1
|
Bhave S, Swain L, Qiao X, Martin G, Aryaputra T, Everett K, Kapur NK. ALK1 Deficiency Impairs the Wound-Healing Process and Increases Mortality in Murine Model of Myocardial Infarction. J Cardiovasc Transl Res 2024; 17:496-504. [PMID: 38064044 DOI: 10.1007/s12265-023-10471-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 11/24/2023] [Indexed: 07/03/2024]
Abstract
The functional role of TGFβ type I receptor, activin-like kinase (ALK)-1 in post-myocardial infarction (MI) cardiac remodeling is unknown. We hypothesize that reduced ALK1 activity reduces survival and promotes cardiac fibrosis after MI. MI was induced in wild-type (WT), and ALK+/- mice by left coronary ligation. After 14 days ALK1+/- mice had reduced survival with a higher rate of cardiac rupture compared to WT mice. ALK1+/- left ventricles (LVs) had increased volumes at the end of systole and at the end of diastole. After MI ALK1+/- LVs had increased profibrotic SMAD3 signaling, type 1 collagen, and fibrosis as well as increased levels of TGFβ1 co-receptor, endoglin, VEGF, and ALK1 ligands BMP9 and BMP10. ALK1+/- LVs had decreased levels of stromal-derived factor 1α. These data identify the critical role of ALK1 in post-MI survival and cardiac remodeling and implicate ALK1 as a potential therapeutic target to improve survival after MI.
Collapse
Affiliation(s)
- Shreyas Bhave
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, 02111, USA
| | - Lija Swain
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, 02111, USA
| | - Xiaoying Qiao
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, 02111, USA
| | - Gregory Martin
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, 02111, USA
| | - Tejasvi Aryaputra
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, 02111, USA
| | - Kay Everett
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, 02111, USA
| | - Navin K Kapur
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, 02111, USA.
| |
Collapse
|
2
|
Yaghoobi A, Rezaee M, Behnoush AH, Khalaji A, Mafi A, Houjaghan AK, Masoudkabir F, Pahlavan S. Role of long noncoding RNAs in pathological cardiac remodeling after myocardial infarction: An emerging insight into molecular mechanisms and therapeutic potential. Biomed Pharmacother 2024; 172:116248. [PMID: 38325262 DOI: 10.1016/j.biopha.2024.116248] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/29/2024] [Accepted: 02/01/2024] [Indexed: 02/09/2024] Open
Abstract
Myocardial infarction (MI) is the leading cause of heart failure (HF), accounting for high mortality and morbidity worldwide. As a consequence of ischemia/reperfusion injury during MI, multiple cellular processes such as oxidative stress-induced damage, cardiomyocyte death, and inflammatory responses occur. In the next stage, the proliferation and activation of cardiac fibroblasts results in myocardial fibrosis and HF progression. Therefore, developing a novel therapeutic strategy is urgently warranted to restrict the progression of pathological cardiac remodeling. Recently, targeting long non-coding RNAs (lncRNAs) provided a novel insight into treating several disorders. In this regard, numerous investigations have indicated that several lncRNAs could participate in the pathogenesis of MI-induced cardiac remodeling, suggesting their potential therapeutic applications. In this review, we summarized lncRNAs displayed in the pathophysiology of cardiac remodeling after MI, emphasizing molecular mechanisms. Also, we highlighted the possible translational role of lncRNAs as therapeutic targets for this condition and discussed the potential role of exosomes in delivering the lncRNAs involved in post-MI cardiac remodeling.
Collapse
Affiliation(s)
- Alireza Yaghoobi
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Malihe Rezaee
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Behnoush
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirmohammad Khalaji
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Mafi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Farzad Masoudkabir
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
3
|
Ding J, Lian J, Wang J, Yang S, Li H, Shen H, Sun Q, Li X, Chen G. The role of Tenascin C in intracerebral hemorrhage-induced secondary brain injury in rats via induction of neuronal cell death and neuroinflammation. J Chem Neuroanat 2022; 125:102147. [PMID: 36028204 DOI: 10.1016/j.jchemneu.2022.102147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 08/18/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Spontaneous intracerebral hemorrhage (ICH) is a major cause of stroke that causes high rates of disability and mortality in adults. Tenascin C (TNC) protein, one of the matricellular proteins associated with platelet-derived growth factor receptor (PDGFR) activation, has been reported to induce neuronal apoptosis. However, the role and underlying mechanisms of TNC in ICH-induced secondary brain injury (SBI) have not yet been fully explained. The main purpose of this study was to explore the role of TNC and its potential mechanisms in ICH. METHODS An ICH model was established by injecting autologous blood into the right basal ganglia in male Sprague Dawley (SD) rats, and imatinib, an inhibitor of PDGFR, was used to inhibit the release of TNC. RESULTS We found that TNC protein was significantly increased in the brain tissues after ICH and expressed in both neurons and microglia. We also found that the TNC level was elevated in the cerebrospinal fluid (CSF) after ICH. Additionally, we observed that the infiltration of activated microglia and the release of TNFα and IL-1β induced by ICH were decreased after inhibition of the protein levels of TNC and cleaved-TNC by a chemical inhibitor (imatinib). Furthermore, imatinib improved neuronal cell death and neurobehavioral abnormalities induced by ICH. CONCLUSION In summary, our study revealed that TNC protein plays an important role in ICH-induced SBI, and inhibition of TNC could alleviate ICH-induced neuroinflammation, neuronal cell death, and neurobehaviour. Therefore, TNC may be a potential therapeutic target for ICH-induced SBI.
Collapse
Affiliation(s)
- Jiasheng Ding
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Institute of Stroke Research, Soochow University, China
| | - Jinrong Lian
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Institute of Stroke Research, Soochow University, China
| | - Jiahe Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Institute of Stroke Research, Soochow University, China
| | - Siyuan Yang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Institute of Stroke Research, Soochow University, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Institute of Stroke Research, Soochow University, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Institute of Stroke Research, Soochow University, China
| | - Qing Sun
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Institute of Stroke Research, Soochow University, China.
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Institute of Stroke Research, Soochow University, China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Institute of Stroke Research, Soochow University, China
| |
Collapse
|
4
|
Feng D, Gao P, Henley N, Dubuissez M, Chen N, Laurin LP, Royal V, Pichette V, Gerarduzzi C. SMOC2 promotes an epithelial-mesenchymal transition and a pro-metastatic phenotype in epithelial cells of renal cell carcinoma origin. Cell Death Dis 2022; 13:639. [PMID: 35869056 PMCID: PMC9307531 DOI: 10.1038/s41419-022-05059-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 06/22/2022] [Accepted: 07/01/2022] [Indexed: 01/21/2023]
Abstract
Renal Cell Carcinoma (RCC) is the most common form of all renal cancer cases, and well-known for its highly aggressive metastatic behavior. SMOC2 is a recently described non-structural component of the extracellular matrix (ECM) that is highly expressed during tissue remodeling processes with emerging roles in cancers, yet its role in RCC remains elusive. Using gene expression profiles from patient samples, we identified SMOC2 as being significantly expressed in RCC tissue compared to normal renal tissue, which correlated with shorter RCC patient survival. Specifically, de novo protein synthesis of SMOC2 was shown to be much higher in the tubular epithelial cells of patients with biopsy-proven RCC. More importantly, we provide evidence of SMOC2 triggering kidney epithelial cells into an epithelial-to-mesenchymal transition (EMT), a phenotype known to promote metastasis. We found that SMOC2 induced mesenchymal-like morphology and activities in both RCC and non-RCC kidney epithelial cell lines. Mechanistically, treatment of RCC cell lines ACHN and 786-O with SMOC2 (recombinant and enforced expression) caused a significant increase in EMT-markers, -matrix production, -proliferation, and -migration, which were inhibited by targeting SMOC2 by siRNA. We further characterized SMOC2 activation of EMT to occur through the integrin β3, FAK and paxillin pathway. The proliferation and metastatic potential of SMOC2 overexpressing ACHN and 786-O cell lines were validated in vivo by their significantly higher tumor growth in kidneys and systemic dissemination into other organs when compared to their respective controls. In principle, understanding the impact that SMOC2 has on EMT may lead to more evidence-based treatments and biomarkers for RCC metastasis.
Collapse
Affiliation(s)
- Daniel Feng
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Centre affilié à l'Université de Montréal, Montréal, Québec, Canada
| | - Peng Gao
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Centre affilié à l'Université de Montréal, Montréal, Québec, Canada
| | - Nathalie Henley
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Centre affilié à l'Université de Montréal, Montréal, Québec, Canada
| | - Marion Dubuissez
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Centre affilié à l'Université de Montréal, Montréal, Québec, Canada
| | - Nan Chen
- Faculty of Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - Louis-Philippe Laurin
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Centre affilié à l'Université de Montréal, Montréal, Québec, Canada
| | - Virginie Royal
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Centre affilié à l'Université de Montréal, Montréal, Québec, Canada
| | - Vincent Pichette
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Centre affilié à l'Université de Montréal, Montréal, Québec, Canada
- Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Casimiro Gerarduzzi
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada.
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Centre affilié à l'Université de Montréal, Montréal, Québec, Canada.
- Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
5
|
He Z, Zeng X, Zhou D, Liu P, Han D, Xu L, Bu T, Wang J, Ke M, Pan X, Du Y, Xue H, Lu D, Luo B. LncRNA Chaer Prevents Cardiomyocyte Apoptosis From Acute Myocardial Infarction Through AMPK Activation. Front Pharmacol 2021; 12:649398. [PMID: 34335241 PMCID: PMC8322763 DOI: 10.3389/fphar.2021.649398] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/04/2021] [Indexed: 11/30/2022] Open
Abstract
Long non-coding RNA (lncRNA) is widely reported to be involved in cardiac (patho)physiology. Acute myocardial infarction, in which cardiomyocyte apoptosis plays an important role, is a life-threatening disease. Here, we report the lncRNA Chaer that is anti-apoptotic in cardiomyocytes during Acute myocardial infarction. Importantly, lncRNA Chaer is significantly downregulated in both oxygen-glucose deprivation (oxygen-glucose deprivation)-treated cardiomyocytes in vitro and AMI heart. In vitro, overexpression of lncRNA Chaer with adeno virus reduces cardiomyocyte apoptosis induced by OGD-treated while silencing of lncRNA Chaer increases cardiomyocyte apoptosis instead. In vivo, forced expression of lncRNA Chaer with AAV9 attenuates cardiac apoptosis, reduces infarction area and improves mice heart function in AMI. Interestingly, overexpression of lncRNA Chaer promotes the phosphorylation of AMPK, and AMPK inhibitor Compound C reverses the overexpression of lncRNA Chaer effect of reducing cardiomyocyte apoptosis under OGD-treatment. In summary, we identify the novel ability of lncRNA Chaer in regulating cardiomyocyte apoptosis by promoting phosphorylation of AMPK in AMI.
Collapse
Affiliation(s)
- Zhiyu He
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaojun Zeng
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Deke Zhou
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Traditional Chinese Medicine Hospital of Gaozhou, Department of Cardiology, Gaozhou, China
| | - Peiying Liu
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dunzheng Han
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lingling Xu
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Tong Bu
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jinping Wang
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mengmeng Ke
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiudi Pan
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yipeng Du
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hao Xue
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dongfeng Lu
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bihui Luo
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
6
|
Kuo LT, Huang APH. The Pathogenesis of Hydrocephalus Following Aneurysmal Subarachnoid Hemorrhage. Int J Mol Sci 2021; 22:ijms22095050. [PMID: 34068783 PMCID: PMC8126203 DOI: 10.3390/ijms22095050] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/29/2021] [Accepted: 04/29/2021] [Indexed: 12/11/2022] Open
Abstract
Hydrocephalus is a common complication of aneurysmal subarachnoid hemorrhage (aSAH) and reportedly contributes to poor neurological outcomes. In this review, we summarize the molecular and cellular mechanisms involved in the pathogenesis of hydrocephalus following aSAH and summarize its treatment strategies. Various mechanisms have been implicated for the development of chronic hydrocephalus following aSAH, including alterations in cerebral spinal fluid (CSF) dynamics, obstruction of the arachnoid granulations by blood products, and adhesions within the ventricular system. Regarding molecular mechanisms that cause chronic hydrocephalus following aSAH, we carried out an extensive review of animal studies and clinical trials about the transforming growth factor-β/SMAD signaling pathway, upregulation of tenascin-C, inflammation-dependent hypersecretion of CSF, systemic inflammatory response syndrome, and immune dysregulation. To identify the ideal treatment strategy, we discuss the predictive factors of shunt-dependent hydrocephalus between surgical clipping and endovascular coiling groups. The efficacy and safety of other surgical interventions including the endoscopic removal of an intraventricular hemorrhage, placement of an external ventricular drain, the use of intraventricular or cisternal fibrinolysis, and an endoscopic third ventriculostomy on shunt dependency following aSAH were also assessed. However, the optimal treatment is still controversial, and it necessitates further investigations. A better understanding of the pathogenesis of acute and chronic hydrocephalus following aSAH would facilitate the development of treatments and improve the outcome.
