101
|
Ríos-Navarro C, Hueso L, Miñana G, Núñez J, Ruiz-Saurí A, Sanz MJ, Cànoves J, Chorro FJ, Piqueras L, Bodí V. El suero de seno coronario tras un infarto de miocardio induce angiogénesis y reparación de la obstrucción microvascular. Implicación del factor inducible por hipoxia-1A. Rev Esp Cardiol 2018. [DOI: 10.1016/j.recesp.2017.06.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
102
|
Ozturk S, Yetkin E, Waltenberger J. Molecular and cellular insights into the pathogenesis of coronary artery ectasia. Cardiovasc Pathol 2018; 35:37-47. [PMID: 29772448 DOI: 10.1016/j.carpath.2018.04.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/17/2018] [Accepted: 04/12/2018] [Indexed: 01/17/2023] Open
Abstract
Coronary artery ectasia describes a local or diffuse dilatation of the epicardial coronary arteries. This review summarizes the molecular and cellular mechanisms involved in the pathogenesis of coronary artery ectasia. Better identification of the pathophysiologic steps will shed light into the clinical significance and may have direct implications for the management strategies of this disease. Additionally, understanding the underlying etiology may help to improve treatment modalities specific to coronary artery ectasia.
Collapse
Affiliation(s)
- Selcuk Ozturk
- Ankara Education and Research Hospital, Department of Cardiology, Ankara 06230, Turkey.
| | - Ertan Yetkin
- Private Yenisehir Hospital, Department of Cardiology, Mersin, Turkey
| | - Johannes Waltenberger
- University of Münster, Faculty of Medicine, Münster, Germany; Cells-in-Motion Cluster of Excellence (EXC 1003-CiM), University of Münster, Münster, Germany
| |
Collapse
|
103
|
Tao J, Barnett JV, Watanabe M, Ramírez-Bergeron D. Hypoxia Supports Epicardial Cell Differentiation in Vascular Smooth Muscle Cells through the Activation of the TGFβ Pathway. J Cardiovasc Dev Dis 2018; 5:jcdd5020019. [PMID: 29652803 PMCID: PMC6023394 DOI: 10.3390/jcdd5020019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/04/2018] [Accepted: 04/04/2018] [Indexed: 12/11/2022] Open
Abstract
Epicardium-derived cells (EPDCs) are an important pool of multipotent cardiovascular progenitor cells. Through epithelial-to-mesenchymal-transition (EMT), EPDCs invade the subepicardium and myocardium and further differentiate into several cell types required for coronary vessel formation. We previously showed that epicardial hypoxia inducible factor (HIF) signaling mediates the invasion of vascular precursor cells critical for patterning the coronary vasculature. Here, we examine the regulatory role of hypoxia (1% oxygen) on EPDC differentiation into vascular smooth muscle cells (VSMCs). Results: Hypoxia stimulates EMT and enhances expression of several VSMC markers in mouse epicardial cell cultures. This stimulation is specifically blocked by inhibiting transforming growth factor-beta (TGFβ) receptor I. Further analyses indicated that hypoxia increases the expression level of TGFβ-1 ligand and phosphorylation of TGFβ receptor II, suggesting an indispensable role of the TGFβ pathway in hypoxia-stimulated VSMC differentiation. We further demonstrate that the non-canonical RhoA/Rho kinase (ROCK) pathway acts as the main downstream effector of TGFβ to modulate hypoxia’s effect on VSMC differentiation. Conclusion: Our results reveal a novel role of epicardial HIF in mediating coronary vasculogenesis by promoting their differentiation into VSMCs through noncanonical TGFβ signaling. These data elucidate that patterning of the coronary vasculature is influenced by epicardial hypoxic signals.
Collapse
Affiliation(s)
- Jiayi Tao
- Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Joey V Barnett
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Michiko Watanabe
- Department of Pediatrics, Rainbow Babies and Children's Hospital, The Congenital Heart Collaborative, Cleveland, OH 44106, USA.
| | - Diana Ramírez-Bergeron
- Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
- University Hospitals Harrington-McLaughlin Heart & Vascular Institute, Cleveland, OH 44106, USA.
| |
Collapse
|
104
|
Jakus IA, Jakus D, Aračić N, Stipić I, Vilović K. Immunohistochemical expression of hypoxia-inducible factor-1α in stromal cells of vaginal tissue in post-menopausal women with pelvic organ prolapse. Indian J Med Res 2018; 146:S63-S67. [PMID: 29578197 PMCID: PMC5890598 DOI: 10.4103/ijmr.ijmr_388_16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background & objectives: Pelvic organ prolapse (POP) is a common medical condition that affects adult women of different ages. The support of a normal pelvic floor is the result of complex interactions between ligaments, muscles, connective tissue and vaginal walls. Hypoxia and oxidative stress can reduce protein synthesis in the pelvic muscles that may contribute to muscular atrophy. Hypoxia-inducible factor-1α (HIF-1α) is a transcriptional activator which, expressed in response to hypoxia, activates a number of genes involved in cellular response to hypoxia. However, a potential role of hypoxia and oxidative stress in pathogenesis of POP is not known. This study was aimed to compare the level of HIF-1α immunohistochemical expression in the vaginal stromal cells of postmenopausal women with and without POP. Methods: Samples of the vaginal tissue from 120 menopausal women were obtained during surgery, and immunohistochemical expression of HIF-1α was assessed. There were 60 women with POP while 60 women in the control group were without prolapse but with benign gynaecological diseases. Results: In post-menopausal women with prolapse, significant differences were observed in the number of HIF-1α-positive stromal cells in the vaginal tissue compared to the control group. There was a significant increase in the number of HIF-1α in the stromal cells of the vaginal tissue in women with prolapse. Interpretation & conclusions: Difference in expression of HIF-1α in stromal cells of the vaginal tissue in the post-menopausal women with and without POP suggests that prolonged hypoxia probably has an important role in the aetiopathogenesis of POP.
Collapse
Affiliation(s)
- Ivana Alujević Jakus
- Clinical Department of Gynecology & Obstetrics, Split University Hospital Center, Split, Croatia
| | - Dora Jakus
- School of Medicine, University of Split, Split, Croatia
| | - Nađa Aračić
- Clinical Department of Gynecology & Obstetrics, Split University Hospital Center, Split, Croatia
| | - Ivica Stipić
- Clinical Department of Gynecology & Obstetrics, Split University Hospital Center, Split, Croatia
| | - Katarina Vilović
- Clinical Department of Pathology, Split University Hospital Center, Split, Croatia
| |
Collapse
|
105
|
Aljakna A, Lauer E, Lenglet S, Grabherr S, Fracasso T, Augsburger M, Sabatasso S, Thomas A. Multiplex quantitative imaging of human myocardial infarction by mass spectrometry-immunohistochemistry. Int J Legal Med 2018; 132:1675-1684. [PMID: 29556718 DOI: 10.1007/s00414-018-1813-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 02/22/2018] [Indexed: 02/01/2023]
Abstract
Simultaneous assessment of a panel of protein markers is becoming essential in order to enhance biomarker research and improve diagnostics. Specifically, postmortem diagnostics of early myocardial ischemia in sudden cardiac death cases could benefit from a multiplex marker assessment in the same tissue section. Current analytical antibody-based techniques (immunohistochemistry and immunofluorescence) limit multiplex analysis usually to not more than three antibodies. In this study, mass spectrometry-immunohistochemistry (MS-IHC) was performed by combining laser ablation inductively coupled plasma mass spectrometry (LA-ICP-MS) with rare-metal-isotope-tagged antibodies as a technique for multiplex analysis of human postmortem myocardial tissue samples. Tissue sections with myocardial infarction were simultaneously analyzed for seven primary, rare-metal-isotope-tagged antibodies (troponin T, myoglobin, fibronectin, C5b-9, unphosphorylated connexin 43, VEGF-B, and JunB). Comparison between the MS-IHC approach and chromogenic IHC showed similar patterns in ionic and optical images. In addition, absolute quantification was performed by MS-IHC, providing a proportional relationship between the signal intensity and the local marker concentration in tissue sections. These data demonstrated that LA-ICP-MS combined with rare-metal-isotope-tagged antibodies is an efficient strategy for simultaneous testing of multiple markers and allows not only visualization of molecules within the tissue but also quantification of the signal. Such imaging approach has a great potential in both diagnostics and pathology-related research.
Collapse
Affiliation(s)
- Aleksandra Aljakna
- University Center of Legal Medicine, Lausanne-Geneva, Rue Michel-Servet, 11211, Geneva, Switzerland
| | - Estelle Lauer
- University Center of Legal Medicine, Lausanne-Geneva, Rue Michel-Servet, 11211, Geneva, Switzerland
| | - Sébastien Lenglet
- University Center of Legal Medicine, Lausanne-Geneva, Rue Michel-Servet, 11211, Geneva, Switzerland
| | - Silke Grabherr
- University Center of Legal Medicine, Lausanne-Geneva, Rue Michel-Servet, 11211, Geneva, Switzerland
| | - Tony Fracasso
- University Center of Legal Medicine, Lausanne-Geneva, Rue Michel-Servet, 11211, Geneva, Switzerland
| | - Marc Augsburger
- University Center of Legal Medicine, Lausanne-Geneva, Rue Michel-Servet, 11211, Geneva, Switzerland
| | - Sara Sabatasso
- University Center of Legal Medicine, Lausanne-Geneva, Rue Michel-Servet, 11211, Geneva, Switzerland
| | - Aurélien Thomas
- University Center of Legal Medicine, Lausanne-Geneva, Rue Michel-Servet, 11211, Geneva, Switzerland.
- Faculty of Biology and Medicine, University of Lausanne, Vulliette 04, 1000, Lausanne 25, Switzerland.
| |
Collapse
|
106
|
Seo Y, Jung Y, Kim SH. Decellularized heart ECM hydrogel using supercritical carbon dioxide for improved angiogenesis. Acta Biomater 2018; 67:270-281. [PMID: 29223704 DOI: 10.1016/j.actbio.2017.11.046] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 11/08/2017] [Accepted: 11/28/2017] [Indexed: 12/30/2022]
Abstract
Initial angiogenesis within the first 3 days is critical for healing ischemic diseases such as myocardial infarction. Recently, decellularized extracellular matrix (dECM) has been reported to provide tissue-derived ECM components and can be used as a scaffold for cell delivery for angiogenesis in tissue engineering. Decellularization by various detergents such as sodium dodecyl sulfate (SDS) and triton X-100 can remove the cell nuclei in tissue organs. However, this leads to ECM structure denaturation, decreased presence of various ECM proteins and cytokines, and loss of mechanical properties. To overcome these limitations, in this study, we developed a supercritical carbon dioxide and ethanol co-solvent (scCO2-EtOH) decellularization method, which is a detergent-free system that prevents ECM structure disruption and retains various angiogenic proteins in the heart dECM, and tested on rat heart tissues. The heart tissue was placed into the scCO2 reactor and decellularized at 37 °C and 350 bar. After scCO2-EtOH treatment, the effects were evaluated by DNA, collagen, and glycosaminoglycan (GAG) quantification and hematoxylin and eosin and immunofluorescence staining to determine the absence of nucleic acids and preservation of heart ECM components. Similar to the native group, the scCO2-EtOH group contained more ECM components such as collagen, GAGs, collagen I, laminin, and fibronectin and angiogenic factors including vascular endothelial growth factor, fibroblast growth factor, and platelet-derived growth factor and others in comparison to the detergent group. In addition, to estimate angiogenesis of the dECM hydrogels, the neutralized dECM solution was injected in a rat subcutaneous layer (n = 6 in each group: collagen, scCO2-EOH, and detergent group), after which the solution naturally formed gelation in the subcutaneous layer. After 3 days, the gels were harvested and estimated by immunofluorescence staining and the ImageJ program for angiogenesis analysis. Consequently, blood vessel formation and density of vWF and α-SMA in the scCO2-EtOH group were significantly greater than that in the collagen group. Here we suggest that heart-derived decellularized extracellular matrix (dECM) with scCO2-EtOH treatment is a highly promising angiogenic material for healing in ischemic disease. STATEMENT OF SIGNIFICANCE Supercritical carbon dioxide (scCO2) in a supercritical phase has low viscosity and high diffusivity between gas and liquid properties and is known to be affordable, non-toxic, and eco-friendly. Therefore, scCO2 extraction technology has been extensively used in commercial and industrial fields. Recently, decellularized extracellular matrix (dECM) was applied to tissue engineering and regenerative medicine as a scaffold, therapeutic material, and bio-ink for 3D printing. Moreover, the general decellularization method using detergents has limitations including eliminating tissue-derived ECM components and disrupting their structures after decellularization. To overcome these limitations, heart tissues were treated with scCO2-EtOH for decellularization, resulting in preserving of tissue due to the various ECM and angiogenic factors derived. In addition, initiation of angiogenesis was highly induced even after 3 days of injection.
Collapse
Affiliation(s)
- Yoojin Seo
- NBIT, KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea; Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Youngmee Jung
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Department of Biomedical Engineering, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea.
| | - Soo Hyun Kim
- NBIT, KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea; Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Department of Biomedical Engineering, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea.
| |
Collapse
|
107
|
Gentile MT, Russo R, Pastorino O, Cioffi S, Barbieri F, Illingworth EA, Grieco M, Chambery A, Colucci-D'Amato L. Ruta graveolens water extract inhibits cell-cell network formation in human umbilical endothelial cells via MEK-ERK1/2 pathway. Exp Cell Res 2018; 364:50-58. [PMID: 29366810 DOI: 10.1016/j.yexcr.2018.01.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 01/10/2018] [Accepted: 01/19/2018] [Indexed: 01/01/2023]
Abstract
Angiogenesis is a process encompassing several steps such as endothelial cells proliferation, differentiation and migration to form a vascular network, involving different signal transduction pathways. Among these, ERK1/2 signaling mediates VEGF-dependent signaling pathway. Here we report that the water extract of Ruta graveolens (RGWE), widely known as a medicinal plant, is able to impair in a dose-dependent manner, cell network formation without affecting cell viability. Biochemical analysis showed that the major component of RGWE is rutin, unable to reproduce RGWE effect. We found that RGWE inhibits ERK1/2 phosphorylation and that this event is crucial in cell network formation since the transfection of HUVEC with a constitutively active MEK (caMEK), the ERK1/2 activator, induces a robust cell network formation as compared to untransfected and/or mock transfected cells and, more importantly, caMEK transfected cells became unresponsive to RGWE. Moreover, RGWE inhibits VEGF and nestin gene expression, necessary for vessel formation, and the caMEK transfection induces their higher expression. In conclusion, we report that RGWE is able to significantly impair vessels network formation without affecting cell viability, preventing ERK1/2 activation and, in turn, down-regulating VEGF and nestin expression. These findings point to RGWE as a potential therapeutic tool capable to interfere with pathologic angiogenesis.
