1
|
Huang H, Huang G, Li R, Wei L, Yuan Z, Huang W. Exercise Training After Myocardial Infarction Enhances Endothelial Progenitor Cells Function via NRG-1 Signaling. Cardiovasc Toxicol 2025; 25:411-426. [PMID: 39893285 DOI: 10.1007/s12012-025-09967-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 01/24/2025] [Indexed: 02/04/2025]
Abstract
Vascular regeneration after myocardial infarction (MI) is essential to improve myocardial ischemia, delay post-infarction ventricular remodeling, and improve the long-term prognosis of MI. Endothelial progenitor cells (EPCs) play important roles in the functional repair and homeostatic maintenance of the vascular endothelium. Exercise training stimulates EPC mobilization and increases the number of circulating EPCs, which has beneficial effects on the restoration of vascular integrity and hemodynamic reconstitution. After post-MI exercise training, cardiac function, the myocardial infarct area, and capillary density in the peri-infarct zone were measured. Bone marrow-derived EPCs were isolated from mice to measure the proliferation, migration, and in vitro angiogenesis of EPCs after myocardial infarction exercise. The expression of NRG-1/ErbB4 signaling factor and related proteins in downstream PI3K/AKT signaling pathway were detected, and the level of autocrine NRG-1 in EPCs was detected. Post-MI resistance training, aerobic exercise training, and combined exercise training increased EPC mobilization and proliferation, migration, and tube-forming capacity, promoted myocardial vascular regeneration, improved cardiac function, and reduced infarct size. Exercise training upregulated NRG-1 expression in EPCs, and NRG-1/ErbB4 signaling activated the downstream PI3K/Akt signaling pathway. Moreover, EPCs may have a positive feedback autocrine loop with NRG-1 to improve the function of EPCs and promote vascular repair and regeneration in mice with MI. Exercise training after MI promotes the function of bone marrow-derived EPCs through NRG-1/ErbB4/PI3K/AKT signaling, thus exerting a role in angiogenesis.
Collapse
Affiliation(s)
- Huai Huang
- Department of Cardiology & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Guoqiang Huang
- Department of Cardiology, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, 528400, China
| | - Ruojun Li
- Department of Cardiology & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Liqin Wei
- Department of Cardiology & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Zhu Yuan
- Department of Cardiology & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Weiqiang Huang
- Department of Cardiology & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
2
|
Klapproth S, Richter K, Türk C, Bock T, Bromberger T, Dominik J, Huck K, Pfaller K, Hess MW, Reichel CA, Krüger M, Nakchbandi IA, Moser M. Low kindlin-3 levels in osteoclasts of kindlin-3 hypomorphic mice result in osteopetrosis due to leaky sealing zones. J Cell Sci 2021; 134:272627. [PMID: 34704600 DOI: 10.1242/jcs.259040] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 10/19/2021] [Indexed: 12/14/2022] Open
Abstract
Osteoclasts form special integrin-mediated adhesion structures called sealing zones that enable them to adhere to and resorb bone. Sealing zones consist of densely packed podosomes tightly interconnected by actin fibers. Their formation requires the presence of the hematopoietic integrin regulator kindlin-3 (also known as Fermt3). In this study, we investigated osteoclasts and their adhesion structures in kindlin-3 hypomorphic mice expressing only 5-10% of the kindlin-3 level of wild-type mice. Low kindlin-3 expression reduces integrin activity, results in impaired osteoclast adhesion and signaling, and delays cell spreading. Despite these defects, in vitro-generated kindlin-3-hypomorphic osteoclast-like cells arrange their podosomes into adhesion patches and belts, but their podosome and actin organization is abnormal. Remarkably, kindlin-3-hypomorphic osteoclasts form sealing zones when cultured on calcified matrix in vitro and on bone surface in vivo. However, functional assays, immunohistochemical staining and electron micrographs of bone sections showed that they fail to seal the resorption lacunae properly, which is required for secreted proteinases to digest bone matrix. This results in mild osteopetrosis. Our study reveals a new, hitherto understudied function of kindlin-3 as an essential organizer of integrin-mediated adhesion structures, such as sealing zones.
