndecan-4-MLP interaction and its potential involvement in MLP-associated cardiomyopathy, we have here investigated the syndecan-4-MLP interaction in primary adult rat cardiomyocytes and the H9c2 cell line. The binding of syndecan-4 and MLP was analyzed in total lysates and subcellular fractions of primary adult rat cardiomyocytes, and baseline and differentiated H9c2 cells by immunoprecipitation. MLP and syndecan-4 localization were determined by confocal microscopy, and MLP oligomerization was determined by immunoblotting under native conditions. Syndecan-4-MLP binding, as well as MLP self-association, were also analyzed by ELISA and peptide arrays. Our results showed that MLP-WT and syndecan-4 co-localized in many subcellular compartments; however, their binding was only detected in nuclear-enriched fractions of isolated adult cardiomyocytes. In vitro, syndecan-4 bound to MLP at three sites, and this binding was reduced in some HCM-associated MLP mutations. While MLP and syndecan-4 also co-localized in many subcellular fractions of H9c2 cells, these proteins did not bind at baseline or after differentiation into cardiomyocyte-resembling cells. Independently of syndecan-4, mutated MLP proteins had an altered subcellular localization in H9c2 cells, compared to MLP-WT. The DCM- and HCM-associated MLP mutations, W4R, L44P, C58G, R64C, Y66C, K69R, G72R, and Q91L, affected the oligomerization of MLP with an increase in monomeric at the expense of trimeric and tetrameric recombinant MLP protein. Lastly, two crucial sites for MLP self-association were identified, which were reduced in most MLP mutations. Our data indicate that the syndecan-4-MLP interaction was present in nuclear-enriched fractions of isolated adult cardiomyocytes and that this interaction was disrupted by some HCM-associated MLP mutations. MLP mutations were also linked to changes in MLP oligomerization and self-association, which may be essential for its interaction with syndecan-4 and a critical molecular mechanism of MLP-associated cardiomyopathy.
Collapse
Affiliation(s)
- Thea Parsberg Støle
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway; (M.L.); (G.C.); (W.E.L.); (C.R.C.)
| | - Marianne Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway; (M.L.); (G.C.); (W.E.L.); (C.R.C.)
- K.G. Jebsen Center for Cardiac Research, University of Oslo, 0313 Oslo, Norway
| | - Katja Gehmlich
- Institute for Cardiovascular Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK;
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford OX3 9DU, UK
| | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway; (M.L.); (G.C.); (W.E.L.); (C.R.C.)
- K.G. Jebsen Center for Cardiac Research, University of Oslo, 0313 Oslo, Norway
| | - William E. Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway; (M.L.); (G.C.); (W.E.L.); (C.R.C.)
- K.G. Jebsen Center for Cardiac Research, University of Oslo, 0313 Oslo, Norway
| | - Cathrine Rein Carlson
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway; (M.L.); (G.C.); (W.E.L.); (C.R.C.)
| |
Collapse
|
3
|
Jiang J, Ni L, Zhang X, Chatterjee E, Lehmann HI, Li G, Xiao J. Keeping the Heart Healthy: The Role of Exercise in Cardiac Repair and Regeneration. Antioxid Redox Signal 2023; 39:1088-1107. [PMID: 37132606 DOI: 10.1089/ars.2023.0301] [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] [Indexed: 05/04/2023]
Abstract
Significance: Heart failure is often accompanied by a decrease in the number of cardiomyocytes. Although the adult mammalian hearts have limited regenerative capacity, the rate of regeneration is extremely low and decreases with age. Exercise is an effective means to improve cardiovascular function and prevent cardiovascular diseases. However, the molecular mechanisms of how exercise acts on cardiomyocytes are still not fully elucidated. Therefore, it is important to explore the role of exercise in cardiomyocytes and cardiac regeneration. Recent Advances: Recent advances have shown that the effects of exercise on cardiomyocytes are critical for cardiac repair and regeneration. Exercise can induce cardiomyocyte growth by increasing the size and number. It can induce physiological cardiomyocyte hypertrophy, inhibit cardiomyocyte apoptosis, and promote cardiomyocyte proliferation. In this review, we have discussed the molecular mechanisms and recent studies of exercise-induced cardiac regeneration, with a focus on its effects on cardiomyocytes. Critical Issues: There is no effective way to promote cardiac regeneration. Moderate exercise can keep the heart healthy by encouraging adult cardiomyocytes to survive and regenerate. Therefore, exercise could be a promising tool for stimulating the regenerative capability of the heart and keeping the heart healthy. Future Directions: Although exercise is an important measure to promote cardiomyocyte growth and subsequent cardiac regeneration, more studies are needed on how to do beneficial exercise and what factors are involved in cardiac repair and regeneration. Thus, it is important to clarify the mechanisms, pathways, and other critical factors involved in the exercise-mediated cardiac repair and regeneration. Antioxid. Redox Signal. 39, 1088-1107.
Collapse
Affiliation(s)
- Jizong Jiang
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| | - Lingyan Ni
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| | - Xinxin Zhang
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| | - Emeli Chatterjee
- Cardiovascular Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - H Immo Lehmann
- Cardiovascular Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Guoping Li
- Cardiovascular Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| |
Collapse
|
4
|
Akbari A, McIntyre CW. Recent Advances in Sodium Magnetic Resonance Imaging and Its Future Role in Kidney Disease. J Clin Med 2023; 12:4381. [PMID: 37445416 PMCID: PMC10342976 DOI: 10.3390/jcm12134381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Sodium imbalance is a hallmark of chronic kidney disease (CKD). Excess tissue sodium in CKD is associated with hypertension, inflammation, and cardiorenal disease. Sodium magnetic resonance imaging (23Na MRI) has been increasingly utilized in CKD clinical trials especially in the past few years. These studies have demonstrated the association of excess sodium tissue accumulation with declining renal function across whole CKD spectrum (early- to end-stage), biomarkers of systemic inflammation, and cardiovascular dysfunction. In this article, we review recent advances of 23Na MRI in CKD and discuss its future role with a focus on the skin, the heart, and the kidney itself.
Collapse
Affiliation(s)
- Alireza Akbari
- Robarts Research Institute, Western University, London, ON N6A 3K7, Canada;
- Lilibeth Caberto Kidney Clinic Research Unit, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - Christopher W. McIntyre
- Robarts Research Institute, Western University, London, ON N6A 3K7, Canada;
- Lilibeth Caberto Kidney Clinic Research Unit, London Health Sciences Centre, London, ON N6A 5W9, Canada
- Departments of Medicine, Pediatrics and Medical Biophysics, Western University, London, ON N6A 3K7, Canada
| |
Collapse
|
5
|
Strand ME, Vanhaverbeke M, Henkens MTHM, Sikking MA, Rypdal KB, Braathen B, Almaas VM, Tønnessen T, Christensen G, Heymans S, Lunde IG. Inflammation and Syndecan-4 Shedding from Cardiac Cells in Ischemic and Non-Ischemic Heart Disease. Biomedicines 2023; 11:biomedicines11041066. [PMID: 37189684 DOI: 10.3390/biomedicines11041066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 04/05/2023] Open
Abstract
Circulating biomarkers reflecting cardiac inflammation are needed to improve the diagnostics and guide the treatment of heart failure patients. The cardiac production and shedding of the transmembrane proteoglycan syndecan-4 is upregulated by innate immunity signaling pathways. Here, we investigated the potential of syndecan-4 as a blood biomarker of cardiac inflammation. Serum syndecan-4 was measured in patients with (i) non-ischemic, non-valvular dilated cardiomyopathy (DCM), with (n = 71) or without (n = 318) chronic inflammation; (ii) acute myocarditis (n = 15), acute pericarditis (n = 3) or acute perimyocarditis (23) and (iii) acute myocardial infarction (MI) at day 0, 3 and 30 (n = 119). Syndecan-4 was investigated in cultured cardiac myocytes and fibroblasts (n = 6–12) treated with the pro-inflammatory cytokines interleukin (IL)-1β and its inhibitor IL-1 receptor antagonist (IL-1Ra), or tumor necrosis factor (TNF)α and its specific inhibitor infliximab, an antibody used in treatment of autoimmune diseases. The levels of serum syndecan-4 were comparable in all subgroups of patients with chronic or acute cardiomyopathy, independent of inflammation. Post-MI, syndecan-4 levels were increased at day 3 and 30 vs. day 0. IL-1Ra attenuated IL-1β-induced syndecan-4 production and shedding in vitro, while infliximab had no effect. In conclusion, syndecan-4 shedding from cardiac myocytes and fibroblasts was attenuated by immunomodulatory therapy. Although its circulating levels were increased post-MI, syndecan-4 did not reflect cardiac inflammatory status in patients with heart disease.
Collapse
Affiliation(s)
- Mari E. Strand
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway
| | | | - Michiel T. H. M. Henkens
- Netherlands Heart Institute (NLHI), 3511 EP Utrecht, The Netherlands
- Department of Pathology, CARIM, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands
- Department of Cardiology, CARIM, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands
| | - Maurits A. Sikking
- Department of Cardiology, CARIM, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands
| | - Karoline B. Rypdal
- Institute of Clinical Medicine, University of Oslo, 0315 Oslo, Norway
- K.G. Jebsen Center for Cardiac Biomarkers, University of Oslo, 0315 Oslo, Norway
- Division of Diagnostics and Technology, Akershus University Hospital, 1478 Lørenskog, Norway
| | - Bjørn Braathen
- Department of Cardiothoracic Surgery, Oslo University Hospital Ullevål, 0450 Oslo, Norway
| | - Vibeke M. Almaas
- Department of Cardiology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
| | - Theis Tønnessen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway
- Department of Cardiothoracic Surgery, Oslo University Hospital Ullevål, 0450 Oslo, Norway
| | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway
| | - Stephane Heymans
- Department of Cardiovascular Science, University of Leuven, 3000 Leuven, Belgium
| | - Ida G. Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway
- K.G. Jebsen Center for Cardiac Biomarkers, University of Oslo, 0315 Oslo, Norway
- Division of Diagnostics and Technology, Akershus University Hospital, 1478 Lørenskog, Norway
| |
Collapse
|
6
|
Lunde IG, Aronsen JM, Melleby AO, Strand ME, Skogestad J, Bendiksen BA, Ahmed MS, Sjaastad I, Attramadal H, Carlson CR, Christensen G. Cardiomyocyte-specific overexpression of syndecan-4 in mice results in activation of calcineurin-NFAT signalling and exacerbated cardiac hypertrophy. Mol Biol Rep 2022; 49:11795-11809. [PMID: 36205855 PMCID: PMC9712407 DOI: 10.1007/s11033-022-07985-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 09/24/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND Cardiomyocyte hypertrophy is a hallmark of cardiac dysfunction in patients with aortic stenosis (AS), and can be triggered by left ventricular (LV) pressure overload in mice by aortic banding (AB). Syndecan-4 is a transmembrane heparan sulphate proteoglycan which is found increased in the myocardium of AS patients and AB mice. The role of syndecan-4 in cardiomyocyte hypertrophy is not well understood. PURPOSE OF THE STUDY We developed mice with cardiomyocyte-specific overexpression of syndecan-4 (Sdc4-Tg) and subjected these to AB to examine the role of syndecan-4 in hypertrophy and activation of the pro-hypertrophic calcineurin-NFAT signalling pathway. METHODS AND RESULTS Sdc4-Tg mice showed exacerbated cardiac remodelling upon AB compared to wild type (WT). At 2-6 weeks post-AB, Sdc4-Tg and WT mice showed similar hypertrophic growth, while at 20 weeks post-AB, exacerbated hypertrophy and dysfunction were evident in Sdc4-Tg mice. After cross-breeding of Sdc4-Tg mice with NFAT-luciferase reporter mice, we found increased NFAT activation in Sdc4-Tg hearts after AB. Immunoprecipitation showed that calcineurin bound to syndecan-4 in Sdc4-Tg hearts. Isolated cardiomyocytes from Sdc4-Tg mice showed alterations in Ca2+ fluxes, suggesting that syndecan-4 regulated Ca2+ levels, and thereby, activating the syndecan-4-calcineurin complex resulting in NFAT activation and hypertrophic growth. Similarly, primary cardiomyocyte cultures from neonatal rats showed increased calcineurin-NFAT-dependent hypertrophic growth upon viral Sdc4 overexpression. CONCLUSION Our study of mice with cardiomyocyte-specific overexpression of Sdc4 have revealed that syndecan-4 is important for activation of the Ca2+-dependent calcineurin-NFAT signalling pathway, hypertrophic remodelling and dysfunction in cardiomyocytes in response to pressure overload.
Collapse
Affiliation(s)
- Ida G Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.
- KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway.
- Division of Diagnostics and Technology, Akershus University Hospital, Lørenskog, Norway.
- Institute for Experimental Medical Research (IEMR), Oslo University Hospital Ullevaal, Building 7, 4th floor, Kirkeveien 166, 0407, Oslo, Norway.
| | - J Magnus Aronsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Institute for Medical Biosciences, University of Oslo, Oslo, Norway
| | - A Olav Melleby
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Institute for Medical Biosciences, University of Oslo, Oslo, Norway
| | - Mari E Strand
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Jonas Skogestad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Institute for Medical Biosciences, University of Oslo, Oslo, Norway
| | - Bård A Bendiksen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - M Shakil Ahmed
- Institute for Surgical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Håvard Attramadal
- Institute for Surgical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Cathrine R Carlson
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| |
Collapse
|
7
|
Thota LNR, Chignalia AZ. The role of the glypican and syndecan families of heparan sulfate proteoglycans in cardiovascular function and disease. Am J Physiol Cell Physiol 2022; 323:C1052-C1060. [PMID: 35993518 DOI: 10.1152/ajpcell.00018.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Heparan sulfate proteoglycans (HSPGs) are proteoglycans formed by a core protein to which one or multiple heparan sulfate chains are covalently bound. They are ubiquitously expressed in cellular surfaces and can be found in the extracellular matrix and secretory vesicles. The cellular effects of HSPGs comprehend multiple functionalities that include 1) the interaction with other membrane surface proteins to act as a substrate for cellular migration, 2) acting as a binding site for circulating molecules, 3) to have a receptor role for proteases, 4) to act as a coreceptor that can provide finetuning of growth factor receptor activity threshold, and 5) to activate intracellular signaling pathways (Sarrazin S, Lamanna WC, Esko JD. Cold Spring Harb Perspect Biol 3: a004952, 2011). Among the different families of HSPGs, the syndecan and glypican families of HSPGs have gained increased attention in relation to their effects on cardiovascular cells and potential role in disease progression. In this review, we will summarize the effects of syndecan and glypican homologs on the different cardiovascular cell types and discuss their contribution to common processes found in cardiovascular diseases (inflammation, hypertrophy, and vascular remodeling) as well as their potential role in the development and progression of specific diseases including hypertension, heart failure, and atherosclerosis.