Collapse
Affiliation(s)
- Maria Teresa Gentile
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Rosita Russo
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Olga Pastorino
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Sara Cioffi
- Dipartimento di Chimica e Biologia "Adolfo Zambelli", Università degli Studi di Salerno, Italy; Institute of Genetics and Biophysics "ABT", CNR, Naples, Italy
| | | | - Elisabeth Anne Illingworth
- Dipartimento di Chimica e Biologia "Adolfo Zambelli", Università degli Studi di Salerno, Italy; Institute of Genetics and Biophysics "ABT", CNR, Naples, Italy
| | - Michele Grieco
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Angela Chambery
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Luca Colucci-D'Amato
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy.
| |
Collapse
|
108
|
Renko O, Tolonen AM, Rysä J, Magga J, Mustonen E, Ruskoaho H, Serpi R. SDF1 gradient associates with the distribution of c-Kit+ cardiac cells in the heart. Sci Rep 2018; 8:1160. [PMID: 29348441 PMCID: PMC5773575 DOI: 10.1038/s41598-018-19417-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 12/29/2017] [Indexed: 12/11/2022] Open
Abstract
Identification of the adult cardiac stem cells (CSCs) has offered new therapeutic possibilities for treating ischemic myocardium. CSCs positive for the cell surface antigen c-Kit are known as the primary source for cardiac regeneration. Accumulating evidence shows that chemokines play important roles in stem cell homing. Here we investigated molecular targets to be utilized in modulating the mobility of endogenous CSCs. In a four week follow-up after experimental acute myocardial infarction (AMI) with ligation of the left anterior descending (LAD) coronary artery of Sprague-Dawley rats c-Kit+ CSCs redistributed in the heart. The number of c-Kit+ CSCs in the atrial c-Kit niche was diminished, whereas increased amount was observed in the left ventricle and apex. This was associated with increased expression of stromal cell-derived factor 1 alpha (SDF1α), and a significant positive correlation was found between c-Kit+ CSCs and SDF1α expression in the heart. Moreover, the migratory capacity of isolated c-Kit+ CSCs was induced by SDF1 treatment in vitro. We conclude that upregulation of SDF1α after AMI associates with increased expression of endogenous c-Kit+ CSCs in the injury area, and show induced migration of c-Kit+ cells by SDF1.
Collapse
Affiliation(s)
- Outi Renko
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Anna-Maria Tolonen
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Jaana Rysä
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Johanna Magga
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Erja Mustonen
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Heikki Ruskoaho
- Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki, Finland
| | - Raisa Serpi
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu Center for Cell-Matrix Research, University of Oulu, Oulu, Finland.
| |
Collapse
|
109
|
Mechanisms of 2,3,7,8-tetrachlorodibenzo-p-dioxin- induced cardiovascular toxicity: An overview. Chem Biol Interact 2018; 282:1-6. [PMID: 29317249 DOI: 10.1016/j.cbi.2018.01.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 12/05/2017] [Accepted: 01/05/2018] [Indexed: 11/21/2022]
Abstract
2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is an environmental contaminant and its toxicity is mediated by the aryl hydrocarbon receptor (AHR). Mechanisms of TCDD cardiovascular toxicity consist of oxidative stress, growth factor modulation, and ionic current alteration. It is indicated that the rodent cardiovascular system is a target for TCDD cardiomyopathy. Here, our understanding of TCDD cardiovascular toxicity is reviewed.
Collapse
|
110
|
Jia Z, Lian W, Shi H, Cao C, Han S, Wang K, Li M, Zhang X. Ischemic Postconditioning Protects Against Intestinal Ischemia/Reperfusion Injury via the HIF-1α/miR-21 Axis. Sci Rep 2017; 7:16190. [PMID: 29170412 PMCID: PMC5700993 DOI: 10.1038/s41598-017-16366-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 11/12/2017] [Indexed: 12/30/2022] Open
Abstract
Intestinal ischemia/reperfusion (I/R) can lead to tissue damage associated with inflammation and mucosal apoptosis. Ischemic postconditioning (IPostC), a series of repeated, brief, intermittent periods of ischemia and reperfusion, has beneficial effects against I/R-induced injury in the heart and intestine, although the underlying mechanisms for these effects remain unclear. We evaluated the involvement of microRNA-21 (miR-21) in the protective effects of IPostC in a rat model of I/R induced by superior mesenteric artery occlusion and reopening. IPostC decreased I/R injury and suppressed apoptosis in the intestinal tissues concomitant with the induction of hypoxia inducible factor 1 alpha (HIF-1α) and the upregulation of miR-21. In vitro experiments in the intestinal epithelial cell line IEC-6 showed that hypoxia induced miR-21 and this effect was abolished by silencing HIF1-α, confirming the induction of miR-21 by HIF1-α, HIF1-α or miR-21 inhibition exacerbated I/R induced apoptosis, and programmed cell death 4 (PDCD4) and Fas-L was involved in miR-21 mediated anti-apoptotic effects on intestinal epithelial cells. Knockdown of miR-21 or inhibition of HIF1-α abolished the IPostC-mediated attenuation of intestinal injury and apoptosis and the downregulation of PDCD4 and Fas-L. A potential mechanism underlying the protective effect of IPostC may therefore involve the induction of miR-21 by HIF1-α and the attenuation of apoptosis via the downregulation of PDCD4 and Fas-L.
Collapse
Affiliation(s)
- Zhongzhi Jia
- Department of Interventional Radiology, No. 2 People's Hospital of Changzhou, Nanjing Medical University, Chang zhou, 213003, China
| | - Weishuai Lian
- Department of Interventional Radiology, Shanghai Tenth People's Hospital, Shanghai, 200072, China
| | - Haifeng Shi
- Department of Interventional Radiology, No. 2 People's Hospital of Changzhou, Nanjing Medical University, Chang zhou, 213003, China
| | - Chuanwu Cao
- Department of Interventional Radiology, Shanghai Tenth People's Hospital, Shanghai, 200072, China
| | - Shilong Han
- Department of Interventional Radiology, Shanghai Tenth People's Hospital, Shanghai, 200072, China
| | - Kai Wang
- Department of Interventional Radiology, No. 2 People's Hospital of Changzhou, Nanjing Medical University, Chang zhou, 213003, China
| | - Maoquan Li
- Department of Interventional Radiology, Shanghai Tenth People's Hospital, Shanghai, 200072, China. .,Institute of Medical Intervention Engineering, Tongji University, Shanghai, 200072, China.
| | - Xiaoping Zhang
- Institute of Medical Intervention Engineering, Tongji University, Shanghai, 200072, China.
| |
Collapse
|
111
|
Zhang W, Zhao X, Xiao Y, Chen J, Han P, Zhang J, Fu H, James Kang Y. The association of depressed angiogenic factors with reduced capillary density in the Rhesus monkey model of myocardial ischemia. Metallomics 2017; 8:654-62. [PMID: 26852735 DOI: 10.1039/c5mt00332f] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Depressed capillary density is associated with myocardial ischemic infarction, in which hypoxia-inducible factor 1α (HIF-1α) is increased. The present study was undertaken to examine changes in the angiogenic factors whose expression is regulated by HIF-1 and their relation to the depressed capillary density in the Rhesus monkey model of myocardial ischemic infarction. Male Rhesus monkeys 2-3 years old were subjected to myocardial ischemia by permanent ligation of left anterior descending (LAD) artery leading to the development of myocardial infarction. Eight weeks after LAD ligation, copper concentrations, myocardial histological changes and capillary density were examined, along with Western blot and immunohistochemical analysis of angiogenic factors and detection of HIF-1 activity. Capillary density was significantly decreased but the concentrations of HIF-1α and HIF-1β were significantly increased in the infarct area. However, the levels of mRNA and protein for VEGF and VEGFR1 were significantly decreased. Other HIF-1 regulated angiogenic factors, including Tie-2, Ang-1 and FGF-1, were also significantly depressed, but vascular destabilizing factor Ang-2 was significantly increased. Copper concentrations were depressed in the infarct area. Copper-independent HIF-1 activity was increased shown by the elevated mRNA level of IGF-2, a HIF-1 target gene. Removal of copper by a copper chelator, tetraethylenepentamine, from primary cultures of neonatal rat cardiomyocytes also suppressed the expression of HIF-1 regulated VEGF and BNIP3, but not IGF-2. The data suggest that under ischemic conditions, copper loss suppressed the expression of critical angiogenic genes regulated by HIF-1, but did not affect copper-independent HIF-1 activation of gene expression. This copper-dependent dysregulation of angiogenic gene expression would contribute to the pathogenesis of myocardial ischemic infarction.
Collapse
Affiliation(s)
- Wenjing Zhang
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, China.
| | - Xinmei Zhao
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, China.
| | - Ying Xiao
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, China.
| | - Jianmin Chen
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, China.
| | - Pengfei Han
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, China.
| | - Jingyao Zhang
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, China.
| | - Haiying Fu
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, China.
| | - Y James Kang
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, China. and Department of Pharmacology and Toxicology, University of Louisville, School of Medicine, Louisville, Kentucky 40202, USA
| |
Collapse
|
112
|
Franchi F, Peterson KM, Paulmurugan R, Folmes C, Lanza IR, Lerman A, Rodriguez-Porcel M. Noninvasive Monitoring of the Mitochondrial Function in Mesenchymal Stromal Cells. Mol Imaging Biol 2017; 18:510-8. [PMID: 26865378 DOI: 10.1007/s11307-016-0929-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE Mitochondria are a gatekeeper of cell survival and mitochondrial function can be used to monitor cell stress. Here we validate a pathway-specific reporter gene to noninvasively image the mitochondrial function of stem cells. PROCEDURES We constructed a mitochondrial sensor with the firefly luciferase (Fluc) reporter gene driven by the NQO1 enzyme promoter. The sensor was introduced in stem cells and validated in vitro and in vivo, in a mouse model of myocardial ischemia/reperfusion (IR). RESULTS The sensor activity showed an inverse relationship with mitochondrial function (R (2) = -0.975, p = 0.025) and showed specificity and sensitivity for mitochondrial dysfunction. In vivo, NQO1-Fluc activity was significantly higher in IR animals vs. controls, indicative of mitochondrial dysfunction, and was corroborated by ex vivo luminometry. CONCLUSIONS Reporter gene imaging allows assessment of the biology of transplanted mesenchymal stromal cells (MSCs), providing important information that can be used to improve the phenotype and survival of transplanted stem cells.
Collapse
Affiliation(s)
- Federico Franchi
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Karen M Peterson
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Ramasamy Paulmurugan
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA, USA
| | - Clifford Folmes
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Ian R Lanza
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Amir Lerman
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Martin Rodriguez-Porcel
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA.
| |
Collapse
|
113
|
Durymanov M, Kamaletdinova T, Lehmann SE, Reineke J. Exploiting passive nanomedicine accumulation at sites of enhanced vascular permeability for non-cancerous applications. J Control Release 2017. [DOI: 10.1016/j.jconrel.2017.06.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
114
|
Dynamics and implications of circulating anti-angiogenic VEGF-A 165b isoform in patients with ST-elevation myocardial infarction. Sci Rep 2017; 7:9962. [PMID: 28855597 PMCID: PMC5577291 DOI: 10.1038/s41598-017-10505-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 08/09/2017] [Indexed: 12/22/2022] Open
Abstract
Angiogenesis is crucial to restore microvascular perfusion in the jeopardized myocardium in the weeks following reperfused ST-segment elevation myocardial infarction (STEMI). (VEGF)-A165b, an anti-angiogenic factor, has been identified as a regulator of vascularization; however, it has not been previously implicated in acute myocardial infarction. We sought to investigate the dynamics of circulating VEGF-A165b and its association with cardiac magnetic resonance-derived infarct size and left ventricular ejection fraction (LVEF). 50 STEMI patients and 23 controls were included. Compared with control individuals, serum VEGF-A165b was elevated in STEMI patients prior to primary percutaneous coronary intervention (PCI). Following PCI, serum VEGF-A165b increased further, reaching a maximum level at 24 h and decreased one month after reperfusion. VEGF-A165b levels at 24 h were associated with a large infarct size and inversely related to LVEF. VEGF-A165b expression was increased in myocardial infarct areas from patients with previous history of AMI. An ex vivo assay using serum from STEMI patients showed that neutralization of VEGF-A165b increased tubulogenesis. Overall, the study suggests that VEGF-A165b might play a deleterious role after AMI as an inhibitor of angiogenesis in the myocardium. Accordingly, neutralization of VEGF-A165b could represent a novel pro-angiogenic therapy for reperfusion of myocardium in STEMI.
