1
|
Li J, Huang N, Zhang X, Sun C, Chen J, Wei Q. Changes of collagen content in lung tissues of plateau yak and its mechanism of adaptation to hypoxia. PeerJ 2024; 12:e18250. [PMID: 39372716 PMCID: PMC11451445 DOI: 10.7717/peerj.18250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/16/2024] [Indexed: 10/08/2024] Open
Abstract
Collagen is crucial for tissue structure, functional maintenance, and cellular processes such as proliferation and differentiation. However, the specific changes in collagen expression and its associated genes in the lung tissues of yaks at high altitudes and their relationship with environmental adaptation remain poorly understood. Studying differences in the content of collagen fibers and gene expression between yaks at high (4,500 m) and low (2,600 m) altitudes, as well as between cattle at low altitudes (2,600 m). Using Masson staining, we found that the collagen fiber content in the lung tissues of yaks at low altitude was significantly higher compared to yaks at high altitude and cattle at the same altitude (P < 0.05). It was revealed through transcriptomic analyses that genes differentially expressed between high and low altitude yaks, as well as between low altitude yaks and cattle, were notably enriched in pathways related to cell adhesion, collagen synthesis, focal adhesion, and ECM-receptor interactions. Specifically, genes involved in mesenchymal collagen synthesis (e.g., COL1A1, COL1A2, COL3A1), basement membrane collagen synthesis (e.g., COL4A1, COL4A2, COL4A4, COL4A6), and peripheral collagen synthesis (e.g., COL5A1, COL6A1, COL6A2, COL6A3) were significantly upregulated in the lung tissues of yaks at low altitude compared to their high altitude counterparts and cattle (P < 0.05). In conclusion, yaks at lower altitudes exhibit increased collagen synthesis by upregulating collagen gene expression, which contributes to maintaining alveolar stability and septal flexibility. Conversely, the expression of collagen genes in yak lung tissues was down-regulated with the increase in altitude, and it was speculated that the decrease in collagen may be used to constrain the function of elastic fibers that are more abundant at high altitude, so as to enable them to adapt to the harsh environment with hypoxia and high altitude. This adaptation mechanism highlights the role of collagen in environmental acclimatization and contributes to our understanding of how altitude and species influence collagen-related physiological processes in yaks.
Collapse
Affiliation(s)
- Jingyi Li
- College of Eco-Environmental Engineering, Qinghai University, Xining, Qinghai, China
| | - Nating Huang
- College of Eco-Environmental Engineering, Qinghai University, Xining, Qinghai, China
| | - Xun Zhang
- College of Eco-Environmental Engineering, Qinghai University, Xining, Qinghai, China
| | - Ci Sun
- College of Eco-Environmental Engineering, Qinghai University, Xining, Qinghai, China
| | - Jiarui Chen
- College of Eco-Environmental Engineering, Qinghai University, Xining, Qinghai, China
| | - Qing Wei
- College of Eco-Environmental Engineering, Qinghai University, Xining, Qinghai, China
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, Qinghai, China
| |
Collapse
|
2
|
Halsey G, Sinha D, Dhital S, Wang X, Vyavahare N. Role of elastic fiber degradation in disease pathogenesis. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166706. [PMID: 37001705 PMCID: PMC11659964 DOI: 10.1016/j.bbadis.2023.166706] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 03/31/2023]
Abstract
Elastin is a crucial extracellular matrix protein that provides structural integrity to tissues. Crosslinked elastin and associated microfibrils, named elastic fiber, contribute to biomechanics by providing the elasticity required for proper function. During aging and disease, elastic fiber can be progressively degraded and since there is little elastin synthesis in adults, degraded elastic fiber is not regenerated. There is substantial evidence linking loss or damage of elastic fibers to the clinical manifestation and pathogenesis of a variety of diseases. Disruption of elastic fiber networks by hereditary mutations, aging, or pathogenic stimuli results in systemic ailments associated with the production of elastin degradation products, inflammatory responses, and abnormal physiology. Due to its longevity, unique mechanical properties, and widespread distribution in the body, elastic fiber plays a central role in homeostasis of various physiological systems. While pathogenesis related to elastic fiber degradation has been more thoroughly studied in elastic fiber rich tissues such as the vasculature and the lungs, even tissues containing relatively small quantities of elastic fibers such as the eyes or joints may be severely impacted by elastin degradation. Elastic fiber degradation is a common observation in certain hereditary, age, and specific risk factor exposure induced diseases representing a converging point of pathological clinical phenotypes which may also help explain the appearance of co-morbidities. In this review, we will first cover the role of elastic fiber degradation in the manifestation of hereditary diseases then individually explore the structural role and degradation effects of elastic fibers in various tissues and organ systems. Overall, stabilizing elastic fiber structures and repairing lost elastin may be effective strategies to reverse the effects of these diseases.
Collapse
Affiliation(s)
- Gregory Halsey
- Department of Bioengineering, Clemson University, SC 29634, United States of America
| | - Dipasha Sinha
- Department of Bioengineering, Clemson University, SC 29634, United States of America
| | - Saphala Dhital
- Department of Bioengineering, Clemson University, SC 29634, United States of America
| | - Xiaoying Wang
- Department of Bioengineering, Clemson University, SC 29634, United States of America
| | - Naren Vyavahare
- Department of Bioengineering, Clemson University, SC 29634, United States of America.
| |
Collapse
|
3
|
Hume RD, Kanagalingam S, Deshmukh T, Chen S, Mithieux SM, Rashid FN, Roohani I, Lu J, Doan T, Graham D, Clayton ZE, Slaughter E, Kizana E, Stempien-Otero AS, Brown P, Thomas L, Weiss AS, Chong JJ. Tropoelastin Improves Post-Infarct Cardiac Function. Circ Res 2023; 132:72-86. [PMID: 36453283 PMCID: PMC9829044 DOI: 10.1161/circresaha.122.321123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
BACKGROUND Myocardial infarction (MI) is among the leading causes of death worldwide. Following MI, necrotic cardiomyocytes are replaced by a stiff collagen-rich scar. Compared to collagen, the extracellular matrix protein elastin has high elasticity and may have more favorable properties within the cardiac scar. We sought to improve post-MI healing by introducing tropoelastin, the soluble subunit of elastin, to alter scar mechanics early after MI. METHODS AND RESULTS We developed an ultrasound-guided direct intramyocardial injection method to administer tropoelastin directly into the left ventricular anterior wall of rats subjected to induced MI. Experimental groups included shams and infarcted rats injected with either PBS vehicle control or tropoelastin. Compared to vehicle treated controls, echocardiography assessments showed tropoelastin significantly improved left ventricular ejection fraction (64.7±4.4% versus 46.0±3.1% control) and reduced left ventricular dyssynchrony (11.4±3.5 ms versus 31.1±5.8 ms control) 28 days post-MI. Additionally, tropoelastin reduced post-MI scar size (8.9±1.5% versus 20.9±2.7% control) and increased scar elastin (22±5.8% versus 6.2±1.5% control) as determined by histological assessments. RNA sequencing (RNAseq) analyses of rat infarcts showed that tropoelastin injection increased genes associated with elastic fiber formation 7 days post-MI and reduced genes associated with immune response 11 days post-MI. To show translational relevance, we performed immunohistochemical analyses on human ischemic heart disease cardiac samples and showed an increase in tropoelastin within fibrotic areas. Using RNA-seq we also demonstrated the tropoelastin gene ELN is upregulated in human ischemic heart disease and during human cardiac fibroblast-myofibroblast differentiation. Furthermore, we showed by immunocytochemistry that human cardiac fibroblast synthesize increased elastin in direct response to tropoelastin treatment. CONCLUSIONS We demonstrate for the first time that purified human tropoelastin can significantly repair the infarcted heart in a rodent model of MI and that human cardiac fibroblast synthesize elastin. Since human cardiac fibroblasts are primarily responsible for post-MI scar synthesis, our findings suggest exciting future clinical translation options designed to therapeutically manipulate this synthesis.
Collapse
Affiliation(s)
- Robert D. Hume
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.).,Sydney Medical School, University of Sydney, NSW, Australia (R.D.H., T.D., F.R., Z.E.C., E.K., J.J.H.C.)
| | - Shaan Kanagalingam
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.)
| | - Tejas Deshmukh
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.).,Department of Cardiology, Westmead Hospital, NSW, Australia (T.D., J.L., E.K., P.B., L.T., J.J.H.C.).,Sydney Medical School, University of Sydney, NSW, Australia (R.D.H., T.D., F.R., Z.E.C., E.K., J.J.H.C.)
| | - Siqi Chen
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.)
| | - Suzanne M. Mithieux
- Charles Perkins Centre, University of Sydney, NSW, Australia (S.M.M., A.S.W.).,School of Life and Environmental Sciences, University of Sydney, NSW, Australia (S.M.M., A.S.W.)
| | - Fairooj N. Rashid
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.)
| | - Iman Roohani
- School of Biomedical Engineering, University of Sydney, NSW, Australia (I.R.).,School of Chemistry, University of New South Wales, Australia (I.R.)
| | - Juntang Lu
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.).,Department of Cardiology, Westmead Hospital, NSW, Australia (T.D., J.L., E.K., P.B., L.T., J.J.H.C.)
| | - Tram Doan
- Centre for Cancer Research, Westmead Institute for Medical Research, NSW, Australia (T.D.‚ D.G.)