Collapse
Affiliation(s)
| | - Andreia Zago Chignalia
- Department of Anesthesiology, College of Medicine-Tucson, University of Arizona, Tucson, Arizona.,Department of Physiology, College of Medicine-Tucson, University of Arizona, Tucson, Arizona.,Department of Pharmacology and Toxicology, College of Pharmacy-Tucson, University of Arizona, Tucson, Arizona.,Sarver Heart Center, College of Medicine-Tucson, University of Arizona, Tucson, Arizona
| |
Collapse
|
8
|
Dai W, Liu Y, Yao F, Li W, Liu J, Li C, Liu D. Syndecan-4 is More Sensitive in Detecting Hypertensive Left Ventricular Diastolic Dysfunction in 2K2C Rats. Int J Hypertens 2022; 2022:1447425. [PMID: 36248198 PMCID: PMC9553681 DOI: 10.1155/2022/1447425] [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: 02/11/2022] [Revised: 08/24/2022] [Accepted: 09/01/2022] [Indexed: 11/23/2022] Open
Abstract
Objective The aim of this study was to investigate the changes of syndecan-4 (SDC-4) during the hypertensive period in two kidney-two clip (2K2C) hypertension rats and compare them to brain natriuretic peptide (BNP) and the echocardiographic parameters for diastolic function evaluation in the rat model of 2K2C hypertension. Methods A total of 36 Sprague-Dawley (SD) rats were used in this study. Hypertension was induced in 21 by 2K2C surgery, and 15 were sham-operated. Both the 2K2C hypertension group (n = 21) and the sham-operated group (n = 15) were equally divided into 3 subgroups according to the schedules (week 4, week 8, and week 12). Serum SDC-4 and BNP were detected by ELISA, and echocardiography indexes were acquired. Results The level of SDC-4 and cardiac fibrosis increased gradually as the experiment was processed, and BNP, Tei index, and E/E' followed to be raised as high blood pressure was maintained after four weeks in the 2K2C hypertension rats. In the earlier 4 weeks, only SDC-4 and cardiac fibrosis were significantly increased in 2K2C hypertensive rats in comparison with normotensive rats. And it was shown that SDC-4 was positively correlated with BNP level during the entire study (r = 0.762, p < 0.01). Conclusion SDC-4 increases gradually during the process of diastolic dysfunction in 2K2C hypertensive rats. SDC-4 is the earliest biomarker reflecting diastolic dysfunction in this model, superior to E/E' and the Tei index. Our results indicate that serum SDC-4 could act as an early biomarker to show diastolic dysfunction.
Collapse
Affiliation(s)
- Wenyue Dai
- Department of Medical Ultrasonics, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
- Department of Medical Ultrasonics, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518033, China
| | - Yanqiu Liu
- Department of Medical Ultrasonics, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Fengjuan Yao
- Department of Medical Ultrasonics, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Wei Li
- Department of Medical Ultrasonics, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Jia Liu
- Department of Medical Ultrasonics, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Cuiling Li
- Department of Medical Ultrasonics, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Donghong Liu
- Department of Medical Ultrasonics, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| |
Collapse
|
9
|
Støle TP, Lunde M, Shen X, Martinsen M, Lunde PK, Li J, Lockwood F, Sjaastad I, Louch WE, Aronsen JM, Christensen G, Carlson CR. The female syndecan-4−/− heart has smaller cardiomyocytes, augmented insulin/pSer473-Akt/pSer9-GSK-3β signaling, and lowered SCOP, pThr308-Akt/Akt and GLUT4 levels. Front Cell Dev Biol 2022; 10:908126. [PMID: 36092718 PMCID: PMC9452846 DOI: 10.3389/fcell.2022.908126] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/06/2022] [Indexed: 11/13/2022] Open
Abstract
Background: In cardiac muscle, the ubiquitously expressed proteoglycan syndecan-4 is involved in the hypertrophic response to pressure overload. Protein kinase Akt signaling, which is known to regulate hypertrophy, has been found to be reduced in the cardiac muscle of exercised male syndecan-4−/− mice. In contrast, we have recently found that pSer473-Akt signaling is elevated in the skeletal muscle (tibialis anterior, TA) of female syndecan-4−/− mice. To determine if the differences seen in Akt signaling are sex specific, we have presently investigated Akt signaling in the cardiac muscle of sedentary and exercised female syndecan-4−/− mice. To get deeper insight into the female syndecan-4−/− heart, alterations in cardiomyocyte size, a wide variety of different extracellular matrix components, well-known syndecan-4 binding partners and associated signaling pathways have also been investigated.Methods: Left ventricles (LVs) from sedentary and exercise trained female syndecan-4−/− and WT mice were analyzed by immunoblotting and real-time PCR. Cardiomyocyte size and phosphorylated Ser473-Akt were analyzed in isolated adult cardiomyocytes from female syndecan-4−/− and WT mice by confocal imaging. LV and skeletal muscle (TA) from sedentary male syndecan-4−/− and WT mice were immunoblotted with Akt antibodies for comparison. Glucose levels were measured by a glucometer, and fasting blood serum insulin and C-peptide levels were measured by ELISA.Results: Compared to female WT hearts, sedentary female syndecan-4−/− LV cardiomyocytes were smaller and hearts had higher levels of pSer473-Akt and its downstream target pSer9-GSK-3β. The pSer473-Akt inhibitory phosphatase PHLPP1/SCOP was lowered, which may be in response to the elevated serum insulin levels found in the female syndecan-4−/− mice. We also observed lowered levels of pThr308-Akt/Akt and GLUT4 in the female syndecan-4−/− heart and an increased LRP6 level after exercise. Otherwise, few alterations were found. The pThr308-Akt and pSer473-Akt levels were unaltered in the cardiac and skeletal muscles of sedentary male syndecan-4−/− mice.Conclusion: Our data indicate smaller cardiomyocytes, an elevated insulin/pSer473-Akt/pSer9-GSK-3β signaling pathway, and lowered SCOP, pThr308-Akt/Akt and GLUT4 levels in the female syndecan-4−/− heart. In contrast, cardiomyocyte size, and Akt signaling were unaltered in both cardiac and skeletal muscles from male syndecan-4−/− mice, suggesting important sex differences.
Collapse
Affiliation(s)
- Thea Parsberg Støle
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- *Correspondence: Thea Parsberg Støle,
| | - Marianne Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Xin Shen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Marita Martinsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Per Kristian Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Jia Li
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Francesca Lockwood
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - William Edward Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Jan Magnus Aronsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Cathrine Rein Carlson
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| |
Collapse
|
10
|
Romaine A, Melleby AO, Alam J, Lobert VH, Lu N, Lockwood FE, Hasic A, Lunde IG, Sjaastad I, Stenmark H, Herum KM, Gullberg D, Christensen G. Integrin α11β1 and syndecan-4 dual receptor ablation attenuates cardiac hypertrophy in the pressure overloaded heart. Am J Physiol Heart Circ Physiol 2022; 322:H1057-H1071. [PMID: 35522553 DOI: 10.1152/ajpheart.00635.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Pathological myocardial hypertrophy in response to an increase in left ventricular (LV) afterload may ultimately lead to heart failure. Cell surface receptors bridge the interface between the cell and the ECM in cardiac myocytes and cardiac fibroblasts, and have been suggested to be important mediators of pathological myocardial hypertrophy. We identify for the first time that integrin α11 (α11) is preferentially upregulated amongst integrin beta 1 heterodimer-forming α subunits in response to increased afterload induced by aortic banding (AB) in wild type mice (WT). Mice were anesthetized in a chamber with 4% isoflurane and 95% oxygen before being intubated and ventilated with 2.5% isoflurane and 97% oxygen. For pre- and post-operative analgesia, animals were administered 0.02 mL buprenorphine (0.3 mg/mL) subcutaneously. Surprisingly, mice lacking α11 develop myocardial hypertrophy following AB comparable to WT. In the mice lacking α11, we further show a compensatory increase in the expression of another mechanoreceptor, syndecan-4, following AB compared to WT AB mice, indicating that syndecan-4 compensated for lack of α11. Intriguingly, mice lacking mechanoreceptors α11 and syndecan-4 show ablated myocardial hypertrophy following AB compared to WT mice. Expression of the main cardiac collagen isoforms col1a2 and col3a1 was significantly reduced in AB mice lacking mechanoreceptors α11 and syndecan-4 compared to WT AB.
Collapse
Affiliation(s)
- Andreas Romaine
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Arne Olav Melleby
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway.,Section of Physiology, Department of Molecular Medicine, Institute for Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Jahedul Alam
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - Ning Lu
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Francesca E Lockwood
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Almira Hasic
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Ida G Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Harald Stenmark
- Institute for Cancer Research, Oslo University Hospital, Norway
| | - Kate M Herum
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway.,Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Donald Gullberg
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
11
|
Haryono A, Ikeda K, Nugroho DB, Ogata T, Tsuji Y, Matoba S, Moriwaki K, Kitagawa H, Igarashi M, Hirata KI, Emoto N. ChGn-2 Plays a Cardioprotective Role in Heart Failure Caused by Acute Pressure Overload. J Am Heart Assoc 2022; 11:e023401. [PMID: 35322673 PMCID: PMC9075488 DOI: 10.1161/jaha.121.023401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Cardiac extracellular matrix is critically involved in cardiac homeostasis, and accumulation of chondroitin sulfate glycosaminoglycans (CS-GAGs) was previously shown to exacerbate heart failure by augmenting inflammation and fibrosis at the chronic phase. However, the mechanism by which CS-GAGs affect cardiac functions remains unclear, especially at the acute phase. Methods and Results We explored a role of CS-GAG in heart failure using mice with target deletion of ChGn-2 (chondroitin sulfate N-acetylgalactosaminyltransferase-2) that elongates CS chains of glycosaminoglycans. Heart failure was induced by transverse aortic constriction in mice. The role of CS-GAG derived from cardiac fibroblasts in cardiomyocyte death was analyzed. Cardiac fibroblasts were subjected to cyclic mechanical stretch that mimics increased workload in the heart. Significant CS-GAGs accumulation was detected in the heart of wild-type mice after transverse aortic constriction, which was substantially reduced in ChGn-2-/- mice. Loss of ChGn-2 deteriorated the cardiac dysfunction caused by pressure overload, accompanied by augmented cardiac hypertrophy and increased cardiomyocyte apoptosis. Cyclic mechanical stretch increased ChGn-2 expression and enhanced glycosaminoglycan production in cardiac fibroblasts. Conditioned medium derived from the stretched cardiac fibroblasts showed cardioprotective effects, which was abolished by CS-GAGs degradation. We found that CS-GAGs elicits cardioprotective effects via dual pathway; direct pathway through interaction with CD44, and indirect pathway through binding to and activating insulin-like growth factor-1. Conclusions Our data revealed the cardioprotective effects of CS-GAGs; therefore, CS-GAGs may play biphasic role in the development of heart failure; cardioprotective role at acute phase despite its possible unfavorable role in the advanced phase.
Collapse
Affiliation(s)
- Andreas Haryono
- Division of Cardiovascular Medicine Department of Internal Medicine Kobe University Graduate School of Medicine Kobe Japan.,Laboratory of Clinical Pharmaceutical Science Kobe Pharmaceutical University Kobe Japan
| | - Koji Ikeda
- Laboratory of Clinical Pharmaceutical Science Kobe Pharmaceutical University Kobe Japan.,Department of Epidemiology for Longevity and Regional Health Kyoto Prefectural University of Medicine Kyoto Japan.,Department of Cardiology Kyoto Prefectural University of Medicine Kyoto Japan
| | - Dhite Bayu Nugroho
- Department of Internal Medicine Faculty of Medicine, Public Health, and Nursing Gadjah Mada University Indonesia
| | - Takehiro Ogata
- Department of Pathology and Cell Regulation Kyoto Prefectural University of Medicine Kyoto Japan
| | - Yumika Tsuji
- Department of Cardiology Kyoto Prefectural University of Medicine Kyoto Japan
| | - Satoaki Matoba
- Department of Cardiology Kyoto Prefectural University of Medicine Kyoto Japan
| | - Kensuke Moriwaki
- Comprehensive Unit for Health Economic Evidence Review and Decision Support (CHEERS) Research Organization of Science and TechnologyRitsumeikan University Kyoto Japan
| | - Hiroshi Kitagawa
- Laboratory of Biochemistry Kobe Pharmaceutical University Kobe Japan
| | - Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology Graduate School of Medical and Dental Sciences and Trans-disciplinary Program Niigata University Niigata Japan
| | - Ken-Ichi Hirata
- Division of Cardiovascular Medicine Department of Internal Medicine Kobe University Graduate School of Medicine Kobe Japan
| | - Noriaki Emoto
- Division of Cardiovascular Medicine Department of Internal Medicine Kobe University Graduate School of Medicine Kobe Japan.,Laboratory of Clinical Pharmaceutical Science Kobe Pharmaceutical University Kobe Japan
| |
Collapse
|
12
|
Trinh K, Julovi SM, Rogers NM. The Role of Matrix Proteins in Cardiac Pathology. Int J Mol Sci 2022; 23:ijms23031338. [PMID: 35163259 PMCID: PMC8836004 DOI: 10.3390/ijms23031338] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/15/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023] Open
Abstract
The extracellular matrix (ECM) and ECM-regulatory proteins mediate structural and cell-cell interactions that are crucial for embryonic cardiac development and postnatal homeostasis, as well as organ remodeling and repair in response to injury. These proteins possess a broad functionality that is regulated by multiple structural domains and dependent on their ability to interact with extracellular substrates and/or cell surface receptors. Several different cell types (cardiomyocytes, fibroblasts, endothelial and inflammatory cells) within the myocardium elaborate ECM proteins, and their role in cardiovascular (patho)physiology has been increasingly recognized. This has stimulated robust research dissecting the ECM protein function in human health and disease and replicating the genetic proof-of-principle. This review summarizes recent developments regarding the contribution of ECM to cardiovascular disease. The clear importance of this heterogeneous group of proteins in attenuating maladaptive repair responses provides an impetus for further investigation into these proteins as potential pharmacological targets in cardiac diseases and beyond.
