1
|
Saraste A, Ståhle M, Roivainen A, Knuuti J. Molecular Imaging of Heart Failure: An Update and Future Trends. Semin Nucl Med 2024; 54:674-685. [PMID: 38609753 DOI: 10.1053/j.semnuclmed.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 03/19/2024] [Indexed: 04/14/2024]
Abstract
Molecular imaging can detect and quantify pathophysiological processes underlying heart failure, complementing evaluation of cardiac structure and function with other imaging modalities. Targeted tracers have enabled assessment of various cellular and subcellular mechanisms of heart failure aiming for improved phenotyping, risk stratification, and personalized therapy. This review outlines the current status of molecular imaging in heart failure, accompanied with discussion on novel developments. The focus is on radionuclide methods with data from clinical studies. Imaging of myocardial metabolism can identify left ventricle dysfunction caused by myocardial ischemia that may be reversible after revascularization in the presence of viable myocardium. In vivo imaging of active inflammation and amyloid deposition have an established role in the detection of cardiac sarcoidosis and transthyretin amyloidosis. Innervation imaging has well documented prognostic value in predicting heart failure progression and arrhythmias. Tracers specific for inflammation, angiogenesis and myocardial fibrotic activity are in earlier stages of development, but have demonstrated potential value in early characterization of the response to myocardial injury and prediction of cardiac function over time. Early detection of disease activity is a key for transition from medical treatment of clinically overt heart failure towards a personalized approach aimed at supporting repair and preventing progressive cardiac dysfunction.
Collapse
Affiliation(s)
- Antti Saraste
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland; Heart Center, Turku University Hospital and University of Turku, Turku, Finland.
| | - Mia Ståhle
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| | - Anne Roivainen
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| | - Juhani Knuuti
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| |
Collapse
|
2
|
Lv J, Fu Z, Zheng H, Song Q. Global research trends and emerging opportunities for integrin adhesion complexes in cardiac repair: a scientometric analysis. Front Cardiovasc Med 2024; 11:1308763. [PMID: 38699584 PMCID: PMC11063371 DOI: 10.3389/fcvm.2024.1308763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 04/04/2024] [Indexed: 05/05/2024] Open
Abstract
Objective Cardiac regenerative medicine has gained significant attention in recent years, and integrins are known to play a critical role in mediating cardiac development and repair, especially after an injury from the myocardial infarction (MI). Given the extensive research history and interdisciplinary nature of this field, a quantitative retrospective analysis and visualization of related topics is necessary. Materials and methods We performed a scientometric analysis of published papers on cardiac integrin adhesion complexes (IACs), including analysis of annual publications, disciplinary evolution, keyword co-occurrence, and literature co-citation. Results A total of 2,664 publications were finally included in the past 20 years. The United States is the largest contributor to the study and is leading this area of research globally. The journal Circulation Research attracts the largest number of high-quality publications. The study of IACs in cardiac repair/regenerative therapies involves multiple disciplines, particularly in materials science and developmental biology. Keywords of research frontiers were represented by Tenasin-C (2019-2023) and inflammation (2020-2023). Conclusion Integrins are topics with ongoing enthusiasm in biological development and tissue regeneration. The rapidly emerging role of matricellular proteins and non-protein components of the extracellular matrix (ECM) in regulating matrix structure and function may be a further breakthrough point in the future; the emerging role of IACs and their downstream molecular signaling in cardiac repair are also of great interest, such as induction of cardiac proliferation, differentiation, maturation, and metabolism, fibroblast activation, and inflammatory modulation.
Collapse
Affiliation(s)
- Jiayu Lv
- Department of General Internal Medicine, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhenyue Fu
- Department of General Internal Medicine, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- College of Traditional Chinese Medicine, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Haoran Zheng
- Department of General Internal Medicine, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- College of Traditional Chinese Medicine, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Qingqiao Song
- Department of General Internal Medicine, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
3
|
Chen YW, Cheng PP, Yin YF, Cai H, Chen JZ, Feng MH, Guo W, Zhao P, Zhang C, Shan XL, Chen HH, Guo S, Lu Y, Xu M. Integrin αV mediated activation of myofibroblast via mechanoparacrine of transforming growth factor β1 in promoting fibrous scar formation after myocardial infarction. Biochem Biophys Res Commun 2024; 692:149360. [PMID: 38081108 DOI: 10.1016/j.bbrc.2023.149360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/17/2023] [Accepted: 12/04/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND Myocardial infarction (MI) dramatically changes the mechanical stress, which is intensified by the fibrotic remodeling. Integrins, especially the αV subunit, mediate mechanical signal and mechanoparacrine of transforming growth factor β1 (TGF-β1) in various organ fibrosis by activating CFs into myofibroblasts (MFBs). We investigated a possible role of integrin αV mediated mechanoparacrine of TGF-β1 in MFBs activation for fibrous reparation in mice with MI. METHODS Heart samples from MI, sham, or MI plus cilengitide (14 mg/kg, specific integrin αV inhibitor) treated mice, underwent functional and morphological assessments by echocardiography, and histochemistry on 7, 14 and 28 days post-surgery. The mechanical and ultrastructural changes of the fibrous scar were further evaluated by atomic mechanics microscope (AFM), immunofluorescence, second harmonic generation (SHG) imaging, polarized light and scanning electron microscope, respectively. Hydroxyproline assay was used for total collagen content, and western blot for protein expression profile examination. Fibroblast bioactivities, including cell shape, number, Smad2/3 signal and expression of extracellular matrix (ECM) related proteins, were further evaluated by microscopic observation and immunofluorescence in polyacrylamide (PA) hydrogel with adjustable stiffness, which was re-explored in fibroblast cultured on stiff matrix after silencing of integrin αV. The content of total and free TGF-β1 was tested by enzyme-linked immunosorbent assay (ELISA) in both infarcted tissue and cell samples. RESULT Increased stiffness with heterogeneity synchronized with integrin αV and alpha smooth muscle actin (α-SMA) positive MFBs accumulation in those less mature fibrous areas. Cilengitide abruptly reduced collagen content and disrupted collagen alignment, which also decreased TGF-β1 bioavailability, Smad2/3 phosphorylation, and α-SMA expression in the fibrous area. Accordingly, fibroblast on stiff but not soft matrix exhibited obvious MFB phenotype, as evidenced by enlarged cell, hyperproliferation, well-developed α-SMA fibers, and elevated ECM related proteins, while silencing of integrin αV almost abolished this switch via attenuating paracrine of TGF-β1 and nuclear translocation of Smad2/3. CONCLUSION This study illustrated that increased tissue stiffness activates CFs into MFBs by integrin αV mediated mechanoparacrine of TGF-β1, especially in immature scar area, which ultimately promotes fibrous scar maturation.
Collapse
Affiliation(s)
- Yu-Wen Chen
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Pei-Pei Cheng
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuan-Feng Yin
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
| | - Hong Cai
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
| | - Jing-Zhi Chen
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ming-Hui Feng
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Guo
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Pei Zhao
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chen Zhang
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-Li Shan
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hui-Hua Chen
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shuo Guo
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yi Lu
- Minhang Hospital, Fu Dan University, Shanghai, China.
| | - Ming Xu
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
4
|
Nammas W, Paunonen C, Teuho J, Siekkinen R, Luoto P, Käkelä M, Hietanen A, Viljanen T, Dietz M, Prior JO, Li XG, Roivainen A, Knuuti J, Saraste A. Imaging of Myocardial α vβ 3 Integrin Expression for Evaluation of Myocardial Injury After Acute Myocardial Infarction. J Nucl Med 2024; 65:132-138. [PMID: 37973184 DOI: 10.2967/jnumed.123.266148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/27/2023] [Indexed: 11/19/2023] Open
Abstract
[68Ga]Ga-NODAGA-Arg-Gly-Asp (RGD) is a PET tracer targeting αvβ3 integrin, which is upregulated during angiogenesis soon after acute myocardial infarction (AMI). We prospectively evaluated determinants of myocardial uptake of [68Ga]Ga-NODAGA-RGD and its associations with left ventricular (LV) function in patients after AMI. Methods: Myocardial blood flow and [68Ga]Ga-NODAGA-RGD uptake (60 min after injection) were evaluated by PET in 31 patients 7.7 ± 3.8 d after primary percutaneous coronary intervention for ST-elevation AMI. Transthoracic echocardiography of LV function was performed on the day of PET and at the 6-mo follow-up. Results: PET images showed increased uptake of [68Ga]Ga-NODAGA-RGD in the ischemic area at risk (AAR), predominantly in injured myocardial segments. The SUV in the segment with the highest uptake (SUVmax) in the ischemic AAR was higher than the SUVmean of the remote myocardium (0.73 ± 0.16 vs. 0.51 ± 0.11, P < 0.001). Multivariable predictors of [68Ga]Ga-NODAGA-RGD uptake in the AAR included high peak N-terminal pro-B-type natriuretic peptide (P < 0.001), low LV ejection fraction, low global longitudinal strain (P = 0.01), and low longitudinal strain in the AAR (P = 0.01). [68Ga]Ga-NODAGA-RGD uptake corrected for myocardial blood flow and perfusable tissue fraction in the AAR predicted improvement in global longitudinal strain at follow-up (P = 0.002), independent of peak troponin, N-terminal pro-B-type natriuretic peptide, and LV ejection fraction. Conclusion: [68Ga]Ga-NODAGA-RGD uptake shows increased αvβ3 integrin expression in the ischemic AAR early after AMI that is associated with regional and global systolic dysfunction, as well as increased LV filling pressure. Increased [68Ga]Ga-NODAGA-RGD uptake predicts improvement of global LV function 6 mo after AMI.
Collapse
Affiliation(s)
- Wail Nammas
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
- Heart Center, Turku University Hospital, University of Turku, Turku, Finland
| | - Christian Paunonen
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| | - Jarmo Teuho
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| | - Reetta Siekkinen
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
- Department of Medical Physics, Turku University Hospital, Turku, Finland
| | - Pauliina Luoto
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| | - Meeri Käkelä
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| | - Ari Hietanen
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| | - Tapio Viljanen
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| | - Matthieu Dietz
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| | - John O Prior
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| | - Xiang-Guo Li
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
- Department of Chemistry, University of Turku, Turku, Finland; and
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Anne Roivainen
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Juhani Knuuti
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Antti Saraste
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland;
- Heart Center, Turku University Hospital, University of Turku, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| |
Collapse
|
5
|
Bentsen S, Jensen JK, Christensen E, Petersen LR, Grandjean CE, Follin B, Madsen JS, Christensen C, Clemmensen A, Binderup T, Hasbak P, Ripa RS, Kjaer A. [ 68Ga]Ga-NODAGA-E[(cRGDyK)] 2 angiogenesis PET following myocardial infarction in an experimental rat model predicts cardiac functional parameters and development of heart failure. J Nucl Cardiol 2023; 30:2073-2084. [PMID: 37127725 PMCID: PMC10558373 DOI: 10.1007/s12350-023-03265-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 03/11/2023] [Indexed: 05/03/2023]
Abstract
BACKGROUND Angiogenesis has increasingly been a target for imaging and treatment over the last decade. The integrin αvβ3 is highly expressed in cells during angiogenesis and are therefore a promising target for imaging. In this study, we aimed to investigate the PET tracer [68Ga]Ga-RGD as a marker of angiogenesis following MI and its ability to predict cardiac functional parameters. METHODS First, the real-time interaction between [68Ga]Ga-RGD and integrin αvβ3 was investigated using surface plasmon resonance (SPR). Second, an animal study was performed to investigate the [68Ga]Ga-RGD uptake in the infarcted area after one and four weeks following MI in a rat model (MI = 68, sham surgery = 36). Finally, the specificity of the [68Ga]Ga-RGD tracer was evaluated ex vivo using histology, autoradiography, gamma counting and flow cytometry. RESULTS SPR showed that [68Ga]Ga-RGD has a high affinity for integrin αvβ3, forming a strong and stable binding. PET/CT showed a significantly higher uptake of [68Ga]Ga-RGD in the infarcted area compared to sham one week (p < 0.001) and four weeks (p < 0.001) after MI. The uptake of [68Ga]Ga-RGD after one week correlated to end diastolic volume (r = 0.74, p < 0.001) and ejection fraction (r = - 0.71, p < 0.001) after four weeks. CONCLUSION This study demonstrates that [68Ga]Ga-RGD has a high affinity for integrin αvβ3, which enables the evaluation of angiogenesis and remodeling. The [68Ga]Ga-RGD uptake after one week indicates that [68Ga]Ga-RGD may be used as an early predictor of cardiac functional parameters and possible development of heart failure after MI. These encouraging data supports the clinical translation and future use in MI patients.