Collapse
|
115
|
Ljubojevic-Holzer S. The unexpected intelligence: what is the naked mole–rat’s secret to surviving oxygen deprivation? Cardiovasc Res 2017; 113:e27-e28. [DOI: 10.1093/cvr/cvx114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
116
|
Ríos-Navarro C, Hueso L, Miñana G, Núñez J, Ruiz-Saurí A, Sanz MJ, Cànoves J, Chorro FJ, Piqueras L, Bodí V. Coronary Serum Obtained After Myocardial Infarction Induces Angiogenesis and Microvascular Obstruction Repair. Role of Hypoxia-inducible Factor-1A. ACTA ACUST UNITED AC 2017; 71:440-449. [PMID: 28751164 DOI: 10.1016/j.rec.2017.06.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 06/21/2017] [Indexed: 11/28/2022]
Abstract
INTRODUCTION AND OBJECTIVES Microvascular obstruction (MVO) exerts deleterious effects following acute myocardial infarction (AMI). We investigated coronary angiogenesis induced by coronary serum and the role of hypoxia-inducible factor-1A (HIF-1A) in MVO repair. METHODS Myocardial infarction was induced in swine by transitory 90-minute coronary occlusion. The pigs were divided into a control group and 4 AMI groups: no reperfusion, 1minute, 1 week and 1 month after reperfusion. Microvascular obstruction and microvessel density were quantified. The proangiogenic effect of coronary serum drawn from coronary sinus on endothelial cells was evaluated using an in vitro tubulogenesis assay. Circulating and myocardial HIF-1A levels and the effect of in vitro blockade of HIF-1A was assessed. RESULTS Compared with control myocardium, microvessel density decreased at 90-minute ischemia, and MVO first occurred at 1minute after reperfusion. Both peaked at 1 week and almost completely resolved at 1 month. Coronary serum exerted a neoangiogenic effect on coronary endothelial cells in vitro, peaking at ischemia and 1minute postreperfusion (32 ± 4 and 41 ± 9 tubes vs control: 3 ± 3 tubes; P < .01). Hypoxia-inducible factor-1A increased in serum during ischemia (5-minute ischemia: 273 ± 52 pg/mL vs control: 148 ± 48 pg/mL; P < .01) being present on microvessels of all AMI groups (no reperfusion: 67% ± 5% vs control: 15% ± 17%; P < .01). In vitro blockade of HIF-1A reduced the angiogenic response induced by serum. CONCLUSIONS Coronary serum represents a potent neoangiogenic stimulus even before reperfusion; HIF-1A might be crucial. Coronary neoangiogenesis induced by coronary serum can contribute to understanding the pathophysiology of AMI.
Collapse
Affiliation(s)
| | - Luisa Hueso
- Instituto de Investigación Sanitaria-INCLIVA, Valencia, Spain
| | - Gema Miñana
- Instituto de Investigación Sanitaria-INCLIVA, Valencia, Spain; Servicio de Cardiología, Hospital Clínico Universitario, Valencia, Spain; Departamento de Medicina, Universidad de Valencia, Valencia, Spain
| | - Julio Núñez
- Instituto de Investigación Sanitaria-INCLIVA, Valencia, Spain; Servicio de Cardiología, Hospital Clínico Universitario, Valencia, Spain; Departamento de Medicina, Universidad de Valencia, Valencia, Spain
| | - Amparo Ruiz-Saurí
- Instituto de Investigación Sanitaria-INCLIVA, Valencia, Spain; Departamento de Patología, Universidad de Valencia, Valencia, Spain
| | - María Jesús Sanz
- Instituto de Investigación Sanitaria-INCLIVA, Valencia, Spain; Departamento de Farmacología, Universidad de Valencia, Valencia, Spain
| | - Joaquin Cànoves
- Instituto de Investigación Sanitaria-INCLIVA, Valencia, Spain; Servicio de Cardiología, Hospital Clínico Universitario, Valencia, Spain; Departamento de Medicina, Universidad de Valencia, Valencia, Spain; Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Francisco J Chorro
- Instituto de Investigación Sanitaria-INCLIVA, Valencia, Spain; Servicio de Cardiología, Hospital Clínico Universitario, Valencia, Spain; Departamento de Medicina, Universidad de Valencia, Valencia, Spain; Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Laura Piqueras
- Instituto de Investigación Sanitaria-INCLIVA, Valencia, Spain.
| | - Vicente Bodí
- Instituto de Investigación Sanitaria-INCLIVA, Valencia, Spain; Servicio de Cardiología, Hospital Clínico Universitario, Valencia, Spain; Departamento de Medicina, Universidad de Valencia, Valencia, Spain; Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
| |
Collapse
|
117
|
Binó L, Procházková J, Radaszkiewicz KA, Kučera J, Kudová J, Pacherník J, Kubala L. Hypoxia favors myosin heavy chain beta gene expression in an Hif-1alpha-dependent manner. Oncotarget 2017; 8:83684-83697. [PMID: 29137374 PMCID: PMC5663546 DOI: 10.18632/oncotarget.19016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 06/18/2017] [Indexed: 11/25/2022] Open
Abstract
The potentiation of the naturally limited regenerative capacity of the heart is dependent on an understanding of the mechanisms that are activated in response to pathological conditions such as hypoxia. Under these conditions, the expression of genes suggested to support cardiomyocyte survival and heart adaptation is triggered. Particularly important are changes in the expression of myosin heavy chain (MHC) isoforms. We propose here that alterations in the expression profiles of MHC genes are induced in response to hypoxia and are primarily mediated by hypoxia inducible factor (HIF). In in vitro models of mouse embryonic stem cell-derived cardiomyocytes, we showed that hypoxia (1% O2) or the pharmacological stabilization of HIFs significantly increased MHCbeta (Myh7) gene expression. The key role of HIF-1alpha is supported by the absence of these effects in HIF-1alpha-deficient cells, even in the presence of HIF-2alpha. Interestingly, ChIP analysis did not confirm the direct interaction of HIF-1alpha with putative HIF response elements predicted in the MHCalpha and beta encoding DNA region. Further analyses showed the significant effect of the mTOR signaling inhibitor rapamycin in inducing Myh7 expression and a hypoxia-triggered reduction in the levels of antisense RNA transcripts associated with the Myh7 gene locus. Overall, the recognized and important role of HIF in the regulation of heart regenerative processes could be highly significant for the development of novel therapeutic interventions in heart failure.
Collapse
Affiliation(s)
- Lucia Binó
- Institute of Biophysics of the CAS, Brno, Czech Republic.,Institute of Experimental Biology, Department of Physiology and Immunology of Animals, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Jiřina Procházková
- Institute of Biophysics of the CAS, Brno, Czech Republic.,Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Katarzyna Anna Radaszkiewicz
- Institute of Experimental Biology, Department of Physiology and Immunology of Animals, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Jan Kučera
- Institute of Experimental Biology, Department of Physiology and Immunology of Animals, Faculty of Science, Masaryk University, Brno, Czech Republic.,International Clinical Research Center, Center of Biomolecular and Cellular Engineering, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Jana Kudová
- Institute of Biophysics of the CAS, Brno, Czech Republic.,Institute of Experimental Biology, Department of Physiology and Immunology of Animals, Faculty of Science, Masaryk University, Brno, Czech Republic.,International Clinical Research Center, Center of Biomolecular and Cellular Engineering, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Jiří Pacherník
- Institute of Experimental Biology, Department of Physiology and Immunology of Animals, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Lukáš Kubala
- Institute of Biophysics of the CAS, Brno, Czech Republic.,Institute of Experimental Biology, Department of Physiology and Immunology of Animals, Faculty of Science, Masaryk University, Brno, Czech Republic.,International Clinical Research Center, Center of Biomolecular and Cellular Engineering, St. Anne's University Hospital Brno, Brno, Czech Republic
| |
Collapse
|
118
|
Hasin T, Iakobishvili Z, Weisz G. Associated Risk of Malignancy in Patients with Cardiovascular Disease: Evidence and Possible Mechanism. Am J Med 2017; 130:780-785. [PMID: 28344133 DOI: 10.1016/j.amjmed.2017.02.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 02/10/2017] [Accepted: 02/13/2017] [Indexed: 12/31/2022]
Abstract
Cardiovascular disease and malignancy are leading causes of morbidity and mortality. Increased risk of malignancy was identified in patients with cardiovascular disease, including patients with heart failure, heart failure after myocardial infarction, patients undergoing cardiac intervention, and patients after a thrombotic event. Common risk factors and biological pathways can explain this association and are explored in this review. Further research is needed to establish the causes of malignancy in this population and direct possible intervention.
Collapse
Affiliation(s)
- Tal Hasin
- Department of Cardiology, Shaare Zedek Medical Center, Jerusalem, Israel.
| | - Zaza Iakobishvili
- Department of Cardiology, Rabin Medical Center, Petach Tiqwa, Israel
| | - Giora Weisz
- Department of Cardiology, Shaare Zedek Medical Center, Jerusalem, Israel
| |
Collapse
|
119
|
Wen SY, Tamilselvi S, Shen CY, Day CH, Chun LC, Cheng LY, Ou HC, Chen RJ, Viswanadha VP, Kuo WW, Huang CY. Protective effect of HDL on NADPH oxidase-derived super oxide anion mediates hypoxia-induced cardiomyocyte apoptosis. PLoS One 2017; 12:e0179492. [PMID: 28617849 PMCID: PMC5472312 DOI: 10.1371/journal.pone.0179492] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 05/31/2017] [Indexed: 11/20/2022] Open
Abstract
Cardiovascular diseases are the leading cause of death of death in Taiwan. Atherosclerosis can lead to serious problems, including heart attack, stroke, or even death. Coronary heart disease (CHD) occurs when plaque builds up in the coronary arteries to cause the ischemic heart disease which will enhance myocardial remodeling and also induce myocardial hypoxia. High density lipoprotein (HDL) has been proposed to have cardio-protective effects. Under hypoxic conditions (1%O2 for 24hr), in H9c2 cells, reactive oxygen species (ROS) is induced which leads to cardiomyocyte apoptosis and cardiac dysfunction. Therefore, the present study described the protective effect of HDL on hypoxia-induced cardiomyocyte damage. We investigated the NADPH oxidase-produced ROS-related signaling pathways and apoptosis in cardiomyocytes under hypoxia conditions. Results showed that the ROS mediated cardiac damage might occur via AT1 and PKC activation. Furthermore, hypoxia downregulated the survival protein (p-AKTser473) and anti-apoptotic protein (BCL2), whereas pro-apoptotic protein, Bax and caspase 3 were upregulated. These detrimental effects by ROS and apoptosis were prevented by HDL pretreatment. Our findings revealed the underlying molecular mechanism by which HDL suppresses the hypoxia-induced cardiomyocyte dysfunction. Further, we elucidated the role of HDL on preventing hypoxia induced cardiomyocyte apoptosis is mediated through the inhibition of NADPH oxidase-derived ROS.
Collapse
Affiliation(s)
- Su-Ying Wen
- Department of Dermatology, Taipei City Hospital, Renai Branch, Taipei, Taiwan
- Center for General Education, Mackay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan
| | - Shanmugam Tamilselvi
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Chia-Yao Shen
- Department of Nursing, MeiHo University, Pingtung, Taiwan
| | | | - Li-Chin Chun
- Department of Hospital and Health Care Administration, Chia Nan University of Pharmacy & Science, Tainan County, Taiwan
| | - Li-Yi Cheng
- Department of Biological Science and Technology, Asia University, Taichung, Taiwan
| | - Hsiu-Chung Ou
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Ray-Jade Chen
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | | | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- Department of Biological Science and Technology, Asia University, Taichung, Taiwan
- Graduate Institute of Chinese Medical Science, China Medical University, Taichung, Taiwan
- * E-mail:
| |
Collapse
|
120
|
Yu J, Zhang X, Zhang Y. Astragaloside attenuates myocardial injury in a rat model of acute myocardial infarction by upregulating hypoxia inducible factor‑1α and Notch1/Jagged1 signaling. Mol Med Rep 2017; 15:4015-4020. [PMID: 28487976 PMCID: PMC5436283 DOI: 10.3892/mmr.2017.6522] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 02/16/2017] [Indexed: 12/12/2022] Open
Abstract
The present study aimed to investigate the mechanisms underlying the cardioprotective effect of Astragaloside against myocardial injury following myocardial infarction (MI) in a rat model. Male Wistar rats were subjected to left anterior descending branch ligation. The rats that survived 24 h (n=18) were randomly and equally assigned to three groups: MI model group, and 2.5 and 10 mg/kg/day Astragaloside group. A further six rats underwent identical surgical procedures without artery ligation, serving as sham controls. Following 28 days of treatment, the left ventricle was harvested for morphological analysis, and mRNA and protein expression levels of hypoxia inducible factor‑1α (HIF‑1α), Notch1 and Jagged1 were measured. Treatment with Astragaloside attenuated pathological changes in the myocardium. Compared with untreated MI rats, rats treated with Astragaloside exhibited significantly increased mRNA expression levels of HIF‑1α, Notch1 and Jagged1 (all P<0.01). HIF‑1α demonstrated a dose‑dependent effect (P<0.05). Astragaloside (10 mg/kg/day) significantly increased HIF‑1α (P<0.05), Notch1 (P<0.01) and Jagged1 (P<0.01) protein expression levels. Additionally, 2.5 mg/kg Astragaloside significantly increased Jagged1 protein expression levels compared with untreated MI rats. Furthermore, there was a dose‑dependent effect of Astragaloside treatment (P<0.01). These findings suggested that the cardioprotective effects of Astragaloside against myocardial injury following MI may involve upregulation of HIF‑α, Notch1 and Jagged1 signaling, implicating these molecules as therapeutic targets for the treatment of MI.