| | - Dinny Graham
- Centre for Cancer Research, Westmead Institute for Medical Research, NSW, Australia (T.D.‚ D.G.).,Westmead Breast Cancer Institute, NSW, Australia (D.G.).,Westmead Clinical School, University of Sydney, NSW, Australia (D.G., L.T.)
| | - Zoe E. Clayton
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.).,Sydney Medical School, University of Sydney, NSW, Australia (R.D.H., T.D., F.R., Z.E.C., E.K., J.J.H.C.)
| | | | - Eddy Kizana
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.).,Department of Cardiology, Westmead Hospital, NSW, Australia (T.D., J.L., E.K., P.B., L.T., J.J.H.C.).,Sydney Medical School, University of Sydney, NSW, Australia (R.D.H., T.D., F.R., Z.E.C., E.K., J.J.H.C.)
| | - April S. Stempien-Otero
- Department of Medicine, Division of Cardiology, University of Washington School of Medicine, Seattle, WA (A.S.S.-O.)
| | - Paula Brown
- Department of Cardiology, Westmead Hospital, NSW, Australia (T.D., J.L., E.K., P.B., L.T., J.J.H.C.)
| | - Liza Thomas
- Department of Cardiology, Westmead Hospital, NSW, Australia (T.D., J.L., E.K., P.B., L.T., J.J.H.C.).,Westmead Clinical School, University of Sydney, NSW, Australia (D.G., L.T.)
| | | | - James J.H. Chong
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.).,Department of Cardiology, Westmead Hospital, NSW, Australia (T.D., J.L., E.K., P.B., L.T., J.J.H.C.).,Sydney Medical School, University of Sydney, NSW, Australia (R.D.H., T.D., F.R., Z.E.C., E.K., J.J.H.C.)
| |
Collapse
|
4
|
Shi X, Zhang S, Liu Y, Brazile B, Cooley J, Butler JR, McMahan SR, Perez KL, Xu J, Eastep T, Nguyen KT, Bajona P, Peltz M, Gao H, Hong Y, Liao J. Spatial distribution and network morphology of epicardial, endocardial, interstitial, and Purkinje cell-associated elastin fibers in porcine left ventricle. Bioact Mater 2023; 19:348-359. [PMID: 35892002 PMCID: PMC9301607 DOI: 10.1016/j.bioactmat.2022.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/07/2022] [Accepted: 04/18/2022] [Indexed: 11/19/2022] Open
Abstract
Cardiac extracellular matrices (ECM) play crucial functional roles in cardiac biomechanics. Previous studies have mainly focused on collagen, the major structural ECM in heart wall. The role of elastin in cardiac mechanics, however, is poorly understood. In this study, we investigated the spatial distribution and microstructural morphologies of cardiac elastin in porcine left ventricles. We demonstrated that the epicardial elastin network had location- and depth-dependency, and the overall epicardial elastin fiber mapping showed certain correlation with the helical heart muscle fiber architecture. When compared to the epicardial layer, the endocardial layer was thicker and has a higher elastin-collagen ratio and a denser elastin fiber network; moreover, the endocardial elastin fibers were finer and more wavy than the epicardial elastin fibers, all suggesting various interface mechanics. The myocardial interstitial elastin fibers co-exist with the perimysial collagen to bind the cardiomyocyte bundles; some of the interstitial elastin fibers showed a locally aligned, hinge-like structure to connect the adjacent cardiomyocyte bundles. This collagen-elastin combination reflects an optimal design in which the collagen provides mechanical strength and elastin fibers facilitate recoiling during systole. Moreover, cardiac elastin fibers, along with collagen network, closely associated with the Purkinje cells, indicating that this ECM association could be essential in organizing cardiac Purkinje cells into "fibrous" and "branching" morphologies and serving as a protective feature when Purkinje fibers experience large deformations in vivo. In short, our observations provide a structural basis for future in-depth biomechanical investigations and biomimicking of this long-overlooked cardiac ECM component.
Collapse
Affiliation(s)
- Xiaodan Shi
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, 76010, USA
| | - Song Zhang
- College of Engineering and College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, 39762, USA
| | - Yue Liu
- School of Engineering, Brown University, Providence, RI, 02912, USA
| | - Bryn Brazile
- College of Engineering and College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, 39762, USA
| | - Jim Cooley
- College of Engineering and College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, 39762, USA
| | - J. Ryan Butler
- College of Engineering and College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, 39762, USA
| | - Sara R. McMahan
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, 76010, USA
| | - Karla L. Perez
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, 76010, USA
| | - Jiazhu Xu
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, 76010, USA
| | - Timothy Eastep
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, 76010, USA
| | - Kytai T. Nguyen
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, 76010, USA
| | - Pietro Bajona
- Department of Cardiovascular and Thoracic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Allegheny Health Network-Drexel University College of Medicine, Pittsburgh, PA, 15212, USA
| | - Matthias Peltz
- Department of Cardiovascular and Thoracic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Huajian Gao
- School of Engineering, Brown University, Providence, RI, 02912, USA
- College of Engineering, Nanyang Technical University, 308232, Singapore
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, 76010, USA
| | - Jun Liao
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, 76010, USA
| |
Collapse
|
5
|
Gonzalez BA, Herrera A, Ponce C, Gonzalez Perez M, Hsu CPD, Mirza A, Perez M, Ramaswamy S. Stem Cell-Secreted Allogeneic Elastin-Rich Matrix with Subsequent Decellularization for the Treatment of Critical Valve Diseases in the Young. Bioengineering (Basel) 2022; 9:bioengineering9100587. [PMID: 36290556 PMCID: PMC9598163 DOI: 10.3390/bioengineering9100587] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/07/2022] [Accepted: 10/12/2022] [Indexed: 11/30/2022] Open
Abstract
Critical valve diseases in infants have a very poor prognosis for survival. Particularly challenging is for the valve replacement to support somatic growth. From a valve regenerative standpoint, bio-scaffolds have been extensively investigated recently. While bio-scaffold valves facilitate acute valve functionality, their xenogeneic properties eventually induce a hostile immune response. Our goal was to investigate if a bio-scaffold valve could be deposited with tissues derived from allogeneic stem cells, with a specific dynamic culture protocol to enhance the extracellular matrix (ECM) constituents, with subsequent stem cell removal. Porcine small intestinal submucosa (PSIS) tubular-shaped bio-scaffold valves were seeded with human bone marrow-derived mesenchymal stem cells (hBMMSCs), cultured statically for 8 days, and then exposed to oscillatory fluid-induced shear stresses for two weeks. The valves were then safely decellularized to remove the hBMMSCs while retaining their secreted ECM. This de novo ECM was found to include significantly higher (p < 0.05) levels of elastin compared to the ECM produced by the hBMMSCs under standard rotisserie culture. The elastin-rich valves consisted of ~8% elastin compared to the ~10% elastin composition of native heart valves. Allogeneic elastin promotes chemotaxis thereby accelerating regeneration and can support somatic growth by rapidly integrating with the host following implantation. As a proof-of-concept of accelerated regeneration, we found that valve interstitial cells (VICs) secreted significantly more (p < 0.05) collagen on the elastin-rich matrix compared to the raw PSIS bio-scaffold.
Collapse
|
6
|
Elkenhans B, Protti A, Shah A, Onthank D, Botnar R. Visualization of elastin using cardiac magnetic resonance imaging after myocardial infarction as inflammatory response. Sci Rep 2021; 11:11004. [PMID: 34040032 PMCID: PMC8155029 DOI: 10.1038/s41598-021-90092-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 04/29/2021] [Indexed: 12/16/2022] Open
Abstract
The aim of this study was to investigate the merits of magnetic resonance imaging (MRI) using an elastin-binding contrast agent after myocardial infarction in mouse models with deletions of monocyte populations. Permanent ligation of the left anterior descending (LAD) artery was conducted in 10 wild-type mice and 10 each of three knockout models: CX3CR-/-, CCR2-/-, and MCP-1-/-. At 7 days and 30 days after permanent ligation, cardiac MRI was performed with a 7 T-Bruker horizontal scanner for in vivo detection of elastin with an elastin/tropoelastin-specific contrast agent (ESMA). Histology was performed with staining for elastin, collagen I and III, and F4/80. Real-time PCR was conducted to quantify the expression of genes for collagen I and III, F4/80, and tumor necrosis factor alpha (TNFα). Histological and ESMA-indicated elastin areas were strongly correlated (r = 0.8). 30 days after permanent ligation, CCR2-deficient mice demonstrated higher elastin levels in the scar relative to MCP-1-/- (p < 0.04) and wild-type mice (p < 0.02). The ejection fraction was lower in CCR2-deficient mice. In vivo MRI in mouse models of MI can detect elastin deposition after myocardial infarction, highlighting the pivotal role of elastin in myocardial remodeling in mouse models with deletions of monocyte populations.