Collapse
Affiliation(s)
- Simon Bentsen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Jacob Kildevang Jensen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Esben Christensen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
- Department of Health Technology, Section for Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark, Copenhagen, Denmark
| | - Lars Ringgaard Petersen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
- Department of Health Technology, Section for Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark, Copenhagen, Denmark
| | - Constance Eline Grandjean
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Bjarke Follin
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
- Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Johanne Straarup Madsen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Camilla Christensen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Andreas Clemmensen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Tina Binderup
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Philip Hasbak
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Rasmus Sejersten Ripa
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Zhang Y, Wang Y, Li J, Li C, Liu W, Long X, Wang Z, Zhao R, Ge J, Shi B. ANNEXIN A2 FACILITATES NEOVASCULARIZATION TO PROTECT AGAINST MYOCARDIAL INFARCTION INJURY VIA INTERACTING WITH MACROPHAGE YAP AND ENDOTHELIAL INTEGRIN Β3. Shock 2023; 60:573-584. [PMID: 37832154 DOI: 10.1097/shk.0000000000002198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2023]
Abstract
ABSTRACT Cardiac macrophages with different polarization phenotypes regulate ventricular remodeling and neovascularization after myocardial infarction (MI). Annexin A2 (ANXA2) promotes macrophage polarization to the repair phenotype and regulates neovascularization. However, whether ANXA2 plays any role in post-MI remodeling and its underlying mechanism remains obscure. In this study, we observed that expression levels of ANXA2 were dynamically altered in mouse hearts upon MI and peaked on the second day post-MI. Using adeno-associated virus vector-mediated overexpression or silencing of ANXA2 in the heart, we also found that elevation of ANXA2 in the infarcted myocardium significantly improved cardiac function, reduced cardiac fibrosis, and promoted peri-infarct angiogenesis, compared with controls. By contrast, reduction of cardiac ANXA2 exhibited opposite effects. Furthermore, using in vitro coculture system, we found that ANXA2-engineered macrophages promoted cardiac microvascular endothelial cell (CMEC) proliferation, migration, and neovascularization. Mechanistically, we identified that ANXA2 interacted with yes-associated protein (YAP) in macrophages and skewed them toward pro-angiogenic phenotype by inhibiting YAP activity. In addition, ANXA2 directly interacted with integrin β3 in CMECs and enhanced their growth, migration, and tubule formation. Our results indicate that increased expression of ANXA2 could confer protection against MI-induced injury by promoting neovascularization in the infarcted area, partly through the inhibition of YAP in macrophages and activation of integrin β3 in endothelial cells. Our study provides new therapeutic strategies for the treatment of MI injury.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Yan Wang
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Jiao Li
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Chaofu Li
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Weiwei Liu
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Xianping Long
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Zhenglong Wang
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Ranzun Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Bei Shi
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Guizhou, China
| |
Collapse
|
7
|
Hwang HS, Kahmini AR, Prascak J, Cejas-Carbonell A, Valera IC, Champion S, Corrigan M, Mumbi F, Parvatiyar MS. Sarcospan Deficiency Increases Oxidative Stress and Arrhythmias in Hearts after Acute Ischemia-Reperfusion Injury. Int J Mol Sci 2023; 24:11868. [PMID: 37511627 PMCID: PMC10380899 DOI: 10.3390/ijms241411868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/14/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
The protein sarcospan (SSPN) is an integral member of the dystrophin-glycoprotein complex (DGC) and has been shown to be important in the heart during the development and the response to acute stress. In this study, we investigated the role of SSPN in the cardiac response to acute ischemia-reperfusion (IR) injury in SSPN-deficient (SSPN-/-) mice. First, the hemodynamic response of SSPN-/- mice was tested and was similar to SSPN+/+ (wild-type) mice after isoproterenol injection. Using the in situ Langendorff perfusion method, SSPN-/- hearts were subjected to IR injury and found to have increased infarct size and arrhythmia susceptibility compared to SSPN+/+. Ca2+ handling was assessed in single cardiomyocytes and diastolic Ca2+ levels were increased after acute β-AR stimulation in SSPN+/+ but not SSPN-/-. It was also found that SSPN-/- cardiomyocytes had reduced Ca2+ SR content compared to SSPN+/+ but similar SR Ca2+ release. Next, we used qRT-PCR to examine gene expression of Ca2+ handling proteins after acute IR injury. SSPN-/- hearts showed a significant decrease in L-type Ca2+ channels and a significant increase in Ca2+ release channel (RyR2) expression. Interestingly, under oxidizing conditions reminiscent of IR, SSPN-/- cardiomyocytes, had increased H2O2-induced reactive oxygen species production compared to SSPN+/+. Examination of oxidative stress proteins indicated that NADPH oxidase 4 and oxidized CAMKII were increased in SSPN-/- hearts after acute IR injury. These results suggest that increased arrhythmia susceptibility in SSPN-/- hearts post-IR injury may arise from alterations in Ca2+ handling and a reduced capacity to regulate oxidative stress pathways.
Collapse
Affiliation(s)
- Hyun Seok Hwang
- Department of Nutrition and Integrative Physiology, Florida State University, 107 Chieftan Way, Biomedical Research Facility, Tallahassee, FL 32306-1490, USA
| | - Aida Rahimi Kahmini
- Department of Nutrition and Integrative Physiology, Florida State University, 107 Chieftan Way, Biomedical Research Facility, Tallahassee, FL 32306-1490, USA
| | - Julia Prascak
- Department of Nutrition and Integrative Physiology, Florida State University, 107 Chieftan Way, Biomedical Research Facility, Tallahassee, FL 32306-1490, USA
| | - Alexis Cejas-Carbonell
- Department of Nutrition and Integrative Physiology, Florida State University, 107 Chieftan Way, Biomedical Research Facility, Tallahassee, FL 32306-1490, USA
| | - Isela C Valera
- Department of Nutrition and Integrative Physiology, Florida State University, 107 Chieftan Way, Biomedical Research Facility, Tallahassee, FL 32306-1490, USA
| | - Samantha Champion
- Department of Nutrition and Integrative Physiology, Florida State University, 107 Chieftan Way, Biomedical Research Facility, Tallahassee, FL 32306-1490, USA
| | - Mikayla Corrigan
- Department of Nutrition and Integrative Physiology, Florida State University, 107 Chieftan Way, Biomedical Research Facility, Tallahassee, FL 32306-1490, USA
| | - Florence Mumbi
- Department of Nutrition and Integrative Physiology, Florida State University, 107 Chieftan Way, Biomedical Research Facility, Tallahassee, FL 32306-1490, USA
| | - Michelle S Parvatiyar
- Department of Nutrition and Integrative Physiology, Florida State University, 107 Chieftan Way, Biomedical Research Facility, Tallahassee, FL 32306-1490, USA
| |
Collapse
|
8
|
Kim J, Zimmerman MA, Shin WY, Boettcher BT, Lee JS, Park JI, Ali M, Yang M, Mishra J, Hagen CE, McGraw JE, Mathison A, Woehlck HJ, Lomberk G, Camara AKS, Urrutia RA, Stowe DF, Hong JC. Effects of Subnormothermic Regulated Hepatic Reperfusion on Mitochondrial and Transcriptomic Profiles in a Porcine Model. Ann Surg 2023; 277:e366-e375. [PMID: 34387201 PMCID: PMC8840998 DOI: 10.1097/sla.0000000000005156] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE We sought to investigate the biological effects of pre-reperfusion treatments of the liver after warm and cold ischemic injuries in a porcine donation after circulatory death model. SUMMARY OF BACKGROUND DATA Donation after circulatory death represents a severe form of liver ischemia and reperfusion injury that has a profound impact on graft function after liver transplantation. METHODS Twenty donor pig livers underwent 60 minutes of in situ warm ischemia after circulatory arrest and 120 minutes of cold static preservation prior to simulated transplantation using an ex vivo perfusion machine. Four reperfusion treatments were compared: Control-Normothermic (N), Control- Subnormothermic (S), regulated hepatic reperfusion (RHR)-N, and RHR-S (n = 5 each). The biochemical, metabolic, and transcriptomic profiles, as well as mitochondrial function were analyzed. RESULTS Compared to the other groups, RHR-S treated group showed significantly lower post-reperfusion aspartate aminotransferase levels in the reperfusion effluent and histologic findings of hepatocyte viability and lesser degree of congestion and necrosis. RHR-S resulted in a significantly higher mitochondrial respiratory control index and calcium retention capacity. Transcriptomic profile analysis showed that treatment with RHR-S activated cell survival and viability, cellular homeostasis as well as other biological functions involved in tissue repair such as cytoskeleton or cytoplasm organization, cell migration, transcription, and microtubule dynamics. Furthermore, RHR-S inhibited organismal death, morbidity and mortality, necrosis, and apoptosis. CONCLUSION Subnormothermic RHR mitigates IRI and preserves hepatic mitochondrial function after warm and cold hepatic ischemia. This organ resuscitative therapy may also trigger the activation of protective genes against IRI. Sub- normothermic RHR has potential applicability to clinical liver transplantation.
Collapse
Affiliation(s)
- Joohyun Kim
- Division of Transplant Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee WI
- Transplant Center, Froedtert & the Medical College of Wisconsin, and Children's Wisconsin, Milwaukee, WI
| | - Michael A Zimmerman
- Division of Transplant Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee WI
- Transplant Center, Froedtert & the Medical College of Wisconsin, and Children's Wisconsin, Milwaukee, WI
| | - Woo Young Shin
- Department of Surgery, inha University School of Medicine, incheon, South Korea
| | - Brent T Boettcher
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI
| | - Ju-Seog Lee
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jong-In Park
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI
| | - Muhammed Ali
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI
| | - Meiying Yang
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI
| | - Jyotsna Mishra
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI
| | | | - Joseph E McGraw
- Department of Pharmacology, Concordia University, Mequon, WI
| | - Angela Mathison
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI; and
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee WI
| | - Harvey J Woehlck
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI
| | - Gwen Lomberk
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI; and
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee WI
| | - Amadou K S Camara
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI
| | - Raul A Urrutia
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI; and
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee WI
| | - David F Stowe
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI
| | - Johnny C Hong
- Division of Transplant Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee WI
- Transplant Center, Froedtert & the Medical College of Wisconsin, and Children's Wisconsin, Milwaukee, WI
| |
Collapse
|
9
|
Li R, Frangogiannis NG. Integrins in cardiac fibrosis. J Mol Cell Cardiol 2022; 172:1-13. [PMID: 35872324 DOI: 10.1016/j.yjmcc.2022.07.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 07/10/2022] [Accepted: 07/18/2022] [Indexed: 12/14/2022]
Abstract
Cells sense mechanical stress and changes in their matrix environment through the integrins, a family of heterodimeric surface receptors that bind to extracellular matrix ligands and trigger cytoskeletal remodeling, while transducing a wide range of intracellular signals. Integrins have been extensively implicated in regulation of inflammation, repair and fibrosis in many different tissues. This review manuscript discusses the role of integrin-mediated cascades in myocardial fibrosis. In vitro studies have demonstrated that β1 and αv integrins play an important role in fibrogenic conversion of cardiac fibroblast, acting through direct stimulation of FAK/Src cascades, or via accentuation of growth factor signaling. Fibrogenic actions of αv integrins may be mediated, at least in part, through pericellular activation of latent TGF-β stores. In vivo evidence supporting the role of integrin heterodimers in fibrotic cardiac remodeling is limited to associative evidence, and to experiments using pharmacologic inhibitors, or global loss-of-function approaches. Studies documenting in vivo actions of integrins on fibroblasts using cell-specific strategies are lacking. Integrin effects on leukocytes may also contribute to the pathogenesis of fibrotic myocardial responses by mediating recruitment and activation of fibrogenic macrophages. The profile and role of integrins in cardiac fibrosis may be dependent on the underlying pathologic condition. Considering their cell surface localization and the availability of small molecule inhibitors, integrins may be attractive therapeutic targets for patients with heart failure associated with prominent fibrotic remodeling.
Collapse
Affiliation(s)
- Ruoshui Li
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, United States of America.
| |
Collapse
|
10
|
Abdul-Ghani S, Skeffington KL, Kim M, Moscarelli M, Lewis PA, Heesom K, Fiorentino F, Emanueli C, Reeves BC, Punjabi PP, Angelini GD, Suleiman MS. Effect of cardioplegic arrest and reperfusion on left and right ventricular proteome/phosphoproteome in patients undergoing surgery for coronary or aortic valve disease. Int J Mol Med 2022; 49:77. [PMID: 35425992 PMCID: PMC9083849 DOI: 10.3892/ijmm.2022.5133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 02/15/2022] [Indexed: 11/18/2022] Open
Abstract
Our earlier work has shown inter‑disease and intra‑disease differences in the cardiac proteome between right (RV) and left (LV) ventricles of patients with aortic valve stenosis (AVS) or coronary artery disease (CAD). Whether disease remodeling also affects acute changes occuring in the proteome during surgical intervention is unknown. This study investigated the effects of cardioplegic arrest on cardiac proteins/phosphoproteins in LV and RV of CAD (n=6) and AVS (n=6) patients undergoing cardiac surgery. LV and RV biopsies were collected during surgery before ischemic cold blood cardioplegic arrest (pre) and 20 min after reperfusion (post). Tissues were snap frozen, proteins extracted, and the extracts were used for proteomic and phosphoproteomic analysis using Tandem Mass Tag (TMT) analysis. The results were analysed using QuickGO and Ingenuity Pathway Analysis softwares. For each comparision, our proteomic analysis identified more than 3,000 proteins which could be detected in both the pre and Post samples. Cardioplegic arrest and reperfusion were associated with significant differential expression of 24 (LV) and 120 (RV) proteins in the CAD patients, which were linked to mitochondrial function, inflammation and cardiac contraction. By contrast, AVS patients showed differential expression of only 3 LV proteins and 2 RV proteins, despite a significantly longer duration of ischaemic cardioplegic arrest. The relative expression of 41 phosphoproteins was significantly altered in CAD patients, with 18 phosphoproteins showing altered expression in AVS patients. Inflammatory pathways were implicated in the changes in phosphoprotein expression in both groups. Inter‑disease comparison for the same ventricular chamber at both timepoints revealed differences relating to inflammation and adrenergic and calcium signalling. In conclusion, the present study found that ischemic arrest and reperfusion trigger different changes in the proteomes and phosphoproteomes of LV and RV of CAD and AVS patients undergoing surgery, with markedly more changes in CAD patients despite a significantly shorter ischaemic period.
Collapse
Affiliation(s)
- Safa Abdul-Ghani
- Bristol Heart Institute and Bristol Medical School, University of Bristol, Bristol BS2 8HW, UK
- Department of Physiology, Faculty of Medicine, Al-Quds University, Abu-Dis, Palestine
| | - Katie L. Skeffington
- Bristol Heart Institute and Bristol Medical School, University of Bristol, Bristol BS2 8HW, UK
| | - Minjoo Kim
- Bristol Heart Institute and Bristol Medical School, University of Bristol, Bristol BS2 8HW, UK
| | - Marco Moscarelli
- National Heart and Lung Institute, Imperial College, London SW3 6LY, UK
- GVM Care and Research, Anthea Hospital, I-70124 Bari, Italy
| | - Philip A. Lewis
- University of Bristol Proteomics/Bioinformatics Facility, University of Bristol, Bristol BS8 1TD, UK
| | - Kate Heesom
- University of Bristol Proteomics/Bioinformatics Facility, University of Bristol, Bristol BS8 1TD, UK
| | | | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College, London SW3 6LY, UK
| | - Barnaby C. Reeves
- Bristol Heart Institute and Bristol Medical School, University of Bristol, Bristol BS2 8HW, UK
| | | | - Gianni D. Angelini
- Bristol Heart Institute and Bristol Medical School, University of Bristol, Bristol BS2 8HW, UK
| | - M-Saadeh Suleiman
- Bristol Heart Institute and Bristol Medical School, University of Bristol, Bristol BS2 8HW, UK
| |
Collapse
|
11
|
Boteanu RM, Suica VI, Uyy E, Ivan L, Cerveanu-Hogas A, Mares RG, Simionescu M, Schiopu A, Antohe F. Short-Term Blockade of Pro-Inflammatory Alarmin S100A9 Favorably Modulates Left Ventricle Proteome and Related Signaling Pathways Involved in Post-Myocardial Infarction Recovery. Int J Mol Sci 2022; 23:ijms23095289. [PMID: 35563680 PMCID: PMC9103348 DOI: 10.3390/ijms23095289] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/05/2022] [Accepted: 05/08/2022] [Indexed: 02/01/2023] Open
Abstract
Prognosis after myocardial infarction (MI) varies greatly depending on the extent of damaged area and the management of biological processes during recovery. Reportedly, the inhibition of the pro-inflammatory S100A9 reduces myocardial damage after MI. We hypothesize that a S100A9 blockade induces changes of major signaling pathways implicated in post-MI healing. Mass spectrometry-based proteomics and gene analyses of infarcted mice left ventricle were performed. The S100A9 blocker (ABR-23890) was given for 3 days after coronary ligation. At 3 and 7 days post-MI, ventricle samples were analyzed versus control and Sham-operated mice. Blockade of S100A9 modulated the expressed proteins involved in five biological processes: leukocyte cell–cell adhesion, regulation of the muscle cell apoptotic process, regulation of the intrinsic apoptotic signaling pathway, sarcomere organization and cardiac muscle hypertrophy. The blocker induced regulation of 36 proteins interacting with or targeted by the cellular tumor antigen p53, prevented myocardial compensatory hypertrophy, and reduced cardiac markers of post-ischemic stress. The blockade effect was prominent at day 7 post-MI when the quantitative features of the ventricle proteome were closer to controls. Blockade of S100A9 restores key biological processes altered post-MI. These processes could be valuable new pharmacological targets for the treatment of ischemic heart. Mass spectrometry data are available via ProteomeXchange with identifier PXD033683.