Collapse
Affiliation(s)
- Sarah Klapproth
- Institute of Experimental Hematology, School of Medicine, Technical University Munich, D-81675 Munich, Germany
| | - Karsten Richter
- Central Unit Electron Microscopy, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
| | - Clara Türk
- CECAD Research Center, Institute for Genetics, University of Cologne, D-50931 Cologne, Germany
| | - Theresa Bock
- CECAD Research Center, Institute for Genetics, University of Cologne, D-50931 Cologne, Germany
| | - Thomas Bromberger
- Institute of Experimental Hematology, School of Medicine, Technical University Munich, D-81675 Munich, Germany
| | - Julian Dominik
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-University Munich, 81377 Munich, Germany.,Department of Otorhinolaryngology, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| | - Kathrin Huck
- Institute of Immunology, University of Heidelberg, D-69120 Heidelberg, Germany
| | - Kristian Pfaller
- Institute of Histology and Embryology, Medical University Innsbruck, A-6020 Innsbruck, Austria
| | - Michael W Hess
- Institute of Histology and Embryology, Medical University Innsbruck, A-6020 Innsbruck, Austria
| | - Christoph A Reichel
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-University Munich, 81377 Munich, Germany.,Department of Otorhinolaryngology, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| | - Marcus Krüger
- CECAD Research Center, Institute for Genetics, University of Cologne, D-50931 Cologne, Germany.,Center for Molecular Medicine (CMMC), University of Cologne, D-50931 Cologne, Germany
| | - Inaam A Nakchbandi
- Institute of Immunology, University of Heidelberg, D-69120 Heidelberg, Germany.,Max-Planck Institute of Biochemistry, D-82152 Martinsried, Germany
| | - Markus Moser
- Institute of Experimental Hematology, School of Medicine, Technical University Munich, D-81675 Munich, Germany.,Max-Planck Institute of Biochemistry, D-82152 Martinsried, Germany
| |
Collapse
|
3
|
Role of PI3K/Akt signaling pathway in cardiac fibrosis. Mol Cell Biochem 2021; 476:4045-4059. [PMID: 34244974 DOI: 10.1007/s11010-021-04219-w] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/29/2021] [Indexed: 12/26/2022]
Abstract
Heart failure (HF) is considered as a severe health problem worldwide, while cardiac fibrosis is one of the main driving factors for the progress of HF. Cardiac fibrosis was characterized by changes in cardiomyocytes, cardiac fibroblasts, ratio of collagen (COL) I/III, and the excessive production and deposition of extracellular matrix (ECM), thus forming a scar tissue, which leads to pathological process of cardiac structural changes and systolic as well as diastolic dysfunction. Cardiac fibrosis is a common pathological change of many advanced cardiovascular diseases including ischemic heart disease, hypertension, and HF. Accumulated studies have proven that phosphoinositol-3 kinase (PI3K)/Akt signaling pathway is involved in regulating the occurrence, progression and pathological formation of cardiac fibrosis via regulating cell survival, apoptosis, growth, cardiac contractility and even the transcription of related genes through a series of molecules including mammalian target of rapamycin (mTOR), glycogen synthase kinase 3 (GSK-3), forkhead box proteins O1/3 (FoxO1/3), and nitric oxide synthase (NOS). Thus, the review focuses on the role of PI3K/Akt signaling pathway in the cardiac fibrosis. The information reviewed here should be significant in understanding the role of PI3K/Akt in cardiac fibrosis and contribute to the design of further studies related to PI3K/Akt and the cardiac fibrotic response, as well as sought to shed light on a potential treatment for cardiac fibrosis.
Collapse
|
4
|
Lupieri A, Smirnova NF, Solinhac R, Malet N, Benamar M, Saoudi A, Santos-Zas I, Zeboudj L, Ait-Oufella H, Hirsch E, Ohayon P, Lhermusier T, Carrié D, Arnal JF, Ramel D, Gayral S, Laffargue M. Smooth muscle cells-derived CXCL10 prevents endothelial healing through PI3Kγ-dependent T cells response. Cardiovasc Res 2020; 116:438-449. [PMID: 31106375 DOI: 10.1093/cvr/cvz122] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 02/25/2019] [Accepted: 05/15/2019] [Indexed: 12/14/2022] Open
Abstract
AIMS Defects in efficient endothelial healing have been associated with complication of atherosclerosis such as post-angioplasty neoatherosclerosis and plaque erosion leading to thrombus formation. However, current preventive strategies do not consider re-endothelialization in their design. Here, we investigate mechanisms linking immune processes and defect in re-endothelialization. We especially evaluate if targeting phosphoinositide 3-kinase γ immune processes could restore endothelial healing and identify immune mediators responsible for these defects. METHODS AND RESULTS Using in vivo model of endovascular injury, we showed that both ubiquitous genetic inactivation of PI3Kγ and hematopoietic cell-specific PI3Kγ deletion improved re-endothelialization and that CD4+ T-cell population drives this effect. Accordingly, absence of PI3Kγ activity correlates with a decrease in local IFNγ secretion and its downstream interferon-inducible chemokine CXCL10. CXCL10 neutralization promoted re-endothelialization in vivo as the same level than those observed in absence of PI3Kγ suggesting a role of CXCL10 in re-endothelialization defect. Using a new established ex vivo model of carotid re-endothelialization, we showed that blocking CXCL10 restore the IFNγ-induced inhibition of endothelial healing and identify smooth muscle cells as the source of CXCL10 secretion in response to Th1 cytokine. CONCLUSION Altogether, these findings expose an unforeseen cellular cross-talk within the arterial wall whereby a PI3Kγ-dependent T-cell response leads to CXCL10 production by smooth muscle cells which in turn inhibits endothelial healing. Therefore, both PI3Kγ and the IFNγ/CXCL10 axis provide novel strategies to promote endothelial healing.