Collapse
|
7
|
Schotman MJG, Peters MMC, Krijger GC, van Adrichem I, de Roos R, Bemelmans JLM, Pouderoijen MJ, Rutten MGTA, Neef K, Chamuleau SAJ, Dankers PYW. In Vivo Retention Quantification of Supramolecular Hydrogels Engineered for Cardiac Delivery. Adv Healthc Mater 2021; 10:e2001987. [PMID: 33586317 PMCID: PMC11468640 DOI: 10.1002/adhm.202001987] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/30/2020] [Indexed: 12/15/2022]
Abstract
Recent advances in the field of cardiac regeneration show great potential in the use of injectable hydrogels to reduce immediate flush-out of injected factors, thereby increasing the effectiveness of the encapsulated drugs. To establish a relation between cardiac function and retention of the drug-encapsulating hydrogel, a quantitative in vivo imaging method is required. Here, the supramolecular ureido-pyrimidinone modified poly(ethylene glycol) (UPy-PEG) material is developed into a bioactive hydrogel for radioactive imaging in a large animal model. A radioactive label is synthesized, being a ureido-pyrimidinone moiety functionalized with a chelator (UPy-DOTA) complexed with the radioactive isotope indium-111 (UPy-DOTA-111 In) that is mixed with the hydrogel. Additionally, bioactive and adhesive properties of the UPy-PEG hydrogel are increased by supramolecular introduction of a UPy-functionalized recombinant collagen type 1-based material (UPy-PEG-RCPhC1). This method enables in vivo tracking of the nonbioactive and bioactive supramolecular hydrogels and quantification of hydrogel retention in a porcine heart. In a small pilot, cardiac retention values of 8% for UPy-PEG and 16% for UPy-PEG-RCPhC1 hydrogel are observed 4 h postinjection. This work highlights the importance of retention quantification of hydrogels in vivo, where elucidation of hydrogel quantity at the target site is proposed to strongly influence efficacy of the intended therapy.
Collapse
Affiliation(s)
- Maaike J. G. Schotman
- Institute for Complex Molecular SystemsLaboratory of Chemical BiologyDepartment of Biomedical EngineeringEindhoven University of TechnologyGroene Loper 7Eindhoven5612 AZThe Netherlands
| | - Marijn M. C. Peters
- Department of CardiologyExperimental Cardiology LaboratoryUMC Utrecht Regenerative Medicine CentreUniversity Medical Centre UtrechtUniversity UtrechtHeidelberglaan 100Utrecht3584 CXThe Netherlands
| | - Gerard C. Krijger
- Department of Nuclear MedicineUniversity Medical Centre UtrechtUtrecht3584 CXThe Netherlands
| | - Iris van Adrichem
- Department of CardiologyExperimental Cardiology LaboratoryUMC Utrecht Regenerative Medicine CentreUniversity Medical Centre UtrechtUniversity UtrechtHeidelberglaan 100Utrecht3584 CXThe Netherlands
| | - Remmert de Roos
- Department of Nuclear MedicineUniversity Medical Centre UtrechtUtrecht3584 CXThe Netherlands
| | - John L. M. Bemelmans
- Department of Nuclear MedicineUniversity Medical Centre UtrechtUtrecht3584 CXThe Netherlands
| | | | - Martin G. T. A. Rutten
- Institute for Complex Molecular SystemsLaboratory of Chemical BiologyDepartment of Biomedical EngineeringEindhoven University of TechnologyGroene Loper 7Eindhoven5612 AZThe Netherlands
| | - Klaus Neef
- Department of CardiologyExperimental Cardiology LaboratoryUMC Utrecht Regenerative Medicine CentreUniversity Medical Centre UtrechtUniversity UtrechtHeidelberglaan 100Utrecht3584 CXThe Netherlands
| | - Steven A. J. Chamuleau
- Department of CardiologyExperimental Cardiology LaboratoryUMC Utrecht Regenerative Medicine CentreUniversity Medical Centre UtrechtUniversity UtrechtHeidelberglaan 100Utrecht3584 CXThe Netherlands
| | - Patricia Y. W. Dankers
- Institute for Complex Molecular SystemsLaboratory of Chemical BiologyLaboratory for Cell and Tissue EngineeringDepartment of Biomedical EngineeringEindhoven University of TechnologyGroene Loper 7Eindhoven5612 AZThe Netherlands
| |
Collapse
|
8
|
Huizer K, Zhu C, Chirifi I, Krist B, Zorgman D, van der Weiden M, van den Bosch TPP, Dumas J, Cheng C, Kros JM, Mustafa DA. Periostin Is Expressed by Pericytes and Is Crucial for Angiogenesis in Glioma. J Neuropathol Exp Neurol 2021; 79:863-872. [PMID: 32647861 DOI: 10.1093/jnen/nlaa067] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/12/2020] [Accepted: 06/11/2020] [Indexed: 12/13/2022] Open
Abstract
The expression of the matricellular protein periostin has been associated with glioma progression. In previous work we found an association of periostin with glioma angiogenesis. Here, we screen gliomas for POSTN expression and identify the cells that express periostin in human gliomas. In addition, we study the role of periostin in an in vitro model for angiogenesis. The expression of periostin was investigated by RT-PCR and by immunohistochemistry. In addition, we used double labeling and in situ RNA techniques to identify the expressing cells. To investigate the function of periostin, we silenced POSTN in a 3D in vitro angiogenesis model. Periostin expression was elevated in pilocytic astrocytoma and glioblastoma, but not in grade II/III astrocytomas and oligodendrogliomas. The expression of periostin colocalized with PDGFRβ+ cells, but not with OLIG2+/SOX2+ glioma stem cells. Silencing of periostin in pericytes in coculture experiments resulted in attenuation of the numbers and the length of the vessels formation and in a decrease in endothelial junction formation. We conclude that pericytes are the main source of periostin in human gliomas and that periostin plays an essential role in the growth and branching of blood vessels. Therefore, periostin should be explored as a novel target for developing anti-angiogenic therapy for glioma.
Collapse
Affiliation(s)
- Karin Huizer
- From the Laboratory for Tumor Immunopathology, Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Changbin Zhu
- From the Laboratory for Tumor Immunopathology, Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ihsan Chirifi
- Laboratory for Experimental Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Bart Krist
- From the Laboratory for Tumor Immunopathology, Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Denise Zorgman
- From the Laboratory for Tumor Immunopathology, Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marcel van der Weiden
- From the Laboratory for Tumor Immunopathology, Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Thierry P P van den Bosch
- From the Laboratory for Tumor Immunopathology, Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jasper Dumas
- From the Laboratory for Tumor Immunopathology, Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Caroline Cheng
- Laboratory for Experimental Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Johan M Kros
- From the Laboratory for Tumor Immunopathology, Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Dana A Mustafa
- From the Laboratory for Tumor Immunopathology, Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
9
|
de Wit L, Fang J, Neef K, Xiao J, A. Doevendans P, Schiffelers RM, Lei Z, Sluijter JP. Cellular and Molecular Mechanism of Cardiac Regeneration: A Comparison of Newts, Zebrafish, and Mammals. Biomolecules 2020; 10:biom10091204. [PMID: 32825069 PMCID: PMC7564143 DOI: 10.3390/biom10091204] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/06/2020] [Accepted: 08/17/2020] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease is the leading cause of death worldwide. Current palliative treatments can slow the progression of heart failure, but ultimately, the only curative treatment for end-stage heart failure is heart transplantation, which is only available for a minority of patients due to lack of donors' hearts. Explorative research has shown the replacement of the damaged and lost myocardium by inducing cardiac regeneration from preexisting myocardial cells. Lower vertebrates, such as the newt and zebrafish, can regenerate lost myocardium through cardiomyocyte proliferation. The preexisting adult cardiomyocytes replace the lost cells through subsequent dedifferentiation, proliferation, migration, and re-differentiation. Similarly, neonatal mice show complete cardiac regeneration post-injury; however, this regenerative capacity is remarkably diminished one week after birth. In contrast, the adult mammalian heart presents a fibrotic rather than a regenerative response and only shows signs of partial pathological cardiomyocyte dedifferentiation after injury. In this review, we explore the cellular and molecular responses to myocardial insults in different adult species to give insights for future interventional directions by which one can promote or activate cardiac regeneration in mammals.
Collapse
Affiliation(s)
- Lousanne de Wit
- Department of Cardiology, Experimental Cardiology Laboratory, UMC Utrecht, 3584CX Utrecht, The Netherlands; (L.d.W.); (J.F.); (K.N.); (P.A.D.)
| | - Juntao Fang
- Department of Cardiology, Experimental Cardiology Laboratory, UMC Utrecht, 3584CX Utrecht, The Netherlands; (L.d.W.); (J.F.); (K.N.); (P.A.D.)
| | - Klaus Neef
- Department of Cardiology, Experimental Cardiology Laboratory, UMC Utrecht, 3584CX Utrecht, The Netherlands; (L.d.W.); (J.F.); (K.N.); (P.A.D.)
- UMC Utrecht RM Center, Circulatory Health Laboratory, 3584CT Utrecht, The Netherlands
| | - Junjie Xiao
- Institute of Cardiovascular Sciences, Shanghai University, Shanghai 200444, China;
| | - Pieter A. Doevendans
- Department of Cardiology, Experimental Cardiology Laboratory, UMC Utrecht, 3584CX Utrecht, The Netherlands; (L.d.W.); (J.F.); (K.N.); (P.A.D.)
- Utrecht University, 3584CS Utrecht, The Netherlands
- Netherlands Heart Institute (NHI), Central Military Hospital (CMH), 3511EP Utrecht, The Netherlands
| | | | - Zhiyong Lei
- Department of Cardiology, Experimental Cardiology Laboratory, UMC Utrecht, 3584CX Utrecht, The Netherlands; (L.d.W.); (J.F.); (K.N.); (P.A.D.)
- Division LAB, CDL Research, UMC Utrecht, 3584CX Utrecht, The Netherlands;
- Correspondence: (Z.L.); (J.P.G.S.)
| | - Joost P.G. Sluijter
- Department of Cardiology, Experimental Cardiology Laboratory, UMC Utrecht, 3584CX Utrecht, The Netherlands; (L.d.W.); (J.F.); (K.N.); (P.A.D.)
- UMC Utrecht RM Center, Circulatory Health Laboratory, 3584CT Utrecht, The Netherlands
- Utrecht University, 3584CS Utrecht, The Netherlands
- Correspondence: (Z.L.); (J.P.G.S.)
| |
Collapse
|
10
|
Dragasevic N, Jakovljevic V, Zivkovic V, Draginic N, Andjic M, Bolevich S, Jovic S. The role of aldosterone inhibitors in cardiac ischemia-reperfusion injury. Can J Physiol Pharmacol 2020; 99:18-29. [PMID: 32799671 DOI: 10.1139/cjpp-2020-0276] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Myocardial ischaemia-reperfusion (I/R) injury is a well-known term for exacerbation of cellular destruction and dysfunction after the restoration of blood flow to a previously ischaemic heart. A vast number of studies that have demonstrated that the role of mineralocorticoids in cardiovascular diseases is based on the use of pharmacological mineralocorticoid receptor (MR) antagonists. This review paper aimed to summarize current knowledge on the effects of MR antagonists on myocardial I/R injury as well as postinfarction remodeling. Animal models, predominantly the Langendorff technique and left anterior descending coronary artery occlusion, have confirmed the potency of MR antagonists as preconditioning and postconditioning agents in limiting infarct size and postinfarction remodeling. Several preclinical studies in rodents have established and proved possible mechanisms of cardioprotection by MR antagonists, such as reduction of oxidative stress, reduction of inflammation, and apoptosis, therefore limiting the infarct zone. However, the results of some clinical trials are inconsistent, since they reported no benefit of MR antagonists in acute myocardial infarction. Due to this, further studies and the results of ongoing clinical trials regarding MR antagonist administration in patients with acute myocardial infarction are being awaited with great interest.