Collapse
Affiliation(s)
- Britta Elkenhans
- Department of Cardiology, Pneumology, and Angiology, University Hospital Aachen, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany.
| | - Andrea Protti
- Harvard Medical School, Department of Imaging, Lurie Family Imaging Center, Boston, USA
| | - Ajay Shah
- Cardiovascular Division, King's College London, London, UK
| | | | - René Botnar
- Cardiovascular Division, King's College London, London, UK
| |
Collapse
|
7
|
Frangogiannis NG, Kovacic JC. Extracellular Matrix in Ischemic Heart Disease, Part 4/4: JACC Focus Seminar. J Am Coll Cardiol 2020; 75:2219-2235. [PMID: 32354387 DOI: 10.1016/j.jacc.2020.03.020] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 02/27/2020] [Accepted: 03/03/2020] [Indexed: 02/06/2023]
Abstract
Myocardial ischemia and infarction, both in the acute and chronic phases, are associated with cardiomyocyte loss and dramatic changes in the cardiac extracellular matrix (ECM). It has long been appreciated that these changes in the cardiac ECM result in altered mechanical properties of ischemic or infarcted myocardial segments. However, a growing body of evidence now clearly demonstrates that these alterations of the ECM not only affect the structural properties of the ischemic and post-infarct heart, but they also play a crucial and sometimes direct role in mediating a range of biological pathways, including the orchestration of inflammatory and reparative processes, as well as the pathogenesis of adverse remodeling. This final part of a 4-part JACC Focus Seminar reviews the evidence on the role of the ECM in relation to the ischemic and infarcted heart, as well as its contribution to cardiac dysfunction and adverse clinical outcomes.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York.
| | - Jason C Kovacic
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Victor Chang Cardiac Research Institute and St. Vincent's Clinical School, University of New South Wales, Darlinghurst, New South Wales, Australia.
| |
Collapse
|
8
|
Zhang S, Chen R, Chakrabarti S, Su Z. Resident macrophages as potential therapeutic targets for cardiac ageing and injury. Clin Transl Immunology 2020; 9:e1167. [PMID: 32874584 PMCID: PMC7450172 DOI: 10.1002/cti2.1167] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 07/21/2020] [Accepted: 08/01/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiac‐resident macrophages (CRMs) play critical roles in maintaining cardiac homoeostasis and removing senescent and dying cells. Recent preclinical data have re‐energised the area of cardioimmunology and provided improved understanding of the modulation of compositional and functional phenotypes of CRMs. These data can aid in achieving improved cardiac regeneration, repair and functional remodelling following cardiac injury. In this review, we discuss the composition and renewal of various subsets of CRMs. Specific attention has been given to delineate the roles of various CRM subsets with respect to (1) facilitation of cardiac development and maintenance of physiological function such as electrical conduction and rhythm; (2) promotion of cardiac regeneration, inflammation resolution and functional remodelling following a cardiac injury; and (3) therapeutic potential. We have also highlighted the relationship between CRM replenishment and cardiomyocyte senescence as well as cardiovascular diseases development. Finally, we have addressed future perspectives and directions in basic research and potentially clinical applications of CRMs.
Collapse
Affiliation(s)
- Shiqing Zhang
- International Genome Center Jiangsu University Zhenjiang China.,Department of Immunology Jiangsu University Zhenjiang China
| | - Rong Chen
- International Genome Center Jiangsu University Zhenjiang China.,Department of Immunology Jiangsu University Zhenjiang China
| | | | - Zhaoliang Su
- International Genome Center Jiangsu University Zhenjiang China.,Department of Immunology Jiangsu University Zhenjiang China.,Laboratory Center The Fourth Affiliated Hospital of Jiangsu University Zhenjiang China
| |
Collapse
|
9
|
Meng Q, Sun Z, Gu H, Luo J, Wang J, Wang C, Han S. Expression profiles of long noncoding RNAs and messenger RNAs in the border zone of myocardial infarction in rats. Cell Mol Biol Lett 2019; 24:63. [PMID: 31827539 PMCID: PMC6889673 DOI: 10.1186/s11658-019-0185-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 10/30/2019] [Indexed: 11/13/2022] Open
Abstract
Background The participation of long noncoding RNAs (lncRNAs) in myocardial infarction has recently been noted. However, their underlying roles in the border zone of myocardial infarction remain unclear. This study uses microarrays to determine the profiles of lncRNAs and mRNAs in the border zone. Methods Bioinformatics methods were employed to uncover their underlying roles. Highly dysregulated lncRNAs was further validated via PCR. Results Four hundred seven lncRNAs and 752 mRNAs were upregulated, while 132 lncRNAs and 547 mRNAs were downregulated in the border zone of myocardial infarction. A circos graph was constructed to visualize the chromosomal distribution and classification of the dysregulated lncRNAs and mRNAs. The upregulated mRNAs in the border zone were most highly enriched in cytokine activity, binding, cytokine receptor binding and related processes, as ascertained through Go analysis. Pathway analysis of the upregulated mRNAs showed the most significant changes were in the TNF signaling pathway, cytokine–cytokine receptor interaction and chemokine signaling pathway and similar pathways and interactions. An lncRNA–mRNA co-expression network was established to probe into the underlying functions of the 10 most highly dysregulated lncRNAs based on their co-expressed mRNAs. In the co-expression network, we found 16 genes directly involved in myocardial infarction, including Alox5ap, Itgb2 and B4galt1. The lncRNAs AY212271, EF424788 and MRAK088538, among others, might be associated with myocardial infarction. BC166504 is probably a key lncRNA in the border zone of myocardial infarction. Conclusions The results may have revealed some aberrantly expressed lncRNAs and mRNAs that contribute to the underlying pathophysiological mechanisms of myocardial infarction.
Collapse
Affiliation(s)
- Qingkun Meng
- Shengjing Hospital, China Medical University, Shenyang, China
| | - Zhijun Sun
- Shengjing Hospital, China Medical University, Shenyang, China
| | - Hui Gu
- Shengjing Hospital, China Medical University, Shenyang, China
| | - Jiaying Luo
- Shengjing Hospital, China Medical University, Shenyang, China
| | - Jingjing Wang
- Shengjing Hospital, China Medical University, Shenyang, China
| | - Chuanhe Wang
- Shengjing Hospital, China Medical University, Shenyang, China
| | - Su Han
- Shengjing Hospital, China Medical University, Shenyang, China
| |
Collapse
|
10
|
O'Rourke SA, Dunne A, Monaghan MG. The Role of Macrophages in the Infarcted Myocardium: Orchestrators of ECM Remodeling. Front Cardiovasc Med 2019; 6:101. [PMID: 31417911 PMCID: PMC6685361 DOI: 10.3389/fcvm.2019.00101] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 07/09/2019] [Indexed: 12/13/2022] Open
Abstract
Myocardial infarction is the most common form of acute cardiac injury attributing to heart failure. While there have been significant advances in current therapies, mortality and morbidity remain high. Emphasis on inflammation and extracellular matrix remodeling as key pathological factors has brought to light new potential therapeutic targets including macrophages which are central players in the inflammatory response following myocardial infarction. Blood derived and tissue resident macrophages exhibit both a pro- and anti-inflammatory phenotype, essential for removing injured tissue and facilitating repair, respectively. Sustained activation of pro-inflammatory macrophages evokes extensive remodeling of cardiac tissue through secretion of matrix proteases and activation of myofibroblasts. As the heart continues to employ methods of remodeling and repair, a destructive cycle prevails ultimately leading to deterioration of cardiac function and heart failure. This review summarizes not only the traditionally accepted role of macrophages in the heart but also recent advances that have deepened our understanding and appreciation of this dynamic cell. We discuss the role of macrophages in normal and maladaptive matrix remodeling, as well as studies to date which have aimed to target the inflammatory response in combatting excessive matrix deposition and subsequent heart failure.
Collapse
Affiliation(s)
- Sinead A O'Rourke
- Department of Mechanical and Manufacturing Engineering, Trinity College Dublin, Dublin, Ireland.,School of Biochemistry & Immunology and School of Medicine, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland.,Trinity Centre for Bioengineering, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Aisling Dunne
- School of Biochemistry & Immunology and School of Medicine, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Michael G Monaghan
- Department of Mechanical and Manufacturing Engineering, Trinity College Dublin, Dublin, Ireland.,Trinity Centre for Bioengineering, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland.,Advanced Materials for BioEngineering Research (AMBER) Centre, Trinity College Dublin and Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
11
|
Hodges MM, Zgheib C, Xu J, Hu J, Dewberry LC, Hilton SA, Allukian MW, Gorman JH, Gorman RC, Liechty KW. Differential Expression of Transforming Growth Factor-β1 Is Associated With Fetal Regeneration After Myocardial Infarction. Ann Thorac Surg 2019; 108:59-66. [PMID: 30690019 DOI: 10.1016/j.athoracsur.2018.12.042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 11/12/2018] [Accepted: 12/17/2018] [Indexed: 11/16/2022]
Abstract
BACKGROUND Global extracellular matrix (ECM)-related gene expression is decreased after myocardial infarction (MI) in fetal sheep when compared with adult sheep. Transforming growth factor (TGF)-β1 is a key regulator of ECM; therefore we hypothesize that TGF-β1 is differentially expressed in adult and fetal infarcts after MI. METHODS Adult and fetal sheep underwent MI via ligation of the left anterior descending coronary artery. Expression of TGF-β1 and ECM-related genes was evaluated by ovine-specific microarray and quantitative polymerase chain reaction. Fibroblasts from the left ventricle of adult and fetal hearts were treated with TGF-β1 or a TGF-β1 receptor inhibitor (LY36497) to evaluate the effect of TGF-β1 on ECM-related genes. RESULTS Col1a1, col3a1, and MMP9 expression were increased in adult infarcts 3 and 30 days after MI but were upregulated in fetal infarcts only 3 days after MI. Three days after MI elastin expression was increased in adult infarcts. Despite upregulation in adult infarcts both 3 and 30 days after MI, TGF-β1 was not upregulated in fetal infarcts at any time point. Inhibition of the TGF-β1 receptor in adult cardiac fibroblasts decreased expression of col1a1, col3a1, MMP9, elastin, and TIMP1, whereas treatment of fetal cardiac fibroblasts with TGF-β1 increased expression of these genes. CONCLUSIONS TGF-β1 is increased in adult infarcts compared with regenerative, fetal infarcts after MI. Although treatment of fetal cardiac fibroblasts with TGF-β1 conveys an adult phenotype, inhibition of TGF-β1 conveys a fetal phenotype to adult cardiac fibroblasts. Decreasing TGF-β1 after MI may facilitate myocardial regeneration by "fetalizing" the otherwise fibrotic, adult response to MI.