Collapse
Affiliation(s)
- Katie Trinh
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; (K.T.); (S.M.J.)
- Faculty of Medicine and Health Sydney, School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Sohel M. Julovi
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; (K.T.); (S.M.J.)
- Faculty of Medicine and Health Sydney, School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Natasha M. Rogers
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; (K.T.); (S.M.J.)
- Faculty of Medicine and Health Sydney, School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia
- Renal and Transplantation Medicine, Westmead Hospital, Westmead, NSW 2145, Australia
- Correspondence:
| |
Collapse
|
13
|
Ginsenoside Rg1 attenuates mechanical stress-induced cardiac injury via calcium sensing receptor-related pathway. J Ginseng Res 2021; 45:683-694. [PMID: 34764723 PMCID: PMC8569322 DOI: 10.1016/j.jgr.2021.03.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 03/02/2021] [Accepted: 03/21/2021] [Indexed: 11/25/2022] Open
Abstract
Background Ginsenoside Rg1 (Rg1) has been well documented to be effective against various cardiovascular disease. The aim of this study is to evaluate the effect of Rg1 on mechanical stress-induced cardiac injury and its possible mechanism with a focus on the calcium sensing receptor (CaSR) signaling pathway. Methods Mechanical stress was implemented on rats through abdominal aortic constriction (AAC) procedure and on cardiomyocytes and cardiac fibroblasts by mechanical stretching with Bioflex Collagen I plates. The effects of Rg1 on cell hypertrophy, fibrosis, cardiac function, [Ca2+]i, and the expression of CaSR and calcineurin (CaN) were assayed both on rat and cellular level. Results Rg1 alleviated cardiac hypertrophy and fibrosis, and improved cardiac decompensation induced by AAC in rat myocardial tissue and cultured cardiomyocytes and cardiac fibroblasts. Importantly, Rg1 treatment inhibited CaSR expression and increase of [Ca2+]i, which similar to the CaSR inhibitor NPS2143. In addition, Rg1 treatment inhibited CaN and TGF-β1 pathways activation. Mechanistic analysis showed that the CaSR agonist GdCl3 could not further increase the [Ca2+]i and CaN pathway related protein expression induced by mechanical stretching in cultured cardiomyocytes. CsA, an inhibitor of CaN, inhibited cardiac hypertrophy, cardiac fibrosis, [Ca2+]i and CaN signaling but had no effect on CaSR expression. Conclusion The activation of CaN pathway and the increase of [Ca2+]i mediated by CaSR are involved in cardiac hypertrophy and fibrosis, that may be the target of cardioprotection of Rg1 against myocardial injury.
Collapse
|
14
|
Sadoughi F, Hallajzadeh J, Mirsafaei L, Asemi Z, Zahedi M, Mansournia MA, Yousefi B. Cardiac fibrosis and curcumin: a novel perspective on this natural medicine. Mol Biol Rep 2021; 48:7597-7608. [PMID: 34648140 DOI: 10.1007/s11033-021-06768-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 09/10/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND According to WHO statistics, cardiovascular disease are the leading causes of death in the world. One of the main factors which is causing heart failure, systolic and diastolic dysfunction, and arrythmias is a condition named cardiac fibrosis. This condition is defined by the accumulation of fibroblast-produced ECM in myocardium layer of the heart. OBJECTIVE Accordingly, the current review aims to depict the role of curcumin in the regulation of different signaling pathways that are involved in cardiac fibrosis. RESULTS A great number of cellular and molecular mechanisms such as oxidative stress, inflammation, and mechanical stress are acknowledged to be involved in cardiac fibrosis. Despite the available therapeutic procedures which are designed to target these mechanisms in order to prevent cardiac fibrosis, still, effective therapeutic methods are needed. Curcumin is a natural Chinese medicine which currently has been declared to have therapeutic properties such as anti-oxidant and immunomodulatory activities. In this review, we have gathered several experimental studies in order to represent diverse impacts of this turmeric derivative on pathogenic factors of cardiac fibrosis. CONCLUSION Curcumin might open new avenues in the field of cardiovascular treatment.
Collapse
Affiliation(s)
- Fatemeh Sadoughi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Jamal Hallajzadeh
- Department of Biochemistry and Nutrition, Medicinal Plants Research Center, Maragheh University of Medical Sciences, Maragheh, Iran.
| | - Liaosadat Mirsafaei
- Department of Cardiology, Ramsar Campus, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Mahdi Zahedi
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgān, Iran.
| | - Mohammad Ali Mansournia
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
15
|
Yu ZY, Gong H, Wu J, Dai Y, Kesteven SH, Fatkin D, Martinac B, Graham RM, Feneley MP. Cardiac Gq Receptors and Calcineurin Activation Are Not Required for the Hypertrophic Response to Mechanical Left Ventricular Pressure Overload. Front Cell Dev Biol 2021; 9:639509. [PMID: 33659256 PMCID: PMC7917224 DOI: 10.3389/fcell.2021.639509] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/26/2021] [Indexed: 01/19/2023] Open
Abstract
Rationale Gq-coupled receptors are thought to play a critical role in the induction of left ventricular hypertrophy (LVH) secondary to pressure overload, although mechano-sensitive channel activation by a variety of mechanisms has also been proposed, and the relative importance of calcineurin- and calmodulin kinase II (CaMKII)-dependent hypertrophic pathways remains controversial. Objective To determine the mechanisms regulating the induction of LVH in response to mechanical pressure overload. Methods and Results Transgenic mice with cardiac-targeted inhibition of Gq-coupled receptors (GqI mice) and their non-transgenic littermates (NTL) were subjected to neurohumoral stimulation (continuous, subcutaneous angiotensin II (AngII) infusion for 14 days) or mechanical pressure overload (transverse aortic arch constriction (TAC) for 21 days) to induce LVH. Candidate signaling pathway activation was examined. As expected, LVH observed in NTL mice with AngII infusion was attenuated in heterozygous (GqI+/-) mice and absent in homozygous (GqI-/-) mice. In contrast, LVH due to TAC was unaltered by either heterozygous or homozygous Gq inhibition. Gene expression of atrial natriuretic peptide (ANP), B-type natriuretic peptide (BNP) and α-skeletal actin (α-SA) was increased 48 h after AngII infusion or TAC in NTL mice; in GqI mice, the increases in ANP, BNP and α-SA in response to AngII were completely absent, as expected, but all three increased after TAC. Increased nuclear translocation of nuclear factor of activated T-cells c4 (NFATc4), indicating calcineurin pathway activation, occurred in NTL mice with AngII infusion but not TAC, and was prevented in GqI mice infused with AngII. Nuclear and cytoplasmic CaMKIIδ levels increased in both NTL and GqI mice after TAC but not AngII infusion, with increased cytoplasmic phospho- and total histone deacetylase 4 (HDAC4) and increased nuclear myocyte enhancer factor 2 (MEF2) levels. Conclusion Cardiac Gq receptors and calcineurin activation are required for neurohumorally mediated LVH but not for LVH induced by mechanical pressure overload (TAC). Rather, TAC-induced LVH is associated with activation of the CaMKII-HDAC4-MEF2 pathway.
Collapse
Affiliation(s)
- Ze-Yan Yu
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,Cardiology Department, St Vincent's Hospital, Darlinghurst, NSW, Australia.,Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Hutao Gong
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Jianxin Wu
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Yun Dai
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Scott H Kesteven
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Diane Fatkin
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,Cardiology Department, St Vincent's Hospital, Darlinghurst, NSW, Australia.,Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Boris Martinac
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Robert M Graham
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,Cardiology Department, St Vincent's Hospital, Darlinghurst, NSW, Australia.,Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Michael P Feneley
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,Cardiology Department, St Vincent's Hospital, Darlinghurst, NSW, Australia.,Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
16
|
Mathiesen SB, Lunde M, Stensland M, Martinsen M, Nyman TA, Christensen G, Carlson CR. The Cardiac Syndecan-2 Interactome. Front Cell Dev Biol 2020; 8:792. [PMID: 32984315 PMCID: PMC7483480 DOI: 10.3389/fcell.2020.00792] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/28/2020] [Indexed: 12/31/2022] Open
Abstract
The extracellular matrix (ECM) is important in cardiac remodeling and syndecans have gained increased interest in this process due to their ability to convert changes in the ECM to cell signaling. In particular, syndecan-4 has been shown to be important for cardiac remodeling, whereas the role of its close relative syndecan-2 is largely unknown in the heart. To get more insight into the role of syndecan-2, we here sought to identify interaction partners of syndecan-2 in rat left ventricle. By using three different affinity purification methods combined with mass spectrometry (MS) analysis, we identified 30 novel partners and 9 partners previously described in the literature, which together make up the first cardiac syndecan-2 interactome. Eleven of the novel partners were also verified in HEK293 cells (i.e., AP2A2, CAVIN2, DDX19A, EIF4E, JPH2, MYL12A, NSF, PFDN2, PSMC5, PSMD11, and RRAD). The cardiac syndecan-2 interactome partners formed connections to each other and grouped into clusters mainly involved in cytoskeletal remodeling and protein metabolism, but also into a cluster consisting of a family of novel syndecan-2 interaction partners, the CAVINs. MS analyses revealed that although syndecan-2 was significantly enriched in fibroblast fractions, most of its partners were present in both cardiomyocytes and fibroblasts. Finally, a comparison of the cardiac syndecan-2 and -4 interactomes revealed surprisingly few protein partners in common.
Collapse
Affiliation(s)
- Sabrina Bech Mathiesen
- Institute for Experimental Medical Research and Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Marianne Lunde
- Institute for Experimental Medical Research and Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Maria Stensland
- Department of Immunology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Marita Martinsen
- Institute for Experimental Medical Research and Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Tuula A Nyman
- Department of Immunology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Geir Christensen
- Institute for Experimental Medical Research and Oslo University Hospital, University of Oslo, Oslo, Norway.,K.G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Cathrine Rein Carlson
- Institute for Experimental Medical Research and Oslo University Hospital, University of Oslo, Oslo, Norway
| |
Collapse
|
17
|
Kazeminia M, Daneshkhah A, Jalali R, Vaisi-Raygani A, Salari N, Mohammadi M. The Effect of Exercise on the Older Adult's Blood Pressure Suffering Hypertension: Systematic Review and Meta-Analysis on Clinical Trial Studies. Int J Hypertens 2020; 2020:2786120. [PMID: 33014449 PMCID: PMC7512073 DOI: 10.1155/2020/2786120] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/28/2020] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Senescence refers to spontaneous and progressive irreversible degenerative changes in which both the physical and psychological power diminish significantly. Hypertension is the most common cardiovascular disease in the elderly. Several studies have been conducted regarding the effect of exercise on reducing the blood pressure of the elderly, which have found contradictory results. One of the uses of meta-analysis study is responding to these assumptions and resolving the discrepancies. Accordingly, the aim of the present study is to determine the impact of exercise on the blood pressure of older adults. METHOD In this research, in order to find electronic published papers from 1992 to 2019, the papers published in both domestic and foreign databases including SID, MagIran, IranMedex, IranDox, Gogole Scholar, Cohrane, Embase, Science Direct, Scopus, PubMed, and Web of Science (ISI) were used. Heterogeneity index between the studies was determined based on Cochran test Q(c) and I 2. Considering existence of heterogeneity, random effects model was employed to estimate the standardized subtraction of the mean exercise test score for reduction of blood pressure in the older adults across the intervention group before and after the test. RESULTS In this meta-analysis and systematic review, eventually 69 papers met the inclusion criteria. The total number of participants was 2272 in the pre- and postintervention groups when examining the systolic changes and 2252 subjects in the pre- and postintervention groups when inspecting the diastolic changes. The standardized mean difference in examining the systolic changes before the intervention was 137.1 ± 8.09 and 132.98 ± 0.96 after the intervention; when exploring the diastolic changes, the pre- and postintervention values were 80.3 ± 0.85 and 76.0 ± 6.56, respectively, where these differences were statistically significant (P < 0.01). CONCLUSION The results of this study indicated that exercise leads to significant reduction in both systolic and diastolic blood pressure. Accordingly, regular exercise can be part of the treatment plan for hypertensive elderly.