Collapse
Affiliation(s)
- Raluca Maria Boteanu
- Department of Proteomics, Institute of Cellular Biology and Pathology “N. Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (R.M.B.); (V.-I.S.); (E.U.); (L.I.); (A.C.-H.); (M.S.)
| | - Viorel-Iulian Suica
- Department of Proteomics, Institute of Cellular Biology and Pathology “N. Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (R.M.B.); (V.-I.S.); (E.U.); (L.I.); (A.C.-H.); (M.S.)
| | - Elena Uyy
- Department of Proteomics, Institute of Cellular Biology and Pathology “N. Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (R.M.B.); (V.-I.S.); (E.U.); (L.I.); (A.C.-H.); (M.S.)
| | - Luminita Ivan
- Department of Proteomics, Institute of Cellular Biology and Pathology “N. Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (R.M.B.); (V.-I.S.); (E.U.); (L.I.); (A.C.-H.); (M.S.)
| | - Aurel Cerveanu-Hogas
- Department of Proteomics, Institute of Cellular Biology and Pathology “N. Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (R.M.B.); (V.-I.S.); (E.U.); (L.I.); (A.C.-H.); (M.S.)
| | - Razvan Gheorghita Mares
- Department of Pathophysiology, University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania; (R.G.M.); (A.S.)
| | - Maya Simionescu
- Department of Proteomics, Institute of Cellular Biology and Pathology “N. Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (R.M.B.); (V.-I.S.); (E.U.); (L.I.); (A.C.-H.); (M.S.)
| | - Alexandru Schiopu
- Department of Pathophysiology, University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania; (R.G.M.); (A.S.)
- Department of Clinical Sciences Malmö, Lund University, 21428 Malmö, Sweden
| | - Felicia Antohe
- Department of Proteomics, Institute of Cellular Biology and Pathology “N. Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (R.M.B.); (V.-I.S.); (E.U.); (L.I.); (A.C.-H.); (M.S.)
- Correspondence: ; Tel.: +40-213-192-737
| |
Collapse
|
12
|
Possible Treatment of Myocardial Infarct Based on Tissue Engineering Using a Cellularized Solid Collagen Scaffold Functionalized with Arg-Glyc-Asp (RGD) Peptide. Int J Mol Sci 2021; 22:ijms222212563. [PMID: 34830447 PMCID: PMC8620820 DOI: 10.3390/ijms222212563] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/23/2021] [Accepted: 11/15/2021] [Indexed: 12/13/2022] Open
Abstract
Currently, the clinical impact of cell therapy after a myocardial infarction (MI) is limited by low cell engraftment due to low cell retention, cell death in inflammatory and poor angiogenic infarcted areas, secondary migration. Cells interact with their microenvironment through integrin mechanoreceptors that control their survival/apoptosis/differentiation/migration and proliferation. The association of cells with a three-dimensional material may be a way to improve interactions with their integrins, and thus outcomes, especially if preparations are epicardially applied. In this review, we will focus on the rationale for using collagen as a polymer backbone for tissue engineering of a contractile tissue. Contractilities are reported for natural but not synthetic polymers and for naturals only for: collagen/gelatin/decellularized-tissue/fibrin/Matrigel™ and for different material states: hydrogels/gels/solids. To achieve a thick/long-term contractile tissue and for cell transfer, solid porous compliant scaffolds are superior to hydrogels or gels. Classical methods to produce solid scaffolds: electrospinning/freeze-drying/3D-printing/solvent-casting and methods to reinforce and/or maintain scaffold properties by reticulations are reported. We also highlight the possibility of improving integrin interaction between cells and their associated collagen by its functionalizing with the RGD-peptide. Using a contractile patch that can be applied epicardially may be a way of improving ventricular remodeling and limiting secondary cell migration.
Collapse
|
13
|
Simons P, Rinaldi DA, Bondu V, Kell AM, Bradfute S, Lidke DS, Buranda T. Integrin activation is an essential component of SARS-CoV-2 infection. Sci Rep 2021; 11:20398. [PMID: 34650161 PMCID: PMC8516859 DOI: 10.1038/s41598-021-99893-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/30/2021] [Indexed: 02/07/2023] Open
Abstract
SARS-CoV-2 infection depends on binding its spike (S) protein to angiotensin-converting enzyme 2 (ACE2). The S protein expresses an RGD motif, suggesting that integrins may be co-receptors. Here, we UV-inactivated SARS-CoV-2 and fluorescently labeled the envelope membrane with octadecyl rhodamine B (R18) to explore the role of integrin activation in mediating cell entry and productive infection. We used flow cytometry and confocal microscopy to show that SARS-CoV-2R18 particles engage basal-state integrins. Furthermore, we demonstrate that Mn2+, which induces integrin extension, enhances cell entry of SARS-CoV-2R18. We also show that one class of integrin antagonist, which binds to the αI MIDAS site and stabilizes the inactive, closed conformation, selectively inhibits the engagement of SARS-CoV-2R18 with basal state integrins, but is ineffective against Mn2+-activated integrins. RGD-integrin antagonists inhibited SARS-CoV-2R18 binding regardless of integrin activation status. Integrins transmit signals bidirectionally: 'inside-out' signaling primes the ligand-binding function of integrins via a talin-dependent mechanism, and 'outside-in' signaling occurs downstream of integrin binding to macromolecular ligands. Outside-in signaling is mediated by Gα13. Using cell-permeable peptide inhibitors of talin and Gα13 binding to the cytoplasmic tail of an integrin's β subunit, we demonstrate that talin-mediated signaling is essential for productive infection.
Collapse
Affiliation(s)
- Peter Simons
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, 87131, USA
| | - Derek A Rinaldi
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, 87131, USA
| | - Virginie Bondu
- Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, 87131, USA
| | - Alison M Kell
- Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, 87131, USA
- Center for Infectious Diseases and Immunity, University of New Mexico School of Medicine, Albuquerque, NM, 87131, USA
| | - Steven Bradfute
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, 87131, USA
- Center for Infectious Diseases and Immunity, University of New Mexico School of Medicine, Albuquerque, NM, 87131, USA
| | - Diane S Lidke
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, 87131, USA
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Tione Buranda
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, 87131, USA.
- Center for Infectious Diseases and Immunity, University of New Mexico School of Medicine, Albuquerque, NM, 87131, USA.
| |
Collapse
|
14
|
Ma SQ, Guo Z, Liu FY, Hasan SG, Yang D, Tang N, An P, Wang MY, Wu HM, Yang Z, Fan D, Tang QZ. 6-Gingerol protects against cardiac remodeling by inhibiting the p38 mitogen-activated protein kinase pathway. Acta Pharmacol Sin 2021; 42:1575-1586. [PMID: 33462378 PMCID: PMC8463710 DOI: 10.1038/s41401-020-00587-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 11/20/2020] [Indexed: 02/02/2023] Open
Abstract
6-Gingerol, a pungent ingredient of ginger, has been reported to possess anti-inflammatory and antioxidant activities, but the effect of 6-gingerol on pressure overload-induced cardiac remodeling remains inconclusive. In this study, we investigated the effect of 6-gingerol on cardiac remodeling in in vivo and in vitro models, and to clarify the underlying mechanisms. C57BL/6 mice were subjected to transverse aortic constriction (TAC), and treated with 6-gingerol (20 mg/kg, ig) three times a week (1 week in advance and continued until the end of the experiment). Four weeks after TAC surgery, the mice were subjected to echocardiography, and then sacrificed to harvest the hearts for analysis. For in vitro study, neonatal rat cardiomyocytes and cardiac fibroblasts were used to validate the protective effects of 6-gingerol in response to phenylephrine (PE) and transforming growth factor-β (TGF-β) challenge. We showed that 6-gingerol administration protected against pressure overload-induced cardiac hypertrophy, fibrosis, inflammation, and dysfunction in TAC mice. In the in vitro study, we showed that treatment with 6-gingerol (20 μM) blocked PE-induced-cardiomyocyte hypertrophy and TGF-β-induced cardiac fibroblast activation. Furthermore, 6-gingerol treatment significantly decreased mitogen-activated protein kinase p38 (p38) phosphorylation in response to pressure overload in vivo and extracellular stimuli in vitro, which was upregulated in the absence of 6-gingerol treatment. Moreover, transfection with mitogen-activated protein kinase kinase 6 expressing adenoviruses (Ad-MKK6), which specifically activated p38, abolished the protective effects of 6-gingerol in both in vitro and in vivo models. In conclusion, 6-gingerol improves cardiac function and alleviates cardiac remodeling induced by pressure overload in a p38-dependent manner. The present study demonstrates that 6-gingerol is a promising agent for the intervention of pathological cardiac remodeling.
Collapse
Affiliation(s)
- Shu-Qing Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Zhen Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Fang-Yuan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Shahzad-Gul Hasan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
- Department of Medicine, Bahawal Victoria Hospital, Bahawalpur, 63100, Pakistan
| | - Dan Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Nan Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Peng An
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Ming-Yu Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Hai-Ming Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Zheng Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Di Fan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China.
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China.
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China.
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China.
| |
Collapse
|
15
|
Balogh V, MacAskill MG, Hadoke PWF, Gray GA, Tavares AAS. Positron Emission Tomography Techniques to Measure Active Inflammation, Fibrosis and Angiogenesis: Potential for Non-invasive Imaging of Hypertensive Heart Failure. Front Cardiovasc Med 2021; 8:719031. [PMID: 34485416 PMCID: PMC8416043 DOI: 10.3389/fcvm.2021.719031] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/22/2021] [Indexed: 12/11/2022] Open
Abstract
Heart failure, which is responsible for a high number of deaths worldwide, can develop due to chronic hypertension. Heart failure can involve and progress through several different pathways, including: fibrosis, inflammation, and angiogenesis. Early and specific detection of changes in the myocardium during the transition to heart failure can be made via the use of molecular imaging techniques, including positron emission tomography (PET). Traditional cardiovascular PET techniques, such as myocardial perfusion imaging and sympathetic innervation imaging, have been established at the clinical level but are often lacking in pathway and target specificity that is important for assessment of heart failure. Therefore, there is a need to identify new PET imaging markers of inflammation, fibrosis and angiogenesis that could aid diagnosis, staging and treatment of hypertensive heart failure. This review will provide an overview of key mechanisms underlying hypertensive heart failure and will present the latest developments in PET probes for detection of cardiovascular inflammation, fibrosis and angiogenesis. Currently, selective PET probes for detection of angiogenesis remain elusive but promising PET probes for specific targeting of inflammation and fibrosis are rapidly progressing into clinical use.
Collapse
Affiliation(s)
- Viktoria Balogh
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom.,Edinburgh Imaging, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Mark G MacAskill
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom.,Edinburgh Imaging, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Patrick W F Hadoke
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Gillian A Gray
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Adriana A S Tavares
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom.,Edinburgh Imaging, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
16
|
Schussler O, Chachques JC, Alifano M, Lecarpentier Y. Key Roles of RGD-Recognizing Integrins During Cardiac Development, on Cardiac Cells, and After Myocardial Infarction. J Cardiovasc Transl Res 2021; 15:179-203. [PMID: 34342855 DOI: 10.1007/s12265-021-10154-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022]
Abstract
Cardiac cells interact with the extracellular matrix (ECM) proteins through integrin mechanoreceptors that control many cellular events such as cell survival, apoptosis, differentiation, migration, and proliferation. Integrins play a crucial role in cardiac development as well as in cardiac fibrosis and hypertrophy. Integrins recognize oligopeptides present on ECM proteins and are involved in three main types of interaction, namely with collagen, laminin, and the oligopeptide RGD (Arg-Gly-Asp) present on vitronectin and fibronectin proteins. To date, the specific role of integrins recognizing the RGD has not been addressed. In this review, we examine their role during cardiac development, their role on cardiac cells, and their upregulation during pathological processes such as heart fibrosis and hypertrophy. We also examine their role in regenerative and angiogenic processes after myocardial infarction (MI) in the peri-infarct area. Specific targeting of these integrins may be a way of controlling some of these pathological events and thereby improving medical outcomes.
Collapse
Affiliation(s)
- Olivier Schussler
- Thoracic Surgery Department, Cochin Hospital, APHP Centre, University of Paris, Paris, France.
| | - Juan C Chachques
- Department of Cardiac Surgery Pompidou Hospital, Laboratory of Biosurgical Research, Carpentier Foundation, University Paris Descartes, 75015, Paris, France
| | - Marco Alifano
- Thoracic Surgery Department, Cochin Hospital, APHP Centre, University of Paris, Paris, France.,INSERM U1138 Team "Cancer, Immune Control, and Escape", Cordeliers Research Center, University of Paris, Paris, France
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien, Meaux, France
| |
Collapse
|
17
|
Simons P, Rinaldi DA, Bondu V, Kell AM, Bradfute S, Lidke D, Buranda T. Integrin activation is an essential component of SARS-CoV-2 infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 34312625 DOI: 10.1101/2021.07.20.453118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cellular entry of coronaviruses depends on binding of the viral spike (S) protein to a specific cellular receptor, the angiotensin-converting enzyme 2 (ACE2). Furthermore, the viral spike protein expresses an RGD motif, suggesting that cell surface integrins may be attachment co-receptors. However, using infectious SARS-CoV-2 requires a biosafety level 3 laboratory (BSL-3), which limits the techniques that can be used to study the mechanism of cell entry. Here, we UV-inactivated SARS-CoV-2 and fluorescently labeled the envelope membrane with octadecyl rhodamine B (R18) to explore the role of integrin activation in mediating both cell entry and productive infection. We used flow cytometry and confocal fluorescence microscopy to show that fluorescently labeled SARS-CoV-2 R18 particles engage basal-state integrins. Furthermore, we demonstrate that Mn 2+ , which activates integrins and induces integrin extension, enhances cell binding and entry of SARS-CoV-2 R18 in proportion to the fraction of integrins activated. We also show that one class of integrin antagonist, which binds to the αI MIDAS site and stabilizes the inactive, closed conformation, selectively inhibits the engagement of SARS-CoV-2 R18 with basal state integrins, but is ineffective against Mn 2+ -activated integrins. At the same time, RGD-integrin antagonists inhibited SARS-CoV-2 R18 binding regardless of integrin activity state. Integrins transmit signals bidirectionally: 'inside-out' signaling primes the ligand binding function of integrins via a talin dependent mechanism and 'outside-in' signaling occurs downstream of integrin binding to macromolecular ligands. Outside-in signaling is mediated by Gα 13 and induces cell spreading, retraction, migration, and proliferation. Using cell-permeable peptide inhibitors of talin, and Gα 13 binding to the cytoplasmic tail of an integrin's β subunit, we further demonstrate that talin-mediated signaling is essential for productive infection by SARS-CoV-2.