Collapse
Affiliation(s)
- Junmin Yu
- Department of Geriatrics, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xiaobo Zhang
- Department of Geriatrics, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yina Zhang
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| |
Collapse
|
121
|
Yamada Y, Kinoshita I, Kohashi K, Yamamoto H, Kuma Y, Ito T, Koda K, Kisanuki A, Kurosawa M, Yoshimura M, Furue M, Oda Y. HIF-1α, MDM2, CDK4, and p16 expression in ischemic fasciitis, focusing on its ischemic condition. Virchows Arch 2017; 471:117-122. [PMID: 28477272 DOI: 10.1007/s00428-017-2122-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 03/02/2017] [Accepted: 04/04/2017] [Indexed: 11/28/2022]
Abstract
Ischemic fasciitis is a benign myofibroblastic lesion, occurring in the sacral region or proximal thigh of elderly or bedridden individuals. The pathogenesis of ischemic fasciitis is thought to be based on ischemic condition; however, it has never been demonstrated. In this study, we examined the expression of ischemia-associated proteins in ischemic fasciitis by immunohistochemical and genetic methods. Specifically, this study aimed to reveal the expression of HIF-1α, MDM2, CDK4, p16, and gene amplification of MDM2 gene. Seven cases of ischemic fasciitis from among the soft-tissue tumors registered at our institution were retrieved. Histopathological findings were as follows: poorly demarcated nodular masses, a proliferation of spindle-shaped fibroblastic or myofibroblastic cells with oval nuclei and eosinophilic or pale cytoplasm, zonal fibrinous deposition, pseudocystic degeneration, granulation-like proliferation of capillary vessels, ganglion-like cells, myxoid or hyalinized stroma, and chronic inflammatory infiltration. Immunohistochemically, the spindle cells were positive for HIF-1α (7/7 cases), MDM2 (4/7 cases), CDK4 (4/7 cases), p16 (7/7 cases), p53 (2/7 case), cyclin D1 (7/7 cases), and alpha-smooth muscle actin (6/7 cases). Neither MDM2 gene amplification nor USP6 gene split signal was detected in any case. Overexpression of the above proteins may be associated with the pathogenic mechanism of ischemic fasciitis. It is noted that the immunohistochemical positivity of MDM2, CDK4, and p16 do not necessarily indicate malignant neoplasm such as dedifferentiated liposarcoma.
Collapse
Affiliation(s)
- Yuichi Yamada
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka-ken, 812-8582, Japan
| | - Izumi Kinoshita
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka-ken, 812-8582, Japan
| | - Kenichi Kohashi
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka-ken, 812-8582, Japan
| | - Hidetaka Yamamoto
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka-ken, 812-8582, Japan
| | - Yuki Kuma
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka-ken, 812-8582, Japan
| | - Takamichi Ito
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka-ken, 812-8582, Japan
| | - Kenji Koda
- Department of Pathology, Fujieda Municipal General Hospital, 4-1-11 Surugadai, Fujieda-shi, Shizuoka-ken, 426-8677, Japan
| | - Atsushi Kisanuki
- Department of Pathology, Nichinan Prefectural Miyazaki Hospital, 1-9-5 Kiyama, Nichinan-shi, Miyazaki-ken, 887-0013, Japan
| | - Manabu Kurosawa
- Department of Pathology, Nagahama City Hospital, 313 Ooinui-cho, Nagahama-shi, Shiga-ken, 526-8580, Japan
| | - Michiko Yoshimura
- Department of Pathology, Belland Hospital, 500-3 Higashiyama, Naka-ku, Sakai-shi, Osaka, 599-8247, Japan
| | - Masutaka Furue
- Department of Dermatology, Graduate School of Medical Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka-ken, 812-8582, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka-ken, 812-8582, Japan.
| |
Collapse
|
122
|
Guo N, Zhang N, Yan L, Cao X, Wang J, Wang Y. Correlation between genetic polymorphisms within the MAPK1/HIF-1/HO-1 signaling pathway and risk or prognosis of perimenopausal coronary artery disease. Clin Cardiol 2017; 40:597-604. [PMID: 28444966 DOI: 10.1002/clc.22708] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 02/27/2017] [Accepted: 02/28/2017] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Mitogen-activated protein kinase-1 (MAPK1), as well as its downstream factors of hypoxia-inducible factor-1 (HIF-1) and heme oxygenase-1 (HO-1), have been documented to be involved in modulating development of coronary artery disease (CAD). HYPOTHESIS Genetic mutations within the MAPK1/HIF-1/HO-1 signaling pathway could alter the risk of perimenopausal CAD in Chinese patients. METHODS Peripheral blood samples were gathered from 589 CAD patients and 860 healthy controls, and 12 potential single-nucleotide polymorphisms (SNPs) were obtained from HapMap database and previously published studies. Genotyping of SNPs was implemented with the TaqMan SNP Genotyping Assays. Odds ratios (OR) and 95% confidence intervals (CI) were utilized to evaluate the correlations between SNPs and CAD risk. RESULTS Regarding MAPK1 , rs6928 (OR: 1.71, 95% CI: 1.47-1.98, P < 0.05), rs9340 (OR: 0.85, 95% CI: 0.73-0.99, P < 0.05), and rs11913721 (OR: 0.70, 95% CI: 0.52-0.95, P < 0.05) were remarkably associated with susceptibility to perimenopausal CAD. Of these, rs9340 and rs11913721 were also regarded as protective factors for perimenopausal CAD patients. Moreover, results of HIF-1 indicated noticeable correlations between combined SNPs of rs1087314 and rs2057482 and risk of perimenopausal CAD (OR: 1.24, 95% CI: 1.01-1.53, P < 0.05; and OR: 0.71, 95% CI: 0.55-0.91, P < 0.05, respectively). Nonetheless, rs2071746 in HO-1 was found to be only associated with perimenopausal CAD risk (OR: 0.67, 95% CI: 0.58-0.78, P < 0.05). CONCLUSIONS The genetic mutations within MAPK1 (rs6928, rs9340, rs11913721), HIF-1 (rs1087314, rs2057482), and HO-1 (rs2071746) could alter susceptibility to perimenopausal CAD in this Chinese population.
Collapse
Affiliation(s)
- Nan Guo
- Department of Cardiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, P. R. China
| | - Nan Zhang
- Department of Cardiothoracic Surgery, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, P. R. China
| | - Liqiu Yan
- Department of Cardiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, P. R. China
| | - Xufen Cao
- Department of Cardiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, P. R. China
| | - Jiawang Wang
- Department of Cardiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, P. R. China
| | - Yunfei Wang
- Department of Cardiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, P. R. China
| |
Collapse
|
123
|
E2/ER β Enhances Calcineurin Protein Degradation and PI3K/Akt/MDM2 Signal Transduction to Inhibit ISO-Induced Myocardial Cell Apoptosis. Int J Mol Sci 2017; 18:ijms18040892. [PMID: 28441761 PMCID: PMC5412471 DOI: 10.3390/ijms18040892] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 03/28/2017] [Accepted: 04/11/2017] [Indexed: 12/31/2022] Open
Abstract
Secretion of multifunctional estrogen and its receptor has been widely considered as the reason for markedly higher frequency of heart disease in men than in women. 17β-Estradiol (E2), for instance, has been reported to prevent development of cardiac apoptosis via activation of estrogen receptors (ERs). In addition, protein phosphatase such as protein phosphatase 1 (PP1) and calcineurin (PP2B) are also involved in cardiac hypertrophy and cell apoptosis signaling. However, the mechanism by which E2/ERβ suppresses apoptosis is not fully understood, and the role of protein phosphatase in E2/ERβ action also needs further investigation. In this study, we observed that E2/ERβ inhibited isoproterenol (ISO)-induced myocardial cell apoptosis, cytochrome c release and downstream apoptotic markers. Moreover, we found that E2/ERβ blocks ISO-induced apoptosis in H9c2 cells through the enhancement of calcineurin protein degradation through PI3K/Akt/MDM2 signaling pathway. Our results suggest that supplementation with estrogen and/or overexpression of estrogen receptor β gene may prove to be effective means to treat stress-induced myocardial damage.
Collapse
|
124
|
Yang L, Gregorich ZR, Cai W, Zhang P, Young B, Gu Y, Zhang J, Ge Y. Quantitative Proteomics and Immunohistochemistry Reveal Insights into Cellular and Molecular Processes in the Infarct Border Zone One Month after Myocardial Infarction. J Proteome Res 2017; 16:2101-2112. [PMID: 28347137 DOI: 10.1021/acs.jproteome.7b00107] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Postinfarction remodeling and expansion of the peri-infarct border zone (BZ) directly correlate with mortality following myocardial infarction (MI); however, the cellular and molecular mechanisms underlying remodeling processes in the BZ remain unclear. Herein, we utilized a label-free quantitative proteomics approach in combination with immunohistochemical analyses to gain a better understanding of processes contributing to postinfarction remodeling of the peri-infarct BZ in a swine model of MI with reperfusion. Our analysis uncovered a significant down-regulation of proteins involved in energy metabolism, indicating impaired myocardial energetics and possibly mitochondrial dysfunction, in the peri-scar BZ. An increase in endothelial and vascular smooth muscles cells, as well as up-regulation of proteins implicated in vascular endothelial growth factor (VEGF) signaling and marked changes in the expression of extracellular matrix and subendothelial basement membrane proteins, is indicative of active angiogenesis in the infarct BZ. A pronounced increase in macrophages in the peri-infarct BZ was also observed, and proteomic analysis uncovered evidence of persistent inflammation in this tissue. Additional evidence suggested an increase in cellular proliferation that, concomitant with increased nestin expression, indicates potential turnover of endogenous stem cells in the BZ. A marked up-regulation of pro-apoptotic proteins, as well as the down-regulation of proteins important for adaptation to mechanical, metabolic, and oxidative stress, likely contributes to increased apoptosis in the peri-infarct BZ. The cellular processes and molecular pathways identified herein may have clinical utility for therapeutic intervention aimed at limiting remodeling and expansion of the BZ myocardium and preventing the development of heart failure post-MI.
Collapse
Affiliation(s)
- Libang Yang
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School , Minneapolis, Minnesota 55455, United States
| | | | | | - Patrick Zhang
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School , Minneapolis, Minnesota 55455, United States
| | - Bernice Young
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School , Minneapolis, Minnesota 55455, United States
| | | | - Jianyi Zhang
- Department of Biomedical Engineering, School of Engineering, School of Medicine, University of Alabama at Birmingham , Birmingham, Alabama 35294, United States
| | | |
Collapse
|
125
|
Strange Bedfellows: Nuclear Factor, Erythroid 2-Like 2 (Nrf2) and Hypoxia-Inducible Factor 1 (HIF-1) in Tumor Hypoxia. Antioxidants (Basel) 2017; 6:antiox6020027. [PMID: 28383481 PMCID: PMC5488007 DOI: 10.3390/antiox6020027] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 03/31/2017] [Accepted: 04/03/2017] [Indexed: 12/14/2022] Open
Abstract
The importance of the tumor microenvironment for cancer progression and therapeutic resistance is an emerging focus of cancer biology. Hypoxia, or low oxygen, is a hallmark of solid tumors that promotes metastasis and represents a significant obstacle to successful cancer therapy. In response to hypoxia, cancer cells activate a transcriptional program that allows them to survive and thrive in this harsh microenvironment. Hypoxia-inducible factor 1 (HIF-1) is considered the main effector of the cellular response to hypoxia, stimulating the transcription of genes involved in promoting angiogenesis and altering cellular metabolism. However, growing evidence suggests that the cellular response to hypoxia is much more complex, involving coordinated signaling through stress response pathways. One key signaling molecule that is activated in response to hypoxia is nuclear factor, erythroid 2 like-2 (Nrf2). Nrf2 is a transcription factor that controls the expression of antioxidant-response genes, allowing the cell to regulate reactive oxygen species. Nrf2 is also activated in various cancer types due to genetic and epigenetic alterations, and is associated with poor survival and resistance to therapy. Emerging evidence suggests that coordinated signaling through Nrf2 and HIF-1 is critical for tumor survival and progression. In this review, we discuss the distinct and overlapping roles of HIF-1 and Nrf2 in the cellular response to hypoxia, with a focus on how targeting Nrf2 could provide novel chemotherapeutic modalities for treating solid tumors.
Collapse
|
126
|
Moreira DM, Lueneberg ME, da Silva RL, Fattah T, Gottschall CAM. MethotrexaTE THerapy in ST-Segment Elevation MYocardial InfarctionS. J Cardiovasc Pharmacol Ther 2017; 22:538-545. [DOI: 10.1177/1074248417699884] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Purpose: Methotrexate is an anti-inflammatory drug that has been shown to have anti-ischemic effects. Our aim was to evaluate if methotrexate could reduce infarct size in patients with ST-segment elevation myocardial infarction (STEMI). Methods: We randomly assigned patients with STEMI to receive either methotrexate or placebo. Primary outcome was infarct size determined by calculating the area under the curve (AUC) for creatine kinase (CK) release. Secondary outcomes were AUC of CK MB (CK-MB) and AUC of troponin I; peak CK, peak CK-MB, and troponin I; B-type natriuretic peptide (BNP) level, high-sensitivity C-reactive protein (hsCRP) result, and erythrocyte sedimentation rate (ESR); left ventricular ejection fraction (LVEF); thrombolysis in myocardial infarction (TIMI) frame count; Killip score; mortality and reinfarction incidence; and incidence of adverse reactions. Results: We included 84 patients. Median AUC of CK was 78 861.0 in the methotrexate group and 68 088.0 in the placebo group ( P = .10). Patients given methotrexate and placebo exhibited, respectively, median AUC for CK-MB of 9803.4 and 8037.0 ( P = .42); median AUC for troponin of 3691.1 and 2132.6 ( P = .09); peak CK of 2806.0 and 2147.0 ( P = .05); peak CK-MB of 516.0 and 462.3 ( P = .25); and peak troponin of 121.0 and 85.1 ( P = .06). At 3 months, LVEF was lower in patients who received methotrexate (49.0% ± 14.1%) than in patients given placebo (56.4% ± 10.0%; P = .01). There were no differences in hsCRP, ESR, BNP, Killip scores, TIMI frame count, reinfarction, and mortality rates. There was a higher median serum glutamic–pyruvic transaminase levels in the methotrexate group. Conclusion: Methotrexate did not reduce infarction size and worsened LVEF at 3 months ( Clinicaltrials.gov identifier NCT01741558).