Collapse
Affiliation(s)
- Mohsen Kazeminia
- Department of Nursing, School of Nursing and Midwifery, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Alireza Daneshkhah
- School of Computing, Electronics and Maths, Coventry University, London, UK
| | - Rostam Jalali
- Department of Nursing, School of Nursing and Midwifery, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Aliakbar Vaisi-Raygani
- Department of Nursing, School of Nursing and Midwifery, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Nader Salari
- Department of Biostatistics, School of Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Masoud Mohammadi
- Department of Nursing, School of Nursing and Midwifery, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
18
|
Rønning SB, Carlson CR, Aronsen JM, Pisconti A, Høst V, Lunde M, Liland KH, Sjaastad I, Kolset SO, Christensen G, Pedersen ME. Syndecan-4 -/- Mice Have Smaller Muscle Fibers, Increased Akt/mTOR/S6K1 and Notch/HES-1 Pathways, and Alterations in Extracellular Matrix Components. Front Cell Dev Biol 2020; 8:730. [PMID: 32850844 PMCID: PMC7411008 DOI: 10.3389/fcell.2020.00730] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/15/2020] [Indexed: 12/11/2022] Open
Abstract
Background Extracellular matrix (ECM) remodeling is essential for skeletal muscle development and adaption in response to environmental cues such as exercise and injury. The cell surface proteoglycan syndecan-4 has been reported to be essential for muscle differentiation, but few molecular mechanisms are known. Syndecan-4–/– mice are unable to regenerate damaged muscle, and display deficient satellite cell activation, proliferation, and differentiation. A reduced myofiber basal lamina has also been reported in syndecan-4–/– muscle, indicating possible defects in ECM production. To get a better understanding of the underlying molecular mechanisms, we have here investigated the effects of syndecan-4 genetic ablation on molecules involved in ECM remodeling and muscle growth, both under steady state conditions and in response to exercise. Methods Tibialis anterior (TA) muscles from sedentary and exercised syndecan-4–/– and WT mice were analyzed by immunohistochemistry, real-time PCR and western blotting. Results Compared to WT, we found that syndecan-4–/– mice had reduced body weight, reduced muscle weight, muscle fibers with a smaller cross-sectional area, and reduced expression of myogenic regulatory transcription factors. Sedentary syndecan-4–/– had also increased mRNA levels of syndecan-2, decorin, collagens, fibromodulin, biglycan, and LOX. Some of these latter ECM components were reduced at protein level, suggesting them to be more susceptible to degradation or less efficiently translated when syndecan-4 is absent. At the protein level, TRPC7 was reduced, whereas activation of the Akt/mTOR/S6K1 and Notch/HES-1 pathways were increased. Finally, although exercise induced upregulation of several of these components in WT, a further upregulation of these molecules was not observed in exercised syndecan-4–/– mice. Conclusion Altogether our data suggest an important role of syndecan-4 in muscle development.
Collapse
Affiliation(s)
| | - Cathrine Rein Carlson
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Jan Magnus Aronsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,Bjørknes College, Oslo, Norway
| | - Addolorata Pisconti
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, United States
| | | | - Marianne Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Kristian Hovde Liland
- Nofima AS, Ås, Norway.,Faculty of Sciences and Technology, Norwegian University of Life Sciences, Ås, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,K.G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Svein Olav Kolset
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,K.G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | | |
Collapse
|
19
|
Cole AJ, Iyengar M, Panesso-Gómez S, O'Hayer P, Chan D, Delgoffe GM, Aird KM, Yoon E, Bai S, Buckanovich RJ. NFATC4 promotes quiescence and chemotherapy resistance in ovarian cancer. JCI Insight 2020; 5:131486. [PMID: 32182216 DOI: 10.1172/jci.insight.131486] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 03/11/2020] [Indexed: 12/30/2022] Open
Abstract
Development of chemotherapy resistance is a major problem in ovarian cancer. One understudied mechanism of chemoresistance is the induction of quiescence, a reversible nonproliferative state. Unfortunately, little is known about regulators of quiescence. Here, we identify the master transcription factor nuclear factor of activated T cells cytoplasmic 4 (NFATC4) as a regulator of quiescence in ovarian cancer. NFATC4 is enriched in ovarian cancer stem-like cells and correlates with decreased proliferation and poor prognosis. Treatment of cancer cells with cisplatin resulted in NFATC4 nuclear translocation and activation of the NFATC4 pathway, while inhibition of the pathway increased chemotherapy response. Induction of NFATC4 activity resulted in a marked decrease in proliferation, G0 cell cycle arrest, and chemotherapy resistance, both in vitro and in vivo. Finally, NFATC4 drove a quiescent phenotype in part via downregulation of MYC. Together, these data identify NFATC4 as a driver of quiescence and a potential new target to combat chemoresistance in ovarian cancer.
Collapse
Affiliation(s)
- Alexander J Cole
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mangala Iyengar
- Department of Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Santiago Panesso-Gómez
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Patrick O'Hayer
- Department of Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Daniel Chan
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Greg M Delgoffe
- Tumor Microenvironment Center, UPMC Hillman Cancer Center; and Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Katherine M Aird
- Department of Cellular & Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Euisik Yoon
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, Michigan, USA
| | - Shoumei Bai
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ronald J Buckanovich
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
20
|
Herum KM, Romaine A, Wang A, Melleby AO, Strand ME, Pacheco J, Braathen B, Dunér P, Tønnessen T, Lunde IG, Sjaastad I, Brakebusch C, McCulloch AD, Gomez MF, Carlson CR, Christensen G. Syndecan-4 Protects the Heart From the Profibrotic Effects of Thrombin-Cleaved Osteopontin. J Am Heart Assoc 2020; 9:e013518. [PMID: 32000579 PMCID: PMC7033859 DOI: 10.1161/jaha.119.013518] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 11/05/2019] [Indexed: 01/18/2023]
Abstract
Background Pressure overload of the heart occurs in patients with hypertension or valvular stenosis and induces cardiac fibrosis because of excessive production of extracellular matrix by activated cardiac fibroblasts. This initially provides essential mechanical support to the heart, but eventually compromises function. Osteopontin is associated with fibrosis; however, the underlying signaling mechanisms are not well understood. Herein, we examine the effect of thrombin-cleaved osteopontin on fibrosis in the heart and explore the role of syndecan-4 in regulating cleavage of osteopontin. Methods and Results Osteopontin was upregulated and cleaved by thrombin in the pressure-overloaded heart of mice subjected to aortic banding. Cleaved osteopontin was higher in plasma from patients with aortic stenosis receiving crystalloid compared with blood cardioplegia, likely because of less heparin-induced inhibition of thrombin. Cleaved osteopontin and the specific osteopontin peptide sequence RGDSLAYGLR that is exposed after thrombin cleavage both induced collagen production in cardiac fibroblasts. Like osteopontin, the heparan sulfate proteoglycan syndecan-4 was upregulated after aortic banding. Consistent with a heparan sulfate binding domain in the osteopontin cleavage site, syndecan-4 was found to bind to osteopontin in left ventricles and cardiac fibroblasts and protected osteopontin from cleavage by thrombin. Shedding of the extracellular part of syndecan-4 was more prominent at later remodeling phases, at which time levels of cleaved osteopontin were increased. Conclusions Thrombin-cleaved osteopontin induces collagen production by cardiac fibroblasts. Syndecan-4 protects osteopontin from cleavage by thrombin, but this protection is lost when syndecan-4 is shed in later phases of remodeling, contributing to progression of cardiac fibrosis.
Collapse
Affiliation(s)
- Kate M. Herum
- Institute for Experimental Medical ResearchOslo University Hospital and University of OsloNorway
- KG Jebsen Center for Cardiac ResearchUniversity of OsloNorway
- Center for Heart Failure ResearchOslo University HospitalOsloNorway
- Department of BioengineeringUniversity of California, San DiegoLa JollaCA
- Biotech Research and Innovation CentreUniversity of CopenhagenDenmark
| | - Andreas Romaine
- Institute for Experimental Medical ResearchOslo University Hospital and University of OsloNorway
- KG Jebsen Center for Cardiac ResearchUniversity of OsloNorway
- Center for Heart Failure ResearchOslo University HospitalOsloNorway
| | - Ariel Wang
- Department of BioengineeringUniversity of California, San DiegoLa JollaCA
| | - Arne Olav Melleby
- Institute for Experimental Medical ResearchOslo University Hospital and University of OsloNorway
- KG Jebsen Center for Cardiac ResearchUniversity of OsloNorway
- Center for Heart Failure ResearchOslo University HospitalOsloNorway
| | - Mari E. Strand
- Institute for Experimental Medical ResearchOslo University Hospital and University of OsloNorway
- KG Jebsen Center for Cardiac ResearchUniversity of OsloNorway
- Center for Heart Failure ResearchOslo University HospitalOsloNorway
| | - Julian Pacheco
- Department of BioengineeringUniversity of California, San DiegoLa JollaCA
| | - Bjørn Braathen
- Department of Cardiothoracic SurgeryOslo University HospitalOsloNorway
| | - Pontus Dunér
- Department of Clinical SciencesLund University Diabetes CentreLund UniversityMalmöSweden
| | - Theis Tønnessen
- Institute for Experimental Medical ResearchOslo University Hospital and University of OsloNorway
- Department of Cardiothoracic SurgeryOslo University HospitalOsloNorway
| | - Ida G. Lunde
- Institute for Experimental Medical ResearchOslo University Hospital and University of OsloNorway
- KG Jebsen Center for Cardiac ResearchUniversity of OsloNorway
- Center for Heart Failure ResearchOslo University HospitalOsloNorway
| | - Ivar Sjaastad
- Institute for Experimental Medical ResearchOslo University Hospital and University of OsloNorway
- KG Jebsen Center for Cardiac ResearchUniversity of OsloNorway
- Center for Heart Failure ResearchOslo University HospitalOsloNorway
| | - Cord Brakebusch
- Biotech Research and Innovation CentreUniversity of CopenhagenDenmark
| | - Andrew D. McCulloch
- Department of BioengineeringUniversity of California, San DiegoLa JollaCA
- Department of MedicineUniversity of California, San DiegoLa JollaCA
| | - Maria F. Gomez
- Department of Clinical SciencesLund University Diabetes CentreLund UniversityMalmöSweden
| | - Cathrine R. Carlson
- Institute for Experimental Medical ResearchOslo University Hospital and University of OsloNorway
- KG Jebsen Center for Cardiac ResearchUniversity of OsloNorway
- Center for Heart Failure ResearchOslo University HospitalOsloNorway
| | - Geir Christensen
- Institute for Experimental Medical ResearchOslo University Hospital and University of OsloNorway
- KG Jebsen Center for Cardiac ResearchUniversity of OsloNorway
- Center for Heart Failure ResearchOslo University HospitalOsloNorway
| |
Collapse
|
21
|
The Non-Fibrillar Side of Fibrosis: Contribution of the Basement Membrane, Proteoglycans, and Glycoproteins to Myocardial Fibrosis. J Cardiovasc Dev Dis 2019; 6:jcdd6040035. [PMID: 31547598 PMCID: PMC6956278 DOI: 10.3390/jcdd6040035] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/16/2019] [Accepted: 09/18/2019] [Indexed: 02/07/2023] Open
Abstract
The extracellular matrix (ECM) provides structural support and a microenvironmentfor soluble extracellular molecules. ECM is comprised of numerous proteins which can be broadly classified as fibrillar (collagen types I and III) and non-fibrillar (basement membrane, proteoglycans, and glycoproteins). The basement membrane provides an interface between the cardiomyocytes and the fibrillar ECM, while proteoglycans sequester soluble growth factors and cytokines. Myocardial fibrosis was originally only linked to accumulation of fibrillar collagens, but is now recognized as the expansion of the ECM including the non-fibrillar ECM proteins. Myocardial fibrosis can be reparative to replace the lost myocardium (e.g., ischemic injury or myocardial infarction), or can be reactive resulting from pathological activity of fibroblasts (e.g., dilated or hypertrophic cardiomyopathy). Contribution of fibrillar collagens to fibrosis is well studied, but the role of the non-fibrillar ECM proteins has remained less explored. In this article, we provide an overview of the contribution of the non-fibrillar components of the extracellular space of the heart to highlight the potential significance of these molecules in fibrosis, with direct evidence for some, although not all of these molecules in their direct contribution to fibrosis.
Collapse
|
22
|
Christa M, Weng AM, Geier B, Wörmann C, Scheffler A, Lehmann L, Oberberger J, Kraus BJ, Hahner S, Störk S, Klink T, Bauer WR, Hammer F, Köstler H. Increased myocardial sodium signal intensity in Conn's syndrome detected by 23Na magnetic resonance imaging. Eur Heart J Cardiovasc Imaging 2019; 20:263-270. [PMID: 30307545 PMCID: PMC6383057 DOI: 10.1093/ehjci/jey134] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 05/11/2018] [Accepted: 08/29/2018] [Indexed: 11/12/2022] Open
Abstract
AIMS Sodium intake has been linked to left ventricular hypertrophy independently of blood pressure, but the underlying mechanisms remain unclear. Primary hyperaldosteronism (PHA), a condition characterized by tissue sodium overload due to aldosterone excess, causes accelerated left ventricular hypertrophy compared to blood pressure matched patients with essential hypertension. We therefore hypothesized that the myocardium constitutes a novel site capable of sodium storage explaining the missing link between sodium and left ventricular hypertrophy. METHODS AND RESULTS Using 23Na magnetic resonance imaging, we investigated relative sodium signal intensities (rSSI) in the heart, calf muscle, and skin in 8 PHA patients (6 male, median age 55 years) and 12 normotensive healthy controls (HC) (8 male, median age 61 years). PHA patients had a higher mean systolic 24 h ambulatory blood pressure [152 (140; 163) vs. 125 (122; 130) mmHg, P < 0.001] and higher left ventricular mass index [71.0 (63.5; 106.8) vs. 55.0 (50.3; 66.8) g/m2, P = 0.037] than HC. Compared to HC, PHA patients exhibited significantly higher rSSI in the myocardium [0.31 (0.26; 0.34) vs. 0.24 (0.20; 0.27); P = 0.007], calf muscle [0.19 (0.16; 0.22) vs. 0.14 (0.13; 0.15); P = 0.001] and skin [0.28 (0.25; 0.33) vs. 0.19 (0.17; 0.26); P = 0.014], reflecting a difference of +27%, +38%, and +39%, respectively. Treatment of PHA resulted in significant reductions of the rSSI in the myocardium, calf muscle and skin by -13%, -27%, and -29%, respectively. CONCLUSION Myocardial tissue rSSI is increased in PHA patients and treatment of aldosterone excess effectively reduces rSSI, thus establishing the myocardium as a novel site of sodium storage in addition to skeletal muscle and skin.