Collapse
Affiliation(s)
- Maggie M Hodges
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, Colorado.
| | - Carlos Zgheib
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, Colorado
| | - Junwang Xu
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, Colorado
| | - Junyi Hu
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, Colorado
| | - Lindel C Dewberry
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, Colorado
| | - Sarah A Hilton
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, Colorado
| | - Myron W Allukian
- Department of Pediatric Surgery, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Joseph H Gorman
- Department of Surgery and Gorman Cardiovascular Research Group, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert C Gorman
- Department of Surgery and Gorman Cardiovascular Research Group, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kenneth W Liechty
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, Colorado
| |
Collapse
|
12
|
Ramos IT, Henningsson M, Nezafat M, Lavin B, Lorrio S, Gebhardt P, Protti A, Eykyn TR, Andia ME, Flögel U, Phinikaridou A, Shah AM, Botnar RM. Simultaneous Assessment of Cardiac Inflammation and Extracellular Matrix Remodeling after Myocardial Infarction. Circ Cardiovasc Imaging 2018; 11:e007453. [PMID: 30524648 PMCID: PMC6277008 DOI: 10.1161/circimaging.117.007453] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 08/04/2018] [Indexed: 01/25/2023]
Abstract
Background Optimal healing of the myocardium following myocardial infarction (MI) requires a suitable degree of inflammation and its timely resolution, together with a well-orchestrated deposition and degradation of extracellular matrix (ECM) proteins. Methods and Results MI and SHAM-operated animals were imaged at 3,7,14 and 21 days with 3T magnetic resonance imaging (MRI) using a 19F/1H surface coil. Mice were injected with 19F-perfluorocarbon (PFC) nanoparticles to study inflammatory cell recruitment, and with a gadolinium-based elastin-binding contrast agent (Gd-ESMA) to evaluate elastin content. 19F MRI signal co-localized with infarction areas, as confirmed by late-gadolinium enhancement, and was highest 7days post-MI, correlating with macrophage content (MAC-3 immunohistochemistry) (ρ=0.89,P<0.0001). 19F quantification with in vivo (MRI) and ex vivo nuclear magnetic resonance (NMR) spectroscopy correlated linearly (ρ=0.58,P=0.020). T1 mapping after Gd-ESMA injection showed increased relaxation rate (R1) in the infarcted regions and was significantly higher at 21days compared with 7days post-MI (R1[s-1]:21days=2.8 [IQR,2.69-3.30] vs 7days=2.3 [IQR,2.12-2.5], P<0.05), which agreed with an increased tropoelastin content (ρ=0.89, P<0.0001). The predictive value of each contrast agent for beneficial remodeling was evaluated in a longitudinal proof-of-principle study. Neither R1 nor 19F at day 7 were significant predictors for beneficial remodeling (P=0.68;P=0.062). However, the combination of both measurements (R1<2.34Hz and 0.55≤19F≤1.85) resulted in an odds ratio of 30.0 (CI95%:1.41-638.15;P=0.029) for favorable post-MI remodeling. Conclusions Multinuclear 1H/19F MRI allows the simultaneous assessment of inflammation and elastin remodeling in a murine MI model. The interplay of these biological processes affects cardiac outcome and may have potential for improved diagnosis and personalized treatment.
Collapse
Affiliation(s)
- Isabel T Ramos
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Markus Henningsson
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Maryam Nezafat
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Begoña Lavin
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Silvia Lorrio
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Pierre Gebhardt
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
- Department of Physics of Molecular Imaging Systems, Institute for Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany
| | - Andrea Protti
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Thomas R Eykyn
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Marcelo E Andia
- Radiology Department, School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Ulrich Flögel
- Department of Molecular Cardiology, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Alkystis Phinikaridou
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Ajay M Shah
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - René M Botnar
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
- Escuela de Ingeniería, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
13
|
A heart for fibrillin: spatial arrangement in adult wild-type murine myocardial tissue. Histochem Cell Biol 2018; 150:271-280. [DOI: 10.1007/s00418-018-1686-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2018] [Indexed: 01/03/2023]
|
14
|
Daley MC, Fenn SL, Black LD. Applications of Cardiac Extracellular Matrix in Tissue Engineering and Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1098:59-83. [PMID: 30238366 DOI: 10.1007/978-3-319-97421-7_4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The role of the cardiac extracellular matrix (cECM) in providing biophysical and biochemical cues to the cells housed within during disease and development has become increasingly apparent. These signals have been shown to influence many fundamental cardiac cell behaviors including contractility, proliferation, migration, and differentiation. Consequently, alterations to cell phenotype result in directed remodeling of the cECM. This bidirectional communication means that the cECM can be envisioned as a medium for information storage. As a result, the reprogramming of the cECM is increasingly being employed in tissue engineering and regenerative medicine as a method with which to treat disease. In this chapter, an overview of the composition and structure of the cECM as well as its role in cardiac development and disease will be provided. Additionally, therapeutic modulation of cECM for cardiac regeneration as well as bottom-up and top-down approaches to ECM-based cardiac tissue engineering is discussed. Finally, lingering questions regarding the role of cECM in tissue engineering and regenerative medicine are offered as a catalyst for future research.
Collapse
Affiliation(s)
- Mark C Daley
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Spencer L Fenn
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
- Center for Biomedical Career Development, University of Massachusetts Medical School, Worcester, MA, USA
| | - Lauren D Black
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA.
- Cellular, Molecular and Developmental Biology Program, Sackler School for Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
15
|
Yu Y, Yin G, Bao S, Guo Z. Kinetic alterations of collagen and elastic fibres and their association with cardiac function in acute myocardial infarction. Mol Med Rep 2017; 17:3519-3526. [PMID: 29286107 PMCID: PMC5802149 DOI: 10.3892/mmr.2017.8347] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 09/14/2017] [Indexed: 11/16/2022] Open
Abstract
The present study aimed to investigate kinetic alterations of collagen and elastic fibres and their association with cardiac function in the acute myocardial infarction (AMI) heart. AMI was generated in Sprague-Dawley rats by ligation of the left anterior descending coronary artery. Cardiac function was determined using B-ultrasonography, AMI was verified using histopathology. The kinetics of collagen type I/III and elastic fibre were evaluated using immunohistochemistry and western blotting at 1 week (1 w), 2 weeks (2 w), 3 weeks (3 w) and 4 weeks (4 w) post-AMI. Cardiac function was decreased by 78, 70, 50 and 38% at weeks 1, 2, 3 or 4 post-AMI, respectively, compared with the normal heart. Elastic fibre was decreased gradually, demonstrating alterations of 2, 77, 86 or 97% reduction, respectively, at weeks 1, 2, 3 or 4 in the AMI heart. Collagen I fibre was increased 1.4-, 1.5-, 2.9- or 3.9-fold at weeks 1, 2, 3 or 4 respectively, compared with the normal heart. Similarly, collagen III was increased 1.2-, 1.7-, 2.8- or 3.9-fold, following AMI. Kinetics of increased collagen I/III, in combination with decreased elastic fibre in infarcted area following AMI, provided evidence that compromised cardiac function following AMI was due to graduate wound healing/scar formation in the infarcted zone, increased stiffness and reduced flexibility of the heart.
Collapse
Affiliation(s)
- Yuexin Yu
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Guotian Yin
- Department of Cardiology, Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453000, P.R. China
| | - Shisan Bao
- Discipline of Pathology, School of Medical Sciences and Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Zhikun Guo
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| |
Collapse
|
16
|
Nielsen SH, Mouton AJ, DeLeon-Pennell KY, Genovese F, Karsdal M, Lindsey ML. Understanding cardiac extracellular matrix remodeling to develop biomarkers of myocardial infarction outcomes. Matrix Biol 2017; 75-76:43-57. [PMID: 29247693 DOI: 10.1016/j.matbio.2017.12.001] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 11/02/2017] [Accepted: 12/08/2017] [Indexed: 01/08/2023]
Abstract
Cardiovascular Disease (CVD) is the most common cause of death in industrialized countries, and myocardial infarction (MI) is a major CVD with significant morbidity and mortality. Following MI, the left ventricle (LV) undergoes a wound healing response to ischemia that results in extracellular matrix (ECM) scar formation to replace necrotic myocytes. While ECM accumulation following MI is termed cardiac fibrosis, this is a generic term that does not differentiate between ECM accumulation that occurs in the infarct region to form a scar that is structurally necessary to preserve left ventricle (LV) wall integrity and ECM accumulation that increases LV wall stiffness to exacerbate dilation and stimulate the progression to heart failure. This review focuses on post-MI LV ECM remodeling, targeting the discussion on ECM biomarkers that could be useful for predicting MI outcomes.