Collapse
Affiliation(s)
- Nevena Dragasevic
- University of Kragujevac, Faculty of Medical Sciences, Department of Physiology, Svetozara Markovica 69, 34 000 Kragujevac, Serbia
| | - Vladimir Jakovljevic
- University of Kragujevac, Faculty of Medical Sciences, Department of Physiology, Svetozara Markovica 69, 34 000 Kragujevac, Serbia.,1st Moscow State Medical University IM Sechenov, Department of Human Pathology, Trubetskaya street 8, 119991 Moscow, Russia
| | - Vladimir Zivkovic
- University of Kragujevac, Faculty of Medical Sciences, Department of Physiology, Svetozara Markovica 69, 34 000 Kragujevac, Serbia
| | - Nevena Draginic
- University of Kragujevac, Faculty of Medical Sciences, Department of Pharmacy, Svetozara Markovica 69, 34 000 Kragujevac, Serbia
| | - Marijana Andjic
- University of Kragujevac, Faculty of Medical Sciences, Department of Pharmacy, Svetozara Markovica 69, 34 000 Kragujevac, Serbia
| | - Sergey Bolevich
- 1 Moscow State Medical University IM Sechenov, Department of Human Pathology, Trubetskaya street 8, 119991 Moscow, Russia
| | - Slavoljub Jovic
- University of Belgrade, Department of Physiology and Biochemistry, Faculty of Veterinary Medicine, Bul. Oslobodjenja 18, Belgrade, Serbia
| |
Collapse
|
11
|
Duan FM, Fu LJ, Wang YH, Adu-Gyamfi EA, Ruan LL, Xu ZW, Xiao SQ, Chen XM, Wang YX, Liu TH, Ding YB. THBS1 regulates trophoblast fusion through a CD36-dependent inhibition of cAMP, and its upregulation participates in preeclampsia. Genes Dis 2020; 8:353-363. [PMID: 33997182 PMCID: PMC8093648 DOI: 10.1016/j.gendis.2020.05.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 05/11/2020] [Accepted: 05/29/2020] [Indexed: 02/06/2023] Open
Abstract
Preeclampsia is a pregnancy complication which threatens the survival of mothers and fetuses. It originates from abnormal placentation, especially insufficient fusion of the cytotrophoblast cells to form the syncytiotrophoblast. In this study, we found that THBS1, a matricellular protein that mediates cell-to-cell and cell-to-matrix interactions, is downregulated during the fusion of primary cytotrophoblast and BeWo cells, but upregulated in the placenta of pregnancies complicated by preeclampsia. Also, THBS1 was observed to interact with CD36, a membrane signal receptor and activator of the cAMP signaling pathway, to regulate the fusion of cytotrophoblast cells. Overexpression of THBS1 inhibited the cAMP signaling pathway and reduced the BeWo cells fusion ratio, while the effects of THBS1 were abolished by a CD36-blocking antibody. Our results suggest that THBS1 signals through a CD36-mediated cAMP pathway to regulate syncytialization of the cytotrophoblast cells, and that its upregulation impairs placental formation to cause preeclampsia. Thus, THBS1 can serve as a therapeutic target regarding the mitigation of abnormal syncytialization and preeclampsia.
Collapse
Affiliation(s)
- Fu-Mei Duan
- School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China
| | - Li-Juan Fu
- Department of Herbal Medicine, School of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yong-Heng Wang
- The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, Chongqing, 400016, PR China
| | - Enoch Appiah Adu-Gyamfi
- The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, Chongqing, 400016, PR China
| | - Ling-Ling Ruan
- Department of Herbal Medicine, School of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400016, PR China
| | - Zeng-Wei Xu
- School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China
| | - Shi-Quan Xiao
- The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, Chongqing, 400016, PR China.,Department of Reproductive Medicine, The Third Affiliated Hospital, Chongqing Medical University, Chongqing, 401120, PR China
| | - Xue-Mei Chen
- School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China.,The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, Chongqing, 400016, PR China
| | - Ying-Xiong Wang
- School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China.,The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, Chongqing, 400016, PR China
| | - Tai-Hang Liu
- Department of Bioinformatics, The School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, PR China.,The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, Chongqing, 400016, PR China
| | - Yu-Bin Ding
- School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China.,The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, Chongqing, 400016, PR China
| |
Collapse
|
12
|
Ushakov A, Ivanchenko V, Gagarina A. Regulation of Myocardial Extracellular Matrix Dynamic Changes in Myocardial Infarction and Postinfarct Remodeling. Curr Cardiol Rev 2020; 16:11-24. [PMID: 31072294 PMCID: PMC7393593 DOI: 10.2174/1573403x15666190509090832] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/22/2019] [Accepted: 04/29/2019] [Indexed: 02/07/2023] Open
Abstract
The article represents literature review dedicated to molecular and cellular mechanisms underlying clinical manifestations and outcomes of acute myocardial infarction. Extracellular matrix adaptive changes are described in detail as one of the most important factors contributing to healing of damaged myocardium and post-infarction cardiac remodeling. Extracellular matrix is reviewed as dynamic constantly remodeling structure that plays a pivotal role in myocardial repair. The role of matrix metalloproteinases and their tissue inhibitors in fragmentation and degradation of extracellular matrix as well as in myocardium healing is discussed. This review provides current information about fibroblasts activity, the role of growth factors, particularly transforming growth factor β and cardiotrophin-1, colony-stimulating factors, adipokines and gastrointestinal hormones, various matricellular proteins. In conclusion considering the fact that dynamic transformation of extracellular matrix after myocardial ischemic damage plays a pivotal role in myocardial infarction outcomes and prognosis, we suggest a high importance of further investigation of mechanisms underlying extracellular matrix remodeling and cell-matrix interactions in cardiovascular diseases.
Collapse
Affiliation(s)
- Alexey Ushakov
- Department of Internal Medicine #1 with Clinical Pharmacology Course, Medical Academy named after S.I. Georgievsky of V.I. Vernadsky Crimean Federal University, Simferopol, Russian Federation
| | - Vera Ivanchenko
- Department of Internal Medicine #1 with Clinical Pharmacology Course, Medical Academy named after S.I. Georgievsky of V.I. Vernadsky Crimean Federal University, Simferopol, Russian Federation
| | - Alina Gagarina
- Department of Internal Medicine #1 with Clinical Pharmacology Course, Medical Academy named after S.I. Georgievsky of V.I. Vernadsky Crimean Federal University, Simferopol, Russian Federation
| |
Collapse
|
13
|
Gerarduzzi C, Hartmann U, Leask A, Drobetsky E. The Matrix Revolution: Matricellular Proteins and Restructuring of the Cancer Microenvironment. Cancer Res 2020; 80:2705-2717. [PMID: 32193287 DOI: 10.1158/0008-5472.can-18-2098] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/04/2019] [Accepted: 03/17/2020] [Indexed: 11/16/2022]
Abstract
The extracellular matrix (ECM) surrounding cells is indispensable for regulating their behavior. The dynamics of ECM signaling are tightly controlled throughout growth and development. During tissue remodeling, matricellular proteins (MCP) are secreted into the ECM. These factors do not serve classical structural roles, but rather regulate matrix proteins and cell-matrix interactions to influence normal cellular functions. In the tumor microenvironment, it is becoming increasingly clear that aberrantly expressed MCPs can support multiple hallmarks of carcinogenesis by interacting with various cellular components that are coupled to an array of downstream signals. Moreover, MCPs also reorganize the biomechanical properties of the ECM to accommodate metastasis and tumor colonization. This realization is stimulating new research on MCPs as reliable and accessible biomarkers in cancer, as well as effective and selective therapeutic targets.
Collapse
Affiliation(s)
- Casimiro Gerarduzzi
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, Québec, Canada. .,Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Ursula Hartmann
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Andrew Leask
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Elliot Drobetsky
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, Québec, Canada.,Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
14
|
Understanding the mechanisms that determine extracellular matrix remodeling in the infarcted myocardium. Biochem Soc Trans 2020; 47:1679-1687. [PMID: 31724697 DOI: 10.1042/bst20190113] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 10/22/2019] [Accepted: 10/24/2019] [Indexed: 02/06/2023]
Abstract
Myocardial Infarction (MI) initiates a series of wound healing events that begins with up-regulation of an inflammatory response and culminates in scar formation. The extracellular matrix (ECM) is intricately involved in all stages from initial break down of existing ECM to synthesis of new ECM to form the scar. This review will summarize our current knowledge on the processes involved in ECM remodeling after MI and identify the gaps that still need to be filled.
Collapse
|
15
|
Żak MM, Gkontra P, Clemente C, Squadrito ML, Ferrarini A, Mota RA, Oliver E, Rocha S, Agüero J, Vázquez J, De Palma M, Ibáñez B, Arroyo AG. Sequential Bone-Marrow Cell Delivery of VEGFA/S1P Improves Vascularization and Limits Adverse Cardiac Remodeling After Myocardial Infarction in Mice. Hum Gene Ther 2019; 30:893-905. [DOI: 10.1089/hum.2018.194] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Affiliation(s)
- Magdalena M. Żak
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Polyxeni Gkontra
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Cristina Clemente
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Mario Leonardo Squadrito
- École Polytechnique Federale de Lausanne (EPFL), ISREC-Swiss Institute for Experimental Cancer Research, Lausanne, Switzerland
| | - Alessia Ferrarini
- Proteomics Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Rubén A. Mota
- Animal Facility, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Eduardo Oliver
- Myocardial Pathology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Susana Rocha
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Jaume Agüero
- Myocardial Pathology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER-CV, Madrid, Spain
| | - Jesús Vázquez
- Proteomics Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER-CV, Madrid, Spain
| | - Michele De Palma
- École Polytechnique Federale de Lausanne (EPFL), ISREC-Swiss Institute for Experimental Cancer Research, Lausanne, Switzerland
| | - Borja Ibáñez
- Myocardial Pathology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER-CV, Madrid, Spain
| | - Alicia G. Arroyo
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| |
Collapse
|
16
|
Chilukoti RK, Lendeckel J, Darm K, Bukowska A, Goette A, Sühling M, Utpatel K, Peters B, Homuth G, Völker U, Wolke C, Scharf C, Lendeckel U. Integration of "omics" techniques: Dronedarone affects cardiac remodeling in the infarction border zone. Exp Biol Med (Maywood) 2018; 243:895-910. [PMID: 30105952 PMCID: PMC6108048 DOI: 10.1177/1535370218788517] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/18/2018] [Indexed: 01/15/2023] Open
Abstract
Dronedarone improves microvascular flow during atrial fibrillation and reduces the infarct size in acute models of myocardial infarction. However, dronedarone might be harmful in patients with recent decompensated heart failure and increases mortality in patients with permanent atrial fibrillation. A pathophysiological explanation for these discrepant data is lacking. This study investigated the effects of dronedarone on gene and protein expression in the infarcted area and border zone in pigs subjected to anterior ischemia/reperfusion myocardial infarction. The ischemia/reperfusion myocardial infarction was induced in 16 pigs. Eight pigs were treated with dronedarone for 28 days after myocardial infarction, the remaining pigs served as control. Microarray-based transcriptome profiling and 2D-DIGE-based proteome analysis were used to assess the effects of dronedarone on left ventricular gene expression in healthy (LV), infarcted (MI), and border zone tissue. Selected targets were validated by RT-qPCR or immunoblot analyses, with special emphasize given to the transcriptome/proteome overlap. Combined "omics" analysis was performed to identify most significant disease and function charts affected by dronedarone and to establish an integrated network. The levels of 879 (BZ) or 7 (MI) transcripts and 51 (LV) or 15 (BZ) proteins were significantly altered by dronedarone, pointing to a substantial efficacy of dronedarone in the border zone. Transcriptome and proteome data indicate that dronedarone influences post-infarction remodeling processes and identify matricellular proteins as major targets of dronedarone in this setting. This finding is fully supported by the disease and function charts as well as by the integrated network established by combined "omics". Dronedarone therapy alters myocardial gene expression after acute myocardial infarction with pronounced effects in the border zone. Dronedarone promotes infarct healing via regulation of periostin and might contribute to the limitation of its expansion as well as cardiac rupture. Thus, there are no experimental hints that dronedarone per se has direct harmful effects after MI in ventricular tissue. Impact statement Dronedarone reduced the infarct size in models of acute myocardial infarction (MI). Here, we show that dronedarone attenuates many of the substantial changes in gene expression that are provoked by acute myocardial infarction (AMI) in pigs. Dronedarone modifies the expression of gene panels related to post-infarction cardiac healing and remodeling processes and, most remarkably, this occurs predominantly in the infarction border-zone and much less so in the vital or infarcted myocardium. Combined "omics" identified matricellular proteins and ECM as major dronedarone-regulated targets and emphasizes their relevance for Disease Charts and Tox Function Charts associated with tissue remodeling and cellular movement. The results demonstrate dronedarone's capability of regulating cardiac repair and remodeling processes specifically in the infarction border zone and identify underlying mechanisms and pathways that might be employed in future therapeutic strategies to improve long-term cardiac tissue function and stability.