Collapse
Affiliation(s)
- Daniel Medeiros Moreira
- Instituto de Cardiologia de Santa Catarina, São José, Santa Catarina, Brazil
- Instituto de Cardiologia do Rio Grande do Sul—FUC, Porto Alegre, Rio Grande do Sul, Brazil
| | | | | | - Tammuz Fattah
- Instituto de Cardiologia de Santa Catarina, São José, Santa Catarina, Brazil
| | | |
Collapse
|
127
|
Abstract
The hearts of lower vertebrates such as fish and salamanders display scarless regeneration following injury, although this feature is lost in adult mammals. The remarkable capacity of the neonatal mammalian heart to regenerate suggests that the underlying machinery required for the regenerative process is evolutionarily retained. Recent studies highlight the epicardial covering of the heart as an important source of the signalling factors required for the repair process. The developing epicardium is also a major source of cardiac fibroblasts, smooth muscle, endothelial cells and stem cells. Here, we examine animal models that are capable of scarless regeneration, the role of the epicardium as a source of cells, signalling mechanisms implicated in the regenerative process and how these mechanisms influence cardiomyocyte proliferation. We also discuss recent advances in cardiac stem cell research and potential therapeutic targets arising from these studies.
Collapse
Affiliation(s)
| | - Nadia Rosenthal
- National Heart and Lung Institute, Imperial College London, London, UK Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia The Jackson Laboratory, Bar Harbor, ME, USA
| |
Collapse
|
128
|
Wolff M, Kosyna FK, Dunst J, Jelkmann W, Depping R. Impact of hypoxia inducible factors on estrogen receptor expression in breast cancer cells. Arch Biochem Biophys 2016; 613:23-30. [PMID: 27823907 DOI: 10.1016/j.abb.2016.11.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 10/31/2016] [Accepted: 11/03/2016] [Indexed: 12/29/2022]
Abstract
In women breast cancer is still the most commonly diagnosed cancer. This type of cancer is classified as a hormone-dependent tumor. Estrogen receptor (ER) expression and functional status contribute to breast cancer development and progression. Another important factor associated with cancer is hypoxia which is defined as the state of reduced oxygen availability in tissues. Intratumoral hypoxia results in the activation of the hypoxia inducible factors (HIFs). HIFs are heterodimeric transcription factors involved in the regulation of many cellular processes, such as angiogenesis, anaerobic metabolism, cell proliferation/survival, and promotion of metastasis. In this study we evaluated the interplay between hypoxia, HIF stabilization and the ER-α/β-ratio in several ER-positive breast cancer cell lines. Hypoxia was shown to inhibit ER expression in ER-positive breast cancer cells. Further experiments using the hypoxia mimetic CoCl2 and HIF-1α knockdown cells indicated that the influence of hypoxia on breast cancer cells involves other pathways than the molecular oxygen sensing pathway. Moreover, we demonstrated that MCF-7 cells in long-term culture lost part of their ability to respond to hypoxic incubation. Understanding the relationships between HIF, ER-α and ER-β expression holds the promise of the development of new therapeutic agents and may provide future advances in prognosis.
Collapse
Affiliation(s)
- Matthias Wolff
- Institute of Physiology, Center for Structural and Cell Biology in Medicine, University of Lübeck, Germany; Klinik für Strahlentherapie, Universitätsklinikum Schleswig-Holstein, Campus Lübeck, Germany
| | | | - Jürgen Dunst
- Klinik für Strahlentherapie, Universitätsklinikum Schleswig-Holstein, Campus Lübeck, Germany
| | - Wolfgang Jelkmann
- Institute of Physiology, Center for Structural and Cell Biology in Medicine, University of Lübeck, Germany
| | - Reinhard Depping
- Institute of Physiology, Center for Structural and Cell Biology in Medicine, University of Lübeck, Germany.
| |
Collapse
|
129
|
SIRT3 in cardiovascular diseases: Emerging roles and therapeutic implications. Int J Cardiol 2016; 220:700-5. [DOI: 10.1016/j.ijcard.2016.06.236] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 06/26/2016] [Indexed: 12/17/2022]
|
130
|
Abdel Hamid M, Bakhoum SWG, Sharaf Y, Sabry D, El-Gengehe AT, Abdel-Latif A. Circulating Endothelial Cells and Endothelial Function Predict Major Adverse Cardiac Events and Early Adverse Left Ventricular Remodeling in Patients With ST-Segment Elevation Myocardial Infarction. J Interv Cardiol 2016; 29:89-98. [PMID: 26864952 DOI: 10.1111/joic.12269] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Endothelial progenitor cells (EPCs) and circulating endothelial cells (CECs) are mobilized from the bone marrow and increase in the early phase after ST-elevation myocardial infarction (STEMI). The aim of this study was to assess the prognostic significance of CECs and indices of endothelial dysfunction in patients with STEMI. In 78 patients with acute STEMI, characterization of CD34+/VEGFR2+CECs, and indices of endothelial damage/dysfunction such as brachial artery flow mediated dilatation (FMD) were determined. Blood samples for CECs assessment and quantification were obtained within 24 hours of admission and FMD was assessed during the index hospitalization. At 30 days follow up, the primary composite end point of major adverse cardiac events (MACE) consisting of all-cause mortality, recurrent nonfatal MI, or heart failure and the secondary endpoint of early adverse left ventricular (LV) remodeling were analyzed. The 17 patients (22%) who developed MACE had significantly higher CEC level (P = 0.004), von Willebrand factor (vWF) level (P = 0.028), and significantly lower FMD (P = 0.006) compared to the remaining patients. Logistic regression analysis showed that CECs level and LV ejection fraction were independent predictors of MACE. The areas under the receiver operating characteristic curves (ROC) for CEC level, FMD, and the logistic model with both markers were 0.73, 0.75, and 0.82, respectively, for prediction of the MACE. The 16 patients who developed the secondary endpoint had significantly higher CEC level compared to remaining patients (P = 0.038). In conclusion, increased circulating endothelial cells and endothelial dysfunction predicted the occurrence of major adverse cardiac events and adverse cardiac remodeling in patients with STEMI.
Collapse
Affiliation(s)
| | | | - Yasser Sharaf
- Department of Cardiology, University of Cairo, Cairo, Egypt
| | - Dina Sabry
- Department of Molecular Biology and Biochemistry, University of Cairo, Cairo, Egypt
| | - Ahmed T El-Gengehe
- Department of Molecular Biology and Biochemistry, University of Cairo, Cairo, Egypt
| | - Ahmed Abdel-Latif
- Division of Cardiology, Department of Medicine, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
131
|
Klenke S, Renckhoff K, Engler A, Peters J, Frey UH. Easy-to-use strategy for reference gene selection in quantitative real-time PCR experiments. Naunyn Schmiedebergs Arch Pharmacol 2016; 389:1353-1366. [PMID: 27650728 DOI: 10.1007/s00210-016-1305-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 09/09/2016] [Indexed: 12/19/2022]
Abstract
Real-time PCR is an indispensable technique for mRNA expression analysis but conclusions depend on appropriate reference gene selection. However, while reference gene selection has been a topic of publications, this issue is often disregarded when measuring target mRNA expression. Therefore, we (1) evaluated the frequency of appropriate reference gene selection, (2) suggest an easy-to-use tool for least variability reference gene selection, (3) demonstrate application of this tool, and (4) show effects on target gene expression profiles. All 2015 published articles in Naunyn-Schmiedeberg's Archives of Pharmacology were screened for the use of quantitative real-time PCR analysis and selection of reference genes. Target gene expression (Vegfa, Grk2, Sirt4, and Timp3) in H9c2 cells was analyzed following various interventions (hypoxia, hyperglycemia, and/or isoflurane exposure with and without subsequent hypoxia) in relation to putative reference genes (Actb, Gapdh, B2m, Sdha, and Rplp1) using the least variability method vs. an arbitrarily selected but established reference gene. In the vast majority (18 of 21) of papers, no information was provided regarding selection of an appropriate reference gene. In only 1 of 21 papers, a method of appropriate reference gene selection was described and in 2 papers reference gene selection remains unclear. The method of reference gene selection had major impact on interpretation of target gene expression. With hypoxia, for instance, the least variability gene was Rplp1 and target gene expression (Vefga) heavily showed a 2-fold up-regulation (p = 0.022) but no change (p = 0.3) when arbitrarily using Gapdh. Frequency of appropriate reference gene selection in this journal is low, and we propose our strategy for reference gene selection as an easy tool for proper target gene expression.
Collapse
Affiliation(s)
- Stefanie Klenke
- Klinik für Anästhesiologie & Intensivmedizin, Universität Duisburg-Essen and Universitätsklinikum Essen, Hufelandstr. 55, D-45122, Essen, Germany.
| | - Kristina Renckhoff
- Klinik für Anästhesiologie & Intensivmedizin, Universität Duisburg-Essen and Universitätsklinikum Essen, Hufelandstr. 55, D-45122, Essen, Germany
| | - Andrea Engler
- Klinik für Anästhesiologie & Intensivmedizin, Universität Duisburg-Essen and Universitätsklinikum Essen, Hufelandstr. 55, D-45122, Essen, Germany
| | - Jürgen Peters
- Klinik für Anästhesiologie & Intensivmedizin, Universität Duisburg-Essen and Universitätsklinikum Essen, Hufelandstr. 55, D-45122, Essen, Germany
| | - Ulrich H Frey
- Klinik für Anästhesiologie & Intensivmedizin, Universität Duisburg-Essen and Universitätsklinikum Essen, Hufelandstr. 55, D-45122, Essen, Germany
| |
Collapse
|
132
|
Rebouças JDS, Santos-Magalhães NS, Formiga FR. Cardiac Regeneration using Growth Factors: Advances and Challenges. Arq Bras Cardiol 2016; 107:271-275. [PMID: 27355588 PMCID: PMC5053196 DOI: 10.5935/abc.20160097] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 03/18/2016] [Accepted: 03/23/2016] [Indexed: 12/15/2022] Open
Abstract
Myocardial infarction is the most significant manifestation of ischemic heart disease and is associated with high morbidity and mortality. Novel strategies targeting at regenerating the injured myocardium have been investigated, including gene therapy, cell therapy, and the use of growth factors. Growth factor therapy has aroused interest in cardiovascular medicine because of the regeneration mechanisms induced by these biomolecules, including angiogenesis, extracellular matrix remodeling, cardiomyocyte proliferation, stem-cell recruitment, and others. Together, these mechanisms promote myocardial repair and improvement of the cardiac function. This review aims to address the strategic role of growth factor therapy in cardiac regeneration, considering its innovative and multifactorial character in myocardial repair after ischemic injury. Different issues will be discussed, with emphasis on the regeneration mechanisms as a potential therapeutic resource mediated by growth factors, and the challenges to make these proteins therapeutically viable in the field of cardiology and regenerative medicine. Resumo O infarto do miocárdio representa a manifestação mais significativa da cardiopatia isquêmica e está associado a elevada morbimortalidade. Novas estratégias vêm sendo investigadas com o intuito de regenerar o miocárdio lesionado, incluindo a terapia gênica, a terapia celular e a utilização de fatores de crescimento. A terapia com fatores de crescimento despertou interesse em medicina cardiovascular, devido aos mecanismos de regeneração induzidos por essas biomoléculas, incluindo angiogênese, remodelamento da matriz extracelular, proliferação de cardiomiócitos e recrutamento de células-tronco, dentre outros. Em conjunto, tais mecanismos promovem a reparação do miocárdio e a melhora da função cardíaca. Esta revisão pretende abordar o papel estratégico da terapia, com fatores de crescimento, para a regeneração cardíaca, considerando seu caráter inovador e multifatorial sobre o reparo do miocárdio após dano isquêmico. Diferentes questões serão discutidas, destacando-se os mecanismos de regeneração como recurso terapêutico potencial mediado por fatores de crescimento e os desafios para tornar essas proteínas terapeuticamente viáveis no âmbito da cardiologia e da medicina regenerativa.
Collapse
Affiliation(s)
- Juliana de Souza Rebouças
- Laboratório de Imunopatologia Keizo-Asami - Universidade
Federal de Pernambuco (UFPE), Recife, PE - Brazil
| | | | - Fabio Rocha Formiga
- Programa de Pós-Graduação em Biologia Celular e
Molecular Aplicada - Universidade de Pernambuco (UPE), Recife, PE - Brazil
- Curso de Pós-Graduação em Patologia
(UFBA/FIOCRUZ) - Centro de Pesquisas Gonçalo Moniz, Fundação
Oswaldo Cruz (FIOCRUZ), Salvador, BA - Brazil
| |
Collapse
|
133
|
Wassenaar JW, Gaetani R, Garcia JJ, Braden RL, Luo CG, Huang D, DeMaria AN, Omens JH, Christman KL. Evidence for Mechanisms Underlying the Functional Benefits of a Myocardial Matrix Hydrogel for Post-MI Treatment. J Am Coll Cardiol 2016; 67:1074-1086. [PMID: 26940929 DOI: 10.1016/j.jacc.2015.12.035] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 12/14/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND There is increasing need for better therapies to prevent the development of heart failure after myocardial infarction (MI). An injectable hydrogel derived from decellularized porcine ventricular myocardium has been shown to halt the post-infarction progression of negative left ventricular remodeling and decline in cardiac function in both small and large animal models. OBJECTIVES This study sought to elucidate the tissue-level mechanisms underlying the therapeutic benefits of myocardial matrix injection. METHODS Myocardial matrix or saline was injected into infarcted myocardium 1 week after ischemia-reperfusion in Sprague-Dawley rats. Cardiac function was evaluated by magnetic resonance imaging and hemodynamic measurements at 5 weeks after injection. Whole transcriptome microarrays were performed on RNA isolated from the infarct at 3 days and 1 week after injection. Quantitative polymerase chain reaction and histologic quantification confirmed expression of key genes and their activation in altered pathways. RESULTS Principal component analysis of the transcriptomes showed that samples collected from myocardial matrix-injected infarcts are distinct and cluster separately from saline-injected control subjects. Pathway analysis indicated that these differences are due to changes in several tissue processes that may contribute to improved cardiac healing after MI. Matrix-injected infarcted myocardium exhibits an altered inflammatory response, reduced cardiomyocyte apoptosis, enhanced infarct neovascularization, diminished cardiac hypertrophy and fibrosis, altered metabolic enzyme expression, increased cardiac transcription factor expression, and progenitor cell recruitment, along with improvements in global cardiac function and hemodynamics. CONCLUSIONS These results indicate that the myocardial matrix alters several key pathways after MI creating a pro-regenerative environment, further demonstrating its promise as a potential post-MI therapy.