Collapse
Affiliation(s)
- Martin Christa
- Comprehensive Heart Failure Center, University of Würzburg, Würzburg, Germany.,Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Andreas M Weng
- Department of Diagnostic and Interventional Radiology, University of Würzburg, Würzburg, Germany
| | - Bettina Geier
- Comprehensive Heart Failure Center, University of Würzburg, Würzburg, Germany
| | - Caroline Wörmann
- Comprehensive Heart Failure Center, University of Würzburg, Würzburg, Germany
| | - Anne Scheffler
- Institute of Pharmacy and Food Chemistry, Chair of Food Chemistry, University of Würzburg, Würzburg, Germany
| | - Leane Lehmann
- Institute of Pharmacy and Food Chemistry, Chair of Food Chemistry, University of Würzburg, Würzburg, Germany
| | - Johannes Oberberger
- Comprehensive Heart Failure Center, University of Würzburg, Würzburg, Germany.,Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Bettina J Kraus
- Comprehensive Heart Failure Center, University of Würzburg, Würzburg, Germany.,Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Stefanie Hahner
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Stefan Störk
- Comprehensive Heart Failure Center, University of Würzburg, Würzburg, Germany.,Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Thorsten Klink
- Department of Diagnostic and Interventional Radiology, University of Würzburg, Würzburg, Germany
| | - Wolfgang R Bauer
- Comprehensive Heart Failure Center, University of Würzburg, Würzburg, Germany.,Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Fabian Hammer
- Comprehensive Heart Failure Center, University of Würzburg, Würzburg, Germany.,Department of Internal Medicine, University Greifswald, Greifswald, Germany
| | - Herbert Köstler
- Comprehensive Heart Failure Center, University of Würzburg, Würzburg, Germany.,Department of Diagnostic and Interventional Radiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
23
|
Mohammadzadeh N, Lunde IG, Andenæs K, Strand ME, Aronsen JM, Skrbic B, Marstein HS, Bandlien C, Nygård S, Gorham J, Sjaastad I, Chakravarti S, Christensen G, Engebretsen KVT, Tønnessen T. The extracellular matrix proteoglycan lumican improves survival and counteracts cardiac dilatation and failure in mice subjected to pressure overload. Sci Rep 2019; 9:9206. [PMID: 31235849 PMCID: PMC6591256 DOI: 10.1038/s41598-019-45651-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 06/07/2019] [Indexed: 12/20/2022] Open
Abstract
Left ventricular (LV) dilatation is a key step in transition to heart failure (HF) in response to pressure overload. Cardiac extracellular matrix (ECM) contains fibrillar collagens and proteoglycans, important for maintaining tissue integrity. Alterations in collagen production and cross-linking are associated with cardiac LV dilatation and HF. Lumican (LUM) is a collagen binding proteoglycan with increased expression in hearts of patients and mice with HF, however, its role in cardiac function remains poorly understood. To examine the role of LUM in pressure overload induced cardiac remodeling, we subjected LUM knock-out (LUMKO) mice to aortic banding (AB) and treated cultured cardiac fibroblasts (CFB) with LUM. LUMKO mice exhibited increased mortality 1-14 days post-AB. Echocardiography revealed increased LV dilatation, altered hypertrophic remodeling and exacerbated contractile dysfunction in surviving LUMKO 1-10w post-AB. LUMKO hearts showed reduced collagen expression and cross-linking post-AB. Transcriptional profiling of LUMKO hearts by RNA sequencing revealed 714 differentially expressed transcripts, with enrichment of cardiotoxicity, ECM and inflammatory pathways. CFB treated with LUM showed increased mRNAs for markers of myofibroblast differentiation, proliferation and expression of ECM molecules important for fibrosis, including collagens and collagen cross-linking enzyme lysyl oxidase. In conclusion, we report the novel finding that lack of LUM attenuates collagen cross-linking in the pressure-overloaded heart, leading to increased mortality, dilatation and contractile dysfunction in mice.
Collapse
Affiliation(s)
- Naiyereh Mohammadzadeh
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Ida G Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
- Center for Molecular Medicine Norway, Oslo University Hospital and University of Oslo, Oslo, Norway
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Kine Andenæs
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Mari E Strand
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Jan Magnus Aronsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Bjørknes College, Oslo, Norway
| | - Biljana Skrbic
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | - Henriette S Marstein
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Caroline Bandlien
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | - Ståle Nygård
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
- Department of Informatics, University of Oslo, Oslo, Norway
| | - Joshua Gorham
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Shukti Chakravarti
- Department of Medicine, Johns Hopkins University, Baltimore, PhD, USA
- Department of Ophthalmology and Pathology, NYU Langone Health, Alexandria Life Sciences Center, West Tower, New York, NY, NY10011, USA
| | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Kristin V T Engebretsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
- Department of Surgery, Vestre Viken Hospital, Drammen, Norway
| | - Theis Tønnessen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.
- KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway.
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
24
|
Saucerman JJ, Tan PM, Buchholz KS, McCulloch AD, Omens JH. Mechanical regulation of gene expression in cardiac myocytes and fibroblasts. Nat Rev Cardiol 2019; 16:361-378. [PMID: 30683889 PMCID: PMC6525041 DOI: 10.1038/s41569-019-0155-8] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The intact heart undergoes complex and multiscale remodelling processes in response to altered mechanical cues. Remodelling of the myocardium is regulated by a combination of myocyte and non-myocyte responses to mechanosensitive pathways, which can alter gene expression and therefore function in these cells. Cellular mechanotransduction and its downstream effects on gene expression are initially compensatory mechanisms during adaptations to the altered mechanical environment, but under prolonged and abnormal loading conditions, they can become maladaptive, leading to impaired function and cardiac pathologies. In this Review, we summarize mechanoregulated pathways in cardiac myocytes and fibroblasts that lead to altered gene expression and cell remodelling under physiological and pathophysiological conditions. Developments in systems modelling of the networks that regulate gene expression in response to mechanical stimuli should improve integrative understanding of their roles in vivo and help to discover new combinations of drugs and device therapies targeting mechanosignalling in heart disease.
Collapse
Affiliation(s)
- Jeffrey J Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Philip M Tan
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Kyle S Buchholz
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA
| | - Andrew D McCulloch
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Jeffrey H Omens
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
25
|
Mathiesen SB, Lunde M, Aronsen JM, Romaine A, Kaupang A, Martinsen M, de Souza GA, Nyman TA, Sjaastad I, Christensen G, Carlson CR. The cardiac syndecan-4 interactome reveals a role for syndecan-4 in nuclear translocation of muscle LIM protein (MLP). J Biol Chem 2019; 294:8717-8731. [PMID: 30967474 DOI: 10.1074/jbc.ra118.006423] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 03/28/2019] [Indexed: 01/02/2023] Open
Abstract
Costameres are signaling hubs at the sarcolemma and important contact points between the extracellular matrix and cell interior, sensing and transducing biomechanical signals into a cellular response. The transmembrane proteoglycan syndecan-4 localizes to these attachment points and has been shown to be important in the initial stages of cardiac remodeling, but its mechanistic function in the heart remains insufficiently understood. Here, we sought to map the cardiac interactome of syndecan-4 to better understand its function and downstream signaling mechanisms. By combining two different affinity purification methods with MS analysis, we found that the cardiac syndecan-4 interactome consists of 21 novel and 29 previously described interaction partners. Nine of the novel partners were further validated to bind syndecan-4 in HEK293 cells (i.e. CAVIN1/PTRF, CCT5, CDK9, EIF2S1, EIF4B, MPP7, PARVB, PFKM, and RASIP). We also found that 19 of the 50 interactome partners bind differently to syndecan-4 in the left ventricle lysate from aortic-banded heart failure (ABHF) rats compared with SHAM-operated animals. One of these partners was the well-known mechanotransducer muscle LIM protein (MLP), which showed direct and increased binding to syndecan-4 in ABHF. Nuclear translocation is important in MLP-mediated signaling, and we found less MLP in the nuclear-enriched fractions from syndecan-4-/- mouse left ventricles but increased nuclear MLP when syndecan-4 was overexpressed in a cardiomyocyte cell line. In the presence of a cell-permeable syndecan-4-MLP disruptor peptide, the nuclear MLP level was reduced. These findings suggest that syndecan-4 mediates nuclear translocation of MLP in the heart.
Collapse
Affiliation(s)
- Sabrina Bech Mathiesen
- From the Institute for Experimental Medical Research, Oslo University Hospital, University of Oslo, 0450 Oslo
| | - Marianne Lunde
- From the Institute for Experimental Medical Research, Oslo University Hospital, University of Oslo, 0450 Oslo
| | - Jan Magnus Aronsen
- From the Institute for Experimental Medical Research, Oslo University Hospital, University of Oslo, 0450 Oslo.,the Bjørknes College, 0456 Oslo
| | - Andreas Romaine
- From the Institute for Experimental Medical Research, Oslo University Hospital, University of Oslo, 0450 Oslo.,KG Jebsen Center for Cardiac Research, University of Oslo, 0450 Oslo, and
| | - Anita Kaupang
- From the Institute for Experimental Medical Research, Oslo University Hospital, University of Oslo, 0450 Oslo
| | - Marita Martinsen
- From the Institute for Experimental Medical Research, Oslo University Hospital, University of Oslo, 0450 Oslo
| | - Gustavo Antonio de Souza
- Department of Immunology, Institute of Clinical Medicine, University of Oslo and Rikshospitalet Oslo, 0372 Oslo, Norway
| | - Tuula A Nyman
- Department of Immunology, Institute of Clinical Medicine, University of Oslo and Rikshospitalet Oslo, 0372 Oslo, Norway
| | - Ivar Sjaastad
- From the Institute for Experimental Medical Research, Oslo University Hospital, University of Oslo, 0450 Oslo.,KG Jebsen Center for Cardiac Research, University of Oslo, 0450 Oslo, and
| | - Geir Christensen
- From the Institute for Experimental Medical Research, Oslo University Hospital, University of Oslo, 0450 Oslo.,KG Jebsen Center for Cardiac Research, University of Oslo, 0450 Oslo, and
| | - Cathrine Rein Carlson
- From the Institute for Experimental Medical Research, Oslo University Hospital, University of Oslo, 0450 Oslo,
| |
Collapse
|
26
|
Mitchell A, Pimenta D, Gill J, Ahmad H, Bogle R. Cardiovascular effects of space radiation: implications for future human deep space exploration. Eur J Prev Cardiol 2019; 26:1707-1714. [PMID: 30776915 DOI: 10.1177/2047487319831497] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND A manned mission to Mars has been contemplated by the world's largest space agencies for a number of years. The duration of the trip would necessitate a much longer exposure to deep space radiation than any human has ever been exposed to in the past. Concern regarding cancer risk has thus far stalled the progress of deep space exploration; however, the effect of space radiation on the cardiovascular system is significantly less well understood. DISCUSSION Damage by radiation in space is mediated by a number of sources, including X-rays, protons and heavier charged atomic nuclei (HZE ions, the high-energy component of galactic cosmic rays). Previously, only lunar mission astronauts have been exposed to significant deep space radiation, with all other missions being low earth orbits only. The effect of this radiation on the human body has been inconclusively studied, and the long-term damage caused to the vascular endothelium by this radiation due to the effect of high-energy particles is not well known. CONCLUSION Current radiation shielding technology, which would be viable for use in spacecraft, would not eliminate radiation risk. Similar to how a variety of shielding techniques are used every day by radiographers, again without full risk elimination, we need to explore and better understand the effect of deep space radiation in order to ensure the safety of those on future space missions.
Collapse
Affiliation(s)
- Adam Mitchell
- Department of Cardiology, Royal Free London NHS Foundation Trust, London, UK
| | - Dominic Pimenta
- Department of Cardiology, Royal Free London NHS Foundation Trust, London, UK
| | - Jaspal Gill
- Department of Cardiology, Royal Free London NHS Foundation Trust, London, UK
| | - Haris Ahmad
- Department of Cardiology, Royal Free London NHS Foundation Trust, London, UK
| | - Richard Bogle
- Department of Cardiology, Royal Free London NHS Foundation Trust, London, UK
| |
Collapse
|
27
|
Christensen G, Herum KM, Lunde IG. Sweet, yet underappreciated: Proteoglycans and extracellular matrix remodeling in heart disease. Matrix Biol 2019; 75-76:286-299. [DOI: 10.1016/j.matbio.2018.01.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/22/2017] [Accepted: 01/01/2018] [Indexed: 12/20/2022]
|
28
|
Emerging roles of proteoglycans in cardiac remodeling. Int J Cardiol 2018; 278:192-198. [PMID: 30528626 DOI: 10.1016/j.ijcard.2018.11.125] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 11/12/2018] [Accepted: 11/27/2018] [Indexed: 02/07/2023]
Abstract
Cardiac remodeling is the response of the heart to a range of pathological stimuli. Cardiac remodeling is initially adaptive; however, if sustained, it ultimately causes adverse clinical outcomes. Cardiomyocyte loss or hypertrophy, inflammation and fibrosis are hallmarks of cardiac remodeling. Proteoglycans, which are composed of glycosaminoglycans and a core protein, are a non-structural component of the extracellular matrix. The lack of proteoglycans results in cardiovascular defects during development. Moreover, emerging evidence has indicated that proteoglycans act as significant modifiers in ischemia and pressure overload-related cardiac remodeling. Proteoglycans may also provide novel therapeutic strategies for further improvement in the prognosis of cardiovascular diseases.