Collapse
Affiliation(s)
- Adrien Lupieri
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse F-31432, France
| | - Natalia F Smirnova
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse F-31432, France
| | - Romain Solinhac
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse F-31432, France
| | - Nicole Malet
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse F-31432, France
| | - Mehdi Benamar
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, INSERM, Centre National de la Recherche Scientifique (CNRS), Toulouse, F 31300, France
| | - Abdel Saoudi
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, INSERM, Centre National de la Recherche Scientifique (CNRS), Toulouse, F 31300, France
| | - Icia Santos-Zas
- Paris-Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Institut National de la Santé et de la Recherche Médicale (INSERM), UMR970, Paris, France
| | - Lynda Zeboudj
- Paris-Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Institut National de la Santé et de la Recherche Médicale (INSERM), UMR970, Paris, France
| | - Hafid Ait-Oufella
- Paris-Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Institut National de la Santé et de la Recherche Médicale (INSERM), UMR970, Paris, France
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | - Paul Ohayon
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse F-31432, France.,Department of Cardiology, University Hospital Rangueil, Toulouse, France
| | - Thibault Lhermusier
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse F-31432, France.,Department of Cardiology, University Hospital Rangueil, Toulouse, France
| | - Didier Carrié
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse F-31432, France.,Department of Cardiology, University Hospital Rangueil, Toulouse, France
| | - Jean-François Arnal
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse F-31432, France
| | - Damien Ramel
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse F-31432, France
| | - Stephanie Gayral
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse F-31432, France
| | - Muriel Laffargue
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse F-31432, France
| |
Collapse
|
5
|
Perrotta F, Perna A, Komici K, Nigro E, Mollica M, D’Agnano V, De Luca A, Guerra G. The State of Art of Regenerative Therapy in Cardiovascular Ischemic Disease: Biology, Signaling Pathways, and Epigenetics of Endothelial Progenitor Cells. Cells 2020; 9:E1886. [PMID: 32796767 PMCID: PMC7465688 DOI: 10.3390/cells9081886] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/19/2020] [Accepted: 08/07/2020] [Indexed: 12/18/2022] Open
Abstract
Ischemic heart disease is currently a major cause of mortality and morbidity worldwide. Nevertheless, the actual therapeutic scenario does not target myocardial cell regeneration and consequently, the progression toward the late stage of chronic heart failure is common. Endothelial progenitor cells (EPCs) are bone marrow-derived stem cells that contribute to the homeostasis of the endothelial wall in acute and chronic ischemic disease. Calcium modulation and other molecular pathways (NOTCH, VEGFR, and CXCR4) contribute to EPC proliferation and differentiation. The present review provides a summary of EPC biology with a particular focus on the regulatory pathways of EPCs and describes promising applications for cardiovascular cell therapy.
Collapse
Affiliation(s)
- Fabio Perrotta
- Dipartimento di Medicina e Scienze della Salute “V.Tiberio”, Università del Molise, 86100 Campobasso, Italy; (A.P.); (K.K.); (G.G.)
| | - Angelica Perna
- Dipartimento di Medicina e Scienze della Salute “V.Tiberio”, Università del Molise, 86100 Campobasso, Italy; (A.P.); (K.K.); (G.G.)
| | - Klara Komici
- Dipartimento di Medicina e Scienze della Salute “V.Tiberio”, Università del Molise, 86100 Campobasso, Italy; (A.P.); (K.K.); (G.G.)
| | - Ersilia Nigro
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche, Farmaceutiche, Università della Campania “Luigi Vanvitelli”, 81100 Caserta, Italy;
- CEINGE-Biotecnologie avanzate, 80145 Naples, Italy
| | - Mariano Mollica
- Dipartimento di Scienze Mediche Traslazionali, Università della Campania “Luigi Vanvitelli”, 80131 Naples, Italy; (M.M.); (V.D.)
| | - Vito D’Agnano
- Dipartimento di Scienze Mediche Traslazionali, Università della Campania “Luigi Vanvitelli”, 80131 Naples, Italy; (M.M.); (V.D.)
| | - Antonio De Luca
- Department of Mental and Physical Health and Preventive Medicine, Section of Human Anatomy, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Germano Guerra
- Dipartimento di Medicina e Scienze della Salute “V.Tiberio”, Università del Molise, 86100 Campobasso, Italy; (A.P.); (K.K.); (G.G.)