Collapse
Affiliation(s)
- Jean W Wassenaar
- Department of Bioengineering, University of California, San Diego; Sanford Consortium for Regenerative Medicine
| | - Roberto Gaetani
- Department of Bioengineering, University of California, San Diego; Sanford Consortium for Regenerative Medicine
| | - Julian J Garcia
- Department of Bioengineering, University of California, San Diego; Sanford Consortium for Regenerative Medicine
| | - Rebecca L Braden
- Department of Bioengineering, University of California, San Diego; Sanford Consortium for Regenerative Medicine
| | - Colin G Luo
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Diane Huang
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Anthony N DeMaria
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Jeffrey H Omens
- Department of Bioengineering, University of California, San Diego; Department of Medicine, University of California, San Diego, La Jolla, California
| | - Karen L Christman
- Department of Bioengineering, University of California, San Diego; Sanford Consortium for Regenerative Medicine.
| |
Collapse
|
134
|
Cole MA, Abd Jamil AH, Heather LC, Murray AJ, Sutton ER, Slingo M, Sebag-Montefiore L, Tan SC, Aksentijević D, Gildea OS, Stuckey DJ, Yeoh KK, Carr CA, Evans RD, Aasum E, Schofield CJ, Ratcliffe PJ, Neubauer S, Robbins PA, Clarke K. On the pivotal role of PPARα in adaptation of the heart to hypoxia and why fat in the diet increases hypoxic injury. FASEB J 2016; 30:2684-2697. [PMID: 27103577 PMCID: PMC5072355 DOI: 10.1096/fj.201500094r] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 04/05/2016] [Indexed: 12/21/2022]
Abstract
The role of peroxisome proliferator-activated receptor α (PPARα)-mediated metabolic remodeling in cardiac adaptation to hypoxia has yet to be defined. Here, mice were housed in hypoxia for 3 wk before in vivo contractile function was measured using cine MRI. In isolated, perfused hearts, energetics were measured using (31)P magnetic resonance spectroscopy (MRS), and glycolysis and fatty acid oxidation were measured using [(3)H] labeling. Compared with a normoxic, chow-fed control mouse heart, hypoxia decreased PPARα expression, fatty acid oxidation, and mitochondrial uncoupling protein 3 (UCP3) levels, while increasing glycolysis, all of which served to maintain normal ATP concentrations ([ATP]) and thereby, ejection fractions. A high-fat diet increased cardiac PPARα expression, fatty acid oxidation, and UCP3 levels with decreased glycolysis. Hypoxia was unable to alter the high PPARα expression or reverse the metabolic changes caused by the high-fat diet, with the result that [ATP] and contractile function decreased significantly. The adaptive metabolic changes caused by hypoxia in control mouse hearts were found to have occurred already in PPARα-deficient (PPARα(-/-)) mouse hearts and sustained function in hypoxia despite an inability for further metabolic remodeling. We conclude that decreased cardiac PPARα expression is essential for adaptive metabolic remodeling in hypoxia, but is prevented by dietary fat.-Cole, M. A., Abd Jamil, A. H., Heather, L. C., Murray, A. J., Sutton, E. R., Slingo, M., Sebag-Montefiore, L., Tan, S. C., Aksentijević, D., Gildea, O. S., Stuckey, D. J., Yeoh, K. K., Carr, C. A., Evans, R. D., Aasum, E., Schofield, C. J., Ratcliffe, P. J., Neubauer, S., Robbins, P. A., Clarke, K. On the pivotal role of PPARα in adaptation of the heart to hypoxia and why fat in the diet increases hypoxic injury.
Collapse
Affiliation(s)
- Mark A Cole
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Amira H Abd Jamil
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Lisa C Heather
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Andrew J Murray
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Elizabeth R Sutton
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Mary Slingo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Liam Sebag-Montefiore
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Suat Cheng Tan
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Dunja Aksentijević
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Ottilie S Gildea
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Daniel J Stuckey
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Kar Kheng Yeoh
- Chemistry Research Laboratory, University of Oxford, Oxford, United Kingdom; and
| | - Carolyn A Carr
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Rhys D Evans
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Ellen Aasum
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | | | - Peter J Ratcliffe
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Peter A Robbins
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Kieran Clarke
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom;
| |
Collapse
|
135
|
Feng CC, Lin CC, Lai YP, Chen TS, Marthandam Asokan S, Lin JY, Lin KH, Viswanadha VP, Kuo WW, Huang CY. Hypoxia suppresses myocardial survival pathway through HIF-1α-IGFBP-3-dependent signaling and enhances cardiomyocyte autophagic and apoptotic effects mainly via FoxO3a-induced BNIP3 expression. Growth Factors 2016; 34:73-86. [PMID: 27366871 DOI: 10.1080/08977194.2016.1191480] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The HIF-1α transcriptional factor and the BH-3 only protein BNIP3 are known to play fundamental roles in response to hypoxia. The objective of this research is to investigate the molecular mechanisms and the correlation of HIF-1α, BNIP3 and IGFBP-3 in hypoxia-induced cardiomyocytes injuries. Heart-derived H9c2 cells and neonatal rat ventricular myocytes (NRVMs) were incubated in normoxic or hypoxic conditions. Hypoxia increased HIF-1α expression and activated the downstream BNIP3 and IGFBP-3 thereby triggered mitochondria-dependent apoptosis. Moreover, IGF1R/PI3K/Akt signaling was attenuated by HIF-1α-dependent IGFBP-3 expression to enhance hypoxia-induced apoptosis. Autophagy suppression with 3-methyladenine or siATG5 or siBeclin-1 significantly decreased myocardial apoptosis under hypoxia. Knockdown of FoxO3a or BNIP3 significantly abrogated hypoxia-induced autophagy and mitochondria-dependent apoptosis. Moreover, prolonged-hypoxia induced HIF-1α stimulated BNIP3 and enhanced IGFBP-3 activation to inhibit IGF1R/PI3K/Akt survival pathway and mediate mitochondria-dependent cardiomyocyte apoptosis. HIF-1α and FoxO3a blockage are sufficient to annul the change of excessive hypoxia of hearts.
Collapse
Affiliation(s)
- Chih-Chung Feng
- a Graduate Institute of Clinical Medical Science, Graduate Institute of Basic Medical Science, China Medical University , Taichung , Taiwan
| | - Chien-Chung Lin
- b Orthopaedic Department, Armed Forces General Hospital , Taichung , Taiwan
| | - Yi-Ping Lai
- c Graduate Institute of Basic Medical Science, China Medical University , Taichung , Taiwan
| | - Tung-Sheng Chen
- c Graduate Institute of Basic Medical Science, China Medical University , Taichung , Taiwan
- d Biomaterials Translational Research Center, China Medical University Hospital , Taichung , Taiwan
| | | | - Jing-Ying Lin
- e Department of Nursing , Central Taiwan University of Science and Technology , Taichung , Taiwan
| | - Kuan-Ho Lin
- f Emergency Department, China Medical University Hospital , Taichung , Taiwan
| | | | - Wei-Wen Kuo
- h Department of Biological Science and Technology , China Medical University , Taichung , Taiwan
| | - Chih-Yang Huang
- c Graduate Institute of Basic Medical Science, China Medical University , Taichung , Taiwan
- i Graduate Institute of Chinese Medical Science, China Medical University , Taichung , Taiwan , and
- j Department of Health and Nutrition Biotechnology , Asia University , Taichung , Taiwan
| |
Collapse
|
136
|
Yu B, Meng F, Yang Y, Liu D, Shi K. NOX2 Antisense Attenuates Hypoxia-Induced Oxidative Stress and Apoptosis in Cardiomyocyte. Int J Med Sci 2016; 13:646-652. [PMID: 27499697 PMCID: PMC4974913 DOI: 10.7150/ijms.15177] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 06/25/2016] [Indexed: 01/12/2023] Open
Abstract
Heart ischemia is a hypoxia related disease. NOX2 and HIF-1α proteins were increased in cardiomyocytes after acute myocardial infarction. However, the relationship of the hypoxia-induced HIF-1α. NOX2-derived oxidative stress and apoptosis in cardiomyocyte remains unclear. In the current study, we use NOX2 antisense strategy to investigate the role of NOX2 in hypoxia-induced oxidative stress and apoptosis in rat cardiomyocytes. Here, we show that transduction of ADV-NOX2-AS induces potent silencing of NOX2 in cardiomyocytes, and resulting in attenuation of hypoxia-induced oxidative stress and apoptosis. This study indicates the potential of antisense-based therapies and validates NOX2 as a potent therapeutic candidate for heart ischemia.
Collapse
Affiliation(s)
| | | | | | | | - Kaiyao Shi
- Department of cardiology, China-Japan union hospital of Jilin University, Changchun, Jilin, 130033, P.R. China
| |
Collapse
|
137
|
Hasan A, Waters R, Roula B, Dana R, Yara S, Alexandre T, Paul A. Engineered Biomaterials to Enhance Stem Cell-Based Cardiac Tissue Engineering and Therapy. Macromol Biosci 2016; 16:958-77. [PMID: 26953627 PMCID: PMC4931991 DOI: 10.1002/mabi.201500396] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 01/18/2016] [Indexed: 12/17/2022]
Abstract
Cardiovascular disease is a leading cause of death worldwide. Since adult cardiac cells are limited in their proliferation, cardiac tissue with dead or damaged cardiac cells downstream of the occluded vessel does not regenerate after myocardial infarction. The cardiac tissue is then replaced with nonfunctional fibrotic scar tissue rather than new cardiac cells, which leaves the heart weak. The limited proliferation ability of host cardiac cells has motivated investigators to research the potential cardiac regenerative ability of stem cells. Considerable progress has been made in this endeavor. However, the optimum type of stem cells along with the most suitable matrix-material and cellular microenvironmental cues are yet to be identified or agreed upon. This review presents an overview of various types of biofunctional materials and biomaterial matrices, which in combination with stem cells, have shown promises for cardiac tissue replacement and reinforcement. Engineered biomaterials also have applications in cardiac tissue engineering, in which tissue constructs are developed in vitro by combining stem cells and biomaterial scaffolds for drug screening or eventual implantation. This review highlights the benefits of using biomaterials in conjunction with stem cells to repair damaged myocardium and give a brief description of the properties of these biomaterials that make them such valuable tools to the field.
Collapse
Affiliation(s)
- Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, Qatar
- Biomedical Engineering and Department of Mechanical Engineering, Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Renae Waters
- BioIntel Research Laboratory, Department of Chemical and Petroleum Engineering, Bioengineering Graduate Program, School of Engineering, University of Kansas, Lawrence, KS, USA
| | - Boustany Roula
- Biomedical Engineering and Department of Mechanical Engineering, Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Rahbani Dana
- Biomedical Engineering and Department of Mechanical Engineering, Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Seif Yara
- Biomedical Engineering and Department of Mechanical Engineering, Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Toubia Alexandre
- Biomedical Engineering and Department of Mechanical Engineering, Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Arghya Paul
- BioIntel Research Laboratory, Department of Chemical and Petroleum Engineering, Bioengineering Graduate Program, School of Engineering, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
138
|
Díaz-Trelles R, Scimia MC, Bushway P, Tran D, Monosov A, Monosov E, Peterson K, Rentschler S, Cabrales P, Ruiz-Lozano P, Mercola M. Notch-independent RBPJ controls angiogenesis in the adult heart. Nat Commun 2016; 7:12088. [PMID: 27357444 PMCID: PMC4931341 DOI: 10.1038/ncomms12088] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 05/26/2016] [Indexed: 12/05/2022] Open
Abstract
Increasing angiogenesis has long been considered a therapeutic target for improving heart function after injury such as acute myocardial infarction. However, gene, protein and cell therapies to increase microvascularization have not been successful, most likely because the studies failed to achieve regulated and concerted expression of pro-angiogenic and angiostatic factors needed to produce functional microvasculature. Here, we report that the transcription factor RBPJ is a homoeostatic repressor of multiple pro-angiogenic and angiostatic factor genes in cardiomyocytes. RBPJ controls angiogenic factor gene expression independently of Notch by antagonizing the activity of hypoxia-inducible factors (HIFs). In contrast to previous strategies, the cardiomyocyte-specific deletion of Rbpj increased microvascularization of the heart without adversely affecting cardiac structure or function even into old age. Furthermore, the loss of RBPJ in cardiomyocytes increased hypoxia tolerance, improved heart function and decreased pathological remodelling after myocardial infarction, suggesting that inhibiting RBPJ might be therapeutic for ischaemic injury.