Collapse
|
29
|
Andenæs K, Lunde IG, Mohammadzadeh N, Dahl CP, Aronsen JM, Strand ME, Palmero S, Sjaastad I, Christensen G, Engebretsen KVT, Tønnessen T. The extracellular matrix proteoglycan fibromodulin is upregulated in clinical and experimental heart failure and affects cardiac remodeling. PLoS One 2018; 13:e0201422. [PMID: 30052659 PMCID: PMC6063439 DOI: 10.1371/journal.pone.0201422] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 07/13/2018] [Indexed: 01/01/2023] Open
Abstract
Pressure overload of the heart leads to cardiac remodeling that may progress into heart failure, a common, morbid and mortal condition. Increased mechanistic insight into remodeling is instrumental for development of novel heart failure treatment. Cardiac remodeling comprises cardiomyocyte hypertrophic growth, extracellular matrix alterations including fibrosis, and inflammation. Fibromodulin is a small leucine-rich proteoglycan that regulates collagen fibrillogenesis. Fibromodulin is expressed in the cardiac extracellular matrix, however its role in the heart remains largely unknown. We investigated fibromodulin levels in myocardial biopsies from heart failure patients and mice, subjected fibromodulin knock-out (FMOD-KO) mice to pressure overload by aortic banding, and overexpressed fibromodulin in cultured cardiomyocytes and cardiac fibroblasts using adenovirus. Fibromodulin was 3-10-fold upregulated in hearts of heart failure patients and mice. Both cardiomyocytes and cardiac fibroblasts expressed fibromodulin, and its expression was increased by pro-inflammatory stimuli. Without stress, FMOD-KO mice showed no cardiac phenotype. Upon aortic banding, left ventricles of FMOD-KO mice developed mildly exacerbated hypertrophic remodeling compared to wild-type mice, with increased cardiomyocyte size and altered infiltration of leukocytes. There were no differences in mortality, left ventricle dilatation, dysfunction or expression of heart failure markers. Although collagen amount and cross-linking were comparable in FMOD-KO and wild-type, overexpression of fibromodulin in cardiac fibroblasts in vitro decreased their migratory capacity and expression of fibrosis-associated molecules, i.e. the collagen-cross linking enzyme lysyl oxidase, transglutaminase 2 and periostin. In conclusion, despite a robust fibromodulin upregulation in clinical and experimental heart failure, FMOD-KO mice showed a relatively mild hypertrophic phenotype. In cultured cardiac fibroblasts, fibromodulin has anti-fibrotic effects.
Collapse
Affiliation(s)
- Kine Andenæs
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Ida G. Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway
- Centre for Molecular Medicine Norway, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Naiyereh Mohammadzadeh
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Christen P. Dahl
- KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Department of Cardiology, Oslo University Hospital, Oslo, Norway
| | - Jan Magnus Aronsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Bjørknes College, Oslo, Norway
| | - Mari E. Strand
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Sheryl Palmero
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Kristin V. T. Engebretsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway
- Department of Surgery, Vestre Viken Hospital, Drammen, Norway
| | - Theis Tønnessen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
30
|
Takahashi I, Yamada S, Nata K. Effects of heparan sulfate proteoglycan syndecan-4 on the insulin secretory response in a mouse pancreatic β-cell line, MIN6. Mol Cell Endocrinol 2018; 470:142-150. [PMID: 29042251 DOI: 10.1016/j.mce.2017.10.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 10/04/2017] [Accepted: 10/13/2017] [Indexed: 01/03/2023]
Abstract
Heparan sulfate proteoglycans (HSPGs) comprise a core protein to which extracellular glycosaminoglycan chains are attached. Syndecan-4, one of the major HS-containing core proteins, is distributed on the cell surface, where they interact with various protein ligands and regulate a wide range of biological activities. Here, we propose that the core protein of HSPGs is involved in the insulin secretory response. To investigate the participation of HSPGs in the insulin-secretion mechanism, MIN6 cells, a mouse pancreatic β-cell line, were subcloned. The subcloned MIN6 cells were selected based on their insulin secretory response, the expression of HS and core proteins. The results from these screening experiments indicated that only syndecan-4-expressing subclones are able to secrete insulin in response to glucose. Silencing of syndecan-4 reduced glucose-induced insulin secretion, whereas the overexpression of syndecan-4 increased the insulin secretory response. These data indicate that the HSPG syndecan-4 plays important role(s) in the insulin secretory response.
Collapse
Affiliation(s)
- Iwao Takahashi
- Department of Medical Biochemistry, School of Pharmacy, Iwate Medical University, Yahaba 028-3603, Japan.
| | - Shuhei Yamada
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, Nagoya 468-8503, Japan.
| | - Koji Nata
- Department of Medical Biochemistry, School of Pharmacy, Iwate Medical University, Yahaba 028-3603, Japan.
| |
Collapse
|
31
|
Melleby AO, Romaine A, Aronsen JM, Veras I, Zhang L, Sjaastad I, Lunde IG, Christensen G. A novel method for high precision aortic constriction that allows for generation of specific cardiac phenotypes in mice. Cardiovasc Res 2018; 114:1680-1690. [DOI: 10.1093/cvr/cvy141] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/31/2018] [Indexed: 12/31/2022] Open
Affiliation(s)
- Arne O Melleby
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
- Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Andreas Romaine
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
- Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Jan Magnus Aronsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway
- Bjørknes College, Oslo, Norway
| | - Ioanni Veras
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway
| | - Lili Zhang
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
- Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
- Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Ida G Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
- Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
- Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
32
|
Romaine A, Sørensen IW, Zeltz C, Lu N, Erusappan PM, Melleby AO, Zhang L, Bendiksen B, Robinson EL, Aronsen JM, Herum KM, Danielsen HE, Sjaastad I, Christensen G, Gullberg D. Overexpression of integrin α11 induces cardiac fibrosis in mice. Acta Physiol (Oxf) 2018; 222. [PMID: 28771943 DOI: 10.1111/apha.12932] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 07/25/2017] [Accepted: 07/29/2017] [Indexed: 01/09/2023]
Abstract
AIM To understand the role of the collagen-binding integrin α11 in vivo, we have used a classical approach of creating a mouse strain overexpressing integrin α11. A transgenic mouse strain overexpressing α11 in muscle tissues was analysed in the current study with special reference to the heart tissue. METHODS We generated and phenotyped integrin α11 transgenic (TG) mice by echocardiography, magnetic resonance imaging and histology. Wild-type (WT) mice were subjected to aortic banding (AB) and the expression of integrin α11 was measured in flow cytometry-sorted cardiomyocytes and non-myocytes. RESULTS TG mice developed left ventricular concentric hypertrophy by 6 months, with increased collagen deposition and reactivation of mRNA encoding foetal genes associated with cardiovascular pathological remodelling compared to WT mice. Masson's trichrome staining revealed interstitial fibrosis, confirmed additionally by magnetic resonance imaging and was found to be most prominent in the cardiac septum of TG but not WT mice. TG hearts expressed increased levels of transforming growth factor-β2 and transforming growth factor-β3 and upregulated smooth muscle actin. Macrophage infiltration coincided with increased NF-κB signalling in TG but not WT hearts. Integrin α11 expression was increased in both cardiomyocytes and non-myocyte cells from WT AB hearts compared to sham-operated animals. CONCLUSION We report for the first time that overexpression of integrin α11 induces cardiac fibrosis and left ventricular hypertrophy. This is a result of changes in intracellular hypertrophic signalling and secretion of soluble factors that increase collagen production in the heart.
Collapse
Affiliation(s)
- A. Romaine
- Institute for Experimental Medical Research; Oslo University Hospital and University of Oslo; Oslo Norway
| | - I. W. Sørensen
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - C. Zeltz
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - N. Lu
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - P. M. Erusappan
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - A. O. Melleby
- Institute for Experimental Medical Research; Oslo University Hospital and University of Oslo; Oslo Norway
| | - L. Zhang
- Institute for Experimental Medical Research; Oslo University Hospital and University of Oslo; Oslo Norway
| | - B. Bendiksen
- Institute for Experimental Medical Research; Oslo University Hospital and University of Oslo; Oslo Norway
| | - E. L. Robinson
- Laboratory of Experimental Cardiology; Department of Cardiovascular Sciences; KU Leuven; Leuven Belgium
| | - J. M. Aronsen
- Institute for Experimental Medical Research; Oslo University Hospital and University of Oslo; Oslo Norway
- Bjørknes College; Oslo Norway
| | - K. M. Herum
- Institute for Experimental Medical Research; Oslo University Hospital and University of Oslo; Oslo Norway
| | - H. E. Danielsen
- Institute for Cancer Genetics and Informatics; Oslo University Hospital; Oslo Norway
- Center for Cancer Biomedicine; University of Oslo; Oslo Norway
- Department of Informatics; University of Oslo; Oslo Norway
- Nuffield Division of Clinical Laboratory Sciences; University of Oxford; Oxford UK
| | - I. Sjaastad
- Institute for Experimental Medical Research; Oslo University Hospital and University of Oslo; Oslo Norway
| | - G. Christensen
- Institute for Experimental Medical Research; Oslo University Hospital and University of Oslo; Oslo Norway
| | - D. Gullberg
- Department of Biomedicine; University of Bergen; Bergen Norway
| |
Collapse
|
33
|
Cytoprotective activated protein C averts Nlrp3 inflammasome-induced ischemia-reperfusion injury via mTORC1 inhibition. Blood 2017; 130:2664-2677. [PMID: 28882883 DOI: 10.1182/blood-2017-05-782102] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 09/01/2017] [Indexed: 12/15/2022] Open
Abstract
Cytoprotection by activated protein C (aPC) after ischemia-reperfusion injury (IRI) is associated with apoptosis inhibition. However, IRI is hallmarked by inflammation, and hence, cell-death forms disjunct from immunologically silent apoptosis are, in theory, more likely to be relevant. Because pyroptosis (ie, cell death resulting from inflammasome activation) is typically observed in IRI, we speculated that aPC ameliorates IRI by inhibiting inflammasome activation. Here we analyzed the impact of aPC on inflammasome activity in myocardial and renal IRIs. aPC treatment before or after myocardial IRI reduced infarct size and Nlrp3 inflammasome activation in mice. Kinetic in vivo analyses revealed that Nlrp3 inflammasome activation preceded myocardial injury and apoptosis, corroborating a pathogenic role of the Nlrp3 inflammasome. The constitutively active Nlrp3A350V mutation abolished the protective effect of aPC, demonstrating that Nlrp3 suppression is required for aPC-mediated protection from IRI. In vitro aPC inhibited inflammasome activation in macrophages, cardiomyocytes, and cardiac fibroblasts via proteinase-activated receptor 1 (PAR-1) and mammalian target of rapamycin complex 1 (mTORC1) signaling. Accordingly, inhibiting PAR-1 signaling, but not the anticoagulant properties of aPC, abolished the ability of aPC to restrict Nlrp3 inflammasome activity and tissue damage in myocardial IRI. Targeting biased PAR-1 signaling via parmodulin-2 restricted mTORC1 and Nlrp3 inflammasome activation and limited myocardial IRI as efficiently as aPC. The relevance of aPC-mediated Nlrp3 inflammasome suppression after IRI was corroborated in renal IRI, where the tissue protective effect of aPC was likewise dependent on Nlrp3 inflammasome suppression. These studies reveal that aPC protects from IRI by restricting mTORC1-dependent inflammasome activation and that mimicking biased aPC PAR-1 signaling using parmodulins may be a feasible therapeutic approach to combat IRI.
Collapse
|
34
|
Herum KM, Lunde IG, McCulloch AD, Christensen G. The Soft- and Hard-Heartedness of Cardiac Fibroblasts: Mechanotransduction Signaling Pathways in Fibrosis of the Heart. J Clin Med 2017; 6:jcm6050053. [PMID: 28534817 PMCID: PMC5447944 DOI: 10.3390/jcm6050053] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/04/2017] [Accepted: 05/08/2017] [Indexed: 12/27/2022] Open
Abstract
Cardiac fibrosis, the excessive accumulation of extracellular matrix (ECM), remains an unresolved problem in most forms of heart disease. In order to be successful in preventing, attenuating or reversing cardiac fibrosis, it is essential to understand the processes leading to ECM production and accumulation. Cardiac fibroblasts are the main producers of cardiac ECM, and harbor great phenotypic plasticity. They are activated by the disease-associated changes in mechanical properties of the heart, including stretch and increased tissue stiffness. Despite much remaining unknown, an interesting body of evidence exists on how mechanical forces are translated into transcriptional responses important for determination of fibroblast phenotype and production of ECM constituents. Such mechanotransduction can occur at multiple cellular locations including the plasma membrane, cytoskeleton and nucleus. Moreover, the ECM functions as a reservoir of pro-fibrotic signaling molecules that can be released upon mechanical stress. We here review the current status of knowledge of mechanotransduction signaling pathways in cardiac fibroblasts that culminate in pro-fibrotic gene expression.
Collapse
Affiliation(s)
- Kate M Herum
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway.
- Center for Heart Failure Research, Oslo University Hospital, 0450 Oslo, Norway.
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA.
| | - Ida G Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway.
- Center for Heart Failure Research, Oslo University Hospital, 0450 Oslo, Norway.
| | - Andrew D McCulloch
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA.
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA.
| | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway.
- Center for Heart Failure Research, Oslo University Hospital, 0450 Oslo, Norway.
| |
Collapse
|
35
|
Parra V, Rothermel BA. Calcineurin signaling in the heart: The importance of time and place. J Mol Cell Cardiol 2017; 103:121-136. [PMID: 28007541 PMCID: PMC5778886 DOI: 10.1016/j.yjmcc.2016.12.006] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 12/12/2016] [Accepted: 12/16/2016] [Indexed: 12/20/2022]
Abstract
The calcium-activated protein phosphatase, calcineurin, lies at the intersection of protein phosphorylation and calcium signaling cascades, where it provides an essential nodal point for coordination between these two fundamental modes of intracellular communication. In excitatory cells, such as neurons and cardiomyocytes, that experience rapid and frequent changes in cytoplasmic calcium, calcineurin protein levels are exceptionally high, suggesting that these cells require high levels of calcineurin activity. Yet, it is widely recognized that excessive activation of calcineurin in the heart contributes to pathological hypertrophic remodeling and the progression to failure. How does a calcium activated enzyme function in the calcium-rich environment of the continuously contracting heart without pathological consequences? This review will discuss the wide range of calcineurin substrates relevant to cardiovascular health and the mechanisms calcineurin uses to find and act on appropriate substrates in the appropriate location while potentially avoiding others. Fundamental differences in calcineurin signaling in neonatal verses adult cardiomyocytes will be addressed as well as the importance of maintaining heterogeneity in calcineurin activity across the myocardium. Finally, we will discuss how circadian oscillations in calcineurin activity may facilitate integration with other essential but conflicting processes, allowing a healthy heart to reap the benefits of calcineurin signaling while avoiding the detrimental consequences of sustained calcineurin activity that can culminate in heart failure.