| |
Collapse
|
6
|
Receptor-Ligand Interaction Mediates Targeting of Endothelial Colony Forming Cell-derived Exosomes to the Kidney after Ischemic Injury. Sci Rep 2018; 8:16320. [PMID: 30397255 PMCID: PMC6218514 DOI: 10.1038/s41598-018-34557-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 10/19/2018] [Indexed: 01/15/2023] Open
Abstract
Endothelial colony forming cell (ECFC)-derived exosomes protect mice against ischemic kidney injury, via transfer of microRNA-(miR)-486-5p. Mechanisms mediating exosome recruitment to tissues are unclear. We hypothesized that ECFC exosomes target ischemic kidneys, involving interaction between exosomal CXC chemokine receptor type 4 (CXCR4) and stromal cell-derived factor (SDF)-1α. Ischemia-reperfusion was induced in mice by bilateral renal vascular clamp, with intravenous infusion of exosomes at reperfusion. Optical imaging determined exosome biodistribution, and miR-486-5p was measured by real-time PCR. Human umbilical vein endothelial cells (HUVECs) were cultured to study the CXCR4/SDF-1α interaction. Targeting of administered exosomes to ischemic kidneys was detected 30 min and 4 hrs after reperfusion. Exosomes increased miR-486-5p levels only in kidneys, within proximal tubules, glomeruli, and endothelial cells. Uptake of fluorescently-labeled exosomes into HUVECs, and exosomal transfer of miR-486-5p were enhanced by hypoxia, effects blocked by neutralizing antibody to SDF-1α or by the CXCR4 inhibitor plerixafor. Infusion of ECFC exosomes prevented ischemic kidney injury in vivo, an effect that was not observed when exosomes were pre-incubated with plerixafor. These data indicate that ECFC exosomes selectively target the kidneys after ischemic injury, with rapid cellular transfer of miR486-5p. Targeting of exosomes may involve interaction of CXCR4 with endothelial cell SDF-1α.
Collapse
|
7
|
Liu X, Xu Y, Zhou Q, Chen M, Zhang Y, Liang H, Zhao J, Zhong W, Wang M. PI3K in cancer: its structure, activation modes and role in shaping tumor microenvironment. Future Oncol 2017; 14:665-674. [PMID: 29219001 DOI: 10.2217/fon-2017-0588] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The role of PI3K in cancer has been well established, and mutations of PIK3CA, the gene coding for catalytic subunit p110α of PI3K, are found in approximately 30% human cancers. The hyperactivated PI3K pathway plays a central role in the tumor cell activities such as proliferation, differentiation, chemotaxis, survival, trafficking and metabolism. Besides, PI3K pathway is involved in the regulation of angiogenesis and the host immune response against cancer. Therefore, the inhibition of PI3K pathway can yield multifaceted tumor cell-extrinsic effects that may synergize with chemotherapy, and more importantly, with the newly revived immunotherapy. Here, we review the structures and activation modes of PI3Ks and its implications in angiogenesis, extracellular matrix remodeling and tumor immunity.
Collapse
Affiliation(s)
- Xiaoyan Liu
- Department of Pulmonary Medicine, Lung Cancer Center, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, PR China
| | - Yan Xu
- Department of Pulmonary Medicine, Lung Cancer Center, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, PR China
| | - Qing Zhou
- Department of Pulmonary Medicine, Lung Cancer Center, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, PR China
| | - Minjiang Chen
- Department of Pulmonary Medicine, Lung Cancer Center, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, PR China
| | - Yu Zhang
- Department of Pulmonary Medicine, Lung Cancer Center, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, PR China
| | - Hongge Liang
- Department of Pulmonary Medicine, Lung Cancer Center, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, PR China
| | - Jing Zhao
- Department of Pulmonary Medicine, Lung Cancer Center, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, PR China
| | - Wei Zhong
- Department of Pulmonary Medicine, Lung Cancer Center, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, PR China
| | - Mengzhao Wang
- Department of Pulmonary Medicine, Lung Cancer Center, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, PR China
| |
Collapse
|
8
|