Collapse
|
18
|
Gao Y, Liang X, Tian Z, Ma Y, Sun C. Betalain exerts cardioprotective and anti-inflammatory effects against the experimental model of heart failure. Hum Exp Toxicol 2021; 40:S16-S28. [PMID: 34189972 DOI: 10.1177/09603271211027933] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Betalain is a natural plant pigment known to elicit various biological activities. However, studies on the protective effect of betalain against heart failure have not reported yet. The experimental model of heart failure was created in Wistar rats using isoproterenol (ISO). The animals were randomly assigned into four groups such as sham-control, ISO-induced heart failure, betalain pretreated before ISO induction (50 mg/kg/day), and betalain drug control group were maintained for 6 weeks. At the end of the experimental period, anti-oxidant enzymes, inflammatory markers, matrix proteins, cardiac-specific markers, and micro RNAs were elucidated using RT-PCR, and ELISA analysis. The results demonstrated that the rats induced with ISO displayed an abnormality in cardiac functions, increased oxidative stress markers (p < 0.01), inflammatory cytokines (p < 0.01) while abrogated the expression of miR-18a, and increased miR-199a. While betalain pre-treated rats prevented the cardiac failure significantly (p < 0.01) with improved anti-oxidant enzymes, abrogated the inflammatory signals with restored matrix proteins, cardiac biomarker genes, and attenuated miR-423 and miR-27 compared to heart failure rats. The results of the study suggest that the betalain treatment protected the hearts from failing via microRNA mediated activation the anti-inflammatory signaling and restoring the matrix protein modulation.
Collapse
Affiliation(s)
- Y Gao
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - X Liang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Z Tian
- Frontier Institute of Science and Technology (FIST), Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Y Ma
- Frontier Institute of Science and Technology (FIST), Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - C Sun
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
19
|
Sun T, Wei L, Tian H, Zhan W, Ma H, Nie D, Wang S, Chen X, Tang G. Novel PET/CT tracers for targeted imaging of membrane receptors to evaluate cardiomyocyte apoptosis and tissue repair process in a rat model of myocardial infarction. Apoptosis 2021; 26:460-473. [PMID: 34185202 DOI: 10.1007/s10495-021-01681-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2021] [Indexed: 12/12/2022]
Abstract
The purpose of this study was to employ novel tracers PET imaging approach to define the time course and intensity of myocardial repair after apoptosis and to correlate the imaging signal to immunohistochemical staining in myocardial infarction (MI). We designed novel αVβ3-targeted and radio-functionalized tracers for detection of apoptosis in H9C2 cells and myocardial tissue. MI rats were imaged with [18F]FDG, [18F]ANP-Cin or [18F]ANP-RGD2 using a small-animal PET/CT device. Rats were sacrificed, and tissue samples from viable and injured myocardial areas were sectioned for TUNEL assay and histology. The uncorrected radiochemical yield of [18F]ANP-Cin and [18F]ANP-RGD2 were 41.3 ± 5.4% and 21.17 ± 4.7%, respectively. Two tracers meet many criteria for cardiac imaging, including high stability, high binding, no toxicity, fast renal clearance and excellent biodistribution in rat models. The uptake of [18F]ANP-Cin was significantly higher on the 1st and 3rd day than the 7th or 28th day after MI induction, a timeframe associated with increased cardiomyocyte apoptosis. Higher uptake of [18F]ANP-Cin was observed in MI rats than in N-acetylcysteine (NAC)-treated rats on the 3rd days. In contrast with [18F]ANP-Cin, no hot-spots was observed with [18F]ANP-RGD2 on the 1st day and more hot-spots was observed from the 3rd day to the 7th day, then less on the 28th days in the high apoptotic site. There was no uptake of [18F]FDG in or around the apoptotic region. On the 7th day the uptake of [18F]ANP-RGD2 was higher in NAC-treated rats than MI rats. [18F]ANP-Cin and [18F]ANP-RGD2 are superior to [18F]FDG for PET/CT imaging for evaluation of cardiomyocyte apoptosis and tissue repair processes in the MI rats.
Collapse
Affiliation(s)
- Ting Sun
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Lijiang Wei
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Nanfang PET Center and Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hua Tian
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China.
| | - Wanlin Zhan
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Hui Ma
- Department of Radiotherapy and Medical Imaging, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Dahong Nie
- Department of Radiotherapy and Medical Imaging, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Shilin Wang
- Department of Radiotherapy and Medical Imaging, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Xin Chen
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Ganghua Tang
- Nanfang PET Center and Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China. .,Department of Radiotherapy and Medical Imaging, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China.
| |
Collapse
|
20
|
Meagher PB, Lee XA, Lee J, Visram A, Friedberg MK, Connelly KA. Cardiac Fibrosis: Key Role of Integrins in Cardiac Homeostasis and Remodeling. Cells 2021; 10:cells10040770. [PMID: 33807373 PMCID: PMC8066890 DOI: 10.3390/cells10040770] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 03/30/2021] [Accepted: 03/30/2021] [Indexed: 12/11/2022] Open
Abstract
Cardiac fibrosis is a common finding that is associated with the progression of heart failure (HF) and impacts all chambers of the heart. Despite intense research, the treatment of HF has primarily focused upon strategies to prevent cardiomyocyte remodeling, and there are no targeted antifibrotic strategies available to reverse cardiac fibrosis. Cardiac fibrosis is defined as an accumulation of extracellular matrix (ECM) proteins which stiffen the myocardium resulting in the deterioration cardiac function. This occurs in response to a wide range of mechanical and biochemical signals. Integrins are transmembrane cell adhesion receptors, that integrate signaling between cardiac fibroblasts and cardiomyocytes with the ECM by the communication of mechanical stress signals. Integrins play an important role in the development of pathological ECM deposition. This review will discuss the role of integrins in mechano-transduced cardiac fibrosis in response to disease throughout the myocardium. This review will also demonstrate the important role of integrins as both initiators of the fibrotic response, and modulators of fibrosis through their effect on cardiac fibroblast physiology across the various heart chambers.
Collapse
Affiliation(s)
- Patrick B. Meagher
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (P.B.M.); (X.A.L.); (J.L.); (A.V.)
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Xavier Alexander Lee
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (P.B.M.); (X.A.L.); (J.L.); (A.V.)
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Joseph Lee
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (P.B.M.); (X.A.L.); (J.L.); (A.V.)
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Aylin Visram
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (P.B.M.); (X.A.L.); (J.L.); (A.V.)
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Mark K. Friedberg
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada;
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
- Labatt Family Heart Center and Department of Paediatrics, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Kim A. Connelly
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (P.B.M.); (X.A.L.); (J.L.); (A.V.)
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada;
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
- Correspondence: ; Tel.: +141-686-45201
| |
Collapse
|
21
|
Dorn LE, Lawrence W, Petrosino JM, Xu X, Hund TJ, Whitson BA, Stratton MS, Janssen PML, Mohler PJ, Schlosser A, Sorensen GL, Accornero F. Microfibrillar-Associated Protein 4 Regulates Stress-Induced Cardiac Remodeling. Circ Res 2021; 128:723-737. [PMID: 33530700 DOI: 10.1161/circresaha.120.317146] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Lisa E Dorn
- Physiology and Cell Biology (L.E.D., W.L., J.M.P., M.S.S., P.M.L.J., P.J.M., F.A.), The Ohio State University Wexner Medical Center, Columbus
| | - William Lawrence
- Physiology and Cell Biology (L.E.D., W.L., J.M.P., M.S.S., P.M.L.J., P.J.M., F.A.), The Ohio State University Wexner Medical Center, Columbus
| | - Jennifer M Petrosino
- Physiology and Cell Biology (L.E.D., W.L., J.M.P., M.S.S., P.M.L.J., P.J.M., F.A.), The Ohio State University Wexner Medical Center, Columbus
| | - Xianyao Xu
- Biomedical Engineering, The Ohio State University, Columbus (X.X., T.J.H.)
| | - Thomas J Hund
- Biomedical Engineering, The Ohio State University, Columbus (X.X., T.J.H.)
| | - Bryan A Whitson
- Bob and Corrine Frick Center for Heart Failure and Arrhythmia (B.A.W., P.J.M.), The Ohio State University Wexner Medical Center, Columbus.,Dorothy M. Davis Heart and Lung Research Institute and Surgery (B.A.W.), The Ohio State University Wexner Medical Center, Columbus
| | - Matthew S Stratton
- Physiology and Cell Biology (L.E.D., W.L., J.M.P., M.S.S., P.M.L.J., P.J.M., F.A.), The Ohio State University Wexner Medical Center, Columbus
| | - Paul M L Janssen
- Physiology and Cell Biology (L.E.D., W.L., J.M.P., M.S.S., P.M.L.J., P.J.M., F.A.), The Ohio State University Wexner Medical Center, Columbus
| | - Peter J Mohler
- Physiology and Cell Biology (L.E.D., W.L., J.M.P., M.S.S., P.M.L.J., P.J.M., F.A.), The Ohio State University Wexner Medical Center, Columbus.,Bob and Corrine Frick Center for Heart Failure and Arrhythmia (B.A.W., P.J.M.), The Ohio State University Wexner Medical Center, Columbus
| | - Anders Schlosser
- Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense (A.S., G.L.S.)
| | - Grith L Sorensen
- Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense (A.S., G.L.S.)
| | - Federica Accornero
- Physiology and Cell Biology (L.E.D., W.L., J.M.P., M.S.S., P.M.L.J., P.J.M., F.A.), The Ohio State University Wexner Medical Center, Columbus
| |
Collapse
|
22
|
Bracco Gartner TCL, Stein JM, Muylaert DEP, Bouten CVC, Doevendans PA, Khademhosseini A, Suyker WJL, Sluijter JPG, Hjortnaes J. Advanced In Vitro Modeling to Study the Paradox of Mechanically Induced Cardiac Fibrosis. Tissue Eng Part C Methods 2021; 27:100-114. [PMID: 33407000 DOI: 10.1089/ten.tec.2020.0298] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In heart failure, cardiac fibrosis is the result of an adverse remodeling process. Collagen is continuously synthesized in the myocardium in an ongoing attempt of the heart to repair itself. The resulting collagen depositions act counterproductively, causing diastolic dysfunction and disturbing electrical conduction. Efforts to treat cardiac fibrosis specifically have not been successful and the molecular etiology is only partially understood. The differentiation of quiescent cardiac fibroblasts to extracellular matrix-depositing myofibroblasts is a hallmark of cardiac fibrosis and a key aspect of the adverse remodeling process. This conversion is induced by a complex interplay of biochemical signals and mechanical stimuli. Tissue-engineered 3D models to study cardiac fibroblast behavior in vitro indicate that cyclic strain can activate a myofibroblast phenotype. This raises the question how fibroblast quiescence is maintained in the healthy myocardium, despite continuous stimulation of ultimately profibrotic mechanotransductive pathways. In this review, we will discuss the convergence of biochemical and mechanical differentiation signals of myofibroblasts, and hypothesize how these affect this paradoxical quiescence. Impact statement Mechanotransduction pathways of cardiac fibroblasts seem to ultimately be profibrotic in nature, but in healthy human myocardium, cardiac fibroblasts remain quiescent, despite continuous mechanical stimulation. We propose three hypotheses that could explain this paradoxical state of affairs. Furthermore, we provide suggestions for future research, which should lead to a better understanding of fibroblast quiescence and activation, and ultimately to new strategies for the prevention and treatment of cardiac fibrosis and heart failure.
Collapse
Affiliation(s)
- Thomas C L Bracco Gartner
- Division of Heart and Lungs, Department of Cardiothoracic Surgery, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jeroen M Stein
- Division of Heart and Lungs, Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands.,Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Dimitri E P Muylaert
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Carlijn V C Bouten
- Division of Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Pieter A Doevendans
- Division of Heart and Lungs, Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands.,Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, the Netherlands.,Division of Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands.,University Utrecht, Utrecht, the Netherlands.,Netherlands Heart Institute, Utrecht, the Netherlands.,Central Military Hospital, Utrecht, the Netherlands
| | - Ali Khademhosseini
- Department of Bioengineering, Radiology, Chemical and Biomolecular Engineering, Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, California, USA
| | - Willem J L Suyker
- Division of Heart and Lungs, Department of Cardiothoracic Surgery, University Medical Center Utrecht, Utrecht, the Netherlands.,Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, the Netherlands.,University Utrecht, Utrecht, the Netherlands
| | - Joost P G Sluijter
- Division of Heart and Lungs, Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands.,Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, the Netherlands.,University Utrecht, Utrecht, the Netherlands
| | - Jesper Hjortnaes
- Division of Heart and Lungs, Department of Cardiothoracic Surgery, University Medical Center Utrecht, Utrecht, the Netherlands.,Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, the Netherlands.,University Utrecht, Utrecht, the Netherlands
| |
Collapse
|
23
|
Ishii R, Okumura K, Akazawa Y, Malhi M, Ebata R, Sun M, Fujioka T, Kato H, Honjo O, Kabir G, Kuebler WM, Connelly K, Maynes JT, Friedberg MK. Heart Rate Reduction Improves Right Ventricular Function and Fibrosis in Pulmonary Hypertension. Am J Respir Cell Mol Biol 2021; 63:843-855. [PMID: 32915674 DOI: 10.1165/rcmb.2019-0317oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The potential benefit of heart rate reduction (HRR), independent of β-blockade, on right ventricular (RV) function in pulmonary hypertension (PH) remains undecided. We studied HRR effects on RV fibrosis and function in PH and RV pressure-loading models. Adult rats were randomized to 1) sham controls, 2) monocrotaline (MCT)-induced PH, 3) SU5416 + hypoxia (SUHX)-induced PH, or 4) pulmonary artery banding (PAB). Ivabradine (IVA) (10 mg/kg/d) was administered from 2 weeks after PH induction or PAB. Exercise tolerance, echocardiography, and pressure-volume hemodynamics were obtained at a terminal experiment 3 weeks later. RV myocardial samples were analyzed for putative mechanisms of HRR effects through fibrosis, profibrotic molecular signaling, and Ca++ handling. The effects of IVA versus carvedilol on human induced pluripotent stem cell-derived cardiomyocytes beat rate and relaxation properties were evaluated in vitro. Despite unabated severely elevated RV systolic pressures, IVA improved RV systolic and diastolic function, profibrotic signaling, and RV fibrosis in PH/PAB rats. RV systolic-elastance (control, 121 ± 116; MCT, 49 ± 36 vs. MCT+IVA, 120 ± 54; PAB, 70 ± 20 vs. PAB+IVA, 168 ± 76; SUHX, 86 ± 56 vs. SUHX +IVA, 218 ± 111; all P < 0.05), the time constant of RV relaxation, echo indices of RV function, and fibrosis (fibrosis: control, 4.6 ± 1%; MCT, 13.4 ± 6.5 vs. MCT+IVA, 6.7 ± 2.6%; PAB, 11.4 ± 4.5 vs. PAB+IVA, 6.4 ± 5.1%; SUHX, 10 ± 4.6 vs. SUHX+IVA, 3.9 ± 2.2%; all P < 0.001) were improved by IVA versus controls. IVA had a dose-response effect on induced pluripotent stem cell-derived cardiomyocytes beat rate by delaying Ca++ loss from the cytoplasm. In experimental PH or RV pressure loading, HRR improves RV fibrosis, function, and exercise endurance independent of β-blockade. The balance between adverse tachycardia and bradycardia requires further study, but judicious HRR may provide a promising strategy to improve RV function in clinical PH.