Collapse
Affiliation(s)
- Signe Holm Nielsen
- Fibrosis Biology and Biomarkers, Nordic Bioscience, Herlev, Denmark; Disease Systems Immunology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Alan J Mouton
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Kristine Y DeLeon-Pennell
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA; Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS, USA
| | | | - Morten Karsdal
- Fibrosis Biology and Biomarkers, Nordic Bioscience, Herlev, Denmark
| | - Merry L Lindsey
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA; Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS, USA.
| |
Collapse
|
17
|
Jonsson MKB, Hartman RJG, Ackers-Johnson M, Tan WLW, Lim B, van Veen TAB, Foo RS. A Transcriptomic and Epigenomic Comparison of Fetal and Adult Human Cardiac Fibroblasts Reveals Novel Key Transcription Factors in Adult Cardiac Fibroblasts. JACC Basic Transl Sci 2016; 1:590-602. [PMID: 30167544 PMCID: PMC6113540 DOI: 10.1016/j.jacbts.2016.07.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/18/2016] [Accepted: 07/19/2016] [Indexed: 01/01/2023]
Abstract
The interplay between cardiomyocytes and cardiac fibroblasts is increasingly being recognized as important in cardiac disease. Fetal and adult cardiac fibroblasts influence their neighboring cardiomyocytes in different ways. A genome-wide comparison of the 2 reveals that they share >80% of gene transcripts. Motif analysis of empirical regulatory elements located next to differentially expressed genes led to identification of key differential regulators of fibroblast identity. STAT1 and PLAGL1 were identified and validated as key transcription factors to maintain the adult cardiac fibroblast phenotype. Loss of either factor led to a significant change in phenotype, including smaller cell size, apoptosis, reduced turnover, and down-regulated collagen gene expression.
Cardiovascular disease remains the number one global cause of death and presents as multiple phenotypes in which the interplay between cardiomyocytes and cardiac fibroblasts (CFs) has become increasingly highlighted. Fetal and adult CFs influence neighboring cardiomyocytes in different ways. Thus far, a detailed comparison between the two is lacking. Using a genome-wide approach, we identified and validated 2 crucial players for maintaining the adult primary human CF phenotype. Knockdown of these factors induced significant phenotypical changes, including senescence and reduced collagen gene expression. These may now represent novel therapeutic targets against deleterious functions of CFs in adult cardiovascular disease.
Collapse
Key Words
- ATAC, assay for transposase accessible chromatin
- ATAC-seq, assay for transposase accessible chromatin–sequencing
- CF, cardiac fibroblast
- CM, cardiomyocyte
- ChIP-seq, chromatin immunoprecipitation–sequencing
- ECM, extracellular matrix
- EMT, epithelial-to-mesenchymal transformation
- FGF, fibroblast growth factor
- HCF, human cardiac fibroblast
- IL, interleukin
- IPA, Ingenuity Pathway Analysis
- RNA-seq, ribonucleic acid–sequencing
- RT-qPCR, reverse transcription–quantitative polymerase chain reaction
- TF, transcription factor
- aHCF, adult human cardiac fibroblast
- cardiac fibroblasts
- collagen
- fHCF, fetal human cardiac fibroblast
- histone methylation
- transcriptome
Collapse
Affiliation(s)
| | - Robin J G Hartman
- Human Genetics, Genome Institute of Singapore, Singapore.,University of Utrecht, Utrecht, the Netherlands
| | - Matthew Ackers-Johnson
- Human Genetics, Genome Institute of Singapore, Singapore.,Cardiovascular Research Institute, National University Health Systems, National University of Singapore, Singapore
| | - Wilson L W Tan
- Human Genetics, Genome Institute of Singapore, Singapore.,Cardiovascular Research Institute, National University Health Systems, National University of Singapore, Singapore
| | - Bing Lim
- Merck Research Laboratories, Translational Medicine Research Centre, Singapore.,Cancer Stem Cell Biology, Genome Institute of Singapore, Singapore
| | - Toon A B van Veen
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Roger S Foo
- Human Genetics, Genome Institute of Singapore, Singapore.,Cardiovascular Research Institute, National University Health Systems, National University of Singapore, Singapore
| |
Collapse
|
18
|
Barallobre-Barreiro J, Lynch M, Yin X, Mayr M. Systems biology-opportunities and challenges: the application of proteomics to study the cardiovascular extracellular matrix. Cardiovasc Res 2016; 112:626-636. [PMID: 27635058 PMCID: PMC5157133 DOI: 10.1093/cvr/cvw206] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 08/31/2016] [Accepted: 09/09/2016] [Indexed: 12/29/2022] Open
Abstract
Systems biology approaches including proteomics are becoming more widely used in cardiovascular research. In this review article, we focus on the application of proteomics to the cardiac extracellular matrix (ECM). ECM remodelling is a hallmark of many cardiovascular diseases. Proteomic techniques using mass spectrometry (MS) provide a platform for the comprehensive analysis of ECM proteins without a priori assumptions. Proteomics overcomes various constraints inherent to conventional antibody detection. On the other hand, studies that use whole tissue lysates for proteomic analysis mask the identification of the less abundant ECM constituents. In this review, we first discuss decellularization-based methods that enrich for ECM proteins in cardiac tissue, and how targeted MS allows for accurate protein quantification. The second part of the review will focus on post-translational modifications including hydroxylation and glycosylation and on the release of matrix fragments with biological activity (matrikines), all of which can be interrogated by proteomic techniques.
Collapse
Affiliation(s)
| | - Marc Lynch
- King's British Heart Foundation Centre, King's College London, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Xiaoke Yin
- King's British Heart Foundation Centre, King's College London, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Manuel Mayr
- King's British Heart Foundation Centre, King's College London, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
19
|
Gilbert A, Wyczalkowska-Tomasik A, Zendzian-Piotrowska M, Czarkowska-Paczek B. Training differentially regulates elastin level and proteolysis in skeletal and heart muscles and aorta in healthy rats. Biol Open 2016; 5:556-62. [PMID: 27069251 PMCID: PMC4874357 DOI: 10.1242/bio.017459] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Exercise induces changes in muscle fibers and the extracellular matrix that may depend on elastin content and the activity of proteolytic enzymes. We investigated the influence of endurance training on the gene expression and protein content and/or activity of elastin, elastase, cathepsin K, and plasmin in skeletal and heart muscles and in the aorta. Healthy rats were randomly divided into untrained (n=10) and trained (n=10; 6 weeks of endurance training with increasing load) groups. Gene expression was evaluated via qRT-PCR. Elastin content was measured via enzyme-linked immunosorbent assay and enzyme activity was measured fluorometrically. Elastin content was significantly higher in skeletal (P=0.0014) and heart muscle (P=0.000022) from trained rats versus untrained rats, but not in the aorta. Although mRNA levels in skeletal muscle did not differ between groups, the activities of elastase (P=0.0434), cathepsin K (P=0.0343) and plasmin (P=0.000046) were higher in trained rats. The levels of cathepsin K (P=0.0288) and plasminogen (P=0.0005) mRNA were higher in heart muscle from trained rats, but enzyme activity was not. Enzyme activity in the aorta did not differ between groups. Increased elastin content in muscles may result in better adaption to exercise, as may remodeling of the extracellular matrix in skeletal muscle. Summary: Endurance training increases elastin content in muscles but not in the aorta. The activities of enzymes responsible for ECM remodeling increase only in skeletal muscle. These changes seem to be adaptive.
Collapse
Affiliation(s)
- Anna Gilbert
- Department of Clinical Nursing, Medical University of Warsaw, E. Ciolka Street 27, 01-445 Warsaw, Poland
| | - Aleksandra Wyczalkowska-Tomasik
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Nowogrodzka Street 59, 02-006 Warsaw, Poland
| | | | - Bozena Czarkowska-Paczek
- Department of Clinical Nursing, Medical University of Warsaw, E. Ciolka Street 27, 01-445 Warsaw, Poland
| |
Collapse
|
20
|
Andreu I, Luque T, Sancho A, Pelacho B, Iglesias-García O, Melo E, Farré R, Prósper F, Elizalde MR, Navajas D. Heterogeneous micromechanical properties of the extracellular matrix in healthy and infarcted hearts. Acta Biomater 2014; 10:3235-42. [PMID: 24717359 DOI: 10.1016/j.actbio.2014.03.034] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 03/07/2014] [Accepted: 03/31/2014] [Indexed: 10/25/2022]
Abstract
Infarcted hearts are macroscopically stiffer than healthy organs. Nevertheless, although cell behavior is mediated by the physical features of the cell niche, the intrinsic micromechanical properties of healthy and infarcted heart extracellular matrix (ECM) remain poorly characterized. Using atomic force microscopy, we studied ECM micromechanics of different histological regions of the left ventricle wall of healthy and infarcted mice. Hearts excised from healthy (n=8) and infarcted mice (n=8) were decellularized with sodium dodecyl sulfate and cut into 12 μm thick slices. Healthy ventricular ECM revealed marked mechanical heterogeneity across histological regions of the ventricular wall with the effective Young's modulus ranging from 30.2 ± 2.8 to 74.5 ± 8.7 kPa in collagen- and elastin-rich regions of the myocardium, respectively. Infarcted ECM showed a predominant collagen composition and was 3-fold stiffer than collagen-rich regions of the healthy myocardium. ECM of both healthy and infarcted hearts exhibited a solid-like viscoelastic behavior that conforms to two power-law rheology. Knowledge of intrinsic micromechanical properties of the ECM at the length scale at which cells sense their environment will provide further insight into the cell-scaffold interplay in healthy and infarcted hearts.