Collapse
Affiliation(s)
- Ravi K Chilukoti
- Institute of Medical Biochemistry and Molecular Biology,
University Medicine Greifswald, Greifswald D-17475, Germany
| | - Josefine Lendeckel
- Institute of Medical Biochemistry and Molecular Biology,
University Medicine Greifswald, Greifswald D-17475, Germany
| | - Katrin Darm
- Department of Otorhinolaryngology, Head and Neck Surgery,
University Medicine Greifswald, Greifswald D-17475, Germany
| | - Alicja Bukowska
- Working Group: Molecular Electrophysiology, Otto-von-Guericke
University, University Hospital Magdeburg, Magdeburg D-39120, Germany
| | - Andreas Goette
- Working Group: Molecular Electrophysiology, Otto-von-Guericke
University, University Hospital Magdeburg, Magdeburg D-39120, Germany
- Department of Cardiology and Intensive Care Medicine, St.
Vincenz-Hospital, Paderborn D-33098, Germany
| | - Marc Sühling
- Institute of Medical Biochemistry and Molecular Biology,
University Medicine Greifswald, Greifswald D-17475, Germany
| | - Kirsten Utpatel
- Department of Pathology, University Medicine Greifswald,
Greifswald D-17475, Germany
| | - Barbara Peters
- Institute of Physiology, University Medicine Greifswald,
Karlsburg D-17495, Germany
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics,
University Medicine Greifswald, D-17475 Greifswald, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics,
University Medicine Greifswald, D-17475 Greifswald, Germany
| | - Carmen Wolke
- Institute of Medical Biochemistry and Molecular Biology,
University Medicine Greifswald, Greifswald D-17475, Germany
| | - Christian Scharf
- Department of Otorhinolaryngology, Head and Neck Surgery,
University Medicine Greifswald, Greifswald D-17475, Germany
| | - Uwe Lendeckel
- Institute of Medical Biochemistry and Molecular Biology,
University Medicine Greifswald, Greifswald D-17475, Germany
| |
Collapse
|
17
|
Deng J, Qian X, Li J, Li Y, Li Y, Luo Y. Evaluation of serum cysteine-rich protein 61 levels in patients with coronary artery disease. Biomark Med 2018; 12:329-339. [PMID: 29345157 DOI: 10.2217/bmm-2017-0390] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
AIM The aim is to evaluate serum cysteine-rich protein 61 (Cyr61) levels in patients with coronary artery disease (CAD). PATIENTS & METHODS Serum Cyr61 levels were measured in 180 patients with CAD and 74 participants without CAD. RESULTS Serum Cyr61 levels were significantly higher in CAD patients. Patients with acute coronary syndrome showed significantly higher Cyr61 than those with stable angina pectoris. Serum Cyr61 levels in complex lesion group were significantly higher. Serum Cyr61 was positively correlated with Gensini score and C-reactive protein. Multivariable logistic regression analyses demonstrated that serum Cyr61 levels were independently correlated with the existence of CAD (p = 0.01). CONCLUSION Our study suggested Cyr61 as a potential biomarker in characterizing CAD and therapeutic target for CAD.
Collapse
Affiliation(s)
- Jingang Deng
- Division of Cardiology, Shenzhen Tenth People's Hospital, Shenzhen, Guangdong, China
| | - Xiaoxian Qian
- Division of Cardiology, The Third Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jianping Li
- Division of Cardiology, Shenzhen Tenth People's Hospital, Shenzhen, Guangdong, China
| | - Yanghua Li
- Division of Cardiology, Shenzhen Tenth People's Hospital, Shenzhen, Guangdong, China
| | - Yang Li
- Division of Cardiology, Shenzhen Tenth People's Hospital, Shenzhen, Guangdong, China
| | - Yijun Luo
- Division of Cardiology, Shenzhen Tenth People's Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
18
|
Stujanna EN, Murakoshi N, Tajiri K, Xu D, Kimura T, Qin R, Feng D, Yonebayashi S, Ogura Y, Yamagami F, Sato A, Nogami A, Aonuma K. Rev-erb agonist improves adverse cardiac remodeling and survival in myocardial infarction through an anti-inflammatory mechanism. PLoS One 2017; 12:e0189330. [PMID: 29232411 PMCID: PMC5726719 DOI: 10.1371/journal.pone.0189330] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/22/2017] [Indexed: 11/18/2022] Open
Abstract
Rev-erb α, known as nuclear receptor 1D1 (NR1D1), regulates circadian rhythm, modulates glucose and lipid metabolism, and inflammatory response. However, little is known about the effect of Rev-erb agonist on the progression of myocardial infarction (MI) and heart failure. To investigate it, wild-type male mice underwent sham-operation or permanent ligation of the left anterior descending coronary artery to create MI model. Rev-erb agonist SR9009 (100 mg/kg/day) or vehicle was intraperitoneally administered. Echocardiography was performed to evaluate cardiac function 1 week after surgery. The gene and protein expression levels in the left ventricles (LVs) were determined with real-time PCR, western blotting, and immunofluorescence. Moreover, immune cell infiltration into the LVs was analyzed by flow cytometry. Survival rate and reduced LV function were significantly improved by the treatment with SR9009 after MI. The expression level and plasma concentration of brain natriuretic peptide were significantly lower in MI mice treated with SR9009 (MI+SR) than in MI mice treated with vehicle (MI+V). Moreover, the mRNA expression levels of inflammatory-related molecules such as Il6, Mcp1, Ly6g, Cd11b, matrix metallopeptidase (Mmp)9, and the protein expression levels of phosphorylated NF-κB p65, phosphorylated ERK, and phosphorylated p38 were also significantly lower in MI+SR than in MI+V. Immunofluorescence intensity for MMP-9 was enhanced in the LVs, but was less so in MI+SR than in MI+V. Furthermore, infiltrations of neutrophils and proinflammatory macrophages in the LVs were dramatically increased in MI+V and were significantly suppressed in MI+SR. Rev-erb agonist SR9009 treatment inhibited post-MI mortality and improved cardiac function through modulating inflammation and remodeling process.
Collapse
Affiliation(s)
- Endin Nokik Stujanna
- Department of Cardiology, Faculty of Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Nobuyuki Murakoshi
- Department of Cardiology, Faculty of Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kazuko Tajiri
- Department of Cardiology, Faculty of Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - DongZhu Xu
- Department of Cardiology, Faculty of Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Taizo Kimura
- Department of Cardiology, Faculty of Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Rujie Qin
- Department of Cardiology, Faculty of Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Duo Feng
- Department of Cardiology, Faculty of Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Saori Yonebayashi
- Department of Cardiology, Faculty of Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yukino Ogura
- Department of Cardiology, Faculty of Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Fumi Yamagami
- Department of Cardiology, Faculty of Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akira Sato
- Department of Cardiology, Faculty of Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akihiko Nogami
- Department of Cardiology, Faculty of Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kazutaka Aonuma
- Department of Cardiology, Faculty of Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
19
|
Cardiac Progenitor Cells and the Interplay with Their Microenvironment. Stem Cells Int 2017; 2017:7471582. [PMID: 29075298 PMCID: PMC5623801 DOI: 10.1155/2017/7471582] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/26/2017] [Indexed: 02/06/2023] Open
Abstract
The microenvironment plays a crucial role in the behavior of stem and progenitor cells. In the heart, cardiac progenitor cells (CPCs) reside in specific niches, characterized by key components that are altered in response to a myocardial infarction. To date, there is a lack of knowledge on these niches and on the CPC interplay with the niche components. Insight into these complex interactions and into the influence of microenvironmental factors on CPCs can be used to promote the regenerative potential of these cells. In this review, we discuss cardiac resident progenitor cells and their regenerative potential and provide an overview of the interactions of CPCs with the key elements of their niche. We focus on the interaction between CPCs and supporting cells, extracellular matrix, mechanical stimuli, and soluble factors. Finally, we describe novel approaches to modulate the CPC niche that can represent the next step in recreating an optimal CPC microenvironment and thereby improve their regeneration capacity.
Collapse
|
20
|
Monocyte adhesion to atherosclerotic matrix proteins is enhanced by Asn-Gly-Arg deamidation. Sci Rep 2017; 7:5765. [PMID: 28720870 PMCID: PMC5515959 DOI: 10.1038/s41598-017-06202-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 06/09/2017] [Indexed: 12/25/2022] Open
Abstract
Atherosclerosis arises from leukocyte infiltration and thickening of the artery walls and constitutes a major component of vascular disease pathology, but the molecular events underpinning this process are not fully understood. Proteins containing an Asn-Gly-Arg (NGR) motif readily undergo deamidation of asparagine to generate isoDGR structures that bind to integrin αvβ3 on circulating leukocytes. Here we report the identification of isoDGR motifs in human atherosclerotic plaque components including extracellular matrix (ECM) proteins fibronectin and tenascin C, which have been strongly implicated in human atherosclerosis. We further demonstrate that deamidation of NGR motifs in fibronectin and tenascin C leads to increased adhesion of the monocytic cell line U937 and enhanced binding of primary human monocytes, except in the presence of a αvβ3-blocking antibody or the αv-selective inhibitor cilengitide. In contrast, under the same deamidating conditions monocyte-macrophages displayed only weak binding to the alternative ECM component vitronectin which lacks NGR motifs. Together, these findings confirm a critical role for isoDGR motifs in mediating leukocyte adhesion to the ECM via integrin αvβ3 and suggest that protein deamidation may promote the pathological progression of human atherosclerosis by enhancing monocyte recruitment to developing plaques.
Collapse
|
21
|
Zarrouk-Mahjoub S, Zaghdoudi M, Amira Z, Chebi H, Khabouchi N, Finsterer J, Mechmeche R, Ghazouani E. Pro- and anti-inflammatory cytokines in post-infarction left ventricular remodeling. Int J Cardiol 2016; 221:632-6. [PMID: 27423081 DOI: 10.1016/j.ijcard.2016.07.073] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 07/04/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVES Acute myocardial infarction (MI) leads to molecular, structural, geometric and functional changes in the heart during a process known as ventricular remodeling. Myocardial infarction is followed by an inflammatory response in which pro- and anti-inflammatory cytokines play a crucial role, particularly in left ventricular remodeling. This study aimed at evaluating serum concentrations of interleukin-8 (IL8), tumor-necrosis-factor-alpha (TNFα) and interleukin-10 (IL10), pro- and anti-inflammatory cytokines, and at correlating them with left ventricular remodeling as assessed by echocardiographic parameters. METHODS In a case-control study 30 MI patients were compared with 30 healthy controls. Serum concentrations of IL8, TNFα and IL10 were measured on day 2 and day 30 post-MI by chemiluminescence immunoassay and correlated with echocardiographic parameters. RESULTS There was an increase of IL8, and TNFα together with a decrease of IL10 at both time points. IL8 was negatively correlated with the left ventricular end-diastolic diameter (LVEDD) and positively with left ventricular systolic volume. IL10 was negatively correlated with LVEDD and left atrial volume 30days post-MI. CONCLUSION The increase of pro-inflammatory cytokines TNFα and IL8 was accompanied by decreased anti-inflammatory IL10. This imbalance between pro- and anti-inflammatory cytokines might contribute to the progression of left ventricular remodeling and may lead to heart failure.
Collapse
Affiliation(s)
| | - M Zaghdoudi
- Genomics Platform, Pasteur Institute of Tunis, Tunis, Tunisia
| | - Z Amira
- Hopital la Rabta de Tunis, Tunis, Tunisia
| | - H Chebi
- Genomics Platform, Pasteur Institute of Tunis, Tunis, Tunisia
| | - N Khabouchi
- Genomics Platform, Pasteur Institute of Tunis, Tunis, Tunisia
| | - J Finsterer
- Krankenanstalt Rudolfstiftung, Vienna, Austria.
| | | | | |
Collapse
|
22
|
Daniel LL, Scofield SLC, Thrasher P, Dalal S, Daniels CR, Foster CR, Singh M, Singh K. Ataxia telangiectasia-mutated kinase deficiency exacerbates left ventricular dysfunction and remodeling late after myocardial infarction. Am J Physiol Heart Circ Physiol 2016; 311:H445-52. [PMID: 27288435 DOI: 10.1152/ajpheart.00338.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 06/09/2016] [Indexed: 12/22/2022]
Abstract
Ataxia telangiectasia-mutated kinase (ATM), a cell cycle checkpoint protein, is activated in response to DNA damage and oxidative stress. We have previously shown that ATM deficiency is associated with increased apoptosis and fibrosis and attenuation of cardiac dysfunction early (1-7 days) following myocardial infarction (MI). Here, we tested the hypothesis that enhanced fibrosis and apoptosis, as observed early post-MI during ATM deficiency, exacerbate cardiac dysfunction and remodeling in ATM-deficient mice late post-MI. MIs were induced in wild-type (WT) and ATM heterozygous knockout (hKO) mice by ligation of the left anterior descending artery. Left ventricular (LV) structural and functional parameters were assessed by echocardiography 14 and 28 days post-MI, whereas biochemical parameters were measured 28 days post-MI. hKO-MI mice exhibited exacerbated LV dysfunction as observed by increased LV end-systolic volume and decreased percent fractional shortening and ejection fraction. Infarct size and thickness were not different between the two genotypes. Myocyte cross-sectional area was greater in hKO-MI group. The hKO-MI group exhibited increased fibrosis in the noninfarct and higher expression of α-smooth muscle actin (myofibroblast marker) in the infarct region. Apoptosis and activation of GSK-3β (proapoptotic kinase) were significantly lower in the infarct region of hKO-MI group. Matrix metalloproteinase 2 (MMP-2) expression was not different between the two genotypes. However, MMP-9 expression was significantly lower in the noninfarct region of hKO-MI group. Thus ATM deficiency exacerbates cardiac remodeling late post-MI with effects on cardiac function, fibrosis, apoptosis, and myocyte hypertrophy.