Collapse
Affiliation(s)
- Ryo Ishii
- The Labatt Family Heart Center, Division of Cardiology and Cardiovascular Surgery, Hospital for Sick Children and University of Toronto, Toronto, Canada
| | - Kenichi Okumura
- The Labatt Family Heart Center, Division of Cardiology and Cardiovascular Surgery, Hospital for Sick Children and University of Toronto, Toronto, Canada
| | - Yohei Akazawa
- The Labatt Family Heart Center, Division of Cardiology and Cardiovascular Surgery, Hospital for Sick Children and University of Toronto, Toronto, Canada
| | - Manpreet Malhi
- Department of Anesthesia and Pain Medicine, Hospital for Sick Children, Toronto, Canada
| | - Ryota Ebata
- The Labatt Family Heart Center, Division of Cardiology and Cardiovascular Surgery, Hospital for Sick Children and University of Toronto, Toronto, Canada
| | - Mei Sun
- The Labatt Family Heart Center, Division of Cardiology and Cardiovascular Surgery, Hospital for Sick Children and University of Toronto, Toronto, Canada
| | - Tao Fujioka
- The Labatt Family Heart Center, Division of Cardiology and Cardiovascular Surgery, Hospital for Sick Children and University of Toronto, Toronto, Canada
| | - Hideyuki Kato
- The Labatt Family Heart Center, Division of Cardiology and Cardiovascular Surgery, Hospital for Sick Children and University of Toronto, Toronto, Canada
| | - Osami Honjo
- The Labatt Family Heart Center, Division of Cardiology and Cardiovascular Surgery, Hospital for Sick Children and University of Toronto, Toronto, Canada
| | - Golam Kabir
- The Keenan Research Center for Biomedical Research of St. Michael's Hospital, Toronto, Canada; and
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Kim Connelly
- The Keenan Research Center for Biomedical Research of St. Michael's Hospital, Toronto, Canada; and
| | - Jason T Maynes
- Department of Anesthesia and Pain Medicine, Hospital for Sick Children, Toronto, Canada
| | - Mark K Friedberg
- The Labatt Family Heart Center, Division of Cardiology and Cardiovascular Surgery, Hospital for Sick Children and University of Toronto, Toronto, Canada
| |
Collapse
|
24
|
Congestive Heart Failure. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00050-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
25
|
Abstract
Myocardial fibrosis, the expansion of the cardiac interstitium through deposition of extracellular matrix proteins, is a common pathophysiologic companion of many different myocardial conditions. Fibrosis may reflect activation of reparative or maladaptive processes. Activated fibroblasts and myofibroblasts are the central cellular effectors in cardiac fibrosis, serving as the main source of matrix proteins. Immune cells, vascular cells and cardiomyocytes may also acquire a fibrogenic phenotype under conditions of stress, activating fibroblast populations. Fibrogenic growth factors (such as transforming growth factor-β and platelet-derived growth factors), cytokines [including tumour necrosis factor-α, interleukin (IL)-1, IL-6, IL-10, and IL-4], and neurohumoral pathways trigger fibrogenic signalling cascades through binding to surface receptors, and activation of downstream signalling cascades. In addition, matricellular macromolecules are deposited in the remodelling myocardium and regulate matrix assembly, while modulating signal transduction cascades and protease or growth factor activity. Cardiac fibroblasts can also sense mechanical stress through mechanosensitive receptors, ion channels and integrins, activating intracellular fibrogenic cascades that contribute to fibrosis in response to pressure overload. Although subpopulations of fibroblast-like cells may exert important protective actions in both reparative and interstitial/perivascular fibrosis, ultimately fibrotic changes perturb systolic and diastolic function, and may play an important role in the pathogenesis of arrhythmias. This review article discusses the molecular mechanisms involved in the pathogenesis of cardiac fibrosis in various myocardial diseases, including myocardial infarction, heart failure with reduced or preserved ejection fraction, genetic cardiomyopathies, and diabetic heart disease. Development of fibrosis-targeting therapies for patients with myocardial diseases will require not only understanding of the functional pluralism of cardiac fibroblasts and dissection of the molecular basis for fibrotic remodelling, but also appreciation of the pathophysiologic heterogeneity of fibrosis-associated myocardial disease.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, 1300 Morris Park Avenue Forchheimer G46B, Bronx, NY 10461, USA
| |
Collapse
|
26
|
Kim BS, Das S, Jang J, Cho DW. Decellularized Extracellular Matrix-based Bioinks for Engineering Tissue- and Organ-specific Microenvironments. Chem Rev 2020; 120:10608-10661. [PMID: 32786425 DOI: 10.1021/acs.chemrev.9b00808] [Citation(s) in RCA: 226] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Biomaterials-based biofabrication methods have gained much attention in recent years. Among them, 3D cell printing is a pioneering technology to facilitate the recapitulation of unique features of complex human tissues and organs with high process flexibility and versatility. Bioinks, combinations of printable hydrogel and cells, can be utilized to create 3D cell-printed constructs. The bioactive cues of bioinks directly trigger cells to induce tissue morphogenesis. Among the various printable hydrogels, the tissue- and organ-specific decellularized extracellular matrix (dECM) can exert synergistic effects in supporting various cells at any component by facilitating specific physiological properties. In this review, we aim to discuss a new paradigm of dECM-based bioinks able to recapitulate the inherent microenvironmental niche in 3D cell-printed constructs. This review can serve as a toolbox for biomedical engineers who want to understand the beneficial characteristics of the dECM-based bioinks and a basic set of fundamental criteria for printing functional human tissues and organs.
Collapse
Affiliation(s)
- Byoung Soo Kim
- Future IT Innovation Laboratory, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu,, Pohang, Kyungbuk 37673, Republic of Korea.,POSTECH-Catholic Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea
| | - Sanskrita Das
- Department of Creative IT Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea
| | - Jinah Jang
- Future IT Innovation Laboratory, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu,, Pohang, Kyungbuk 37673, Republic of Korea.,Department of Creative IT Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea.,Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea.,School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea.,POSTECH-Catholic Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea.,Institute of Convergence Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea.,POSTECH-Catholic Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea.,Institute of Convergence Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| |
Collapse
|
27
|
Castillo EA, Lane KV, Pruitt BL. Micromechanobiology: Focusing on the Cardiac Cell-Substrate Interface. Annu Rev Biomed Eng 2020; 22:257-284. [PMID: 32501769 DOI: 10.1146/annurev-bioeng-092019-034950] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Engineered, in vitro cardiac cell and tissue systems provide test beds for the study of cardiac development, cellular disease processes, and drug responses in a dish. Much effort has focused on improving the structure and function of engineered cardiomyocytes and heart tissues. However, these parameters depend critically on signaling through the cellular microenvironment in terms of ligand composition, matrix stiffness, and substrate mechanical properties-that is, matrix micromechanobiology. To facilitate improvements to in vitro microenvironment design, we review how cardiomyocytes and their microenvironment change during development and disease in terms of integrin expression and extracellular matrix (ECM) composition. We also discuss strategies used to bind proteins to common mechanobiology platforms and describe important differences in binding strength to the substrate. Finally, we review example biomaterial approaches designed to support and probe cell-ECM interactions of cardiomyocytes in vitro, as well as open questions and challenges.
Collapse
Affiliation(s)
- Erica A Castillo
- Department of Mechanical Engineering, Stanford University, Stanford, California 94305, USA; .,Department of Mechanical Engineering, University of California, Santa Barbara, California 93106, USA;
| | - Kerry V Lane
- Department of Mechanical Engineering, University of California, Santa Barbara, California 93106, USA;
| | - Beth L Pruitt
- Department of Mechanical Engineering, University of California, Santa Barbara, California 93106, USA; .,Biomolecular Science and Engineering Program, University of California, Santa Barbara, California 93106, USA.,Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, California 93117, USA;
| |
Collapse
|
28
|
Cohen L, Sagi I, Bigelman E, Solomonov I, Aloshin A, Ben-Shoshan J, Rozenbaum Z, Keren G, Entin-Meer M. Cardiac remodeling secondary to chronic volume overload is attenuated by a novel MMP9/2 blocking antibody. PLoS One 2020; 15:e0231202. [PMID: 32271823 PMCID: PMC7145114 DOI: 10.1371/journal.pone.0231202] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 03/18/2020] [Indexed: 12/18/2022] Open
Abstract
Objective Monoclonal antibody derivatives are promising drugs for the treatment of various diseases due to their high matrix metalloproteinases (MMP) active site specificity. We studied the effects of a novel antibody, SDS3, which specifically recognizes the mature active site of MMP9/2 during ventricular remodeling progression in a mouse model of chronic volume overload (VO). Methods VO was induced by creating an aortocaval fistula (ACF) in 10- to 12-week-old C57BL male mice. The VO-induced mice were treated with either vehicle control (PBS) or with SDS3 twice weekly by intraperitoneal (ip) injection. The relative changes in cardiac parameters between baseline (day 1) and end-point (day 30), were evaluated by echocardiography. The effects of SDS3 treatment on cardiac fibrosis, cardiomyocyte volume, and cardiac inflammation were tested by cardiac staining with Masson's trichrome, wheat Germ Agglutinin (WGA), and CD45, respectively. Serum levels of TNFα and IL-6 with and without SDS3 treatment were tested by ELISA. Results SDS3 significantly reduced cardiac dilatation, left ventricular (LV) mass, and cardiomyocyte hypertrophy compared to the vehicle treated animals. The antibody also reduced the heart-to-body weight ratio of the ACF animals to values comparable to those of the controls. Interestingly, the SDS3 group underwent significant reduction of cardiac inflammation and pro-inflammatory cytokine production, indicating a regulatory role for MMP9/2 in tissue remodeling, possibly by tumor necrosis factor alpha (TNFα) activation. In addition, significant changes in the expression of proteins related to mitochondrial function were observed in ACF animals, these changes were reversed following treatment with SDS3. Conclusion The data suggest that MMP9/2 blockage with SDS3 attenuates myocardial remodeling associated with chronic VO by three potential pathways: downregulating the extracellular matrix proteolytic cleavage, reducing the cardiac inflammatory responses, and preserving the cardiac mitochondrial structure and function.
Collapse
Affiliation(s)
- Lena Cohen
- Laboratory of Cardiovascular Research, Tel Aviv Sourasky Medical Center, Tel-Aviv, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Irit Sagi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Einat Bigelman
- Laboratory of Cardiovascular Research, Tel Aviv Sourasky Medical Center, Tel-Aviv, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Inna Solomonov
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Anna Aloshin
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Jeremy Ben-Shoshan
- Laboratory of Cardiovascular Research, Tel Aviv Sourasky Medical Center, Tel-Aviv, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Zach Rozenbaum
- Laboratory of Cardiovascular Research, Tel Aviv Sourasky Medical Center, Tel-Aviv, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Gad Keren
- Laboratory of Cardiovascular Research, Tel Aviv Sourasky Medical Center, Tel-Aviv, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Michal Entin-Meer
- Laboratory of Cardiovascular Research, Tel Aviv Sourasky Medical Center, Tel-Aviv, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- * E-mail:
| |
Collapse
|
29
|
Ward M, Iskratsch T. Mix and (mis-)match - The mechanosensing machinery in the changing environment of the developing, healthy adult and diseased heart. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118436. [PMID: 30742931 PMCID: PMC7042712 DOI: 10.1016/j.bbamcr.2019.01.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/07/2019] [Accepted: 01/29/2019] [Indexed: 01/01/2023]
Abstract
The composition and the stiffness of cardiac microenvironment change during development and/or in heart disease. Cardiomyocytes (CMs) and their progenitors sense these changes, which decides over the cell fate and can trigger CM (progenitor) proliferation, differentiation, de-differentiation or death. The field of mechanobiology has seen a constant increase in output that also includes a wealth of new studies specific to cardiac or cardiomyocyte mechanosensing. As a result, mechanosensing and transduction in the heart is increasingly being recognised as a main driver of regulating the heart formation and function. Recent work has for instance focused on measuring the molecular, physical and mechanical changes of the cellular environment - as well as intracellular contributors to the passive stiffness of the heart. On the other hand, a variety of new studies shed light into the molecular machinery that allow the cardiomyocytes to sense these properties. Here we want to discuss the recent work on this topic, but also specifically focus on how the different components are regulated at various stages during development, in health or disease in order to highlight changes that might contribute to disease progression and heart failure.