Collapse
Affiliation(s)
- Valentina Parra
- Advanced Centre for Chronic Disease (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas, Universidad de Chile, Santiago,Chile; Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Quimicas y Farmaceuticas, Universidad de Chie, Santiago, Chile
| | - Beverly A Rothermel
- Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Centre, Dallas, TX, USA; Department of Molecular Biology, University of Texas Southwestern Medical Centre, Dallas, TX, USA.
| |
Collapse
|
36
|
Wang Y, Baeyens N, Corti F, Tanaka K, Fang JS, Zhang J, Jin Y, Coon B, Hirschi KK, Schwartz MA, Simons M. Syndecan 4 controls lymphatic vasculature remodeling during mouse embryonic development. Development 2016; 143:4441-4451. [PMID: 27789626 DOI: 10.1242/dev.140129] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 10/19/2016] [Indexed: 01/13/2023]
Abstract
The role of fluid shear stress in vasculature development and remodeling is well appreciated. However, the mechanisms regulating these effects remain elusive. We show that abnormal flow sensing in lymphatic endothelial cells (LECs) caused by Sdc4 or Pecam1 deletion in mice results in impaired lymphatic vessel remodeling, including abnormal valve morphogenesis. Ablation of either gene leads to the formation of irregular, enlarged and excessively branched lymphatic vessels. In both cases, lymphatic valve-forming endothelial cells are randomly oriented, resulting in the formation of abnormal valves. These abnormalities are much more pronounced in Sdc4-/-; Pecam1-/- double-knockout mice, which develop severe edema. In vitro, SDC4 knockdown human LECs fail to align under flow and exhibit high expression of the planar cell polarity protein VANGL2. Reducing VANGL2 levels in SDC4 knockdown LECs restores their alignment under flow, while VANGL2 overexpression in wild-type LECs mimics the flow alignment abnormalities seen in SDC4 knockdown LECs. SDC4 thus controls flow-induced LEC polarization via regulation of VANGL2 expression.
Collapse
Affiliation(s)
- Yingdi Wang
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Nicolas Baeyens
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Federico Corti
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Keiichiro Tanaka
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Jennifer S Fang
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Jiasheng Zhang
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Yu Jin
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Brian Coon
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Karen K Hirschi
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA.,Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Martin A Schwartz
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA.,Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA.,Department of Biomedical Engineering, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Michael Simons
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA .,Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
37
|
Melleby AO, Strand ME, Romaine A, Herum KM, Skrbic B, Dahl CP, Sjaastad I, Fiane AE, Filmus J, Christensen G, Lunde IG. The Heparan Sulfate Proteoglycan Glypican-6 Is Upregulated in the Failing Heart, and Regulates Cardiomyocyte Growth through ERK1/2 Signaling. PLoS One 2016; 11:e0165079. [PMID: 27768722 PMCID: PMC5074531 DOI: 10.1371/journal.pone.0165079] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 10/05/2016] [Indexed: 11/18/2022] Open
Abstract
Pressure overload is a frequent cause of heart failure. Heart failure affects millions of patients worldwide and is a major cause of morbidity and mortality. Cell surface proteoglycans are emerging as molecular players in cardiac remodeling, and increased knowledge about their regulation and function is needed for improved understanding of cardiac pathogenesis. Here we investigated glypicans (GPC1-6), a family of evolutionary conserved heparan sulfate proteoglycans anchored to the extracellular leaflet of the cell membrane, in experimental and clinical heart failure, and explored the function of glypican-6 in cardiac cells in vitro. In mice subjected to pressure overload by aortic banding (AB), we observed elevated glypican-6 levels during hypertrophic remodeling and dilated, end-stage heart failure. Consistently, glypican-6 mRNA was elevated in left ventricular myocardium from explanted hearts of patients with end-stage, dilated heart failure with reduced ejection fraction. Glypican-6 levels correlated negatively with left ventricular ejection fraction in patients, and positively with lung weight after AB in mice. Glypican-6 mRNA was expressed in both cardiac fibroblasts and cardiomyocytes, and the corresponding protein displayed different sizes in the two cell types due to tissue-specific glycanation. Importantly, adenoviral overexpression of glypican-6 in cultured cardiomyocytes increased protein synthesis and induced mRNA levels of the pro-hypertrophic signature gene ACTA1 and the hypertrophy and heart failure signature genes encoding natriuretic peptides, NPPA and NPPB. Overexpression of GPC6 induced ERK1/2 phosphorylation, and co-treatment with the ERK inhibitor U0126 attenuated the GPC6-induced increase in NPPA, NPPB and protein synthesis. In conclusion, our data suggests that glypican-6 plays a role in clinical and experimental heart failure progression by regulating cardiomyocyte growth through ERK signaling.
Collapse
Affiliation(s)
- Arne O. Melleby
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Center for Heart Failure Research, University of Oslo, Oslo, Norway
- * E-mail:
| | - Mari E. Strand
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Andreas Romaine
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Kate M. Herum
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Biljana Skrbic
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Center for Heart Failure Research, University of Oslo, Oslo, Norway
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | - Christen P. Dahl
- Center for Heart Failure Research, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Center for Heart Failure Research, University of Oslo, Oslo, Norway
- Division of Molecular and Cellular Biology, Sunnybrook Research Institute and Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Arnt E. Fiane
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | - Jorge Filmus
- Division of Molecular and Cellular Biology, Sunnybrook Research Institute and Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Ida G. Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Center for Heart Failure Research, University of Oslo, Oslo, Norway
| |
Collapse
|
38
|
Jaroszyński AJ, Jaroszyńska A, Przywara S, Zaborowski T, Książek A, Dąbrowski W. Syndecan-4 Is an Independent Predictor of All-Cause as Well as Cardiovascular Mortality in Hemodialysis Patients. PLoS One 2016; 11:e0163532. [PMID: 27685148 PMCID: PMC5042500 DOI: 10.1371/journal.pone.0163532] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 09/10/2016] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Left ventricular hypertrophy is associated withincreased mortality in hemodialysis (HD) patients.Syndecan-4 plays a role in many processes that are involved in the heart fibrosis and hypertrophy.We designed this study to prospectively determine whether syndecan-4 was predictive of mortality in a group of HD patients. METHODS In total, 191 HD patients were included. Clinical, biochemical and echocardiographic parameters were recorded. HD patients were followed-up for 23.18 ± 4.02 months. RESULTS Syndecan-4 levels correlated strongly with geometrical echocardiographic parameters and ejection fraction. Relations with pressure-related parameters were weak and only marginally significant. Using the receiver operating characteristics the optimal cut-off points in predicting all-cause as well as cardiovascular (CV) mortality were evaluated and patients were divided into low and high syndecan-4 groups. A Kaplan-Meier analysis showed that the cumulative incidences of all-cause as well as CV mortality were higher in high serum syndecan-4 group compared with those with low serum syndecan-4 (p<0.001 in both cases).A multivariate Cox proportional hazards regression analysis revealed syndecan-4 concentration to be an independent and significant predictor of all-cause (hazard ratio, 2.99; confidence interval, 2.34 to 3.113; p<0.001)as well as CV mortality (hazard ratio, 2.81;confidence interval, 2.28to3.02; p<0.001). CONCLUSIONS Serum syndecan-4 concentration reflects predominantly geometrical echocardiographic parameters. In HD patients serum syndecan-4 concentration is independently associated with all-cause as well as CV mortality.
Collapse
Affiliation(s)
- Andrzej J. Jaroszyński
- Institute of Medical Sciences, Jan Kochanowski University in Kielce, Kielce, Poland
- Department of Family Medicine, Medical University of Lublin, Lublin, Poland
| | - Anna Jaroszyńska
- Department of Cardiology, Medical University of Lublin, Lublin, Poland
| | - Stanisław Przywara
- Department of Vascular Surgery, Medical University of Lublin, Lublin, Poland
| | - Tomasz Zaborowski
- Department of Family Medicine, Medical University of Lublin, Lublin, Poland
| | - Andrzej Książek
- Department of Nephrology, Medical University of Lublin, Lublin, Poland
| | - Wojciech Dąbrowski
- Department of Anesthesiology and Intensive Care, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
39
|
Eftestøl E, Egner IM, Lunde IG, Ellefsen S, Andersen T, Sjåland C, Gundersen K, Bruusgaard JC. Increased hypertrophic response with increased mechanical load in skeletal muscles receiving identical activity patterns. Am J Physiol Cell Physiol 2016; 311:C616-C629. [PMID: 27488660 DOI: 10.1152/ajpcell.00016.2016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 07/26/2016] [Indexed: 11/22/2022]
Abstract
It is often assumed that mechanical factors are important for effects of exercise on muscle, but during voluntary training and most experimental conditions the effects could solely be attributed to differences in electrical activity, and direct evidence for a mechanosensory pathway has been scarce. We here show that, in rat muscles stimulated in vivo under deep anesthesia with identical electrical activity patterns, isometric contractions induced twofold more hypertrophy than contractions with 50-60% of the isometric force. The number of myonuclei and the RNA levels of myogenin and myogenic regulatory factor 4 were increased with high load, suggesting that activation of satellite cells is mechano dependent. On the other hand, training induced a major shift in fiber type distribution from type 2b to 2x that was load independent, indicating that the electrical signaling rather than mechanosignaling controls fiber type. RAC-α serine/threonine-protein kinase (Akt) and ribosomal protein S6 kinase β-1 (S6K1) were not significantly differentially activated by load, suggesting that the differences in mechanical factors were not important for activating the Akt/mammalian target of rapamycin/S6K1 pathway. The transmembrane molecule syndecan-4 implied in overload hypertrophy in cardiac muscle was not load dependent, suggesting that mechanosignaling in skeletal muscle is different.
Collapse
Affiliation(s)
- Einar Eftestøl
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Ingrid M Egner
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Ida G Lunde
- Department of Genetics, Harvard Medical School, Boston, Massachusetts; Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway; KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Stian Ellefsen
- Section for Sport Sciences, Lillehammer University College, Lillehammer, Norway; and
| | - Tom Andersen
- Department of Biosciences, University of Oslo, Oslo, Norway
| | | | | | - Jo C Bruusgaard
- Department of Biosciences, University of Oslo, Oslo, Norway; Department of Health Sciences, Kristiania University College, Oslo, Norway
| |
Collapse
|
40
|
|
41
|
Abstract
In the heart, Ca(2+) signals regulate a variety of biological functions ranging from contractility to gene expression, cellular hypertrophy and death. In this review, we summarize the role of local Ca(2+) homeostasis in these processes in healthy cardiac muscle cells, and highlight how mismanaged Ca(2+) handling contributes to the pathophysiology of conditions such as cardiac arrhythmia, ischemic heart disease, cardiac hypertrophy and heart failure. Aiming to provide an introduction to the field with a clinical perspective, we also indicate how current and future therapies may modulate cardiomyocytes Ca(2+) handling for the treatment of patients.
Collapse
Affiliation(s)
| | - William E Louch
- b Institute for Experimental Medical Research, Oslo University Hospital Ullevål and University of Oslo , Oslo , Norway
| | - Ivar Sjaastad
- b Institute for Experimental Medical Research, Oslo University Hospital Ullevål and University of Oslo , Oslo , Norway
| |
Collapse
|
42
|
Xie J, He G, Chen Q, Sun J, Dai Q, Lu J, Li G, Wu H, Li R, Chen J, Xu W, Xu B. Syndecan-4 Signaling Is Required for Exercise-Induced Cardiac Hypertrophy. Mol Med 2016; 22:192-201. [PMID: 26835698 DOI: 10.2119/molmed.2015.00026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 01/20/2016] [Indexed: 01/28/2023] Open
Abstract
Cardiac hypertrophy can be broadly classified as either physiological or pathological. Physiological stimuli such as exercise cause adaptive cardiac hypertrophy and normal heart function. Pathological stimuli including hypertension and aortic valvular stenosis cause maladaptive cardiac remodeling and ultimately heart failure. Syndecan-4 (synd4) is a transmembrane proteoglycan identified as being involved in cardiac adaptation after injury, but whether it takes part in physiological cardiac hypertrophy is unclear. We observed upregulation of synd4 in exercise-induced hypertrophic myocardium. To evaluate the role of synd4 in the physiological form of cardiac hypertrophy, mice lacking synd4 (synd4-/-) were exercised by swimming for 4 wks. Ultrasonic cardiogram (UCG) and histological analysis revealed that swimming induced the hypertrophic phenotype but was blunted in synd4-/- compared with wild-type (WT) mice. The swimming-induced activation of Akt, a key molecule in physiological hypertrophy was also more decreased than in WT controls. In cultured cardiomyocytes, synd4 overexpression could induce cell enlargement, protein synthesis and distinct physiological molecular alternation. Akt activation also was observed in synd4-overexpressed cardiomyocytes. Furthermore, inhibition of protein kinase C (PKC) prevented the synd4-induced hypertrophic phenotype and Akt phosphorylation. This study identified an essential role of synd4 in mediation of physiological cardiac hypertrophy.