Collapse
|
21
|
Gishto A, Farrell K, Kothapalli CR. Tuning composition and architecture of biomimetic scaffolds for enhanced matrix synthesis by murine cardiomyocytes. J Biomed Mater Res A 2014; 103:693-708. [PMID: 24798055 DOI: 10.1002/jbm.a.35217] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 04/24/2014] [Accepted: 05/02/2014] [Indexed: 12/31/2022]
Abstract
A major onset of heart failure is myocardial infarction, which causes the myocardium to lose cardiomyocytes and transform into a scar tissue. Since mammalian infarcted cardiac tissue has a limited ability to regenerate, alternative strategies including implantation of tissue-engineered scaffolds at the site of damaged myocardium have been explored. The goal is to enable in situ cardiac reconstruction at the injured myocardium site, replace the lost cardiomyocytes, deliver the required biomolecules, and remodel the extracellular matrix (ECM). ECM synthesis and deposition by cardiomyocytes within such scaffolds remains categorically unexplored. Here, we investigated the survival, ECM synthesis and deposition, and matrix metalloproteinases (MMPs) release by cardiomyocytes within three-dimensional (3D) substrates. Rat cardiomyocytes were cultured for three weeks within two structurally different substrates: 3D collagen hydrogels or polycaprolactone (PCL) nanofibrous scaffolds. The concentration and composition of the hydrogels was varied, while PCL nanofibers were surface-modified with various ECM proteins. Results showed that myocyte attachment and survival was higher within collagen hydrogels, while myocyte alignment and beating was noted only within PCL scaffolds. Total protein synthesis by myocytes within PCL scaffolds was significantly higher compared to that within collagen hydrogels, although more protein was deposited as matrix within hydrogels. Significant ECM synthesis and matrix deposition, TIMP-1, and MMP release were noted within modified collagen hydrogels and PCL nanofiber scaffolds. These results were qualitatively confirmed by imaging techniques. Results attest to the prominent role of scaffold composition and architecture in influencing cardiomyocyte phenotype, matrix synthesis and cytokines release, with significant applications in cardiac tissue remodeling strategies.
Collapse
Affiliation(s)
- Arsela Gishto
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, Ohio, 44115
| | | | | |
Collapse
|
22
|
Wildgruber M, Bielicki I, Aichler M, Kosanke K, Feuchtinger A, Settles M, Onthank DC, Cesati RR, Robinson SP, Huber AM, Rummeny EJ, Walch AK, Botnar RM. Assessment of myocardial infarction and postinfarction scar remodeling with an elastin-specific magnetic resonance agent. Circ Cardiovasc Imaging 2013; 7:321-9. [PMID: 24363356 DOI: 10.1161/circimaging.113.001270] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND To prospectively evaluate an elastin-specific MR contrast agent (ESMA) for in vivo targeting of elastic fibers in myocardial infarction (MI) and postinfarction scar remodeling. METHODS AND RESULTS MI was induced in C57BL/6J mice (n=40) by permanent ligation of the left anterior descending coronary artery. MRI was performed at 7 and 21 days after MI. The merits of gadolinium-based ESMA (Gd-ESMA) were compared with gadopentetic acid (Gd-DTPA) for infarct size determination, contrast-to-noise ratio (CNR), and enhancement kinetics. Specific binding in vivo was evaluated by blocking the molecular target using nonparamagnetic lanthanum-ESMA. In vivo imaging results were confirmed by postmortem triphenyltetrazolium chloride staining, elastica van Gieson staining, and Western blotting. Delayed enhancement MRI revealed prolonged enhancement of Gd-ESMA in the postischemic scar compared with Gd-DTPA. Infarct size measurements showed good agreement between Gd-ESMA and Gd-DTPA and were confirmed by ex vivo triphenyltetrazolium chloride staining. Preinjection of the blocking lanthanum-ESMA resulted in significantly lower CNR of Gd-ESMA at the infarct site (P=0.0019). Although no significant differences in CNR were observed between delayed enhancement imaging and Gd-DTPA between days 7 and 21 (1.8± versus 3.8; P=ns), Gd-ESMA showed markedly higher CNR on day 21 after MI (14.1 versus 4.9; P=0.0032), which correlated with increased synthesis of tropoelastin detected by Western blot analysis and histology. Higher CNR values for Gd-ESMA further correlated with improved ejection fraction of the mice on day 21 after MI. CONCLUSIONS Gd-ESMA enables targeting of elastin within the infarct scar in a mouse model of MI. The imaging properties of Gd-ESMA allow quantification of intrascar elastin content in vivo and thereby provide potential for noninvasive characterization of postinfarction scar remodeling.
Collapse
Affiliation(s)
- Moritz Wildgruber
- Department of Diagnostic and Interventional Radiology, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Bogatyryov Y, Tomanek RJ, Dedkov EI. Structural composition of myocardial infarction scar in middle-aged male and female rats: does sex matter? J Histochem Cytochem 2013; 61:833-48. [PMID: 23867842 DOI: 10.1369/0022155413499794] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The present study was designed to determine whether the structural composition of the scar in middle-aged post-myocardial infraction (MI) rats is affected by the biological sex of the animals. A large MI was induced in 12-month-old male (M-MI) and female (F-MI) Sprague-Dawley rats by ligation of the left coronary artery. Four weeks after the MI, rats with transmural infarctions, greater than 50% of the left ventricular (LV) free wall, were evaluated. The extent of LV remodeling and fractional volumes of fibrillar collagen (FC), myofibroblasts, vascular smooth muscle (SM) cells, and surviving cardiac myocytes (CM) in the scars were compared between the two sexes. The left ventricle of post-MI male and female rats underwent a similar degree of remodeling as evidenced by the analogous scar thinning ratio (0.46 ± 0.02 vs. 0.42 ± 0.05) and infarct expansion index (1.06 ± 0.07 vs. 1.12 ± 0.08), respectively. Most important, the contents of major structural components of the scar revealed no evident difference between M-MI and F-MI rats (interstitial FC, 80.74 ± 2.08 vs. 82.57 ± 4.53; myofibroblasts, 9.59 ± 1.68 vs.9.56 ± 1.15; vascular SM cells, 2.27 ± 0.51 vs. 3.38 ± 0.47; and surviving CM, 3.26 ± 0.39 vs. 3.05 ± 0.38, respectively). Our data are the first to demonstrate that biological sex does not influence the structural composition of a mature scar in middle-aged post-MI rats.
Collapse
Affiliation(s)
- Yevgen Bogatyryov
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York (YB,EID)
| | | | | |
Collapse
|
24
|
Li SH, Sun Z, Guo L, Han M, Wood MFG, Ghosh N, Vitkin IA, Weisel RD, Li RK. Elastin overexpression by cell-based gene therapy preserves matrix and prevents cardiac dilation. J Cell Mol Med 2013; 16:2429-39. [PMID: 22435995 PMCID: PMC3823437 DOI: 10.1111/j.1582-4934.2012.01560.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
After a myocardial infarction, thinning and expansion of the fibrotic scar contribute to progressive heart failure. The loss of elastin is a major contributor to adverse extracellular matrix remodelling of the infarcted heart, and restoration of the elastic properties of the infarct region can prevent ventricular dysfunction. We implanted cells genetically modified to overexpress elastin to re-establish the elastic properties of the infarcted myocardium and prevent cardiac failure. A full-length human elastin cDNA was cloned, subcloned into an adenoviral vector and then transduced into rat bone marrow stromal cells (BMSCs). In vitro studies showed that BMSCs expressed the elastin protein, which was deposited into the extracellular matrix. Transduced BMSCs were injected into the infarcted myocardium of adult rats. Control groups received either BMSCs transduced with the green fluorescent protein gene or medium alone. Elastin deposition in the infarcted myocardium was associated with preservation of myocardial tissue structural integrity (by birefringence of polarized light; P < 0.05 versus controls). As a result, infarct scar thickness and diastolic compliance were maintained and infarct expansion was prevented (P < 0.05 versus controls). Over a 9-week period, rats implanted with BMSCs demonstrated better cardiac function than medium controls; however, rats receiving BMSCs overexpressing elastin showed the greatest functional improvement (P < 0.01). Overexpression of elastin in the infarcted heart preserved the elastic structure of the extracellular matrix, which, in turn, preserved diastolic function, prevented ventricular dilation and preserved cardiac function. This cell-based gene therapy provides a new approach to cardiac regeneration.