Collapse
Key Words
- cm, cardiomyocytes
- hcm, hypertrophic cardiomyopathy
- dcm, dilated cardiomyopathy
- icm, idiopathic cardiomyopathy
- myh, myosin heavy chain
- tnnt, troponin t
- tnni, troponin i
- afm, atomic force microscope
- mre, magnetic resonance elastography
- swe, ultrasound cardiac shear-wave elastography
- lv, left ventricle
- lox, lysyl oxidase
- loxl, lysyl oxidase like protein
- lh, lysyl hydroxylase
- lys, lysin
- lccs, lysald-derived collagen crosslinks
- hlccs, hylald-derived collagen crosslinks
- pka, protein kinase a
- pkc, protein kinase c
- vash1, vasohibin-1
- svbp, small vasohibin binding protein
- tcp, tubulin carboxypeptidase
- ttl, tubulin tyrosine ligase
- mrtf, myocardin-related transcription factor
- gap, gtpase activating protein
- gef, guanine nucleotide exchange factor
Collapse
Affiliation(s)
- Matthew Ward
- Division of Bioengineering, School of Engineering and Materials Science & Institute for Bioengineering, Queen Mary University of London, United Kingdom
| | - Thomas Iskratsch
- Division of Bioengineering, School of Engineering and Materials Science & Institute for Bioengineering, Queen Mary University of London, United Kingdom.
| |
Collapse
|
30
|
Wei L, Zhou Q, Tian H, Su Y, Fu GH, Sun T. Integrin β3 promotes cardiomyocyte proliferation and attenuates hypoxia-induced apoptosis via regulating the PTEN/Akt/mTOR and ERK1/2 pathways. Int J Biol Sci 2020; 16:644-654. [PMID: 32025212 PMCID: PMC6990915 DOI: 10.7150/ijbs.39414] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 12/02/2019] [Indexed: 12/13/2022] Open
Abstract
Objective: Integrin β3 is one of the main integrin heterodimer receptors on the surface of cardiac myocytes. Our previous studies showed that hypoxia induces apoptosis and increases integrin β3 expression in cardiomyocytes. However, the exact mechanism by which integrin β3 protects against apoptosis remains unclear. Hence, the present investigation aimed to explore the mechanism of integrin β3 in cardiomyocyte proliferation and hypoxia-induced cardiomyocyte apoptosis. Methods: Stable cells and in vivo acute and chronic heart failure rat models were generated to reveal the essential role of integrin β3 in cardiomyocyte proliferation and apoptosis. Western blotting and immunohistochemistry were employed to detect the expression of integrin β3 in the stable cells and rat cardiac tissue. Flow cytometer was used to investigate the role of integrin β3 in hypoxia-induced cardiomyocyte apoptosis. Confocal microscopy was used to detect the localization of integrin β3 and integrin αv in cardiomyocytes. Results: A cobaltous chloride-induced hypoxic microenvironment stimulated cardiomyocyte apoptosis and increased integrin β3 expression in H9C2 cells, AC16 cells, and cardiac tissue from acute and chronic heart failure rats. The overexpression of integrin β3 promoted cardiomyocyte proliferation, whereas silencing integrin β3 expression resulted in decreased cell proliferation in vitro. Furthermore, knocking down integrin β3 expression using shRNA or the integrin β3 inhibitor cilengitide exacerbated cobaltous chloride-induced cardiomyocyte apoptosis, whereas overexpression of integrin β3 weakened cobaltous chloride-induced cardiomyocytes apoptosis. We found that integrin β3 promoted cardiomyocytes proliferation through the regulation of the PTEN/Akt/mTOR and ERK1/2 signaling pathways. In addition, we found that knockdown of integrin αv or integrin β1 weakened the effect of integrin β3 in cardiomyocyte proliferation. Conclusion: Our findings revealed the molecular mechanism of the role of integrin β3 in cardiomyocyte proliferation and hypoxia-induced cardiomyocyte apoptosis, providing new insights into the mechanisms underlying myocardial protection.
Collapse
Affiliation(s)
- Lijiang Wei
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine. Shanghai, 200025, China
| | - Qingqing Zhou
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 20032, China
| | - Hua Tian
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 20032, China
| | - Yifan Su
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine. Shanghai, 200025, China
| | - Guo-Hui Fu
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, No.280, South Chong-Qing Road, Shanghai 200025, People's Republic of China
| | - Ting Sun
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine. Shanghai, 200025, China
| |
Collapse
|
31
|
Dukinfield M, Maniati E, Reynolds LE, Aubdool A, Baliga RS, D'Amico G, Maiques O, Wang J, Bedi KC, Margulies KB, Sanz‐Moreno V, Hobbs A, Hodivala‐Dilke K. Repurposing an anti-cancer agent for the treatment of hypertrophic heart disease. J Pathol 2019; 249:523-535. [PMID: 31424556 PMCID: PMC6900130 DOI: 10.1002/path.5340] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 08/14/2019] [Accepted: 08/15/2019] [Indexed: 02/06/2023]
Abstract
Coronary microvascular dysfunction combined with maladaptive cardiomyocyte morphology and energetics is a major contributor to heart failure advancement. Thus, dually enhancing cardiac angiogenesis and targeting cardiomyocyte function to slow, or reverse, the development of heart failure is a logical step towards improved therapy. We present evidence for the potential to repurpose a former anti-cancer Arg-Gly-Asp (RGD)-mimetic pentapeptide, cilengitide, here used at low doses. Cilengitide targets αvβ3 integrin and this protein is upregulated in human dilated and ischaemic cardiomyopathies. Treatment of mice after abdominal aortic constriction (AAC) surgery with low-dose cilengitide (ldCil) enhances coronary angiogenesis and directly affects cardiomyocyte hypertrophy with an associated reduction in disease severity. At a molecular level, ldCil treatment has a direct effect on cardiac endothelial cell transcriptomic profiles, with a significant enhancement of pro-angiogenic signalling pathways, corroborating the enhanced angiogenic phenotype after ldCil treatment. Moreover, ldCil treatment of Angiotensin II-stimulated AngII-stimulated cardiomyocytes significantly restores transcriptomic profiles similar to those found in normal human heart. The significance of this finding is enhanced by transcriptional similarities between AngII-treated cardiomyocytes and failing human hearts. Taken together, our data provide evidence supporting a possible new strategy for improved heart failure treatment using low-dose RGD-mimetics with relevance to human disease. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Matthew Dukinfield
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse SquareLondonUK
| | - Eleni Maniati
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse SquareLondonUK
| | - Louise E Reynolds
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse SquareLondonUK
| | - Aisah Aubdool
- William Harvey Research Institute, Queen Mary University of London, Charterhouse SquareLondonUK
| | - Reshma S Baliga
- William Harvey Research Institute, Queen Mary University of London, Charterhouse SquareLondonUK
| | - Gabriela D'Amico
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse SquareLondonUK
| | - Oscar Maiques
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse SquareLondonUK
| | - Jun Wang
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse SquareLondonUK
| | - Kenneth C Bedi
- Perelman School of MedicineUniversity of Pennsylvania, Translational Research CenterPhiladelphiaPAUSA
| | - Kenneth B Margulies
- Perelman School of MedicineUniversity of Pennsylvania, Translational Research CenterPhiladelphiaPAUSA
| | - Victoria Sanz‐Moreno
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse SquareLondonUK
| | - Adrian Hobbs
- William Harvey Research Institute, Queen Mary University of London, Charterhouse SquareLondonUK
| | - Kairbaan Hodivala‐Dilke
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse SquareLondonUK
| |
Collapse
|
32
|
Yoon Lee J, Chung J, Hwa Kim K, Hyun An S, Yi JE, Ae Kwon K, Kwon K. Extracorporeal shock waves protect cardiomyocytes from doxorubicin-induced cardiomyopathy by upregulating survivin via the integrin-ILK-Akt-Sp1/p53 axis. Sci Rep 2019; 9:12149. [PMID: 31434946 PMCID: PMC6704172 DOI: 10.1038/s41598-019-48470-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 08/02/2019] [Indexed: 12/12/2022] Open
Abstract
Doxorubicin (DOX) is a widely used anti-cancer drug; however, it has limited application due to cardiotoxicity. Extracorporeal shock waves (ESW) have been suggested to treat inflammatory and ischemic diseases, but the concrete effect of ESW in DOX-induced cardiomyopathy remain obscure. After H9c2 cells were subjected to ESW (0.04 mJ/cm2), they were treated with 1 μM DOX. As a result, ESW protected cardiomyocytes from DOX-induced cell death. H9c2 cells treated with DOX downregulated p-Akt and survivin expression, whereas the ESW treatment recovered both, suggesting its anti-apoptotic effect. ESW activated integrin αvβ3 and αvβ5, cardiomyocyte mechanosensors, followed by upregulation of ILK, p-Akt and survivin levels. Further, Sp1 and p53 were determined as key transcriptional factors mediating survivin expression via Akt phosphorylation by ESW. In in vivo acute DOX-induced cardiomyopathy model, the echocardiographic results showed that group subjected to ESW recovered from acute DOX-induced cardiomyopathy; left ventricular function was improved. The immunohistochemical staining results showed increased survivin and Bcl2 expression in ESW + DOX group compared to those in the DOX-injected group. In conclusion, non-invasive shockwaves protect cardiomyocytes from DOX-induced cardiomyopathy by upregulating survivin via integrin-ILK-Akt-Sp1/p53 pathway. In vivo study proposed ESW as a new kind of specific and safe therapy against acute DOX-induced cardiomyopathy.
Collapse
Affiliation(s)
- Ji Yoon Lee
- Medical Research Institute, School of Medicine, Ewha Womans University, Seoul, 158-710, Korea
| | - Jihwa Chung
- Medical Research Institute, School of Medicine, Ewha Womans University, Seoul, 158-710, Korea
| | - Kyoung Hwa Kim
- Medical Research Institute, School of Medicine, Ewha Womans University, Seoul, 158-710, Korea
| | - Shung Hyun An
- Medical Research Institute, School of Medicine, Ewha Womans University, Seoul, 158-710, Korea
| | - Jeong-Eun Yi
- Department of Internal Medicine, Cardiology Division, School of medicine, Ewha Womans University, Seoul, 158-710, Korea
| | - Kyoung Ae Kwon
- Graduate School of Industrial Pharmaceutical Sciences, Ewha Womans University, Seoul, Korea
| | - Kihwan Kwon
- Medical Research Institute, School of Medicine, Ewha Womans University, Seoul, 158-710, Korea. .,Department of Internal Medicine, Cardiology Division, School of medicine, Ewha Womans University, Seoul, 158-710, Korea.
| |
Collapse
|
33
|
Bryson TD, Ross J, Peterson E, Harding P. Prostaglandin E 2 and an EP4 receptor agonist inhibit LPS-Induced monocyte chemotactic protein 5 production and secretion in mouse cardiac fibroblasts via Akt and NF-κB signaling. Prostaglandins Other Lipid Mediat 2019; 144:106349. [PMID: 31229524 DOI: 10.1016/j.prostaglandins.2019.106349] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 06/14/2019] [Accepted: 06/19/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Prostaglandin E2 (PGE2) signals through 4 separate G-protein coupled receptor sub-types to elicit a variety of physiologic and pathophysiological effects. We have previously reported that mice lacking the EP4 receptor in the cardiomyocytes develop heart failure with a phenotype of dilated cardiomyopathy. Also, these mice have increased levels of chemokines, like MCP-5, in their left ventricles. We have recently reported that overexpression of the EP4 receptor could improve cardiac function in the myocardial infarction model. Furthermore, we showed that overexpression of EP4 had an anti-inflammatory effect in the whole left ventricle. It has also been shown that PGE2 can antagonize lipopolysaccharide-induced secretion of chemokines/cytokines in various cell types. We therefore hypothesized that PGE2 inhibits lipopolysaccharide (LPS)-induced MCP-5 secretion in adult mouse cardiac fibroblasts via its EP4 receptor. METHODS AND RESULTS Our hypothesis was tested using isolated mouse adult ventricular fibroblasts (AVF) treated with LPS. Pre-treatment of the cells with PGE2 and the EP4 agonist CAY10598 resulted in reductions of the pro-inflammatory response induced by LPS. Specifically, we observed reductions in MCP-5 secretion. Western blot analysis showed reductions in phosphorylated Akt and IκBα indicating reduced NF-κB activation. The anti-inflammatory effects of PGE2 and EP4 agonist signaling appeared to be independent of cAMP, p-44/42, or p38 pathways. CONCLUSION Exogenous treatment of PGE2 and the EP4 receptor agonist blocked the pro-inflammatory actions of LPS. Mechanistically, this was mediated via reduced Akt phosphorylation and inhibition of NF-κB.
Collapse
Affiliation(s)
- Timothy D Bryson
- Hypertension & Vascular Research Division, Dept. Internal Medicine, USA; Dept. of Physiology, Wayne State University School of Medicine, USA
| | - Jacob Ross
- Hypertension & Vascular Research Division, Dept. Internal Medicine, USA
| | - Edward Peterson
- Dept. of Public Health Sciences Henry Ford Hospital, Detroit, Michigan, USA
| | - Pamela Harding
- Hypertension & Vascular Research Division, Dept. Internal Medicine, USA; Dept. of Physiology, Wayne State University School of Medicine, USA.
| |
Collapse
|
34
|
Serum of patients with acute myocardial infarction prevents inflammation in iPSC-cardiomyocytes. Sci Rep 2019; 9:5651. [PMID: 30948775 PMCID: PMC6449343 DOI: 10.1038/s41598-019-42079-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 03/20/2019] [Indexed: 12/19/2022] Open
Abstract
Acute myocardial infarction (MI) evokes a systemic inflammatory response and locally the degradation of the necrotic tissue, followed by scar formation. The mechanisms for containment of the infarct zone are not studied well. The study aimed to examine the response of healthy cardiomyocytes to serum of patients with myocardial infarction. Human iPSC-cardiomyocytes (iPSC-CM) generated from two healthy donors were incubated with serum of patients with MI with and without ventricular fibrillation (VF) or of healthy controls. Different cell adhesion molecules were studied by flow cytometry and immunostaining. Cellular electrophysiology was studied by patch clamp. The cell adhesion molecules CD54/ICAM-1, CD58/LFA-3 and CD321/JAM-A were expressed on iPSC-CM within the plasma membrane. Incubation with serum of MI patients reduced the levels of expression of CD54/ICAM-1 and CD321/JAM-A by 15–20%. VF serum was less effective than serum of MI patients without VF. MI serum or VF serum did not affect resting potential, action potential duration or maximum depolarization velocity. Myocardial infarction serum exerts anti-inflammatory effects on healthy cardiomyocytes without affecting their electrical activity, thus helping to contain the infarct zone and to protect healthy tissue. Ventricular fibrillation during MI drives healthy cardiomyocytes towards a pro-inflammatory phenotype.