Collapse
Affiliation(s)
- Laura L Daniel
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Stephanie L C Scofield
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Patsy Thrasher
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Suman Dalal
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Christopher R Daniels
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Cerrone R Foster
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee; Department of Biological Sciences, East Tennessee State University, Johnson City, Tennessee
| | - Mahipal Singh
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Krishna Singh
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee; Center for Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, Tennessee; James H. Quillen Veterans Affairs Medical Center, Mountain Home, Johnson City, Tennessee
| |
Collapse
|
23
|
Sawyer AJ, Kyriakides TR. Matricellular proteins in drug delivery: Therapeutic targets, active agents, and therapeutic localization. Adv Drug Deliv Rev 2016; 97:56-68. [PMID: 26763408 DOI: 10.1016/j.addr.2015.12.016] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 12/17/2015] [Accepted: 12/17/2015] [Indexed: 02/06/2023]
Abstract
Extracellular matrix is composed of a complex array of molecules that together provide structural and functional support to cells. These properties are mainly mediated by the activity of collagenous and elastic fibers, proteoglycans, and proteins such as fibronectin and laminin. ECM composition is tissue-specific and could include matricellular proteins whose primary role is to modulate cell-matrix interactions. In adults, matricellular proteins are primarily expressed during injury, inflammation and disease. Particularly, they are closely associated with the progression and prognosis of cardiovascular and fibrotic diseases, and cancer. This review aims to provide an overview of the potential use of matricellular proteins in drug delivery including the generation of therapeutic agents based on the properties and structures of these proteins as well as their utility as biomarkers for specific diseases.
Collapse
|
24
|
Extracellular matrix-mediated cellular communication in the heart. J Mol Cell Cardiol 2016; 91:228-37. [PMID: 26778458 DOI: 10.1016/j.yjmcc.2016.01.011] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 01/10/2016] [Accepted: 01/11/2016] [Indexed: 01/13/2023]
Abstract
The extracellular matrix (ECM) is a complex and dynamic scaffold that maintains tissue structure and dynamics. However, the view of the ECM as an inert architectural support has been increasingly challenged. The ECM is a vibrant meshwork, a crucial organizer of cellular microenvironments. It plays a direct role in cellular interactions regulating cell growth, survival, spreading, proliferation, differentiation and migration through the intricate relationship among cellular and acellular tissue components. This complex interrelationship preserves cardiac function during homeostasis; however it is also responsible for pathologic remodeling following myocardial injury. Therefore, enhancing our understanding of this cross-talk may provide mechanistic insights into the pathogenesis of heart failure and suggest new approaches to novel, targeted pharmacologic therapies. This review explores the implications of ECM-cell interactions in myocardial cell behavior and cardiac function at baseline and following myocardial injury.
Collapse
|
25
|
Thies S, Friess M, Frischknecht L, Korol D, Felley-Bosco E, Stahel R, Vrugt B, Weder W, Opitz I, Soltermann A. Expression of the Stem Cell Factor Nestin in Malignant Pleural Mesothelioma Is Associated with Poor Prognosis. PLoS One 2015; 10:e0139312. [PMID: 26421614 PMCID: PMC4589394 DOI: 10.1371/journal.pone.0139312] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 09/11/2015] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The epithelioid and sarcomatoid histologic variants of malignant pleural mesothelioma (MPM) can be considered as E- and M-parts of the epithelial-mesenchymal transition (EMT) axis; the biphasic being an intermediate. EMT is associated with an increase of stem cell (SC) traits. We correlated the neural crest SC marker nestin and the EMT marker periostin with histology, type of neo-adjuvant chemotherapy (CT) and overall survival (OS) of MPM patients. PATIENTS AND METHODS Tumor tissues of a historic cohort 1 (320 patients) and an intended induction chemotherapy followed by extrapleural pneumonectomy (EPP) cohort 2 (145 patients) were immunohistochemically H-scored (intensity of immunoreactivity multiplied by frequency of stained cells). Paired chemo-naïve biopsies and -treated surgical specimens were available for 105/145 patients. CT included platinum/gemcitabine (Pla/Gem) or platinum/pemetrexed (Pla/Pem). RESULTS Expression of any cytosolic nestin progressively increased from epithelioid to biphasic to sarcomatoid MPM in cohort 1, whereas the diagnostic markers calretinin and podoplanin decreased. In cohort 2, Pla/Pem CT increased the expression level of nestin in comparison to Pla/Gem, whereas the opposite was found for periostin. In Pla/Pem treated patients, nestin was higher in biphasic MPM compared to epithelioid. In addition to non-epithelioid histology, any expression of nestin in chemo-naïve biopsies (median overall survival: 22 vs. 17 months) and chemo-treated surgical specimens (18 vs. 12 months) as well as high periostin in biopsies (23 vs. 15 months) were associated with poor prognosis. In the multivariate survival analysis, any nestin expression in chemo-naïve biopsies proved to be an independent prognosticator against histology. In both pre- and post-CT situations, the combination of nestin or periostin expression with non-epithelioid histology was particularly/ dismal (all p-values <0.05). CONCLUSIONS The SC marker nestin and the EMT marker periostin allow for further prognostic stratification among histologic variants of MPM. Their expression level is influenced by neo-adjuvant chemotherapy.
Collapse
Affiliation(s)
- Svenja Thies
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Martina Friess
- Division of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Lukas Frischknecht
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Dimitri Korol
- Cancer Registry, University Hospital Zurich, Zurich, Switzerland
| | | | - Rolf Stahel
- Clinic of Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Bart Vrugt
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Walter Weder
- Division of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Isabelle Opitz
- Division of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Alex Soltermann
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
- * E-mail:
| |
Collapse
|
26
|
Abstract
The heart pumps blood to maintain circulation and ensure the delivery of oxygenated blood to all the organs of the body. Mechanics play a critical role in governing and regulating heart function under both normal and pathological conditions. Biological processes and mechanical stress are coupled together in regulating myocyte function and extracellular matrix structure thus controlling heart function. Here, we offer a brief introduction to the biomechanics of left ventricular function and then summarize recent progress in the study of the effects of mechanical stress on ventricular wall remodeling and cardiac function as well as the effects of wall mechanical properties on cardiac function in normal and dysfunctional hearts. Various mechanical models to determine wall stress and cardiac function in normal and diseased hearts with both systolic and diastolic dysfunction are discussed. The results of these studies have enhanced our understanding of the biomechanical mechanism in the development and remodeling of normal and dysfunctional hearts. Biomechanics provide a tool to understand the mechanism of left ventricular remodeling in diastolic and systolic dysfunction and guidance in designing and developing new treatments.
Collapse
Affiliation(s)
- Andrew P. Voorhees
- Department of Mechanical Engineering, The University of Texas at San Antonio, Biomedical Engineering Program, UTSA-UTHSCSA
| | - Hai-Chao Han
- Department of Mechanical Engineering, The University of Texas at San Antonio, Biomedical Engineering Program, UTSA-UTHSCSA
| |
Collapse
|
27
|
Richardson WJ, Clarke SA, Quinn TA, Holmes JW. Physiological Implications of Myocardial Scar Structure. Compr Physiol 2015; 5:1877-909. [PMID: 26426470 DOI: 10.1002/cphy.c140067] [Citation(s) in RCA: 194] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Once myocardium dies during a heart attack, it is replaced by scar tissue over the course of several weeks. The size, location, composition, structure, and mechanical properties of the healing scar are all critical determinants of the fate of patients who survive the initial infarction. While the central importance of scar structure in determining pump function and remodeling has long been recognized, it has proven remarkably difficult to design therapies that improve heart function or limit remodeling by modifying scar structure. Many exciting new therapies are under development, but predicting their long-term effects requires a detailed understanding of how infarct scar forms, how its properties impact left ventricular function and remodeling, and how changes in scar structure and properties feed back to affect not only heart mechanics but also electrical conduction, reflex hemodynamic compensations, and the ongoing process of scar formation itself. In this article, we outline the scar formation process following a myocardial infarction, discuss interpretation of standard measures of heart function in the setting of a healing infarct, then present implications of infarct scar geometry and structure for both mechanical and electrical function of the heart and summarize experiences to date with therapeutic interventions that aim to modify scar geometry and structure. One important conclusion that emerges from the studies reviewed here is that computational modeling is an essential tool for integrating the wealth of information required to understand this complex system and predict the impact of novel therapies on scar healing, heart function, and remodeling following myocardial infarction.
Collapse
Affiliation(s)
- William J Richardson
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA.,Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| | - Samantha A Clarke
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - T Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jeffrey W Holmes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA.,Department of Medicine, University of Virginia, Charlottesville, Virginia, USA.,Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
28
|
Horckmans M, Esfahani H, Beauloye C, Clouet S, di Pietrantonio L, Robaye B, Balligand JL, Boeynaems JM, Dessy C, Communi D. Loss of mouse P2Y4 nucleotide receptor protects against myocardial infarction through endothelin-1 downregulation. THE JOURNAL OF IMMUNOLOGY 2015; 194:1874-81. [PMID: 25595790 DOI: 10.4049/jimmunol.1401364] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Nucleotides are released in the heart under pathological conditions, but little is known about their contribution to cardiac inflammation. The present study defines the P2Y4 nucleotide receptor, expressed on cardiac microvascular endothelial cells and involved in postnatal heart development, as an important regulator of the inflammatory response to cardiac ischemia. P2Y4-null mice displayed smaller infarcts in the left descending artery ligation model, as well as reduced neutrophil infiltration and fibrosis. Gene profiling identified inter alia endothelin-1 (ET-1) as one of the target genes of P2Y4 in ischemic heart. The reduced level of ET-1 was correlated with reduction of microvascular hyperpermeability, neutrophil infiltration, and endothelial adhesion molecule expression, and it could be explained by the decreased number of endothelial cells in P2Y4-null mice. Expression analysis of metalloproteinases and their tissue inhibitors in ischemic heart revealed reduced expression of matrix metalloproteinase (MMP)-9, reported to be potentially regulated by ET-1, and MMP-8, considered as neutrophil collagenase, as well as reduction of tissue inhibitor of MMP-1 and tissue inhibitor of MMP-4 in P2Y4-null mice. Reduction of cardiac permeability and neutrophil infiltration was also observed in P2Y4-null mice in LPS-induced inflammation model. Protection against infarction resulting from loss of P2Y4 brings new therapeutic perspectives for cardiac ischemia and remodeling.