Collapse
Affiliation(s)
- Shu-Hong Li
- Division of Cardiovascular Surgery and Toronto General Research Institute, University Health Network, University of Toronto, Toronto, ON, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Transplantation of elastin-secreting myoblast sheets improves cardiac function in infarcted rat heart. Mol Cell Biochem 2012; 368:203-14. [PMID: 22718481 DOI: 10.1007/s11010-012-1361-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 06/07/2012] [Indexed: 10/28/2022]
Abstract
Myoblast sheet transplantation for cardiac failure is a promising therapy to enhance cardiac function via paracrine mechanism. However, their efficacies of treatment showed a gradual decline. The gene modification of the implanted myoblast is important in improving the long-term results of the treatment. Elastin fiber enhances the extensibility of the infarcted wall and can prevent left ventricular dilation. We therefore hypothesized that the elastin gene modification of the implanted myoblast could strengthen and maintain the long-term improvement effects of cardiac function. In this study, we evaluated long-term follow-up benefits of functional myoblast sheets that secrete elastin in an infarcted model. The animal models were divided into three groups: a group transplanted with nontransfected, wild-type, skeletal myoblast-type sheets (WT-rSkM); group transplanted with myoblast sheets that secreted elastin fragments (ELN-rSkM); and a control group (ligation only). Cardiac function was examined by echocardiography, and cardiac remodeling after infarction was evaluated by histological examination. The cardiac function was significantly improved and the left ventricle end-diastolic dimensions were significantly reduced in the ELN-rSkM group. Histological analysis showed that left ventricular remodeling was attenuated in the ELN-rSkM group and that elastic fiber was formed in the epicardial area of ELN-rSkM group. The functionalization of myoblast sheet by elastin gene transfer showed the long-term improvement of cardiac function. Expressed recombinant elastin fiber prevented the dilation of the left ventricular chamber after myocardial infarction. The functional myoblast sheet transplantation maintained the treatment effect by the paracrine effect of myoblast and the formed recombinant elastin.
Collapse
|
26
|
Lichtenauer M, Mildner M, Baumgartner A, Hasun M, Werba G, Beer L, Altmann P, Roth G, Gyöngyösi M, Podesser BK, Ankersmit HJ. Intravenous and intramyocardial injection of apoptotic white blood cell suspensions prevents ventricular remodelling by increasing elastin expression in cardiac scar tissue after myocardial infarction. Basic Res Cardiol 2011; 106:645-55. [PMID: 21416207 PMCID: PMC3105227 DOI: 10.1007/s00395-011-0173-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2010] [Revised: 02/17/2011] [Accepted: 03/09/2011] [Indexed: 11/09/2022]
Abstract
Congestive heart failure developing after acute myocardial infarction (AMI) is a major cause of morbidity and mortality. Clinical trials of cell-based therapy after AMI evidenced only a moderate benefit. We could show previously that suspensions of apoptotic peripheral blood mononuclear cells (PBMC) are able to reduce myocardial damage in a rat model of AMI. Here we experimentally examined the biochemical mechanisms involved in preventing ventricular remodelling and preserving cardiac function after AMI. Cell suspensions of apoptotic cells were injected intravenously or intramyocardially after experimental AMI induced by coronary artery ligation in rats. Administration of cell culture medium or viable PBMC served as controls. Immunohistological analysis was performed to analyse the cellular infiltrate in the ischaemic myocardium. Cardiac function was quantified by echocardiography. Planimetry of the infarcted hearts showed a significant reduction of infarction size and an improvement of post AMI remodelling in rats treated with suspensions of apoptotic PBMC (injected either intravenously or intramoycardially). Moreover, these hearts evidenced enhanced homing of macrophages and cells staining positive for c-kit, FLK-1, IGF-I and FGF-2 as compared to controls. A major finding in this study further was that the ratio of elastic and collagenous fibres within the scar tissue was altered in a favourable fashion in rats injected with apoptotic cells. Intravenous or intramyocardial injection of apoptotic cell suspensions results in attenuation of myocardial remodelling after experimental AMI, preserves left ventricular function, increases homing of regenerative cells and alters the composition of cardiac scar tissue. The higher expression of elastic fibres provides passive energy to the cardiac scar tissue and results in prevention of ventricular remodelling.
Collapse
Affiliation(s)
- Michael Lichtenauer
- Department of Thoracic Surgery, Christian Doppler Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Medical University Vienna, Währinger Gürtel 18-20, Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Holladay CA, O'Brien T, Pandit A. Non-viral gene therapy for myocardial engineering. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2010; 2:232-48. [PMID: 20063367 DOI: 10.1002/wnan.60] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Despite significant advances in surgical and pharmacological techniques, myocardial infarction (MI) remains the main cause of morbidity in the developed world because no remedy has been found for the regeneration of infarcted myocardium. Once the blood supply to the area in question is interrupted, the inflammatory cascade, among other mechanisms, results in the damaged tissue becoming a scar. The goals of cardiac gene therapy are essentially to minimize damage, to promote regeneration, or some combination thereof. While the vector is, in theory, less important than the gene being delivered, the choice of vector can have a significant impact. Viral therapies can have very high transfection efficiencies, but disadvantages include immunogenicity, retroviral-mediated insertional mutagenesis, and the expense and difficulty of manufacture. For these reasons, researchers have focused on non-viral gene therapy as an alternative. In this review, naked plasmid delivery, or the delivery of complexed plasmids, and cell-mediated gene delivery to the myocardium will be reviewed. Pre-clinical and clinical trials in the cardiac tissue will form the core of the discussion. While unmodified stem cells are sometimes considered therapeutic vectors on the basis of paracrine mechanisms of action basic understanding is limited. Thus, only genetically modified cells will be discussed as cell-mediated gene therapy.
Collapse
Affiliation(s)
- Carolyn A Holladay
- Network of Excellence for Functional Biomaterials, National University of Ireland, Galway, Ireland
| | | | | |
Collapse
|
28
|
Akima T, Nakanishi K, Suzuki K, Katayama M, Ohsuzu F, Kawai T. Soluble elastin decreases in the progress of atheroma formation in human aorta. Circ J 2009; 73:2154-62. [PMID: 19755752 DOI: 10.1253/circj.cj-09-0104] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND The serum levels of soluble elastin increase in patients with aortic dissection, but its distribution and characteristics are unclear. METHODS AND RESULTS The 173 aortic specimens were categorized into 4 groups under microscopy (non-atherosclerotic aorta, n=13; fiber-rich plaque, n=77; lipid-rich plaque, n=66; ruptured plaque, n=17). Soluble elastin was abundant within the intima of both the non-atherosclerotic aorta and fiber-rich plaque, rather than in the media, and was decreased within the intima of lipid-rich and ruptured plaques. Soluble elastin levels decreased with progress of atherosclerosis (6.0 +/-0.3 microg/mg protein in non-atherosclerotic aorta; 5.8 +/-0.2 microg/mg protein in fiber-rich plaque; 4.9 +/-0.2 microg/mg protein in lipid-rich plaque; 2.8 +/-0.4 microg/mg protein in ruptured plaque, P<0.05). As well, both matrix metalloprotease-9 activity and elastin mRNA expression showed inverse distribution against soluble elastin (r=0.437, P<0.0001; r=0.186, P<0.05, respectively). Multivariable analysis revealed a decrease in the level of soluble elastin in ruptured plaque (2.8 +/-0.4 microg/mg protein in ruptured plaque, n=18; 5.5 +/-0.2 microg/mg protein in non-ruptured plaque, n=155, P<0.01). Furthermore, western blot showed soluble elastin consists of heterogeneous molecular pattern proteins. CONCLUSIONS Both the synthesis and degradation of elastin may be enhanced in active atherosclerotic lesions.
Collapse
Affiliation(s)
- Takashi Akima
- Department of Pathology and Laboratory Medicine, National Defense Medical College, Tokorozawa, Japan
| | | | | | | | | | | |
Collapse
|
29
|
Antonicelli F, Bellon G, Lorimier S, Hornebeck W. Role of the elastin receptor complex (S-Gal/Cath-A/Neu-1) in skin repair and regeneration. Wound Repair Regen 2009; 17:631-8. [DOI: 10.1111/j.1524-475x.2009.00525.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
30
|
Abramowitch SD, Feola A, Jallah Z, Moalli PA. Tissue mechanics, animal models, and pelvic organ prolapse: a review. Eur J Obstet Gynecol Reprod Biol 2009; 144 Suppl 1:S146-58. [PMID: 19285776 DOI: 10.1016/j.ejogrb.2009.02.022] [Citation(s) in RCA: 152] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pelvic floor disorders such as pelvic organ prolapse, urinary incontinence, and fecal incontinence affect a large number of women each year. The pelvic floor can be thought of as a biomechanical structure due to the complex interaction between the vagina and its supportive structures that are designed to withstand the downward descent of the pelvic organs in response to increases in abdominal pressure. Although previous work has highlighted the biochemical changes that are associated with specific risk factors (i.e. parity, menopause, and genetics), little work has been done to understand the biomechanical changes that occur within the vagina and its supportive structures to prevent the onset of these pelvic floor disorders. Human studies are often limited due to the challenges of obtaining large tissue samples and ethical concerns. Therefore, it is necessary to investigate the use of animal models and their importance in understanding how different risk factors affect the biomechanical properties of the vagina and its supportive structures. In this review paper, we will discuss the different animal models that have been previously used to characterize the biomechanical properties of the vagina: including non-human primates, rodents, rabbits, and sheep. The anatomy and preliminary biomechanical findings are discussed along with the importance of considering experimental conditions, tissue anisotropy, and viscoelasticity when characterizing the biomechanical properties of vaginal tissue. Although there is not a lot of biomechanics research related to the vagina and pelvic floor, the future is exciting due to the significant potential for scientific findings that will improve our understanding of these conditions and hopefully lead to improvements in the prevention and treatment of pelvic disorders.