Collapse
|
35
|
Molecular Imaging to Monitor Left Ventricular Remodeling in Heart Failure. CURRENT CARDIOVASCULAR IMAGING REPORTS 2019. [DOI: 10.1007/s12410-019-9487-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
36
|
Frangogiannis NG. Cardiac fibrosis: Cell biological mechanisms, molecular pathways and therapeutic opportunities. Mol Aspects Med 2018; 65:70-99. [PMID: 30056242 DOI: 10.1016/j.mam.2018.07.001] [Citation(s) in RCA: 521] [Impact Index Per Article: 86.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 07/23/2018] [Indexed: 12/13/2022]
Abstract
Cardiac fibrosis is a common pathophysiologic companion of most myocardial diseases, and is associated with systolic and diastolic dysfunction, arrhythmogenesis, and adverse outcome. Because the adult mammalian heart has negligible regenerative capacity, death of a large number of cardiomyocytes results in reparative fibrosis, a process that is critical for preservation of the structural integrity of the infarcted ventricle. On the other hand, pathophysiologic stimuli, such as pressure overload, volume overload, metabolic dysfunction, and aging may cause interstitial and perivascular fibrosis in the absence of infarction. Activated myofibroblasts are the main effector cells in cardiac fibrosis; their expansion following myocardial injury is primarily driven through activation of resident interstitial cell populations. Several other cell types, including cardiomyocytes, endothelial cells, pericytes, macrophages, lymphocytes and mast cells may contribute to the fibrotic process, by producing proteases that participate in matrix metabolism, by secreting fibrogenic mediators and matricellular proteins, or by exerting contact-dependent actions on fibroblast phenotype. The mechanisms of induction of fibrogenic signals are dependent on the type of primary myocardial injury. Activation of neurohumoral pathways stimulates fibroblasts both directly, and through effects on immune cell populations. Cytokines and growth factors, such as Tumor Necrosis Factor-α, Interleukin (IL)-1, IL-10, chemokines, members of the Transforming Growth Factor-β family, IL-11, and Platelet-Derived Growth Factors are secreted in the cardiac interstitium and play distinct roles in activating specific aspects of the fibrotic response. Secreted fibrogenic mediators and matricellular proteins bind to cell surface receptors in fibroblasts, such as cytokine receptors, integrins, syndecans and CD44, and transduce intracellular signaling cascades that regulate genes involved in synthesis, processing and metabolism of the extracellular matrix. Endogenous pathways involved in negative regulation of fibrosis are critical for cardiac repair and may protect the myocardium from excessive fibrogenic responses. Due to the reparative nature of many forms of cardiac fibrosis, targeting fibrotic remodeling following myocardial injury poses major challenges. Development of effective therapies will require careful dissection of the cell biological mechanisms, study of the functional consequences of fibrotic changes on the myocardium, and identification of heart failure patient subsets with overactive fibrotic responses.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, 1300 Morris Park Avenue, Forchheimer G46B, Bronx, NY, 10461, USA.
| |
Collapse
|
37
|
Sun M, Ishii R, Okumura K, Krauszman A, Breitling S, Gomez O, Hinek A, Boo S, Hinz B, Connelly KA, Kuebler WM, Friedberg MK. Experimental Right Ventricular Hypertension Induces Regional β1-Integrin-Mediated Transduction of Hypertrophic and Profibrotic Right and Left Ventricular Signaling. J Am Heart Assoc 2018; 7:e007928. [PMID: 29599211 PMCID: PMC5907585 DOI: 10.1161/jaha.117.007928] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 02/28/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Development of right ventricular (RV) hypertension eventually contributes to RV and left ventricular (LV) myocardial fibrosis and dysfunction. The molecular mechanisms are not fully elucidated. METHODS AND RESULTS Pulmonary artery banding was used to induce RV hypertension in rats in vivo. Then, we evaluated cardiac function and regional remodeling 6 weeks after pulmonary artery banding. To further elucidate mechanisms responsible for regional cardiac remodeling, we also mimicked RV hypertensive stress by cyclic mechanical stretching applied to confluent cultures of cardiac fibroblasts, isolated from the RV free wall, septal hinge points, and LV free wall. Echocardiography and catheter evaluation demonstrated that rats in the pulmonary artery banding group developed RV hypertension with leftward septal displacement, LV compression, and increased LV end-diastolic pressures. Picrosirius red staining indicated that pulmonary artery banding induced marked RV fibrosis and dysfunction, with prominent fibrosis and elastin deposition at the septal hinge points but less LV fibrosis. These changes were associated with proportionally increased expressions of integrin-β1 and profibrotic signaling proteins, including phosphorylated Smad2/3 and transforming growth factor-β1. Moreover, mechanically stretched fibroblasts also expressed significantly increased levels of α-smooth muscle actin, integrin-β1, transforming growth factor-β1, collagen I deposition, and wrinkle formation on gel assays, consistent with myofibroblast transformation. These changes were not observed in parallel cultures of mechanically stretched fibroblasts, preincubated with the integrin inhibitor (BTT-3033). CONCLUSIONS Experimentally induced RV hypertension triggers regional RV, hinge-point, and LV integrin β1-dependent mechanotransduction signaling pathways that eventually trigger myocardial fibrosis via transforming growth factor-β1 signaling. Reduced LV fibrosis and preserved global function, despite geometrical and pressure aberrations, suggest a possible elastin-mediated protective mechanism at the septal hinge points.
Collapse
MESH Headings
- Animals
- Arterial Pressure
- Cells, Cultured
- Collagen Type I/metabolism
- Disease Models, Animal
- Elastin/metabolism
- Fibrosis
- Heart Ventricles/metabolism
- Heart Ventricles/pathology
- Heart Ventricles/physiopathology
- Hypertension, Pulmonary/complications
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/physiopathology
- Hypertrophy, Left Ventricular/etiology
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Right Ventricular/etiology
- Hypertrophy, Right Ventricular/metabolism
- Hypertrophy, Right Ventricular/pathology
- Hypertrophy, Right Ventricular/physiopathology
- Integrin beta1/metabolism
- Male
- Mechanotransduction, Cellular
- Pulmonary Artery/physiopathology
- Rats, Sprague-Dawley
- Transforming Growth Factor beta1/metabolism
- Ventricular Function, Left
- Ventricular Function, Right
- Ventricular Remodeling
Collapse
Affiliation(s)
- Mei Sun
- Division of Cardiology, Labatt Family Heart Center, Toronto, Ontario, Canada
- Translational Medicine, Hospital for Sick Children and University of Toronto, Ontario, Canada
| | - Ryo Ishii
- Division of Cardiology, Labatt Family Heart Center, Toronto, Ontario, Canada
- Translational Medicine, Hospital for Sick Children and University of Toronto, Ontario, Canada
| | - Kenichi Okumura
- Division of Cardiology, Labatt Family Heart Center, Toronto, Ontario, Canada
- Translational Medicine, Hospital for Sick Children and University of Toronto, Ontario, Canada
| | - Adrienn Krauszman
- The Keenan Research Center for Biomedical Science, St Michael's Hospital, Toronto, Canada
| | - Siegfried Breitling
- The Keenan Research Center for Biomedical Science, St Michael's Hospital, Toronto, Canada
| | - Olga Gomez
- Division of Cardiology, Labatt Family Heart Center, Toronto, Ontario, Canada
- Translational Medicine, Hospital for Sick Children and University of Toronto, Ontario, Canada
| | - Aleksander Hinek
- Translational Medicine, Hospital for Sick Children and University of Toronto, Ontario, Canada
| | - Stellar Boo
- Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Ontario, Canada
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Ontario, Canada
| | - Kim A Connelly
- The Keenan Research Center for Biomedical Science, St Michael's Hospital, Toronto, Canada
| | - Wolfgang M Kuebler
- The Keenan Research Center for Biomedical Science, St Michael's Hospital, Toronto, Canada
| | - Mark K Friedberg
- Division of Cardiology, Labatt Family Heart Center, Toronto, Ontario, Canada
- Translational Medicine, Hospital for Sick Children and University of Toronto, Ontario, Canada
| |
Collapse
|
38
|
Schernthaner C, Paar V, Wernly B, Pistulli R, Rohm I, Jung C, Figulla HR, Yilmaz A, Cadamuro J, Haschke-Becher E, Schulze PC, Hoppe UC, Lichtenauer M, Kretzschmar D. Elevated plasma levels of interleukin-16 in patients with acute myocardial infarction. Medicine (Baltimore) 2017; 96:e8396. [PMID: 29095267 PMCID: PMC5682786 DOI: 10.1097/md.0000000000008396] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Interleukin (IL)-16, a polypeptide cytokine, plays a crucial role in the inflammatory process, acting as a chemoattractant for peripheral immune cells and has been linked to various inflammatory diseases. However, its role in patients with acute myocardial infarction (AMI) is unclear.We retrospectively analyzed serum levels of IL-16 in blood of patients with (STEMI, n = 45) and without ST-segment elevation myocardial infarction (NSTEMI, n = 42) compared with controls with excluded coronary artery disease (n = 55). Furthermore, correlation analysis with inflammatory cells, C-reactive protein (CRP) levels, dendritic cell precursors (DCPs), and other clinical and biochemical markers was performed.Compared with controls, patients with STEMI and NSTEMI evidenced higher levels of IL-16 in pg/mL (STEMI: 759.38 ± 471.54, NSTEMI: 677.77 ± 438.8, control: 500.45 ± 432.21; P = .002). IL-16 correlated with CRP (r = 0.26, P = .001), leucocytes (r = 0.38, P < .001), NT-proBNP (r = 0.20, P = .02) and hsTnT (r = 0.25, P = .004). Circulating myeloid DCPs, plasmacytoid DCPs, and total DCPs showed a significant inverse correlation to IL-16 levels (r = -0.21, P = .01; r = -0.23, P = .005; r = -0.26, P = .002, respectively).Interleukin-16 might play an important role in the inflammatory process of patients suffering from AMI and correlates with inflammatory cell activation and clinical and biochemical markers. The cytokine IL-16 might upregulate the proinflammatory response and recruitment of inflammatory cells into infarcted myocardium.
Collapse
Affiliation(s)
| | - Vera Paar
- Department of Cardiology, Paracelsus Medical University, Salzburg, Austria
| | - Bernhard Wernly
- Department of Cardiology, Paracelsus Medical University, Salzburg, Austria
| | - Rudin Pistulli
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive MedicalCare, Friedrich-Schiller-University Jena, Jena, Germany
| | - Ilonka Rohm
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive MedicalCare, Friedrich-Schiller-University Jena, Jena, Germany
| | - Christian Jung
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Duesseldorf, Duesseldorf, Germany
| | - Hans-Reiner Figulla
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive MedicalCare, Friedrich-Schiller-University Jena, Jena, Germany
| | - Attila Yilmaz
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive MedicalCare, Friedrich-Schiller-University Jena, Jena, Germany
| | - Janne Cadamuro
- Department of Laboratory Medicine, Paracelsus Medical University, Salzburg, Austria
| | | | | | - Uta C. Hoppe
- Department of Cardiology, Paracelsus Medical University, Salzburg, Austria
| | | | - Daniel Kretzschmar
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive MedicalCare, Friedrich-Schiller-University Jena, Jena, Germany
| |
Collapse
|
39
|
Grönman M, Tarkia M, Kiviniemi T, Halonen P, Kuivanen A, Savunen T, Tolvanen T, Teuho J, Käkelä M, Metsälä O, Pietilä M, Saukko P, Ylä-Herttuala S, Knuuti J, Roivainen A, Saraste A. Imaging of α vβ 3 integrin expression in experimental myocardial ischemia with [ 68Ga]NODAGA-RGD positron emission tomography. J Transl Med 2017. [PMID: 28629432 PMCID: PMC5477135 DOI: 10.1186/s12967-017-1245-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background Radiolabeled RGD peptides detect αvβ3 integrin expression associated with angiogenesis and extracellular matrix remodeling after myocardial infarction. We studied whether cardiac positron emission tomography (PET) with [68Ga]NODAGA-RGD detects increased αvβ3 integrin expression after induction of flow-limiting coronary stenosis in pigs, and whether αvβ3 integrin is expressed in viable ischemic or injured myocardium. Methods We studied 8 Finnish landrace pigs 13 ± 4 days after percutaneous implantation of a bottleneck stent in the proximal left anterior descending coronary artery. Antithrombotic therapy was used to prevent stent occlusion. Myocardial uptake of [68Ga]NODAGA-RGD (290 ± 31 MBq) was evaluated by a 62 min dynamic PET scan. The ischemic area was defined as the regional perfusion abnormality during adenosine-induced stress by [15O]water PET. Guided by triphenyltetrazolium chloride staining, tissue samples from viable and injured myocardial areas were obtained for autoradiography and histology. Results Stent implantation resulted in a partly reversible myocardial perfusion abnormality. Compared with remote myocardium, [68Ga]NODAGA-RGD PET showed increased tracer uptake in the ischemic area (ischemic-to-remote ratio 1.3 ± 0.20, p = 0.0034). Tissue samples from the injured areas, but not from the viable ischemic areas, showed higher [68Ga]NODAGA-RGD uptake than the remote non-ischemic myocardium. Uptake of [68Ga]NODAGA-RGD correlated with immunohistochemical detection of αvβ3 integrin that was expressed in the injured myocardial areas. Conclusions Cardiac [68Ga]NODAGA-RGD PET demonstrates increased myocardial αvβ3 integrin expression after induction of flow-limiting coronary stenosis in pigs. Localization of [68Ga]NODAGA-RGD uptake indicates that it reflects αvβ3 integrin expression associated with repair of recent myocardial injury.
Collapse
Affiliation(s)
- Maria Grönman
- Turku PET Centre, University of Turku, 20521, Turku, Finland
| | - Miikka Tarkia
- Turku PET Centre, University of Turku, 20521, Turku, Finland
| | | | - Paavo Halonen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Joensuu, Finland
| | - Antti Kuivanen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Joensuu, Finland
| | - Timo Savunen
- Heart Center, Turku University Hospital, Turku, Finland.,Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Tuula Tolvanen
- Turku PET Centre, Turku University Hospital, Turku, Finland.,Department of Medical Physics, Turku University Hospital and University of Turku, Turku, Finland
| | - Jarmo Teuho
- Turku PET Centre, Turku University Hospital, Turku, Finland.,Department of Medical Physics, Turku University Hospital and University of Turku, Turku, Finland
| | - Meeri Käkelä
- Turku PET Centre, University of Turku, 20521, Turku, Finland
| | - Olli Metsälä
- Turku PET Centre, University of Turku, 20521, Turku, Finland
| | - Mikko Pietilä
- Heart Center, Turku University Hospital, Turku, Finland
| | - Pekka Saukko
- Department of Forensic Medicine, University of Turku, Turku, Finland
| | - Seppo Ylä-Herttuala
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Joensuu, Finland
| | - Juhani Knuuti
- Turku PET Centre, University of Turku, 20521, Turku, Finland.,Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Anne Roivainen
- Turku PET Centre, University of Turku, 20521, Turku, Finland.,Turku PET Centre, Turku University Hospital, Turku, Finland.,Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Antti Saraste
- Turku PET Centre, University of Turku, 20521, Turku, Finland. .,Heart Center, Turku University Hospital, Turku, Finland. .,Turku PET Centre, Turku University Hospital, Turku, Finland. .,Institute of Clinical Medicine, University of Turku, Turku, Finland.
| |
Collapse
|
40
|
Abstract
Cardiac and skeletal striated muscles are intricately designed machines responsible for muscle contraction. Coordination of the basic contractile unit, the sarcomere, and the complex cytoskeletal networks are critical for contractile activity. The sarcomere is comprised of precisely organized individual filament systems that include thin (actin), thick (myosin), titin, and nebulin. Connecting the sarcomere to other organelles (e.g., mitochondria and nucleus) and serving as the scaffold to maintain cellular integrity are the intermediate filaments. The costamere, on the other hand, tethers the sarcomere to the cell membrane. Unique structures like the intercalated disc in cardiac muscle and the myotendinous junction in skeletal muscle help synchronize and transmit force. Intense investigation has been done on many of the proteins that make up these cytoskeletal assemblies. Yet the details of their function and how they interconnect have just started to be elucidated. A vast number of human myopathies are contributed to mutations in muscle proteins; thus understanding their basic function provides a mechanistic understanding of muscle disorders. In this review, we highlight the components of striated muscle with respect to their interactions, signaling pathways, functions, and connections to disease. © 2017 American Physiological Society. Compr Physiol 7:891-944, 2017.