Collapse
Affiliation(s)
- Ramón Díaz-Trelles
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, USA
- Department of Bioengineering, Jacobs School of Engineering, University of California, San Diego, La Jolla, California 92093 USA
| | - Maria Cecilia Scimia
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, USA
- Department of Bioengineering, Jacobs School of Engineering, University of California, San Diego, La Jolla, California 92093 USA
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, California 92093 USA
| | - Paul Bushway
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, USA
- Department of Bioengineering, Jacobs School of Engineering, University of California, San Diego, La Jolla, California 92093 USA
| | - Danh Tran
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, USA
| | - Anna Monosov
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, USA
| | - Edward Monosov
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, USA
| | - Kirk Peterson
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, California 92093 USA
| | - Stacey Rentschler
- Departments of Medicine, Developmental Biology and Biomedical Engineering, Washington University, St Louis, Missouri 63110 USA
| | - Pedro Cabrales
- Department of Bioengineering, Jacobs School of Engineering, University of California, San Diego, La Jolla, California 92093 USA
| | - Pilar Ruiz-Lozano
- Department of Pediatrics, Stanford University, Stanford, California 94305 USA
| | - Mark Mercola
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, USA
- Department of Bioengineering, Jacobs School of Engineering, University of California, San Diego, La Jolla, California 92093 USA
- Stanford Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, California 94305, USA
| |
Collapse
|
139
|
Amoah V, Wrigley B, Holroyd E, Smallwood A, Armesilla AL, Nevill A, Cotton J. Vascular endothelial growth factor and hypoxia-inducible factor-1α gene polymorphisms and coronary collateral formation in patients with coronary chronic total occlusions. SAGE Open Med 2016; 4:2050312116654403. [PMID: 27621802 PMCID: PMC5006805 DOI: 10.1177/2050312116654403] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 05/10/2016] [Indexed: 11/30/2022] Open
Abstract
Introduction: We evaluated the association between two single nucleotide polymorphisms of the vascular endothelial growth factor gene and one of the hypoxia-inducible factor-1α gene and the degree of coronary collateral formation in patients with a coronary chronic total occlusion. Methods: Totally, 98 patients with symptomatic coronary artery disease and a chronic total occlusion observed during coronary angiography were recruited. Genotyping of two vascular endothelial growth factor promoter single nucleotide polymorphisms (−152G>A and −165C>T) and the C1772T single nucleotide polymorphism of hypoxia-inducible factor-1α were performed using polymerase chain reaction and restriction fragment length polymorphism analysis. The presence and extent of collateral vessel filling was scored by blinded observers using the Rentrop grade. Results: We found no association between the vascular endothelial growth factor −152G>A, −165C>T and hypoxia-inducible factor-1α −1772C>T with the presence and filling of coronary collateral vessels. A history of percutaneous coronary intervention and transient ischaemic attack/cerebrovascular accident were associated with the presence of enhanced collateral vessel formation following binary logistic regression analysis. Conclusion: The study findings suggest that coronary collateral formation is not associated with the tested polymorphic variants of vascular endothelial growth factor and hypoxia-inducible factor-1α in patients with symptomatic coronary artery disease and the presence of a chronic total occlusion.
Collapse
Affiliation(s)
- Vincent Amoah
- Department of Cardiology, Heart and Lung Centre, New Cross Hospital, Wolverhampton, UK
| | - Benjamin Wrigley
- Department of Cardiology, Heart and Lung Centre, New Cross Hospital, Wolverhampton, UK
| | - Eric Holroyd
- Department of Cardiology, Heart and Lung Centre, New Cross Hospital, Wolverhampton, UK
| | - Andrew Smallwood
- Department of Cardiology, Heart and Lung Centre, New Cross Hospital, Wolverhampton, UK
| | - Angel L Armesilla
- Research Institute in Healthcare Science, School of Pharmacy, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, UK
| | - Alan Nevill
- Faculty of Education, Health and Wellbeing, University of Wolverhampton, Wolverhampton, UK
| | - James Cotton
- Department of Cardiology, Heart and Lung Centre, New Cross Hospital, Wolverhampton, UK
- James Cotton, Department of Cardiology, Heart and Lung Centre, New Cross Hospital, Wolverhampton, West Midlands WV10 0QP, UK.
| |
Collapse
|
140
|
Yeh YL, Hu WS, Ting WJ, Shen CY, Hsu HH, Chung LC, Tu CC, Chang SH, Day CH, Tsai Y, Huang CY. Hypoxia Augments Increased HIF-1α and Reduced Survival Protein p-Akt in Gelsolin (GSN)-Dependent Cardiomyoblast Cell Apoptosis. Cell Biochem Biophys 2016; 74:221-8. [PMID: 27193608 DOI: 10.1007/s12013-016-0729-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 04/09/2016] [Indexed: 12/25/2022]
Abstract
Cytoskeleton filaments play an important role in cellular functions such as maintaining cell shape, cell motility, intracellular transport, and cell division. Actin-binding proteins (ABPs) have numerous functions including regulation of actin filament nucleation, elongation, severing, capping, cross linking, and actin monomer sequestration. Gelsolin (GSN) is one of the actin-binding proteins. Gelsolin (GSN) is one of the actin-binding proteins that regulate cell morphology, differentiation, movement, and apoptosis. GSN also regulates cell morphology, differentiation, movement, and apoptosis. In this study, we have used H9c2 cardiomyoblast cell and H9c2-GSN stable clones to understand the roles and mechanisms of GSN overexpression in hypoxia-induced cardiomyoblast cell death. The data show that hypoxia or GSN overexpression induces HIF-1α expression and reduces the expression of survival markers p-Akt and Bcl-2 in H9c2 cardiomyoblast cells. Under hypoxic conditions, GSN overexpression further reduces p-Akt expression and elevates total as well as cleaved GSN levels and HIF-1α levels. In addition, GSN overexpression enhances apoptosis in cardiomyoblasts under hypoxia. Hypoxic challenge further induced activated caspase-3 and cell death that was attenuated after GSN knock down, which implies that GSN is a critical therapeutic target against hypoxia-induced cardiomyoblast cell death.
Collapse
Affiliation(s)
- Yu-Lan Yeh
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan
- Department of Medical Technology, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli, Taiwan
| | | | - Wei-Jen Ting
- Graduate Institute of Basic Medical Science, School of Chinese Medicine, China Medical University and Hospital, 91 Hsueh-Shih Road 404, Taichung, Taiwan, ROC
| | - Chia-Yao Shen
- Department of Nursing, MeiHo University, Pingtung, Taiwan
| | - Hsi-Hsien Hsu
- Division of Colorectal Surgery, Mackay Memorial Hospital, Taipei, Taiwan
| | - Li-Chin Chung
- Department of Hospital and Health Care Administration, Chia Nan University of Pharmacy and Science, Tainan County, Taiwan
| | - Chuan-Chou Tu
- Division of Chest Medicine, Department of Internal Medicine, Armed Force Taichung General Hospital, Taichung, Taiwan
| | - Sheng-Huang Chang
- Department of Health, Tsao-Tun Psychiatric Center, Executive Yuan, Nantou, 54249, Taiwan
| | | | - Yuhsin Tsai
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Basic Medical Science, School of Chinese Medicine, China Medical University and Hospital, 91 Hsueh-Shih Road 404, Taichung, Taiwan, ROC.
- School of Chinese Medicine, China Medical University, Taichung, Taiwan.
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan.
| |
Collapse
|
141
|
Gui L, Liu B, Lv G. Hypoxia induces autophagy in cardiomyocytes via a hypoxia-inducible factor 1-dependent mechanism. Exp Ther Med 2016; 11:2233-2239. [PMID: 27284306 PMCID: PMC4887955 DOI: 10.3892/etm.2016.3190] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 08/26/2015] [Indexed: 12/18/2022] Open
Abstract
Hypoxia frequently accompanies such vascular disorders as atherosclerosis, thrombosis and ischemia/reperfusion injury. Myocardial ischemia/reperfusion, in particular, is a major contributor to cardiomyocyte impairment. Autophagy is a dynamic, self-catabolic process that has been implicated in a wide range of physiological processes and the pathogenesis of diverse diseases. The aim of the present study was to investigate the promotion of autophagy by hypoxia in a rat H9c2 heart cell line and determine the regulatory role of hypoxia-inducible factor 1 (HIF-1) in the hypoxia-induced autophagy in H9c2 cells, using quantitative green fluorescent protein-microtubule-associated protein 1 light chain 3 analysis and electron microscopy of autophagic vesicles. In addition, western blot and quantitative polymerase chain reaction analysis of autophagy-associated markers was conducted. In addition, the role of HIF-1-mediated autophagy in the hypoxia-induced impairment of H9c2 cells was examined, as a measure of cellular viability, using an MTT assay. The results demonstrated that autophagy was induced in H9c2 cells under hypoxia, and the autophagy induction triggered by hypoxia could be enhanced by HIF-1α overexpression and inhibited by HIF-1α knockdown. Furthermore, the HIF-1-mediated autophagy ameliorated the reduction in the H9c2 cell viability induced by hypoxia. These findings provide a novel insight into the hypoxic-ischemic injury to cardiomyocytes and give evidence for the occurrence of HIF-1-mediated autophagy in myocardial ischemia.
Collapse
Affiliation(s)
- Lan Gui
- Department of Basic Theory, Sports Institute of Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia 028041, P.R. China; Institute of Mongolian Genome and Genetic Diseases, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia 028041, P.R. China
| | - Batu Liu
- Sports Department, Sports Institute of Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia 028041, P.R. China
| | - Guang Lv
- Institute of Pathogenic Microbiology and Immunology, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia 028041, P.R. China
| |
Collapse
|
142
|
van der Hoeven NW, Hollander MR, Yıldırım C, Jansen MF, Teunissen PF, Horrevoets AJ, van der Pouw Kraan TCTM, van Royen N. The emerging role of galectins in cardiovascular disease. Vascul Pharmacol 2016; 81:31-41. [PMID: 26945624 DOI: 10.1016/j.vph.2016.02.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 12/08/2015] [Accepted: 02/22/2016] [Indexed: 01/06/2023]
Abstract
Galectins are an ancient family of β-galactoside-specific lectins and consist of 15 different types, each with a specific function. They play a role in the immune system, inflammation, wound healing and carcinogenesis. In particular the role of galectin in cancer is widely studied. Lately, the role of galectins in the development of cardiovascular disease has gained attention. Worldwide cardiovascular disease is still the leading cause of death. In ischemic heart disease, atherosclerosis limits adequate blood flow. Angiogenesis and arteriogenesis are highly important mechanisms relieving ischemia by restoring perfusion to the post-stenotic myocardial area. Galectins act ambiguous, both relieving ischemia and accelerating atherosclerosis. Atherosclerosis can ultimately lead to myocardial infarction or ischemic stroke, which are both associated with galectins. There is also a role for galectins in the development of myocarditis by their influence on inflammatory processes. Moreover, galectin acts as a biomarker for the severity of myocardial ischemia and heart failure. This review summarizes the association between galectins and the development of multiple cardiovascular diseases such as myocarditis, ischemic stroke, myocardial infarction, heart failure and atrial fibrillation. Furthermore it focuses on the association between galectin and more general mechanisms such as angiogenesis, arteriogenesis and atherosclerosis.
Collapse
Affiliation(s)
| | - Maurits R Hollander
- Department of Cardiology, VU University Medical Center, Amsterdam, The Netherlands
| | - Cansu Yıldırım
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Matthijs F Jansen
- Department of Cardiology, VU University Medical Center, Amsterdam, The Netherlands
| | - Paul F Teunissen
- Department of Cardiology, VU University Medical Center, Amsterdam, The Netherlands
| | - Anton J Horrevoets
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Niels van Royen
- Department of Cardiology, VU University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
143
|
Chang RL, Lin JW, Kuo WW, Hsieh DJY, Yeh YL, Shen CY, Day CH, Ho TJ, Viswanadha VP, Huang CY. Angiotensin-(1-7) attenuated long-term hypoxia-stimulated cardiomyocyte apoptosis by inhibiting HIF-1α nuclear translocation via Mas receptor regulation. Growth Factors 2016; 34:11-8. [PMID: 27055565 DOI: 10.3109/08977194.2016.1155150] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Extreme hypoxia often leads to myocardial apoptosis and causes heart failure. Angiotensin-(1-7)Ang-(1-7) is well known for its cardio-protective effects. However, the effects of Ang-(1-7) on long-term hypoxia (LTH)-induced apoptosis remain unknown. In this study, we found that Ang-(1-7) reduced myocardial apoptosis caused by hypoxia through the Mas receptor. Activation of the Ang-(1-7)/Mas axis down-regulated the hypoxia pro-apoptotic signaling cascade by decreasing the protein levels of hypoxia-inducible factor 1α (HIF-1α) and insulin-like growth factor binding protein-3 (IGFBP3). Moreover, the Ang-(1-7)/Mas axis further inhibited HIF-1α nuclear translocation. On the other hand, Ang-(1-7) activated the IGF1R/PI3K/Akt signaling pathways, which mediate cell survival. However, the above effects were abolished by A779 treatment or silencing of Mas expression. Taken together, our findings indicate that the Ang-(1-7)/Mas axis protects cardiomyocytes from LTH-stimulated apoptosis. The protective effect of Ang-(1-7) is associated with the inhibition of HIF-1α nuclear translocation and the induction of IGF1R and Akt phosphorylation.