Collapse
Affiliation(s)
- Steven D Abramowitch
- Department of Bioengineering, Musculoskeletal Research Center, University of Pittsburgh, PA 15219, USA.
| | | | | | | |
Collapse
|
31
|
Farahmand P, Lai TYY, Weisel RD, Fazel S, Yau T, Menasche P, Li RK. Skeletal myoblasts preserve remote matrix architecture and global function when implanted early or late after coronary ligation into infarcted or remote myocardium. Circulation 2008; 118:S130-7. [PMID: 18824744 DOI: 10.1161/circulationaha.107.757617] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND The inability of skeletal myoblasts to transdifferentiate into cardiomyocytes suggests that their beneficial effects on cardiac function after a myocardial infarction are mediated by paracrine effects. We evaluated the roles of these factors in the preservation of matrix architecture (in the infarct and remote regions) by varying the timing (postmyocardial infarction) and delivery site of the implanted cells. METHODS AND RESULTS Skeletal myoblasts (5x10(6)) or control media were injected into the infarct or noninfarcted myocardium at 5 or 30 days after coronary artery ligation in rats. Function was assessed by echocardiography before transplantation and 14 and 30 days thereafter and with a Millar catheter at 30 days after transplantation. Ventricular geometry, remote fibrillar collagen architecture, and changes in the matrix metalloproteinase-TIMP system were evaluated. Myoblast implantation in both sites and at both times preserved matrix architecture (length and width of collagen fibers) in the remote myocardium (in association with some decreases in remote myocardial matrix metalloprotease activity), improved global cardiac function, and attenuated the progressive increase in end diastolic volume (P<0.05 for all measures compared with medium controls). Cells delivered into the infarct region preserved scar thickness; cells delivered into the noninfarcted myocardium preserved wall thickness. CONCLUSIONS Regardless of whether the cells were injected into the infarct or the noninfarcted myocardium early after an myocardial infarction or later, skeletal myoblasts improved cardiac function by preventing ventricular dilation and preserving matrix architecture in the remote region, likely mediated by paracrine effects.
Collapse
Affiliation(s)
- Patrick Farahmand
- MaRS Centre, Toronto Medical Discovery Tower, Room #3-702, 101 College Street, Toronto, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
32
|
Bunda S, Liu P, Wang Y, Liu K, Hinek A. Aldosterone induces elastin production in cardiac fibroblasts through activation of insulin-like growth factor-I receptors in a mineralocorticoid receptor-independent manner. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 171:809-19. [PMID: 17724138 PMCID: PMC1959490 DOI: 10.2353/ajpath.2007.070101] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Aldosterone is known to regulate electrolyte homeostasis, but it may also contribute to other processes, including the maladaptive remodeling of postinfarct hearts. Because aldosterone has been implicated in the stimulation of collagen production in the heart, we investigated whether it would also affect elastin deposition in cultures of human cardiac fibroblasts. We first demonstrated that treatment with 1 to 50 nmol/L aldosterone leads to a significant increase in collagen type I mRNA levels and in subsequent collagen fiber deposition. Pretreatment of cells with the mineralocorticoid receptor antagonist spironolactone, but not with the glucocorticoid receptor antagonist RU 486, inhibited collagen synthesis in aldosterone-treated cultures. Most importantly, we demonstrated that aldosterone also increases elastin mRNA levels, tropoelastin synthesis, and elastic fiber deposition in a dose-dependent manner. Strikingly, neither spironolactone nor RU 486 eliminated aldosterone-induced increases in elastin production. We further discovered that the proelastogenic effect of aldosterone involves a rapid increase in tyrosine phosphorylation of the insulin-like growth factor-I receptor and that the insulin-like growth factor-I receptor kinase inhibitor AG1024 or an anti-insulin-like growth factor-I receptor-neutralizing antibody inhibits both insulin-like growth factor-I and aldosterone-induced elastogenesis. Thus, we have demonstrated for the first time that aldosterone, which stimulates collagen production through the mineralocorticoid receptor-dependent pathway, also increases elastogenesis via a parallel mineralocorticoid receptor-independent pathway involving I insulin-like growth factor-I receptor signaling.
Collapse
Affiliation(s)
- Severa Bunda
- Cardiovascular Research Program, University of Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
33
|
Robinet A, Millart H, Oszust F, Hornebeck W, Bellon G. Binding of elastin peptides to S-Gal protects the heart against ischemia/reperfusion injury by triggering the RISK pathway. FASEB J 2007; 21:1968-78. [PMID: 17341689 DOI: 10.1096/fj.06-6477com] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Elastin peptides (EPs) generated by hydrolysis of elastic fibers by elastinolytic enzymes display a wide spectrum of biological activities. Here, we investigated their influence on rat heart ischemia-mediated injury using the Langendorff ex vivo model. EPs, i.e., kappa elastin, at 1.32- and 660-nM concentrations, when administered before the ischemia period, elicited a beneficial influence against ischemia by accelerating the recovery rate of heart contractile parameters and by decreasing significantly creatine kinase release and heart necrosis area when measured at the onset of the reperfusion. All effects were S-Gal-dependent, as being reproduced by (VGVAPG)3 and as being inhibited by receptor antagonists, such as lactose and V14 peptide (VVGSPSAQDEASPL). EPs interaction with S-Gal triggered NO release and activation of PI3-kinase/Akt and ERK1/2 in human coronary endothelial cells (HCAECs) and rat neonatal cardiomyocytes (RCs). This signaling pathway, as designated as RISK, for reperfusion injury salvage kinase pathway, was shown to be responsible for the beneficial influence of EPs on ischemia/reperfusion injury on the basis of its inhibition by specific pharmacological inhibitors. EPs survival activity was attained at a concentration averaging that present into the blood circulation, supporting the contention that these matrikines might offer a natural protection against cardiac injury in young and adult individuals. Such protective effect might be lost with aging, since we found that hearts from 24-month-old rats did not respond to EPs.
Collapse
Affiliation(s)
- Arnaud Robinet
- Laboratoire de Pharmacologie Médicale, Université de Reims-Champagne-Ardenne, Faculté de Médecine, Reims, France
| | | | | | | | | |
Collapse
|
34
|
Duca L, Blanchevoye C, Cantarelli B, Ghoneim C, Dedieu S, Delacoux F, Hornebeck W, Hinek A, Martiny L, Debelle L. The elastin receptor complex transduces signals through the catalytic activity of its Neu-1 subunit. J Biol Chem 2007; 282:12484-91. [PMID: 17327233 DOI: 10.1074/jbc.m609505200] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The binding of elastin peptides on the elastin receptor complex leads to the formation of intracellular signals but how this is achieved remains totally unknown. Using pharmacological inhibitors of the enzymatic activities of its subunits, we show here that the elastin peptide-driven ERK1/2 activation and subsequent pro-MMP-1 production, observed in skin fibroblasts when they are cultured in the presence of these peptides, rely on a membrane-bound sialidase activity. As lactose blocked this effect, the elastin receptor sialidase subunit, Neu-1, seemed to be involved. The use of a catalytically inactive form of Neu-1 and the small interfering RNA-mediated decrease of Neu-1 expression strongly support this view. Finally, we report that N-acetyl neuraminic acid can reproduce the effects of elastin peptides on both ERK1/2 activation and pro-MMP-1 production. Altogether, our results indicate that the enzymatic activity of the Neu-1 subunit of the elastin receptor complex is responsible for its signal transduction, presumably through sialic acid generation from undetermined substrates.
Collapse
Affiliation(s)
- Laurent Duca
- Laboratoire de Biochimie, UMR CNRS 6198, Faculté des Sciences, Moulin de la Housse, BP 1039, 51687 Reims Cedex 2, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Study of human lung elastin degradation by different elastases using high-performance liquid chromatography/mass spectrometry. Anal Biochem 2006; 358:216-24. [PMID: 16962555 DOI: 10.1016/j.ab.2006.07.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2006] [Revised: 07/06/2006] [Accepted: 07/17/2006] [Indexed: 11/30/2022]
Abstract
Elastin is a structural insoluble protein which gives elasticity to tissues and organs. Although its hydrophobic and highly cross-linked nature makes it a very durable polymer, degradation of elastin in relation with several pathological conditions, such as pulmonary emphysema, has been documented. Since different enzymes may be involved in elastolysis, it is of interest to determine which enzyme is responsible for the degradative effects observed in a certain disease. The aim of this work was to study elastin degradation by proteases from different families (serine, cysteine, and metalloproteases) using liquid chromatography coupled to mass spectrometry to characterize the elastin-derived peptides. Incubation of insoluble human elastin with different elastases revealed that, indeed, each protease degrades elastin in a preferential way giving rise to specific peptide patterns. This opens the possibility of using a given set of peptides as biomarkers for disease-related elastolysis.
Collapse
|
36
|
Abstract
Proteomics was established 10 years ago by the analysis of microbial genomes via their protein complement or proteome. Bionics is an ancient art, which converts structures optimized by nature into advanced technical products. Previously, we analyzed survival modalities in nanobacteria and converted the interplay between survival-oriented protein functions and nanoscale mineral shells into models for advanced drug delivery. Exploiting protein functions observed in nature to design biomedical products and therapies could be named proteobionics. Here, we present examples for this new branch of nanoproteomics.
Collapse
Affiliation(s)
- Andrei P Sommer
- Central Institute of Biomedical Engineering, University of Ulm, 89081 Ulm, Germany.
| | | |
Collapse
|