Collapse
Affiliation(s)
- Christine A Henderson
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Christopher G Gomez
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Stefanie M Novak
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Lei Mi-Mi
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Carol C Gregorio
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
41
|
|
42
|
Fibrillar Type I Collagen Enhances the Differentiation and Proliferation of Myofibroblasts by Lowering α2 β1 Integrin Expression in Cardiac Fibrosis. BIOMED RESEARCH INTERNATIONAL 2017; 2017:1790808. [PMID: 28251149 PMCID: PMC5303846 DOI: 10.1155/2017/1790808] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 12/07/2016] [Accepted: 12/25/2016] [Indexed: 01/02/2023]
Abstract
Many studies have shown that α2β1 integrin plays an important role in the development of cardiac fibrosis. However, the mechanism of how α2β1 integrin regulates the differentiation and proliferation of myofibroblasts in cardiac fibrosis through fibrillar collagen (FC) remains uncertain. We established that FC mimicked the 3-dimensional extracellular matrix (ECM) of fibroblasts from post-myocardial infarction (MI) patients in vivo. This allowed us to explore the differentiation and proliferation of cardiac fibroblasts on FC. Here, we report that low expression of α2β1 integrin increased protein kinase B (AKT) activation and α-smooth muscle actin (α-SMA) expression. This occurred due to the instability of phosphatase and tensin homolog (PTEN) in myofibroblasts on FC. We also demonstrated that FC reduced protein phosphatase type 2A (PP2A) activity of myofibroblasts, which was coincident with low α2β1 integrin expression and activation of AKT, but not mitogen-activated protein kinase (ERK). In addition, knock-down of both β1 integrin and PP2A in fibroblasts promoted differentiation and proliferation via AKT activation and increased α-SMA expression. In summary, our study demonstrated that low α2β1 integrin expression regulated its downstream targets PTEN and AKT via crosstalk with PP2A, a critical cell signaling pathway that permits aberrant differentiation and proliferation of myofibroblasts on FC.
Collapse
|
43
|
Civitarese RA, Kapus A, McCulloch CA, Connelly KA. Role of integrins in mediating cardiac fibroblast–cardiomyocyte cross talk: a dynamic relationship in cardiac biology and pathophysiology. Basic Res Cardiol 2016; 112:6. [DOI: 10.1007/s00395-016-0598-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 12/14/2016] [Indexed: 12/16/2022]
|
44
|
Jenkins WSA, Vesey AT, Stirrat C, Connell M, Lucatelli C, Neale A, Moles C, Vickers A, Fletcher A, Pawade T, Wilson I, Rudd JHF, van Beek EJR, Mirsadraee S, Dweck MR, Newby DE. Cardiac α Vβ 3 integrin expression following acute myocardial infarction in humans. Heart 2016; 103:607-615. [PMID: 27927700 PMCID: PMC5566089 DOI: 10.1136/heartjnl-2016-310115] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 09/16/2016] [Accepted: 09/20/2016] [Indexed: 12/31/2022] Open
Abstract
Objective Maladaptive repair contributes towards the development of heart failure following myocardial infarction (MI). The αvβ3 integrin receptor is a key mediator and determinant of cardiac repair. We aimed to establish whether αvβ3 integrin expression determines myocardial recovery following MI. Methods 18F-Fluciclatide (a novel αvβ3-selective radiotracer) positron emission tomography (PET) and CT imaging and gadolinium-enhanced MRI (CMR) were performed in 21 patients 2 weeks after ST-segment elevation MI (anterior, n=16; lateral, n=4; inferior, n=1). CMR was repeated 9 months after MI. 7 stable patients with chronic total occlusion (CTO) of a major coronary vessel and nine healthy volunteers underwent a single PET/CT and CMR. Results 18F-Fluciclatide uptake was increased at sites of acute infarction compared with remote myocardium (tissue-to-background ratio (TBRmean) 1.34±0.22 vs 0.85±0.17; p<0.001) and myocardium of healthy volunteers (TBRmean 1.34±0.22 vs 0.70±0.03; p<0.001). There was no 18F-fluciclatide uptake at sites of established prior infarction in patients with CTO, with activity similar to the myocardium of healthy volunteers (TBRmean 0.71±0.06 vs 0.70±0.03, p=0.83). 18F-Fluciclatide uptake occurred at sites of regional wall hypokinesia (wall motion index≥1 vs 0; TBRmean 0.93±0.31 vs 0.80±0.26 respectively, p<0.001) and subendocardial infarction. Importantly, although there was no correlation with infarct size (r=0.03, p=0.90) or inflammation (C reactive protein, r=−0.20, p=0.38), 18F-fluciclatide uptake was increased in segments displaying functional recovery (TBRmean 0.95±0.33 vs 0.81±0.27, p=0.002) and associated with increase in probability of regional recovery. Conclusion 18F-Fluciclatide uptake is increased at sites of recent MI acting as a biomarker of cardiac repair and predicting regions of recovery. Trial registration number NCT01813045; Post-results.
Collapse
Affiliation(s)
- William S A Jenkins
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Alex T Vesey
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Colin Stirrat
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Martin Connell
- Clinical Research Imaging Center, University of Edinburgh, Edinburgh, UK
| | | | - Anoushka Neale
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Catriona Moles
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Anna Vickers
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Alison Fletcher
- Clinical Research Imaging Center, University of Edinburgh, Edinburgh, UK
| | - Tania Pawade
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Ian Wilson
- Edinburgh Molecular Imaging Ltd, Edinburgh, UK
| | - James H F Rudd
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
| | - Edwin J R van Beek
- Clinical Research Imaging Center, University of Edinburgh, Edinburgh, UK
| | - Saeed Mirsadraee
- Clinical Research Imaging Center, University of Edinburgh, Edinburgh, UK
| | - Marc R Dweck
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - David E Newby
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
45
|
Rodness J, Mihic A, Miyagi Y, Wu J, Weisel RD, Li RK. VEGF-loaded microsphere patch for local protein delivery to the ischemic heart. Acta Biomater 2016; 45:169-181. [PMID: 27619839 DOI: 10.1016/j.actbio.2016.09.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 09/06/2016] [Accepted: 09/08/2016] [Indexed: 12/25/2022]
Abstract
BACKGROUND Revascularization of the heart after myocardial infarction (MI) using growth factors delivered by hydrogel-based microspheres represents a promising therapeutic approach for cardiac regeneration. Microspheres have tuneable degradation properties and support the prolonged release of soluble factors. Cardiac patches provide mechanical restraint, preventing dilatation associated with ventricular remodelling. METHODS We combined these approaches and produced a compacted calcium-alginate microsphere patch, restrained by a chitosan sheet, to deliver vascular endothelial growth factor (VEGF) to the heart after myocardial injury in rats. RESULTS Microspheres had an average diameter of 3.2μm, were nonporous, and characterized by a smooth dimpled surface. Microsphere patches demonstrated prolonged in vitro release characteristics compared to non-compacted microspheres and VEGF supernatants obtained from patches maintained their bioactivity for the 5day duration of the study in vitro. In vivo, patches were assessed with magnetic resonance imaging following MI, and demonstrated 50% degradation 25.6days after implantation. Both VEGF(-) and VEGF(+) microsphere patch-treated hearts had better cardiac function than unpatched (chitosan sheet only) controls. However, VEGF(+) microsphere-patched hearts had thicker scars characterized by higher capillary density in the border zone than did those treated with VEGF(-) patches. VEGF was detected in the patches 4weeks post-implantation. CONCLUSION The condensed microsphere patch represents a new therapeutic platform for cytokine delivery and could be used as an adjuvant to current biomaterial and cell-based therapies to promote localized angiogenesis in the infarcted heart. STATEMENT OF SIGNIFICANCE Following a heart attack, a lack of blood flow to the heart results in loss of heart cells. Growth factors may facilitate growth of blood vessels and heart tissue repair and prevent the onset of heart failure. Determining a way to deliver these growth factors directly to the heart is vital. Here, we combined two biomaterial-based approaches to deliver vascular endothelial growth factor (VEGF) to rat hearts after heart attack: a microsphere for prolonged release of VEGF, and a cardiac patch for mechanical restraint to prevent heart dysfunction. The feasibility of this microsphere patch was demonstrated by surgically implanting it over the infarct region of the heart post-injury. VEGF-patched hearts had better blood vessel growth, tissue repair, and heart function.
Collapse
Affiliation(s)
- Jonathan Rodness
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada; Division of Cardiac Surgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada.
| | - Anton Mihic
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada; Division of Cardiac Surgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada.
| | - Yasuo Miyagi
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada; Division of Cardiac Surgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada.
| | - Jun Wu
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada; Division of Cardiac Surgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada.
| | - Richard D Weisel
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada; Division of Cardiac Surgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada.
| | - Ren-Ke Li
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada; Division of Cardiac Surgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
46
|
Clinical Utility and Future Applications of PET/CT and PET/CMR in Cardiology. Diagnostics (Basel) 2016; 6:diagnostics6030032. [PMID: 27598207 PMCID: PMC5039566 DOI: 10.3390/diagnostics6030032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 08/17/2016] [Accepted: 08/23/2016] [Indexed: 12/15/2022] Open
Abstract
Over the past several years, there have been major advances in cardiovascular positron emission tomography (PET) in combination with either computed tomography (CT) or, more recently, cardiovascular magnetic resonance (CMR). These multi-modality approaches have significant potential to leverage the strengths of each modality to improve the characterization of a variety of cardiovascular diseases and to predict clinical outcomes. This review will discuss current developments and potential future uses of PET/CT and PET/CMR for cardiovascular applications, which promise to add significant incremental benefits to the data provided by each modality alone.
Collapse
|
47
|
Singh RB, Dandekar SP, Elimban V, Gupta SK, Dhalla NS. Role of proteases in the pathophysiology of cardiac disease. Mol Cell Biochem 2016; 263:241-56. [PMID: 27520682 DOI: 10.1023/b:mcbi.0000041865.63445.40] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease is a major cause of death and thus a great deal of effort has been made in salvaging the diseased myocardium. Although various factors have been identified as possible causes of different cardiac diseases such as heart failure and ischemic heart disease, there is a real need to elucidate their role for the better understanding of the cardiac disease pathology and formulation of strategies for developing newer therapeutic interventions. In view of the intimate involvement of different types of proteases in maintaining cellular structure, the role of proteases in various cardiac diseases has become the focus of recent research. Proteases are present in the cytosol as well as are localized in a number of subcellular organelles in the cell. These are known to use extracellular matrix, cytoskeletal, sarcolemmal, sarcoplasmic reticular, mitochondrial and myofibrillar proteins as substrates. Work from different laboratories using a wide variety of techniques has shown that the activation of proteases causes alterations of a number of specific proteins leading to subcellular remodeling and cardiac dysfunction. Inhibition of protease action by different drugs and agents, therefore, has a clinical relevance and is expected to form a part of new treatment paradigm for improving heart function. This review examines the biochemistry and localization of some of the proteases in the cardiac tissue in addition to identification of the sites of action of some protease inhibitors. (Mol Cell Biochem 263: 241-256, 2004).
Collapse
Affiliation(s)
- Raja B Singh
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, Canada R2H 2A6
| | - Sucheta P Dandekar
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, Canada R2H 2A6
| | - Vijayan Elimban
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, Canada R2H 2A6
| | - Suresh K Gupta
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, Canada R2H 2A6
| | - Naranjan S Dhalla
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, Canada R2H 2A6
| |
Collapse
|
48
|
Parkar NA, Bhatt LK, Addepalli V. Efficacy of nobiletin, a citrus flavonoid, in the treatment of the cardiovascular dysfunction of diabetes in rats. Food Funct 2016; 7:3121-9. [DOI: 10.1039/c6fo00294c] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nobiletin, due to its MMP-2 & MMP-9 inhibitory and antioxidant effects, may ameliorate the cardiovascular dysfunction of diabetes.
Collapse
Affiliation(s)
- N. A. Parkar
- Department of Pharmacology
- SPP School of Pharmacy and Technology Management
- NMIMS University
- Mumbai
- India
| | - L. K. Bhatt
- Dr Bhanuben Nanavati College of Pharmacy
- Mumbai
- India
| | - V. Addepalli
- Department of Pharmacology
- SPP School of Pharmacy and Technology Management
- NMIMS University
- Mumbai
- India
| |
Collapse
|
49
|
Chen C, Li R, Ross RS, Manso AM. Integrins and integrin-related proteins in cardiac fibrosis. J Mol Cell Cardiol 2015; 93:162-74. [PMID: 26562414 DOI: 10.1016/j.yjmcc.2015.11.010] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 11/07/2015] [Accepted: 11/07/2015] [Indexed: 12/21/2022]
Abstract
Cardiac fibrosis is one of the major components of the healing mechanism following any injury of the heart and as such may contribute to both systolic and diastolic dysfunction in a range of pathophysiologic conditions. Canonically, it can occur as part of the remodeling process that occurs following myocardial infarction or that follows as a response to pressure overload. Integrins are cell surface receptors which act in both cellular adhesion and signaling. Most importantly, in the context of the continuously contracting myocardium, they are recognized as mechanotransducers. They have been implicated in the development of fibrosis in several organs, including the heart. This review will focus on the involvement of integrins and integrin-related proteins, in cardiac fibrosis, outlining the roles of these proteins in the fibrotic responses in specific cardiac pathologies, discuss some of the common end effectors (angiotensin II, transforming growth factor beta 1 and mechanical stress) through which integrins function and finally discuss how manipulation of this set of proteins may lead to new treatments which could prove useful to alter the deleterious effects of cardiac fibrosis.
Collapse
Affiliation(s)
- Chao Chen
- Department of Medicine, Cardiology, UCSD School of Medicine, La Jolla, CA 92093-0613, USA; Veterans Administration San Diego Healthcare System, San Diego, CA 92161, USA.
| | - Ruixia Li
- Department of Medicine, Cardiology, UCSD School of Medicine, La Jolla, CA 92093-0613, USA; Veterans Administration San Diego Healthcare System, San Diego, CA 92161, USA.
| | - Robert S Ross
- Department of Medicine, Cardiology, UCSD School of Medicine, La Jolla, CA 92093-0613, USA; Veterans Administration San Diego Healthcare System, San Diego, CA 92161, USA.
| | - Ana Maria Manso
- Department of Medicine, Cardiology, UCSD School of Medicine, La Jolla, CA 92093-0613, USA; Veterans Administration San Diego Healthcare System, San Diego, CA 92161, USA.
| |
Collapse
|
50
|
|