Collapse
Affiliation(s)
- Michael Horckmans
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Hrag Esfahani
- Unité de Pharmacologie et de Thérapeutique, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Christophe Beauloye
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Sophie Clouet
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Larissa di Pietrantonio
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Bernard Robaye
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Université Libre de Bruxelles, 6041 Gosselies, Belgium; and
| | - Jean-Luc Balligand
- Unité de Pharmacologie et de Thérapeutique, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Jean-Marie Boeynaems
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Université Libre de Bruxelles, 1070 Brussels, Belgium; Département de Médecine de Laboratoire, Hôpital Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Chantal Dessy
- Unité de Pharmacologie et de Thérapeutique, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Didier Communi
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Université Libre de Bruxelles, 1070 Brussels, Belgium;
| |
Collapse
|
29
|
Suzuki H, Kanamaru K, Shiba M, Fujimoto M, Kawakita F, Imanaka-Yoshida K, Yoshida T, Taki W. Tenascin-C is a possible mediator between initial brain injury and vasospasm-related and -unrelated delayed cerebral ischemia after aneurysmal subarachnoid hemorrhage. ACTA NEUROCHIRURGICA. SUPPLEMENT 2015; 120:117-21. [PMID: 25366610 DOI: 10.1007/978-3-319-04981-6_20] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
INTRODUCTION Tenascin-C (TNC), a matricellular protein, exerts diverse functions, including tissue remodeling and apoptosis, and is induced in cerebrospinal fluid (CSF) after aneurysmal subarachnoid hemorrhage (SAH). The purpose of this study was to examine the relationships among CSF TNC levels, initial brain injury, delayed cerebral ischemia (DCI), and vasospasm after SAH. METHODS CSF TNC levels were measured in 30 patients with aneurysmal SAH of Fisher computed tomography (CT) group III who were treated microsurgically or endovascularly with CSF drainage within 24 h of SAH. Admission World Federation of Neurosurgical Societies (WFNS) grade was supposed to indicate the severity of initial brain injury. Cerebral vasospasm was defined as narrowed (≥ 25 %) cerebral arteries demonstrated by angiography. DCI was defined as any neurological deterioration presumed related to ischemia that persisted for ≥ 1 h. RESULTS Higher CSF TNC levels were correlated with worse admission WFNS grades. Vasospasm was aggravated with higher TNC levels. DCI occurred regardless of the degree of vasospasm but was associated with TNC induction. Multivariate analyses showed that higher TNC levels and vasospasm were independent predictors of DCI occurrence. CONCLUSIONS SAH (initial brain injury) that is more severe induces more TNC, which may cause the subsequent development of both vasospasm and vasospasm-unrelated secondary brain injury, leading to DCI.
Collapse
Affiliation(s)
- Hidenori Suzuki
- Department of Neurosurgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan,
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Shiba M, Fujimoto M, Kawakita F, Imanaka-Yoshida K, Yoshida T, Kanamaru K, Taki W, Suzuki H. Effects of tenascin-C on early brain injury after subarachnoid hemorrhage in rats. ACTA NEUROCHIRURGICA. SUPPLEMENT 2015; 120:69-73. [PMID: 25366602 DOI: 10.1007/978-3-319-04981-6_12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND AND PURPOSE We previously reported that tenascin-C (TNC), a matricellular protein, was involved in the pathogenesis of cerebral vasospasm after subarachnoid hemorrhage (SAH), but the role of TNC in early brain injury (EBI) is unknown. This study assessed whether inhibition of TNC upregulation in brain by imatinib mesylate (imatinib), an inhibitor of the tyrosine kinases of platelet-derived growth factor receptors, prevents EBI after experimental SAH. METHODS Rats were assigned to sham, SAH plus vehicle, and SAH plus imatinib groups (n = 4 per group). Imatinib (50 mg/kg body weight) was administered intraperitoneally to rats undergoing SAH by endovascular perforation, and EBI was evaluated using terminal deoxynucleotidyl transferase-mediated uridine 5-triphosphate-biotin nick end-labeling staining at 24 h after SAH. Imatinib-treated SAH rats were also treated by a cisternal injection of recombinant TNC. RESULTS SAH upregulated TNC and caused EBI. Imatinib treatment suppressed both TNC upregulation and EBI at 24 h. Recombinant TNC reinduced EBI in imatinib-treated SAH rats. CONCLUSIONS TNC may be involved in the pathogenesis of EBI after SAH.
Collapse
Affiliation(s)
- Masato Shiba
- Department of Neurosurgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan,
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Ghasemi O, Ma Y, Lindsey ML, Jin YF. Using systems biology approaches to understand cardiac inflammation and extracellular matrix remodeling in the setting of myocardial infarction. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2014; 6:77-91. [PMID: 24741709 DOI: 10.1002/wsbm.1248] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Inflammation and extracellular matrix (ECM) remodeling are important components regulating the response of the left ventricle to myocardial infarction (MI). Significant cellular- and molecular-level contributors can be identified by analyzing data acquired through high-throughput genomic and proteomic technologies that provide expression levels for thousands of genes and proteins. Large-scale data provide both temporal and spatial information that need to be analyzed and interpreted using systems biology approaches in order to integrate this information into dynamic models that predict and explain mechanisms of cardiac healing post-MI. In this review, we summarize the systems biology approaches needed to computationally simulate post-MI remodeling, including data acquisition, data analysis for biomarker classification and identification, data integration to build dynamic models, and data interpretation for biological functions. An example for applying a systems biology approach to ECM remodeling is presented as a reference illustration.
Collapse
|
32
|
Poggio P, Branchetti E, Grau JB, Lai EK, Gorman RC, Gorman JH, Sacks MS, Bavaria JE, Ferrari G. Osteopontin-CD44v6 interaction mediates calcium deposition via phospho-Akt in valve interstitial cells from patients with noncalcified aortic valve sclerosis. Arterioscler Thromb Vasc Biol 2014; 34:2086-94. [PMID: 25060796 DOI: 10.1161/atvbaha.113.303017] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The activation of valve interstitial cells (VICs) toward an osteogenic phenotype characterizes aortic valve sclerosis, the early asymptomatic phase of calcific aortic valve disease. Osteopontin is a phosphorylated acidic glycoprotein that accumulates within the aortic leaflets and labels VIC activation even in noncalcified asymptomatic patients. Despite this, osteopontin protects VICs against in vitro calcification. Here, we hypothesize that the specific interaction of osteopontin with CD44v6, and the related intracellular pathway, prevents calcium deposition in human-derived VICs from patients with aortic valve sclerosis. APPROACH AND RESULTS On informed consent, 23 patients and 4 controls were enrolled through the cardiac surgery and heart transplant programs. Human aortic valves and VICs were tested for osteogenic transdifferentiation, ex vivo and in vitro. Osteopontin-CD44 interaction was analyzed using proximity ligation assay and the signaling pathways investigated. A murine model based on angiotensin II infusion was used to mimic early pathological remodeling of the aortic valves. We report osteopontin-CD44 functional interaction as a hallmark of early stages of calcific aortic valve disease. We demonstrated that osteopontin-CD44 interaction mediates calcium deposition via phospho-Akt in VICs from patients with noncalcified aortic valve sclerosis. Finally, microdissection analysis of murine valves shows increased cusp thickness in angiotensin II-treated mice versus saline infused along with colocalization of osteopontin and CD44 as seen in human lesions. CONCLUSIONS Here, we unveil a specific protein-protein association and intracellular signaling mechanisms of osteopontin. Understanding the molecular mechanisms of early VIC activation and calcium deposition in asymptomatic stage of calcific aortic valve disease could open new prospective for diagnosis and therapeutic intervention.
Collapse
Affiliation(s)
- Paolo Poggio
- From the Department of Surgery, Perelman School of Medicine at University of Pennsylvania, Philadelphia (P.P., E.B., J.B.G., E.K.L., R.C.G., J.H.G., J.E.B., G.F.); Centro Cardiologico Monzino IRCCS, Milan, Italy (P.P.); Columbia University-Valley Heart Center, Ridgewood, NJ (J.B.G.); and Department of Biomedical Engineering, University of Texas at Austin (M.S.S.)
| | - Emanuela Branchetti
- From the Department of Surgery, Perelman School of Medicine at University of Pennsylvania, Philadelphia (P.P., E.B., J.B.G., E.K.L., R.C.G., J.H.G., J.E.B., G.F.); Centro Cardiologico Monzino IRCCS, Milan, Italy (P.P.); Columbia University-Valley Heart Center, Ridgewood, NJ (J.B.G.); and Department of Biomedical Engineering, University of Texas at Austin (M.S.S.)
| | - Juan B Grau
- From the Department of Surgery, Perelman School of Medicine at University of Pennsylvania, Philadelphia (P.P., E.B., J.B.G., E.K.L., R.C.G., J.H.G., J.E.B., G.F.); Centro Cardiologico Monzino IRCCS, Milan, Italy (P.P.); Columbia University-Valley Heart Center, Ridgewood, NJ (J.B.G.); and Department of Biomedical Engineering, University of Texas at Austin (M.S.S.)
| | - Eric K Lai
- From the Department of Surgery, Perelman School of Medicine at University of Pennsylvania, Philadelphia (P.P., E.B., J.B.G., E.K.L., R.C.G., J.H.G., J.E.B., G.F.); Centro Cardiologico Monzino IRCCS, Milan, Italy (P.P.); Columbia University-Valley Heart Center, Ridgewood, NJ (J.B.G.); and Department of Biomedical Engineering, University of Texas at Austin (M.S.S.)
| | - Robert C Gorman
- From the Department of Surgery, Perelman School of Medicine at University of Pennsylvania, Philadelphia (P.P., E.B., J.B.G., E.K.L., R.C.G., J.H.G., J.E.B., G.F.); Centro Cardiologico Monzino IRCCS, Milan, Italy (P.P.); Columbia University-Valley Heart Center, Ridgewood, NJ (J.B.G.); and Department of Biomedical Engineering, University of Texas at Austin (M.S.S.)
| | - Joseph H Gorman
- From the Department of Surgery, Perelman School of Medicine at University of Pennsylvania, Philadelphia (P.P., E.B., J.B.G., E.K.L., R.C.G., J.H.G., J.E.B., G.F.); Centro Cardiologico Monzino IRCCS, Milan, Italy (P.P.); Columbia University-Valley Heart Center, Ridgewood, NJ (J.B.G.); and Department of Biomedical Engineering, University of Texas at Austin (M.S.S.)
| | - Michael S Sacks
- From the Department of Surgery, Perelman School of Medicine at University of Pennsylvania, Philadelphia (P.P., E.B., J.B.G., E.K.L., R.C.G., J.H.G., J.E.B., G.F.); Centro Cardiologico Monzino IRCCS, Milan, Italy (P.P.); Columbia University-Valley Heart Center, Ridgewood, NJ (J.B.G.); and Department of Biomedical Engineering, University of Texas at Austin (M.S.S.)
| | - Joseph E Bavaria
- From the Department of Surgery, Perelman School of Medicine at University of Pennsylvania, Philadelphia (P.P., E.B., J.B.G., E.K.L., R.C.G., J.H.G., J.E.B., G.F.); Centro Cardiologico Monzino IRCCS, Milan, Italy (P.P.); Columbia University-Valley Heart Center, Ridgewood, NJ (J.B.G.); and Department of Biomedical Engineering, University of Texas at Austin (M.S.S.)
| | - Giovanni Ferrari
- From the Department of Surgery, Perelman School of Medicine at University of Pennsylvania, Philadelphia (P.P., E.B., J.B.G., E.K.L., R.C.G., J.H.G., J.E.B., G.F.); Centro Cardiologico Monzino IRCCS, Milan, Italy (P.P.); Columbia University-Valley Heart Center, Ridgewood, NJ (J.B.G.); and Department of Biomedical Engineering, University of Texas at Austin (M.S.S.).
| |
Collapse
|
33
|
Morris AH, Kyriakides TR. Matricellular proteins and biomaterials. Matrix Biol 2014; 37:183-91. [PMID: 24657843 DOI: 10.1016/j.matbio.2014.03.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 03/12/2014] [Accepted: 03/12/2014] [Indexed: 01/05/2023]
Abstract
Biomaterials are essential to modern medicine as components of reconstructive implants, implantable sensors, and vehicles for localized drug delivery. Advances in biomaterials have led to progression from simply making implants that are nontoxic to making implants that are specifically designed to elicit particular functions within the host. The interaction of implants and the extracellular matrix during the foreign body response is a growing area of concern for the field of biomaterials, because it can lead to implant failure. Expression of matricellular proteins is modulated during the foreign body response and these proteins interact with biomaterials. The design of biomaterials to specifically alter the levels of matricellular proteins surrounding implants provides a new avenue for the design and fabrication of biomimetic biomaterials.
Collapse
Affiliation(s)
- Aaron H Morris
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Themis R Kyriakides
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States; Department of Pathology, Yale University, New Haven, CT, United States; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, United States.
| |
Collapse
|
34
|
Shiba M, Fujimoto M, Imanaka-Yoshida K, Yoshida T, Taki W, Suzuki H. Tenascin-C causes neuronal apoptosis after subarachnoid hemorrhage in rats. Transl Stroke Res 2014; 5:238-47. [PMID: 24481545 DOI: 10.1007/s12975-014-0333-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Revised: 01/15/2014] [Accepted: 01/16/2014] [Indexed: 10/25/2022]
Abstract
The role of tenascin-C (TNC), a matricellular protein, in brain injury is unknown. The aim of this study was to examine if TNC causes neuronal apoptosis after subarachnoid hemorrhage (SAH), a deadly cerebrovascular disorder, using imatinib mesylate (a selective inhibitor of platelet-derived growth factor receptor [PDGFR] that is reported to suppress TNC induction) and recombinant TNC. SAH by endovascular perforation caused caspase-dependent neuronal apoptosis in the cerebral cortex irrespective of cerebral vasospasm development at 24 and 72 h post-SAH, associated with PDGFR activation, mitogen-activated protein kinases (MAPKs) activation, and TNC induction in rats. PDGFR inactivation by an intraperitoneal injection of imatinib mesylate prevented neuronal apoptosis, as well as MAPKs activation and TNC induction in the cerebral cortex at 24 h. A cisternal injection of recombinant TNC reactivated MAPKs and abolished anti-apoptotic effects of imatinib mesylate. The TNC injection also induced TNC itself in SAH brain, which may internally augment neuronal apoptosis after SAH. These findings suggest that TNC upregulation by PDGFR activation causes neuronal apoptosis via MAPK activation, and that the positive feedback mechanisms may exist to augment neuronal apoptosis after SAH. TNC-induced neuronal apoptosis would be a new target to improve outcome after SAH.