Collapse
Affiliation(s)
- Ruey-Lin Chang
- a Graduate Institute of Basic Medical Science, China Medical University , Taichung , Taiwan
- b College of Chinese Medicine, School of Post-Baccalaureate Chinese Medicine, China Medical University , Taichung , Taiwan
| | - Jing-Wei Lin
- a Graduate Institute of Basic Medical Science, China Medical University , Taichung , Taiwan
| | - Wei-Wen Kuo
- c Department of Biological Science and Technology , China Medical University , Taichung , Taiwan
| | - Dennis Jine-Yuan Hsieh
- d School of Medical Laboratory and Biotechnology, Chung Shan Medical University , Taichung , Taiwan
| | - Yu-Lan Yeh
- e Department of Pathology , Changhua Christian Hospital , Changhua , Taiwan
- f en-Teh Junior College of Medicine, Nursing and Management , Miaoli , Taiwan
| | - Chia-Yao Shen
- g Department of Nursing , Mei Ho University , Pingguang Road , Pingtung , Taiwan
| | - Cecilia-Hsuan Day
- g Department of Nursing , Mei Ho University , Pingguang Road , Pingtung , Taiwan
| | - Tsung-Jung Ho
- h Chinese Medicine Department, China Medical University Beigang Hospital , Taichung , Taiwan
| | | | - Chih-Yang Huang
- a Graduate Institute of Basic Medical Science, China Medical University , Taichung , Taiwan
- j Graduate Institute of Chinese Medical Science, China Medical University , Taichung , Taiwan , and
- k Department of Health and Nutrition Biotechnology , Asia University , Taichung , Taiwan
| |
Collapse
|
144
|
Opacic D, van Bragt KA, Nasrallah HM, Schotten U, Verheule S. Atrial metabolism and tissue perfusion as determinants of electrical and structural remodelling in atrial fibrillation. Cardiovasc Res 2016; 109:527-41. [DOI: 10.1093/cvr/cvw007] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/12/2016] [Indexed: 12/14/2022] Open
|
145
|
Mansor LS, Mehta K, Aksentijevic D, Carr CA, Lund T, Cole MA, Le Page L, Sousa Fialho MDL, Shattock MJ, Aasum E, Clarke K, Tyler DJ, Heather LC. Increased oxidative metabolism following hypoxia in the type 2 diabetic heart, despite normal hypoxia signalling and metabolic adaptation. J Physiol 2016; 594:307-20. [PMID: 26574233 PMCID: PMC4713751 DOI: 10.1113/jp271242] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 11/10/2015] [Indexed: 01/13/2023] Open
Abstract
KEY POINTS Adaptation to hypoxia makes the heart more oxygen efficient, by metabolising more glucose. In contrast, type 2 diabetes makes the heart metabolise more fatty acids. Diabetes increases the chances of the heart being exposed to hypoxia, but whether the diabetic heart can adapt and respond is unknown. In this study we show that diabetic hearts retain the ability to adapt their metabolism in response to hypoxia, with functional hypoxia signalling pathways. However, the hypoxia-induced changes in metabolism are additive to abnormal baseline metabolism, resulting in hypoxic diabetic hearts metabolising more fat and less glucose than controls. This stops the diabetic heart being able to recover its function when stressed. These results demonstrate that the diabetic heart retains metabolic flexibility to adapt to hypoxia, but is hindered by the baseline effects of the disease. This increases our understanding of how the diabetic heart is affected by hypoxia-associated complications of the disease. ABSTRACT Hypoxia activates the hypoxia-inducible factor (HIF), promoting glycolysis and suppressing mitochondrial respiration. In the type 2 diabetic heart, glycolysis is suppressed whereas fatty acid metabolism is promoted. The diabetic heart experiences chronic hypoxia as a consequence of increased obstructive sleep apnoea and cardiovascular disease. Given the opposing metabolic effects of hypoxia and diabetes, we questioned whether diabetes affects cardiac metabolic adaptation to hypoxia. Control and type 2 diabetic rats were housed for 3 weeks in normoxia or 11% oxygen. Metabolism and function were measured in the isolated perfused heart using radiolabelled substrates. Following chronic hypoxia, both control and diabetic hearts upregulated glycolysis, lactate efflux and glycogen content and decreased fatty acid oxidation rates, with similar activation of HIF signalling pathways. However, hypoxia-induced changes were superimposed on diabetic hearts that were metabolically abnormal in normoxia, resulting in glycolytic rates 30% lower, and fatty acid oxidation 36% higher, in hypoxic diabetic hearts than hypoxic controls. Peroxisome proliferator-activated receptor α target proteins were suppressed by hypoxia, but activated by diabetes. Mitochondrial respiration in diabetic hearts was divergently activated following hypoxia compared with controls. These differences in metabolism were associated with decreased contractile recovery of the hypoxic diabetic heart following an acute hypoxic insult. In conclusion, type 2 diabetic hearts retain metabolic flexibility to adapt to hypoxia, with normal HIF signalling pathways. However, they are more dependent on oxidative metabolism following hypoxia due to abnormal normoxic metabolism, which was associated with a functional deficit in response to stress.
Collapse
Affiliation(s)
- Latt S Mansor
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Keshavi Mehta
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Dunja Aksentijevic
- British Heart Foundation Centre of Research Excellence, King's College London, The Rayne Institute, London, UK
| | - Carolyn A Carr
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Trine Lund
- Department of Medical Biology, University of Tromso, Norway
| | - Mark A Cole
- University of Nottingham Medical School, Queens Medical Centre, Nottingham, UK
| | - Lydia Le Page
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | | | - Michael J Shattock
- British Heart Foundation Centre of Research Excellence, King's College London, The Rayne Institute, London, UK
| | - Ellen Aasum
- Department of Medical Biology, University of Tromso, Norway
| | - Kieran Clarke
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Damian J Tyler
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Lisa C Heather
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
146
|
Increased mortality and aggravation of heart failure in liver X receptor-α knockout mice after myocardial infarction. Heart Vessels 2016; 31:1370-9. [PMID: 26753692 DOI: 10.1007/s00380-015-0781-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 12/02/2015] [Indexed: 12/23/2022]
Abstract
Liver X receptors, LXRα (NR1H3) and LXRβ (NR1H2), are best known as nuclear oxysterol receptors and physiological master regulators of lipid and cholesterol metabolism. LXRα play a protective role in acute myocardial ischemia/reperfusion (MI/R) injury, but its role in myocardial infarction (MI) is unknown. The present study was undertaken to determine the effect of LXRα knockout on survival and development of chronic heart failure after MI. Wild-type (WT) and LXRα(-/-) mice were subjected to MI followed by serial echocardiographic and histological assessments. Greater myocyte apoptosis and inflammation within the infarcted zones were found in LXRα(-/-) group at 3 days after MI. At 4 weeks post-MI, LXRα(-/-) MI murine hearts demonstrated significantly increased infarct size, reduced ejection fraction (LXRα(-/-) 29.4 % versus WT 34.4 %), aggravated left ventricular (LV) chamber dilation, enhanced fibrosis and reduced angiogenesis. In addition, LXRα(-/-) mice had increased mortality compared with WT mice. LXRα deficiency increases mortality, aggravates pathological injury and LV remodeling induced by MI. Drugs specifically targeting LXRα may be promising in the treatment of MI.
Collapse
|
147
|
Sinning C, Schnabel RB, Zeller T, Seiffert M, Rupprecht HJ, Lackner KJ, Blankenberg S, Bickel C, Westermann D. Prognostic use of soluble fms-like tyrosine kinase-1 and placental growth factor in patients with coronary artery disease. Biomark Med 2016; 10:95-106. [DOI: 10.2217/bmm.15.111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background: Intention of the study is to assess the cardiovascular mortality of patients with coronary artery disease (CAD) with the biomarkers of angiogenesis PlGF and its endogenous inhibitor sFlt-1. Methods: The cohort included n = 1848 patients with CAD and 282 subjects without CAD. In 85 patients cardiovascular mortality, as combination of fatal myocardial infarction or any cardiac death, during a median follow-up duration of 3.9 years was reported. Results: In Kaplan–Meier curve analysis PlGF in rising thirds was not predictive regarding outcome (p = 0.54), the same was shown for sFlt-1 (p = 0.44). Cox regression for the fully adjusted model provided a hazard ratio (HR) of 0.8 (p = 0.18) for PlGF and for sFlt-1 a HR = 1.0 (p = 0.8). Conclusion: Our results point out that these biomarkers reflecting angiogenesis might not be suited to establish prognosis in CAD.
Collapse
Affiliation(s)
- Christoph Sinning
- Department of General & Interventional Cardiology, University Heart Center Hamburg, Germany
| | - Renate B Schnabel
- Department of General & Interventional Cardiology, University Heart Center Hamburg, Germany
| | - Tanja Zeller
- Department of General & Interventional Cardiology, University Heart Center Hamburg, Germany
| | - Moritz Seiffert
- Department of General & Interventional Cardiology, University Heart Center Hamburg, Germany
| | - Hans J Rupprecht
- Department of Internal Medicine II, GPR Klinikum Rüsselsheim, Germany
| | - Karl J Lackner
- Department of Clinical Chemistry & Laboratory Medicine, Johannes Gu-tenberg-University Mainz, Germany
| | - Stefan Blankenberg
- Department of General & Interventional Cardiology, University Heart Center Hamburg, Germany
| | - Christoph Bickel
- Department of Internal Medicine, Federal Armed Forces Central Hospital, Koblenz, Germany
| | - Dirk Westermann
- Department of General & Interventional Cardiology, University Heart Center Hamburg, Germany
| |
Collapse
|
148
|
Janssen R, Zuidwijk MJ, Muller A, van Mil A, Dirkx E, Oudejans CBM, Paulus WJ, Simonides WS. MicroRNA 214 Is a Potential Regulator of Thyroid Hormone Levels in the Mouse Heart Following Myocardial Infarction, by Targeting the Thyroid-Hormone-Inactivating Enzyme Deiodinase Type III. Front Endocrinol (Lausanne) 2016; 7:22. [PMID: 27014189 PMCID: PMC4783388 DOI: 10.3389/fendo.2016.00022] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 02/22/2016] [Indexed: 12/19/2022] Open
Abstract
Cardiac thyroid-hormone signaling is a critical determinant of cellular metabolism and function in health and disease. A local hypothyroid condition within the failing heart in rodents has been associated with the re-expression of the fetally expressed thyroid-hormone-inactivating enzyme deiodinase type III (Dio3). While this enzyme emerges as a common denominator in the development of heart failure, the mechanism underlying its regulation remains largely unclear. In the present study, we investigated the involvement of microRNAs (miRNAs) in the regulation of Dio3 mRNA expression in the remodeling left ventricle (LV) of the mouse heart following myocardial infarction (MI). In silico analysis indicated that of the miRNAs that are differentially expressed in the post-MI heart, miR-214 has the highest potential to target Dio3 mRNA. In accordance, a luciferase reporter assay, including the full-length 3'UTR of mouse Dio3 mRNA, showed a 30% suppression of luciferase activity by miR-214. In the post-MI mouse heart, miR-214 and Dio3 protein were shown to be co-expressed in cardiomyocytes, while time-course analysis revealed that Dio3 mRNA expression precedes miR-214 expression in the post-MI LV. This suggests that a Dio3-induced decrease of T3 levels is involved in the induction of miR-214, which was supported by the finding that cardiac miR-214 expression is down regulated by T3 in mice. In vitro analysis of human DIO3 mRNA furthermore showed that miR-214 is able to suppress both mRNA and protein expression. Dio3 mRNA is a target of miR-214 and the Dio3-dependent stimulation of miR-214 expression in post-MI cardiomyocytes supports the involvement of a negative feedback mechanism regulating Dio3 expression.
Collapse
Affiliation(s)
- Rob Janssen
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, Netherlands
| | - Marian J. Zuidwijk
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, Netherlands
| | - Alice Muller
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, Netherlands
| | - Alain van Mil
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| | - Ellen Dirkx
- Department of Cardiology, Faculty of Health, Medicine and Life Sciences, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands
| | - Cees B. M. Oudejans
- Department of Clinical Chemistry, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, Netherlands
| | - Walter J. Paulus
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, Netherlands
| | - Warner S. Simonides
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, Netherlands
- *Correspondence: Warner S. Simonides,
| |
Collapse
|
149
|
van Zuylen VL, den Haan MC, Geutskens SB, Roelofs H, Fibbe WE, Schalij MJ, Atsma DE. Post-myocardial infarct inflammation and the potential role of cell therapy. Cardiovasc Drugs Ther 2015; 29:59-73. [PMID: 25583678 DOI: 10.1007/s10557-014-6568-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Myocardial infarction triggers reparative inflammatory processes programmed to repair damaged tissue. However, often additional injury to the myocardium occurs through the course of this inflammatory process, which ultimately can lead to heart failure. The potential beneficial effects of cell therapy in treating cardiac ischemic disease, the number one cause of death worldwide, are being studied extensively, both in clinical trials using adult stem cells as well as in fundamental research on cardiac stem cells and regenerative biology. This review summarizes the current knowledge on molecular and cellular processes implicated in post-infarction inflammation and discusses the potential beneficial role cell therapy might play in this process. Due to its immunomodulatory properties, the mesenchymal stromal cell is a candidate to reverse the disease progression of the infarcted heart towards heart failure, and therefore is emphasized in this review.
Collapse
Affiliation(s)
- Vanessa-leigh van Zuylen
- Department of Cardiology, Leiden University Medical Center, P.O. Box 9600, 2300, RC, Leiden, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
150
|
Maladaptive Modulations of NLRP3 Inflammasome and Cardioprotective Pathways Are Involved in Diet-Induced Exacerbation of Myocardial Ischemia/Reperfusion Injury in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:3480637. [PMID: 26788246 PMCID: PMC4691622 DOI: 10.1155/2016/3480637] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 09/09/2015] [Indexed: 12/22/2022]
Abstract
Excessive fatty acids and sugars intake is known to affect the development of cardiovascular diseases, including myocardial infarction. However, the underlying mechanisms are ill defined. Here we investigated the balance between prosurvival and detrimental pathways within the heart of C57Bl/6 male mice fed a standard diet (SD) or a high-fat high-fructose diet (HFHF) for 12 weeks and exposed to cardiac ex vivo ischemia/reperfusion (IR) injury. Dietary manipulation evokes a maladaptive response in heart mice, as demonstrated by the shift of myosin heavy chain isoform content from α to β, the increased expression of the Nlrp3 inflammasome and markers of oxidative metabolism, and the downregulation of the hypoxia inducible factor- (HIF-)2α and members of the Reperfusion Injury Salvage Kinases (RISK) pathway. When exposed to IR, HFHF mice hearts showed greater infarct size and lactic dehydrogenase release in comparison with SD mice. These effects were associated with an exacerbated overexpression of Nlrp3 inflammasome, resulting in marked caspase-1 activation and a compromised activation of the cardioprotective RISK/HIF-2α pathways. The common mechanisms of damage here reported lead to a better understanding of the cross-talk among prosurvival and detrimental pathways leading to the development of cardiovascular disorders associated with metabolic diseases.
Collapse
|