Klotzsche-von Ameln A, Cremer S, Hoffmann J, Schuster P, Khedr S, Korovina I, Troullinaki M, Neuwirth A, Sprott D, Chatzigeorgiou A, Economopoulou M, Orlandi A, Hain A, Zeiher AM, Deussen A, Hajishengallis G, Dimmeler S, Chavakis T, Chavakis E. Endogenous developmental endothelial locus-1 limits ischaemia-related angiogenesis by blocking inflammation. Thromb Haemost 2017; 117:1150-1163. [PMID: 28447099 PMCID: PMC5502105 DOI: 10.1160/th16-05-0354] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 03/05/2017] [Indexed: 12/23/2022]
Abstract
We have recently identified endothelial cell-secreted developmental endothelial locus-1 (Del-1) as an endogenous inhibitor of β2-integrin-dependent leukocyte infiltration. Del-1 was previously also implicated in angiogenesis. Here, we addressed the role of endogenously produced Del-1 in ischaemia-related angiogenesis. Intriguingly, Del-1-deficient mice displayed increased neovascularisation in two independent ischaemic models (retinopathy of prematurity and hind-limb ischaemia), as compared to Del-1-proficient mice. On the contrary, angiogenic sprouting in vitro or ex vivo (aortic ring assay) and physiological developmental retina angiogenesis were not affected by Del-1 deficiency. Mechanistically, the enhanced ischaemic neovascularisation in Del-1-deficiency was linked to higher infiltration of the ischaemic tissue by CD45+ haematopoietic and immune cells. Moreover, Del-1-deficiency promoted β2-integrin-dependent adhesion of haematopoietic cells to endothelial cells in vitro, and the homing of hematopoietic progenitor cells and of immune cell populations to ischaemic muscles in vivo. Consistently, the increased hind limb ischaemia-related angiogenesis in Del-1 deficiency was completely reversed in mice lacking both Del-1 and the β2-integrin LFA-1. Additionally, enhanced retinopathy-associated neovascularisation in Del-1-deficient mice was reversed by LFA-1 blockade. Our data reveal a hitherto unrecognised function of endogenous Del-1 as a local inhibitor of ischaemia-induced angiogenesis by restraining LFA-1-dependent homing of pro-angiogenic haematopoietic cells to ischaemic tissues. Our findings are relevant for the optimisation of therapeutic approaches in the context of ischaemic diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Emmanouil Chavakis
- Emmanouil Chavakis, MD, Dept. of Internal Medicine III, Goethe University of Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany, Tel.: +49 69 6301 4131, +49 69 6301 87965, Fax: +49 69 6301 83462, E-mail:
| |
Collapse
|
9
|
Vázquez-Prado J, Bracho-Valdés I, Cervantes-Villagrana RD, Reyes-Cruz G. Gβγ Pathways in Cell Polarity and Migration Linked to Oncogenic GPCR Signaling: Potential Relevance in Tumor Microenvironment. Mol Pharmacol 2016; 90:573-586. [PMID: 27638873 DOI: 10.1124/mol.116.105338] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 09/14/2016] [Indexed: 02/14/2025] Open
Abstract
Cancer cells and stroma cells in tumors secrete chemotactic agonists that exacerbate invasive behavior, promote tumor-induced angiogenesis, and recruit protumoral bone marrow-derived cells. In response to shallow gradients of chemotactic stimuli recognized by G protein-coupled receptors (GPCRs), Gβγ-dependent signaling cascades contribute to specifying the spatiotemporal assembly of cytoskeletal structures that can dynamically alter cell morphology. This sophisticated process is intrinsically linked to the activation of Rho GTPases and their cytoskeletal-remodeling effectors. Thus, Rho guanine nucleotide exchange factors, the activators of these molecular switches, and their upstream signaling partners are considered participants of tumor progression. Specifically, phosphoinositide-3 kinases (class I PI3Ks, β and γ) and P-Rex1, a Rac-specific guanine nucleotide exchange factor, are fundamental Gβγ effectors in the pathways controlling directionally persistent motility. In addition, GPCR-dependent chemotactic responses often involve endosomal trafficking of signaling proteins; coincidently, endosomes serve as signaling platforms for Gβγ In preclinical murine models of cancer, inhibition of Gβγ attenuates tumor growth, whereas in cancer patients, aberrant overexpression of chemotactic Gβγ effectors and recently identified mutations in Gβ correlate with poor clinical outcome. Here we discuss emerging paradigms of Gβγ signaling in cancer, which are essential for chemotactic cell migration and represent novel opportunities to develop pathway-specific pharmacologic treatments.