Collapse
Affiliation(s)
- Jun Xie
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Guixin He
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, China.,Department of Cardiology, the First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Qinhua Chen
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Jiayin Sun
- Department of VIP, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Qin Dai
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Jianrong Lu
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Guannan Li
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Han Wu
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Ran Li
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Jianzhou Chen
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Wei Xu
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Biao Xu
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| |
Collapse
|
43
|
Strand ME, Aronsen JM, Braathen B, Sjaastad I, Kvaløy H, Tønnessen T, Christensen G, Lunde IG. Shedding of syndecan-4 promotes immune cell recruitment and mitigates cardiac dysfunction after lipopolysaccharide challenge in mice. J Mol Cell Cardiol 2015; 88:133-44. [DOI: 10.1016/j.yjmcc.2015.10.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 09/20/2015] [Accepted: 10/03/2015] [Indexed: 12/24/2022]
|
44
|
Gopal S, Søgaard P, Multhaupt HAB, Pataki C, Okina E, Xian X, Pedersen ME, Stevens T, Griesbeck O, Park PW, Pocock R, Couchman JR. Transmembrane proteoglycans control stretch-activated channels to set cytosolic calcium levels. J Cell Biol 2015; 210:1199-211. [PMID: 26391658 PMCID: PMC4586746 DOI: 10.1083/jcb.201501060] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 08/25/2015] [Indexed: 02/07/2023] Open
Abstract
Syndecans regulate members of the transient receptor potential family to control cytosolic calcium levels with impact on cell adhesion, junction formation, and neuronal guidance. Transmembrane heparan sulfate proteoglycans regulate multiple aspects of cell behavior, but the molecular basis of their signaling is unresolved. The major family of transmembrane proteoglycans is the syndecans, present in virtually all nucleated cells, but with mostly unknown functions. Here, we show that syndecans regulate transient receptor potential canonical (TRPCs) channels to control cytosolic calcium equilibria and consequent cell behavior. In fibroblasts, ligand interactions with heparan sulfate of syndecan-4 recruit cytoplasmic protein kinase C to target serine714 of TRPC7 with subsequent control of the cytoskeleton and the myofibroblast phenotype. In epidermal keratinocytes a syndecan–TRPC4 complex controls adhesion, adherens junction composition, and early differentiation in vivo and in vitro. In Caenorhabditis elegans, the TRPC orthologues TRP-1 and -2 genetically complement the loss of syndecan by suppressing neuronal guidance and locomotory defects related to increases in neuronal calcium levels. The widespread and conserved syndecan–TRPC axis therefore fine tunes cytoskeletal organization and cell behavior.
Collapse
Affiliation(s)
- Sandeep Gopal
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark Biotech Research and Innovation Center, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Pernille Søgaard
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark Biotech Research and Innovation Center, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Hinke A B Multhaupt
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark Biotech Research and Innovation Center, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Csilla Pataki
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark Biotech Research and Innovation Center, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Elena Okina
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark Biotech Research and Innovation Center, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Xiaojie Xian
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark Biotech Research and Innovation Center, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Mikael E Pedersen
- Biotech Research and Innovation Center, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Troy Stevens
- Department of Pharmacology, Center for Lung Biology, University of South Alabama, Mobile, AL 36688 Department of Medicine, Center for Lung Biology, University of South Alabama, Mobile, AL 36688
| | - Oliver Griesbeck
- Max Planck Institute of Neurobiology, 82152 Martinsried, Germany
| | - Pyong Woo Park
- Division of Newborn Medicine, Children's Hospital, Harvard Medical School, Boston, MA 02115 Division of Respiratory Diseases, Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Roger Pocock
- Biotech Research and Innovation Center, University of Copenhagen, 2200 Copenhagen, Denmark
| | - John R Couchman
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark Biotech Research and Innovation Center, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
45
|
Rønning SB, Carlson CR, Stang E, Kolset SO, Hollung K, Pedersen ME. Syndecan-4 Regulates Muscle Differentiation and Is Internalized from the Plasma Membrane during Myogenesis. PLoS One 2015; 10:e0129288. [PMID: 26068620 PMCID: PMC4467083 DOI: 10.1371/journal.pone.0129288] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 05/06/2015] [Indexed: 12/31/2022] Open
Abstract
The cell surface proteoglycan syndecan-4 has been reported to be crucial for muscle differentiation, but the molecular mechanisms still remain to be fully understood. During in vitro differentiation of bovine muscle cells immunocytochemical analyses showed strong labelling of syndecan-4 intracellularly, in close proximity with Golgi structures, in membranes of intracellular vesicles and finally, in the nuclear area including the nuclear envelope. Chase experiments showed that syndecan-4 was internalized from the plasma membrane during this process. Furthermore, when syndecan-4 was knocked down by siRNA more myotubes were formed, and the expression of myogenic transcription factors, β1-integrin and actin was influenced. However, when bovine muscle cells were treated with a cell-penetrating peptide containing the cytoplasmic region of syndecan-4, myoblast fusion and thus myotube formation was blocked, both in normal cells and in syndecan-4 knock down cells. Altogether this suggests that the cytoplasmic domain of syndecan-4 is important in regulation of myogenesis. The internalization of syndecan-4 from the plasma membrane during muscle differentiation and the nuclear localization of syndecan-4 in differentiated muscle cells may be part of this regulation, and is a novel aspect of syndecan biology which merits further studies.
Collapse
Affiliation(s)
| | - Cathrine R. Carlson
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Espen Stang
- Department of Pathology, Oslo University Hospital, Rikshospitalet, P.O. Box 4950 Nydalen, 0424 Oslo, Norway
| | - Svein O. Kolset
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | | | | |
Collapse
|
46
|
Chistiakov DA, Orekhov AN, Bobryshev YV. Vascular smooth muscle cell in atherosclerosis. Acta Physiol (Oxf) 2015; 214:33-50. [PMID: 25677529 DOI: 10.1111/apha.12466] [Citation(s) in RCA: 295] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 02/02/2015] [Accepted: 02/09/2015] [Indexed: 12/30/2022]
Abstract
Vascular smooth muscle cells (VSMCs) exhibit phenotypic and functional plasticity in order to respond to vascular injury. In case of the vessel damage, VSMCs are able to switch from the quiescent 'contractile' phenotype to the 'proinflammatory' phenotype. This change is accompanied by decrease in expression of smooth muscle (SM)-specific markers responsible for SM contraction and production of proinflammatory mediators that modulate induction of proliferation and chemotaxis. Indeed, activated VSMCs could efficiently proliferate and migrate contributing to the vascular wall repair. However, in chronic inflammation that occurs in atherosclerosis, arterial VSMCs become aberrantly regulated and this leads to increased VSMC dedifferentiation and extracellular matrix formation in plaque areas. Proatherosclerotic switch in VSMC phenotype is a complex and multistep mechanism that may be induced by a variety of proinflammatory stimuli and hemodynamic alterations. Disturbances in hemodynamic forces could initiate the proinflammatory switch in VSMC phenotype even in pre-clinical stages of atherosclerosis. Proinflammatory signals play a crucial role in further dedifferentiation of VSMCs in affected vessels and propagation of pathological vascular remodelling.
Collapse
Affiliation(s)
- D. A. Chistiakov
- Research Center for Children's Health; Moscow Russia
- The Mount Sinai Community Clinical Oncology Program; Mount Sinai Comprehensive Cancer Center; Mount Sinai Medical Center; Miami Beach FL USA
| | - A. N. Orekhov
- Institute for Atherosclerosis; Skolkovo Innovative Center; Moscow Russia
- Laboratory of Angiopathology; Institute of General Pathology and Pathophysiology; Russian Academy of Sciences; Moscow Russia
- Department of Biophysics; Biological Faculty; Moscow State University; Moscow Russia
| | - Y. V. Bobryshev
- Institute for Atherosclerosis; Skolkovo Innovative Center; Moscow Russia
- Faculty of Medicine; School of Medical Sciences; University of New South Wales; Kensington Sydney NSW Australia
- School of Medicine; University of Western Sydney; Campbelltown NSW Australia
| |
Collapse
|
47
|
Pataki CA, Couchman JR, Brábek J. Wnt Signaling Cascades and the Roles of Syndecan Proteoglycans. J Histochem Cytochem 2015; 63:465-80. [PMID: 25910817 DOI: 10.1369/0022155415586961] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 04/21/2015] [Indexed: 12/17/2022] Open
Abstract
Wnt signaling comprises a group of pathways emanating from the extracellular environment through cell-surface receptors into the intracellular milieu. Wnt signaling cascades can be divided into two main branches, the canonical/β-catenin pathway and the non-canonical pathways containing the Wnt/planar cell polarity and Wnt/calcium signaling. Syndecans are type I transmembrane proteoglycans with a long evolutionary history, being expressed in all Bilateria and in almost all cell types. Both Wnt pathways have been extensively studied over the past 30 years and shown to have roles during development and in a multitude of diseases. Although the first evidence for interactions between syndecans and Wnts dates back to 1997, the number of studies connecting these pathways is low, and many open questions remained unanswered. In this review, syndecan's involvement in Wnt signaling pathways as well as some of the pathologies resulting from dysregulation of the components of these pathways are summarized.
Collapse
Affiliation(s)
- Csilla A Pataki
- Department of Cell Biology, Charles University in Prague, Czech Republic, University of Copenhagen, Denmark (CAP,JB)
| | - John R Couchman
- Department of Biomedical Sciences and Biotech Research and Innovation Center, University of Copenhagen, Denmark (JRC)
| | - Jan Brábek
- Department of Cell Biology, Charles University in Prague, Czech Republic, University of Copenhagen, Denmark (CAP,JB)
| |
Collapse
|
48
|
Herum KM, Lunde IG, Skrbic B, Louch WE, Hasic A, Boye S, Unger A, Brorson SH, Sjaastad I, Tønnessen T, Linke WA, Gomez MF, Christensen G. Syndecan-4 is a key determinant of collagen cross-linking and passive myocardial stiffness in the pressure-overloaded heart. Cardiovasc Res 2015; 106:217-26. [DOI: 10.1093/cvr/cvv002] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 12/20/2014] [Indexed: 01/02/2023] Open
|
49
|
Kunnas T, Nikkari ST. Contribution of syndecan-4 genetic variants to hypertension, the TAMRISK study. BMC Res Notes 2014; 7:815. [PMID: 25410619 PMCID: PMC4247658 DOI: 10.1186/1756-0500-7-815] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 11/06/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND A human syndecan-4 genetic variant (rs1981429) has previously been associated with lean tissue mass and intra-abdominal fat, and SNP rs4599 with resting energy expenditure in healthy early pubertal children. These variations could thus cause overweight and hypothetically lead to hypertension. Their association with body mass index and blood pressure was therefore studied in a Finnish cohort of adults. METHODS The data was collected from the Tampere adult population cardiovascular risk study (TAMRISK). A total of 279 cases with hypertension and/or coronary artery disease (CAD), and 488 non-hypertensive healthy controls were selected from a Finnish periodic health examination 50-year-old cohort. Information was available also from their 45-year examination. DNA was extracted from buccal swabs and human syndecan-4 gene SNPs were analyzed using KASP genotyping. RESULTS The SNP rs1981429 variant TT was significantly associated with hypertension, as compared to variants TG and GG at the age of 50 years (p=0.015). The variant TT was also associated with increased BMI at the ages of 45 and 50 years (p=0.008 and p=0.026, respectively). In addition, TT genotype associated with increased CAD prevalence (P=0.013). No significant associations between rs4599 variants and hypertension or BMI were found. In haplotype analysis the number of alleles T (rs1981429)/C (rs4599) was linearly associated with CAD prevalence; the highest prevalence (13%) was in haplotype TT/CC and lowest prevalence (1%) in haplotype GG/TT (p=0.01). CONCLUSION Syndecan-4 polymorphisms were associated with essential hypertension, BMI, and CAD prevalence in the TAMRISK study.
Collapse
Affiliation(s)
| | - Seppo T Nikkari
- Department of Medical Biochemistry, University of Tampere Medical School and Fimlab laboratories, Tampere 33014, Finland.
| |
Collapse
|
50
|
Ueland T, Aukrust P, Nymo SH, Kjekshus J, McMurray JJV, Wikstrand J, Block D, Zaugg C, Gullestad L. Novel extracellular matrix biomarkers as predictors of adverse outcome in chronic heart failure: association between biglycan and response to statin therapy in the CORONA trial. J Card Fail 2014; 21:153-9. [PMID: 25451704 DOI: 10.1016/j.cardfail.2014.10.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 10/13/2014] [Accepted: 10/27/2014] [Indexed: 01/15/2023]
Abstract
BACKGROUND The extracellular matrix (ECM) plays an important role in left ventricular remodeling and progression of heart failure (HF). Biglycan and mimecan are ECM proteins that are abundantly expressed in cardiac tissue but have not been evaluated as prognostic markers in HF. We investigated their interaction with statin treatment and association with adverse outcome in chronic HF. METHODS AND RESULTS The association between serum levels of biglycan and mimecan and the primary end point (cardiovascular [CV] death, nonfatal myocardial infarction, nonfatal stroke), all-cause mortality, CV death, the composite of all-cause mortality/hospitalization for worsening of HF, and the coronary end point was evaluated in 1,390 patients >60 years of age with ischemic systolic HF in the Controlled Rosuvastatin Multinational Trial in HF (CORONA) population, randomly assigned to 10 mg rosuvastatin or placebo. Serum biglycan and mimecan added no prognostic information beyond conventional risk factors, including N-terminal pro-B-type natriuretic peptide. However, statin treatment improved all outcomes except CV death in patients with low biglycan levels (ie, lower tertile), even after full multivariable adjustment. CONCLUSIONS Although circulating levels of mimecan and biglycan were of limited predictive value in patients with chronic HF, circulating biglycan could be a useful marker for targeting statin therapy in patients with HF.
Collapse
Affiliation(s)
- Thor Ueland
- Research Institute for Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Pål Aukrust
- Research Institute for Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway; Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Ståle H Nymo
- Research Institute for Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - John Kjekshus
- Department of Cardiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - John J V McMurray
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland, United Kingdom
| | | | | | | | - Lars Gullestad
- Department of Cardiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway; Center for Heart Failure Research, University of Oslo, Oslo, Norway
| |
Collapse
|