Collapse
Affiliation(s)
- Masato Shiba
- Department of Neurosurgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | | | | | | | | | | |
Collapse
|
35
|
Wildgruber M, Bielicki I, Aichler M, Kosanke K, Feuchtinger A, Settles M, Onthank DC, Cesati RR, Robinson SP, Huber AM, Rummeny EJ, Walch AK, Botnar RM. Assessment of myocardial infarction and postinfarction scar remodeling with an elastin-specific magnetic resonance agent. Circ Cardiovasc Imaging 2013; 7:321-9. [PMID: 24363356 DOI: 10.1161/circimaging.113.001270] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND To prospectively evaluate an elastin-specific MR contrast agent (ESMA) for in vivo targeting of elastic fibers in myocardial infarction (MI) and postinfarction scar remodeling. METHODS AND RESULTS MI was induced in C57BL/6J mice (n=40) by permanent ligation of the left anterior descending coronary artery. MRI was performed at 7 and 21 days after MI. The merits of gadolinium-based ESMA (Gd-ESMA) were compared with gadopentetic acid (Gd-DTPA) for infarct size determination, contrast-to-noise ratio (CNR), and enhancement kinetics. Specific binding in vivo was evaluated by blocking the molecular target using nonparamagnetic lanthanum-ESMA. In vivo imaging results were confirmed by postmortem triphenyltetrazolium chloride staining, elastica van Gieson staining, and Western blotting. Delayed enhancement MRI revealed prolonged enhancement of Gd-ESMA in the postischemic scar compared with Gd-DTPA. Infarct size measurements showed good agreement between Gd-ESMA and Gd-DTPA and were confirmed by ex vivo triphenyltetrazolium chloride staining. Preinjection of the blocking lanthanum-ESMA resulted in significantly lower CNR of Gd-ESMA at the infarct site (P=0.0019). Although no significant differences in CNR were observed between delayed enhancement imaging and Gd-DTPA between days 7 and 21 (1.8± versus 3.8; P=ns), Gd-ESMA showed markedly higher CNR on day 21 after MI (14.1 versus 4.9; P=0.0032), which correlated with increased synthesis of tropoelastin detected by Western blot analysis and histology. Higher CNR values for Gd-ESMA further correlated with improved ejection fraction of the mice on day 21 after MI. CONCLUSIONS Gd-ESMA enables targeting of elastin within the infarct scar in a mouse model of MI. The imaging properties of Gd-ESMA allow quantification of intrascar elastin content in vivo and thereby provide potential for noninvasive characterization of postinfarction scar remodeling.
Collapse
Affiliation(s)
- Moritz Wildgruber
- Department of Diagnostic and Interventional Radiology, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Silvestre JS, Smadja DM, Lévy BI. Postischemic revascularization: from cellular and molecular mechanisms to clinical applications. Physiol Rev 2013; 93:1743-802. [PMID: 24137021 DOI: 10.1152/physrev.00006.2013] [Citation(s) in RCA: 181] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
After the onset of ischemia, cardiac or skeletal muscle undergoes a continuum of molecular, cellular, and extracellular responses that determine the function and the remodeling of the ischemic tissue. Hypoxia-related pathways, immunoinflammatory balance, circulating or local vascular progenitor cells, as well as changes in hemodynamical forces within vascular wall trigger all the processes regulating vascular homeostasis, including vasculogenesis, angiogenesis, arteriogenesis, and collateral growth, which act in concert to establish a functional vascular network in ischemic zones. In patients with ischemic diseases, most of the cellular (mainly those involving bone marrow-derived cells and local stem/progenitor cells) and molecular mechanisms involved in the activation of vessel growth and vascular remodeling are markedly impaired by the deleterious microenvironment characterized by fibrosis, inflammation, hypoperfusion, and inhibition of endogenous angiogenic and regenerative programs. Furthermore, cardiovascular risk factors, including diabetes, hypercholesterolemia, hypertension, diabetes, and aging, constitute a deleterious macroenvironment that participates to the abrogation of postischemic revascularization and tissue regeneration observed in these patient populations. Thus stimulation of vessel growth and/or remodeling has emerged as a new therapeutic option in patients with ischemic diseases. Many strategies of therapeutic revascularization, based on the administration of growth factors or stem/progenitor cells from diverse sources, have been proposed and are currently tested in patients with peripheral arterial disease or cardiac diseases. This review provides an overview from our current knowledge regarding molecular and cellular mechanisms involved in postischemic revascularization, as well as advances in the clinical application of such strategies of therapeutic revascularization.
Collapse
|
37
|
Lajiness JD, Conway SJ. Origin, development, and differentiation of cardiac fibroblasts. J Mol Cell Cardiol 2013; 70:2-8. [PMID: 24231799 DOI: 10.1016/j.yjmcc.2013.11.003] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 10/23/2013] [Accepted: 11/04/2013] [Indexed: 01/14/2023]
Abstract
Cardiac fibroblasts are the most abundant cell in the mammalian heart. While they have been historically underappreciated in terms of their functional contributions to cardiac development and physiology, they and their activated form, myofibroblasts, are now known to play key roles in both development and disease through structural, paracrine, and electrical interactions with cardiomyocytes. The lack of specific markers for fibroblasts currently convolutes the study of this dynamic cell lineage, but advances in marker analysis and lineage mapping technologies are continuously being made. Understanding how to best utilize these tools, both individually and in combination, will help to elucidate the functional significance of fibroblast-cardiomyocyte interactions in vivo. Here we review what is currently known about the diverse roles played by cardiac fibroblasts and myofibroblasts throughout development and periods of injury with the intent of emphasizing the duality of their nature. This article is part of a Special Issue entitled "Myocyte-Fibroblast Signalling in Myocardium ".
Collapse
Affiliation(s)
- Jacquelyn D Lajiness
- Developmental Biology and Neonatal Medicine Program, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Simon J Conway
- Developmental Biology and Neonatal Medicine Program, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
38
|
Prevalence and predictors of worsened left ventricular diastolic dysfunction after catheter ablation of atrial fibrillation. Int J Cardiol 2013; 168:3613-5. [DOI: 10.1016/j.ijcard.2013.05.047] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Accepted: 05/04/2013] [Indexed: 01/01/2023]
|
39
|
Minicucci MF, Santos PPD, Rafacho BPM, Gonçalves AF, Ardisson LP, Batista DF, Azevedo PS, Polegato BF, Okoshi K, Pereira EJ, Paiva SAR, Zornoff LAM. Periostin as a modulator of chronic cardiac remodeling after myocardial infarction. Clinics (Sao Paulo) 2013; 68:1344-9. [PMID: 24212842 PMCID: PMC3798673 DOI: 10.6061/clinics/2013(10)09] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 05/16/2013] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVE After acute myocardial infarction, during the cardiac repair phase, periostin is released into the infarct and activates signaling pathways that are essential for the reparative process. However, the role of periostin in chronic cardiac remodeling after myocardial infarction remains to be elucidated. Therefore, the objective of this study was to investigate the relationship between tissue periostin and cardiac variables in the chronic cardiac remodeling induced by myocardial infarction. METHODS Male Wistar rats were assigned to 2 groups: a simulated surgery group (SHAM; n = 8) and a myocardial infarction group (myocardial infarction; n = 13). After 3 months, morphological, functional and biochemical analyses were performed. The data are expressed as means±SD or medians (including the lower and upper quartiles). RESULTS Myocardial infarctions induced increased left ventricular diastolic and systolic areas associated with a decreased fractional area change and a posterior wall shortening velocity. With regard to the extracellular matrix variables, the myocardial infarction group presented with higher values of periostin and types I and III collagen and higher interstitial collagen volume fractions and myocardial hydroxyproline concentrations. In addition, periostin was positively correlated with type III collagen levels (r = 0.673, p = 0.029) and diastolic (r = 0.678, p = 0.036) and systolic (r = 0.795, p = 0.006) left ventricular areas. Considering the relationship between periostin and the cardiac function variables, periostin was inversely correlated with both the fractional area change (r = -0.783, p = 0.008) and the posterior wall shortening velocity (r = -0.767, p = 0.012). CONCLUSIONS Periostin might be a modulator of deleterious cardiac remodeling in the chronic phase after myocardial infarction in rats.
Collapse
Affiliation(s)
- Marcos F Minicucci
- Universidade Estadual Paulista (UNESP), Botucatu Medical School, Internal Medicine Department, BotucatuSP, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Minicucci MF, dos Santos PP, Rafacho BPM, Gonçalves AF, Silva RAC, Chiuso-Minicucci F, Azevedo PS, Polegato BF, Okoshi K, Pereira EJ, Paiva SAR, Zornoff LAM. Mechanisms involved in the beneficial effects of spironolactone after myocardial infarction. PLoS One 2013; 8:e76866. [PMID: 24098808 PMCID: PMC3786966 DOI: 10.1371/journal.pone.0076866] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2013] [Accepted: 08/23/2013] [Indexed: 11/18/2022] Open
Abstract
Introduction Our objective was to analyze the effect of spironolactone on cardiac remodeling after experimental myocardial infarction (MI), assessed by matricellular proteins levels, cardiac collagen amount and distribution, myocardial tissue metalloproteinase inhibitor-1(TIMP-1) concentration, myocyte hypertrophy, left ventricular architecture, and invitro and invivo cardiac function. Methods Wistar rats were assigned to 4 groups: control group, in which animals were submitted to simulated surgery (SHAM group; n=9); group that received spironolactone and in which animals were submitted to simulated surgery (SHAM-S group, n=9); myocardial infarction group, in which animals were submitted to coronary artery ligation (MI group, n=15); and myocardial infarction group with spironolactone supplementation (MI-S group, n=15). The rats were observed for 3 months. Results The MI group had higher values of left cardiac chambers and mass index and lower relative wall thicknesses compared with the SHAM group. In addition, diastolic and systolic functions were worse in the MI groups. However, spironolactone did not influence any of these variables. The MI-S group had a lower myocardial hydroxyproline concentration and myocyte cross-sectional area compared with the MI group. Myocardial periostin and collagen type III were lower in the MI-S group compared with the MI-group. In addition, TIMP-1 concentration in myocardium was higher in the MI-S group compared with the MI group. Conclusions The predominant consequence of spironolactone supplementation after MI is related to reductions in collagens, with discrete attenuation of other remodeling variables. Importantly, this effect may be modulated by periostin and TIMP-1 levels.
Collapse
Affiliation(s)
- Marcos F. Minicucci
- Internal Medicine Department, Botucatu Medical School, University Estadual Paulista, Botucatu, São Paulo, Brazil
- * E-mail:
| | - Priscila P. dos Santos
- Internal Medicine Department, Botucatu Medical School, University Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Bruna P. M. Rafacho
- Internal Medicine Department, Botucatu Medical School, University Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Andrea F. Gonçalves
- Internal Medicine Department, Botucatu Medical School, University Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Renata A. C. Silva
- Internal Medicine Department, Botucatu Medical School, University Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Fernanda Chiuso-Minicucci
- Department of Microbiology and Immunology, Institute of Biosciences, University Estadual, Paulista, Botucatu, São Paulo, Brazil
| | - Paula S. Azevedo
- Internal Medicine Department, Botucatu Medical School, University Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Bertha F. Polegato
- Internal Medicine Department, Botucatu Medical School, University Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Katashi Okoshi
- Internal Medicine Department, Botucatu Medical School, University Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Elenize J. Pereira
- Internal Medicine Department, Botucatu Medical School, University Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Sergio A. R. Paiva
- Internal Medicine Department, Botucatu Medical School, University Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Leonardo A. M. Zornoff
- Internal Medicine Department, Botucatu Medical School, University Estadual Paulista, Botucatu, São Paulo, Brazil
| |
Collapse
|
41
|
Wong VW, Gurtner GC, Longaker MT. Wound healing: a paradigm for regeneration. Mayo Clin Proc 2013; 88:1022-31. [PMID: 24001495 DOI: 10.1016/j.mayocp.2013.04.012] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 04/12/2013] [Indexed: 02/07/2023]
Abstract
Human skin is a remarkably plastic organ that sustains insult and injury throughout life. Its ability to expeditiously repair wounds is paramount to survival and is thought to be regulated by wound components such as differentiated cells, stem cells, cytokine networks, extracellular matrix, and mechanical forces. These intrinsic regenerative pathways are integrated across different skin compartments and are being elucidated on the cellular and molecular levels. Recent advances in bioengineering and nanotechnology have allowed researchers to manipulate these microenvironments in increasingly precise spatial and temporal scales, recapitulating key homeostatic cues that may drive regeneration. The ultimate goal is to translate these bench achievements into viable bedside therapies that address the growing global burden of acute and chronic wounds. In this review, we highlight current concepts in cutaneous wound repair and propose that many of these evolving paradigms may underlie regenerative processes across diverse organ systems.