Collapse
Affiliation(s)
- José Vázquez-Prado
- Departments of Pharmacology (J.V.-P., R.D.C.-V.) and Cell Biology (G.R.-C.). CINVESTAV-IPN, Mexico City, and Department of Pharmacology (I.B.-V.), School of Medicine, UABC, Mexicali, B.C., Mexico
| | - Ismael Bracho-Valdés
- Departments of Pharmacology (J.V.-P., R.D.C.-V.) and Cell Biology (G.R.-C.). CINVESTAV-IPN, Mexico City, and Department of Pharmacology (I.B.-V.), School of Medicine, UABC, Mexicali, B.C., Mexico
| | - Rodolfo Daniel Cervantes-Villagrana
- Departments of Pharmacology (J.V.-P., R.D.C.-V.) and Cell Biology (G.R.-C.). CINVESTAV-IPN, Mexico City, and Department of Pharmacology (I.B.-V.), School of Medicine, UABC, Mexicali, B.C., Mexico
| | - Guadalupe Reyes-Cruz
- Departments of Pharmacology (J.V.-P., R.D.C.-V.) and Cell Biology (G.R.-C.). CINVESTAV-IPN, Mexico City, and Department of Pharmacology (I.B.-V.), School of Medicine, UABC, Mexicali, B.C., Mexico
| |
Collapse
|
10
|
Lupieri A, Smirnova N, Malet N, Gayral S, Laffargue M. PI3K signaling in arterial diseases: Non redundant functions of the PI3K isoforms. Adv Biol Regul 2015; 59:4-18. [PMID: 26238239 DOI: 10.1016/j.jbior.2015.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 06/15/2015] [Accepted: 06/15/2015] [Indexed: 06/04/2023]
Abstract
Cardiovascular diseases are the most common cause of death around the world. This includes atherosclerosis and the adverse effects of its treatment, such as restenosis and thrombotic complications. The development of these arterial pathologies requires a series of highly-intertwined interactions between immune and arterial cells, leading to specific inflammatory and fibroproliferative cellular responses. In the last few years, the study of phosphoinositide 3-kinase (PI3K) functions has become an attractive area of investigation in the field of arterial diseases, especially since inhibitors of specific PI3K isoforms have been developed. The PI3K family includes 8 members divided into classes I, II or III depending on their substrate specificity. Although some of the different isoforms are responsible for the production of the same 3-phosphoinositides, they each have specific, non-redundant functions as a result of differences in expression levels in different cell types, activation mechanisms and specific subcellular locations. This review will focus on the functions of the different PI3K isoforms that are suspected as having protective or deleterious effects in both the various immune cells and types of cell found in the arterial wall. It will also discuss our current understanding in the context of which PI3K isoform(s) should be targeted for future therapeutic interventions to prevent or treat arterial diseases.
Collapse
Affiliation(s)
- Adrien Lupieri
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, F-31300, France
| | - Natalia Smirnova
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, F-31300, France
| | - Nicole Malet
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, F-31300, France
| | - Stéphanie Gayral
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, F-31300, France
| | - Muriel Laffargue
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, F-31300, France.
| |
Collapse
|
11
|
Balakrishnan K, Peluso M, Fu M, Rosin NY, Burger JA, Wierda WG, Keating MJ, Faia K, O'Brien S, Kutok JL, Gandhi V. The phosphoinositide-3-kinase (PI3K)-delta and gamma inhibitor, IPI-145 (Duvelisib), overcomes signals from the PI3K/AKT/S6 pathway and promotes apoptosis in CLL. Leukemia 2015; 29:1811-22. [PMID: 25917267 DOI: 10.1038/leu.2015.105] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 03/26/2015] [Accepted: 04/07/2015] [Indexed: 11/10/2022]
Abstract
The functional relevance of the B-cell receptor (BCR) and the evolution of protein kinases as therapeutic targets have recently shifted the paradigm for treatment of B-cell malignancies. Inhibition of p110δ with idelalisib has shown clinical activity in chronic lymphocytic leukemia (CLL). The dynamic interplay of isoforms p110δ and p110γ in leukocytes support the hypothesis that dual blockade may provide a therapeutic benefit. IPI-145, an oral inhibitor of p110δ and p110γ isoforms, sensitizes BCR-stimulated and/or stromal co-cultured primary CLL cells to apoptosis (median 20%, n=57; P<0.0001) including samples with poor prognostic markers, unmutated IgVH (n=28) and prior treatment (n=15; P<0.0001). IPI-145 potently inhibits the CD40L/IL-2/IL-10 induced proliferation of CLL cells with an IC50 in sub-nanomolar range. A corresponding dose-responsive inhibition of pAKT(Ser473) is observed with an IC50 of 0.36 nM. IPI-145 diminishes the BCR-induced chemokines CCL3 and CCL4 secretion to 17% and 37%, respectively. Pre-treatment with 1 μM IPI-145 inhibits the chemotaxis toward CXCL12; reduces pseudoemperipolesis to median 50%, inferring its ability to interfere with homing capabilities of CLL cells. BCR-activated signaling proteins AKT(Ser473), BAD(Ser112), ERK(Thr202/Tyr204) and S6(Ser235/236) are mitigated by IPI-145. Importantly, for clinical development in hematological malignancies, IPI-145 is selective to CLL B cells, sparing normal B- and T-lymphocytes.
Collapse
Affiliation(s)
- K Balakrishnan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - M Peluso
- Infinity Pharmaceuticals Inc., Cambridge, MA, USA
| | - M Fu
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - N Y Rosin
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - J A Burger
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - W G Wierda
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - M J Keating
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - K Faia
- Infinity Pharmaceuticals Inc., Cambridge, MA, USA
| | - S O'Brien
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - J L Kutok
- Infinity Pharmaceuticals Inc., Cambridge, MA, USA
| | - V Gandhi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
12
|
Choudhury TR, Mathur A. The birth of 'regenerative pharmacology': a clinical perspective. Br J Pharmacol 2014; 169:239-46. [PMID: 23425309 DOI: 10.1111/bph.12128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
13
|
Kobayashi K, Sato K, Kida T, Omori K, Hori M, Ozaki H, Murata T. Stromal cell-derived factor-1α/C-X-C chemokine receptor type 4 axis promotes endothelial cell barrier integrity via phosphoinositide 3-kinase and Rac1 activation. Arterioscler Thromb Vasc Biol 2014; 34:1716-22. [PMID: 24925969 DOI: 10.1161/atvbaha.114.303890] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Although stromal cell-derived factor (SDF)-1αis well known to modulate the mobilization of hematopoietic stem cells and endothelial progenitor cells, its effects on some pre-existing vascular functions remain unknown. We have investigated here the role of SDF-1αsignaling in endothelial barrier function. APPROACH AND RESULTS Treatment with SDF-1α elevated transendothelial electrical resistance and inhibited the dextran hyperpermeability elicited by thrombin in bovine aortic endothelial cells, both indicating an increase in endothelial barrier function. SDF-1α binds to 2 receptors, C-X-C chemokine receptor types 4 and 7 (CXCR4 and CXCR7). Pretreatment with a CXCR4 antagonist or CXCR4 gene depletion by small interfering RNA (siRNA) eliminated SDF-1α-induced endothelial barrier enhancement. In contrast, CXCR7 antagonist or CXCR7 gene depletion by siRNA did not influence SDF-1α-induced barrier enhancement. Pretreatment with a Gi-protein inhibitor, a phosphoinositide 3-kinase (PI3K) inhibitor, or PI3K p110γsubunit gene depletion by siRNA also inhibited SDF-1α-induced barrier enhancement significantly. Western blot analysis revealed that SDF-1α phosphorylated Akt(Ser473) in endothelial cells, suggesting PI3K activation. Immunostaining showed that treatment with SDF-1αformed a cortical actin rim, which was accompanied by Rac1 activation. In vivo, SDF-1αinhibited croton oil-induced vascular leakage indexed by dye extravasation, which is attenuated by a pretreatment with a CXCR4 antagonist. CONCLUSIONS We have identified SDF-1α as a novel suppressor of endothelial permeability. Specifically, SDF-1α stimulates the CXCR4/PI3K/Rac1 signaling pathway and the subsequent cytoskeletal rearrangement.
Collapse
Affiliation(s)
- Koji Kobayashi
- From the Departments of Animal Radiology (K.K., K.O., T.M.) and Veterinary Pharmacology (K.S., T.K., M.H., H.O.), Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Kanako Sato
- From the Departments of Animal Radiology (K.K., K.O., T.M.) and Veterinary Pharmacology (K.S., T.K., M.H., H.O.), Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Taiki Kida
- From the Departments of Animal Radiology (K.K., K.O., T.M.) and Veterinary Pharmacology (K.S., T.K., M.H., H.O.), Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Keisuke Omori
- From the Departments of Animal Radiology (K.K., K.O., T.M.) and Veterinary Pharmacology (K.S., T.K., M.H., H.O.), Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Masatoshi Hori
- From the Departments of Animal Radiology (K.K., K.O., T.M.) and Veterinary Pharmacology (K.S., T.K., M.H., H.O.), Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Ozaki
- From the Departments of Animal Radiology (K.K., K.O., T.M.) and Veterinary Pharmacology (K.S., T.K., M.H., H.O.), Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Takahisa Murata
- From the Departments of Animal Radiology (K.K., K.O., T.M.) and Veterinary Pharmacology (K.S., T.K., M.H., H.O.), Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
14
|
Silvestre JS, Smadja DM, Lévy BI. Postischemic revascularization: from cellular and molecular mechanisms to clinical applications. Physiol Rev 2013; 93:1743-802. [PMID: 24137021 DOI: 10.1152/physrev.00006.2013] [Citation(s) in RCA: 181] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
After the onset of ischemia, cardiac or skeletal muscle undergoes a continuum of molecular, cellular, and extracellular responses that determine the function and the remodeling of the ischemic tissue. Hypoxia-related pathways, immunoinflammatory balance, circulating or local vascular progenitor cells, as well as changes in hemodynamical forces within vascular wall trigger all the processes regulating vascular homeostasis, including vasculogenesis, angiogenesis, arteriogenesis, and collateral growth, which act in concert to establish a functional vascular network in ischemic zones. In patients with ischemic diseases, most of the cellular (mainly those involving bone marrow-derived cells and local stem/progenitor cells) and molecular mechanisms involved in the activation of vessel growth and vascular remodeling are markedly impaired by the deleterious microenvironment characterized by fibrosis, inflammation, hypoperfusion, and inhibition of endogenous angiogenic and regenerative programs. Furthermore, cardiovascular risk factors, including diabetes, hypercholesterolemia, hypertension, diabetes, and aging, constitute a deleterious macroenvironment that participates to the abrogation of postischemic revascularization and tissue regeneration observed in these patient populations. Thus stimulation of vessel growth and/or remodeling has emerged as a new therapeutic option in patients with ischemic diseases. Many strategies of therapeutic revascularization, based on the administration of growth factors or stem/progenitor cells from diverse sources, have been proposed and are currently tested in patients with peripheral arterial disease or cardiac diseases. This review provides an overview from our current knowledge regarding molecular and cellular mechanisms involved in postischemic revascularization, as well as advances in the clinical application of such strategies of therapeutic revascularization.