Collapse
Affiliation(s)
- Victor W Wong
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, Stanford, CA, USA
| | | | | |
Collapse
|
42
|
[Epithelial-mesenchymal transition in non-small cell lung cancer]. DER PATHOLOGE 2013; 33 Suppl 2:311-7. [PMID: 23080026 DOI: 10.1007/s00292-012-1635-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Non-small cell lung carcinoma (NSCLC) is a highly fibrotic malignancy, which exhibits a prominent desmoplastic stroma. Epithelial-mesenchymal transition (EMT) is one of the main modes of carcinoma invasion. We identified the stromal N-glycoprotein periostin by mass spectrometry of lung adenocarcinoma pleural effusions. Validation on a NSCLC tissue microarray and on tumor whole sections by immunohistochemistry indicated that periostin is strongly upregulated at the invasive front in both tumor epithelia and the surrounding matricellular space. In comparison to collagen, elastin and vimentin, periostin was found to be most closely associated with parameters of tumor progression such as larger size and higher stage, with the squamous cell histotype, and with decreased survival. An association with decreased survival was also found for the cell adhesion molecule L1CAM. In conclusion, enlargement of NSCLC tumors is associated with an increase of desmoplastic stroma and concomitant upregulation of EMT markers at the invasive front. The tumor-stroma interface may be a candidate topographic region for stroma- or EMT-directed therapy.
Collapse
|
43
|
He Y, Zhou X, Zheng X, Jiang X. Exogenous high-mobility group box 1 protein prevents postinfarction adverse myocardial remodeling through TGF-β/Smad signaling pathway. J Cell Biochem 2013; 114:1634-41. [PMID: 23355476 DOI: 10.1002/jcb.24505] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2012] [Accepted: 01/10/2013] [Indexed: 11/06/2022]
Affiliation(s)
- Yiyu He
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China
| | | | | | | |
Collapse
|
44
|
Suzuki H, Shiba M, Fujimoto M, Kawamura K, Nanpei M, Tekeuchi E, Matsushima S, Kanamaru K, Imanaka-Yoshida K, Yoshida T, Taki W. Matricellular protein: a new player in cerebral vasospasm following subarachnoid hemorrhage. ACTA NEUROCHIRURGICA. SUPPLEMENT 2013; 115:213-218. [PMID: 22890671 DOI: 10.1007/978-3-7091-1192-5_39] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
INTRODUCTION Matricellular protein (MCP) is a class of nonstructural and secreted extracellular matrix proteins that exert diverse functions, but its role in vascular smooth muscle contraction has not been investigated. MATERIAL AND METHODS First, rat subarachnoid hemorrhage (SAH) models were produced by endovascular perforation and examined for tenascin-C (TNC) and osteopontin (OPN) induction (representatives of MCPs) in vasospastic cerebral arteries using immunostaining. Second, recombinant TNC (r-TNC), recombinant OPN (r-OPN), or both were injected into a cisterna magna in healthy rats, and the effects on the diameter of basilar arteries were determined using India ink angiography. RESULTS In SAH rats, TNC immunoreactivity was markedly induced in the smooth muscle cell layers of spastic cerebral arteries on day 1 but not in control animals. The TNC immunoreactivity decreased on day 3 as vasospasm improved: OPN immunoreactivity, on the other hand, was more induced in the arterial wall on day 3. r-TNC injections caused prolonged contractions of rat basilar arteries, which were reversed by r-OPN, although r-OPN itself had no effect on the vessel diameter. CONCLUSIONS MCPs, including TNC and OPN, may contribute to the pathophysiology of cerebral vasospasm and provide a novel therapeutic approach against it.
Collapse
Affiliation(s)
- Hidenori Suzuki
- Department of Neurosurgery, Mie University Graduate School of Medicine, Mie, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Myocardial infarction is one of the major causes of left ventricular dilatation, frequently leading to heart failure. In the last decade, the wound healing process that takes place in the infarct area after infarction has been recognized as a novel therapeutic target to attenuate left ventricular dilatation and preserve an adequate cardiac function. In this chapter, we discuss the role of Wnt signaling in the wound healing process after infarction, with a specific focus on its modulating effect on myofibroblast characteristics.
Collapse
|
46
|
Barallobre-Barreiro J, Didangelos A, Yin X, Doménech N, Mayr M. A sequential extraction methodology for cardiac extracellular matrix prior to proteomics analysis. Methods Mol Biol 2013; 1005:215-223. [PMID: 23606260 DOI: 10.1007/978-1-62703-386-2_17] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Cardiac fibrosis is characterized by excessive deposition of extracellular matrix (ECM) and is a common complication of various cardiovascular diseases. However, little is known about proteins in the cardiac extracellular space. Proteomics analysis of cardiac ECM can be challenging due to the presence of more abundant intracellular proteins, the low degree of solubility of integral ECM proteins, and the presence of abundant posttranslational modifications. Here we describe an extraction methodology based on tissue decellularization, which allows the biochemical subfractionation of extracellular proteins in cardiac tissue. These relatively low-complexity protein fractions are suitable for analysis by gel-LC-MS/MS and other proteomics techniques.
Collapse
|
47
|
Carpena N, Roselló-Lletí E, Calabuig JR, Tarazón E, González-Juanatey JR, Martínez-Dolz L, Salvador A, Grigorian L, Orosa P, Portolés M, Rivera M. MMP-2 and sTNF-R1 Variability in Patients with Essential Hypertension: 1-Year Follow-Up Study. ISRN CARDIOLOGY 2012; 2012:501894. [PMID: 23008783 PMCID: PMC3449115 DOI: 10.5402/2012/501894] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 08/15/2012] [Indexed: 01/02/2023]
Abstract
The aim of this study is to analyze MMP-2 and sTNF-R1 variability, potent predictors of cardiovascular events, in stable hypertensive patients during a 12-month followup. 234 asymptomatic patients (age 60 ± 13, 136 male) out of 252 patients with essential hypertension were followed up. MMP-2 and sTNF-R1 were measured at baseline and after 12 months (stage I). To compare MMP-2 and sTNF-R1 levels over time interval, we used the statistical method of Bland-Altman. MMP-2 and sTNF-R1 reproducibility was good in our patients for the two intervals with a coefficient of reproducibility of 8.2% and 11.3%, respectively. The percentages of patients within 1.96 × standard deviation of the mean were 93.6% and 92.7%. An elevated coefficient of correlation was obtained for MMP-2, basal versus stage I (r = 0.55, P < 0.0001) and for sTNF-R1 (r = 0.75, P < 0.0001). There is good stability in MMP-2 and sTNF-R1 levels in a followup study of patients with stable hypertension. As a consequence, assessment of its concentrations may be a useful tool for monitoring the follow-up of these patients. Measured variations in MMP-2 and sTNF-R1 levels, exceeding 8.2% and 11.3%, respectively, may indicate an increase in cardiovascular risk, thus, could be used to optimizing treatment than blood pressure control alone.
Collapse
Affiliation(s)
- Núria Carpena
- Cardiocirculatory Unit, Research Center, Hospital Universitario y Politécnico La Fe, 46009 Valencia, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Anwar A, Li M, Frid MG, Kumar B, Gerasimovskaya EV, Riddle SR, McKeon BA, Thukaram R, Meyrick BO, Fini MA, Stenmark KR. Osteopontin is an endogenous modulator of the constitutively activated phenotype of pulmonary adventitial fibroblasts in hypoxic pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2012; 303:L1-L11. [PMID: 22582113 DOI: 10.1152/ajplung.00050.2012] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Increased cell proliferation and migration, of several cell types are key components of vascular remodeling observed in pulmonary hypertension (PH). Our previous data demonstrate that adventitial fibroblasts isolated from pulmonary arteries of chronically hypoxic hypertensive calves (termed PH-Fibs) exhibit a "constitutively activated" phenotype characterized by high proliferative and migratory potential. Osteopontin (OPN) has been shown to promote several cellular activities including growth and migration in cancer cells. We thus tested the hypothesis that elevated OPN expression confers the "activated" highly proproliferative and promigratory/invasive phenotype of PH-Fibs. Our results demonstrate that, both in vivo and ex vivo, PH-Fibs exhibited increased expression of OPN, as well as its cognate receptors, α(V)β(3) and CD44, compared with control fibroblasts (CO-Fibs). Augmented OPN expression in PH-Fibs corresponded to their high proliferative, migratory, and invasive properties and constitutive activation of ERK1/2 and AKT signaling. OPN silencing via small interfering RNA or sequestering OPN production by specific antibodies led to decreased proliferation, migration, invasion, and attenuated ERK1/2, AKT phosphorylation in PH-Fibs. Furthermore, increasing OPN levels in CO-Fibs via recombinant OPN resulted in significant increases in their proliferative, migratory, and invasive capabilities to the levels resembling those of PH-Fibs. Thus our data suggest OPN as an essential contributor to the activated (highly proliferative, migratory, and proinvasive) phenotype of pulmonary adventitial fibroblasts in hypoxic PH.
Collapse
Affiliation(s)
- Adil Anwar
- Department of Pediatrics, University of Colorado Denver, Aurora, CO 80045, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
The mammalian heart loses its regenerative capacity during early postnatal stages; consequently, individuals surviving myocardial infarction are at risk of heart failure due to excessive fibrosis and maladaptive remodeling. There is an urgent need, therefore, to develop novel therapies for myocardial and coronary vascular regeneration. The epicardium-derived cells present a tractable resident progenitor source with the potential to stimulate neovasculogenesis and contribute de novo cardiomyocytes. The ability to revive ordinarily dormant epicardium-derived cells lies in the identification of key stimulatory factors, such as Tβ4, and elucidation of the molecular cues used in the embryo to orchestrate cardiovascular development. myocardial infarction injury signaling reactivates the adult epicardium; understanding the timing and magnitude of these signals will enlighten strategies for myocardial repair.
Collapse
Affiliation(s)
- Nicola Smart
- Molecular Medicine Unit, UCL-Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | | |
Collapse
|
50
|
Nerve sprouting contributes to increased severity of ventricular tachyarrhythmias by upregulating iGluRs in rats with healed myocardial necrotic injury. J Mol Neurosci 2012; 48:448-55. [PMID: 22383217 DOI: 10.1007/s12031-012-9720-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2011] [Accepted: 02/07/2012] [Indexed: 12/12/2022]
Abstract
Sympathetic nerve sprouting in healed myocardial infarction (MI) has been associated with high incidences of lethal arrhythmias, but the underlying mechanisms are largely unknown. This study sought to test that sympathetic hyperinnervation and/or MI remodels the myocardial glutamate signaling and ultimately increases the severity of ventricular tachyarrhythmias. Myocardial necrotic injury (MNI) was created by liquid nitrogen freeze-thawing across an intact diaphragm to mimic MI. Cardiac sympathetic hyperinnervation was induced by chronic subcutaneous injection of 4-methylcatechol, a potent stimulator of nerve growth factor expression. The results showed that sympathetic hyperinnervation with or without MNI upregulated the myocardial expression of ionotropic glutamate receptors (iGluRs), including NMDA receptor (NMDAR) and AMPA receptor (AMPAR), and induced cardiomyocyte apoptosis. Intravenous infusion with either NMDA (12 mg/kg) or AMPA (15 mg/kg) triggered ventricular tachycardia and ventricular fibrillation in rats with healed MNI plus sympathetic hyperinnervation; these arrhythmias were prevented by respective antagonist of NMDAR or AMPAR. We conclude that MNI with sympathetic nerve sprouting upregulates the expression of NMDAR and AMPAR in the myocardium and this impact in turn enhances cardiac responses to stimulations of iGluRs and thus increases the incidence of ventricular tachyarrhythmias.
Collapse
|