Collapse
|
15
|
Hirsch E, Ciraolo E, Franco I, Ghigo A, Martini M. PI3K in cancer-stroma interactions: bad in seed and ugly in soil. Oncogene 2013; 33:3083-90. [PMID: 23893246 DOI: 10.1038/onc.2013.265] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 05/21/2013] [Accepted: 05/21/2013] [Indexed: 02/07/2023]
Abstract
Over the past decade the phosphoinositide-3 kinase (PI3K) signaling pathway emerged as an important player for tumor initiation and growth and, currently, PI3K inhibition constitutes a promising therapeutic approach for solid and hematological tumors. Beside its role in tumor cell evolution, PI3K signaling also provides integral functions for noncancerous cells that reside in healthy tissues surrounding the tumor, also referred as tumor microenvironment (TME). This review will address how PI3K signaling participates to the tumorigenic process and discuss the interaction between tumor cells and the surrounding TME, with particular focus on the role of PI3Ks in tumor-associated immune responses, tumor angiogenesis and metastasis formation.
Collapse
Affiliation(s)
- E Hirsch
- Department of Molecular Biotechnology and Health Sciences-Center for Molecular Biotechnology, University of Torino, Torino, Italy
| | - E Ciraolo
- Department of Molecular Biotechnology and Health Sciences-Center for Molecular Biotechnology, University of Torino, Torino, Italy
| | - I Franco
- Department of Molecular Biotechnology and Health Sciences-Center for Molecular Biotechnology, University of Torino, Torino, Italy
| | - A Ghigo
- Department of Molecular Biotechnology and Health Sciences-Center for Molecular Biotechnology, University of Torino, Torino, Italy
| | - M Martini
- Department of Molecular Biotechnology and Health Sciences-Center for Molecular Biotechnology, University of Torino, Torino, Italy
| |
Collapse
|
16
|
Cubbon RM, Mercer BN, Sengupta A, Kearney MT. Importance of insulin resistance to vascular repair and regeneration. Free Radic Biol Med 2013; 60:246-63. [PMID: 23466555 DOI: 10.1016/j.freeradbiomed.2013.02.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Revised: 02/22/2013] [Accepted: 02/23/2013] [Indexed: 01/14/2023]
Abstract
Metabolic insulin resistance is apparent across a spectrum of clinical disorders, including obesity and diabetes, and is characterized by an adverse clustering of cardiovascular risk factors related to abnormal cellular responses to insulin. These disorders are becoming increasingly prevalent and represent a major global public health concern because of their association with significant increases in atherosclerosis-related mortality. Endogenous repair mechanisms are thought to retard the development of vascular disease, and a growing evidence base supports the adverse impact of the insulin-resistant phenotype upon indices of vascular repair. Beyond the impact of systemic metabolic changes, emerging data from murine studies also provide support for abnormal insulin signaling at the level of vascular cells in retarding vascular repair. Interrelated pathophysiological factors, including reduced nitric oxide bioavailability, oxidative stress, altered growth factor activity, and abnormal intracellular signaling, are likely to act in conjunction to impede vascular repair while also driving vascular damage. Understanding of these processes is shaping novel therapeutic paradigms that aim to promote vascular repair and regeneration, either by recruiting endogenous mechanisms or by the administration of cell-based therapies.
Collapse
Affiliation(s)
- Richard M Cubbon
- Multidisciplinary Cardiovascular Research Centre, LIGHT Laboratories, The University of Leeds, Leeds LS2 9JT, UK.
| | | | | | | |
Collapse
|
17
|
Abstract
PI3Ks are signaling enzymes engaged by different types of membrane receptors and activated in cardiovascular diseases such as hypertension, atherosclerosis, thrombosis and heart failure. Studies performed on genetically modified animals have provided proof-of-concept that general or isoform-specific blockade of these enzymes can modify disease development and progression. Hence, therapeutic inhibition of PI3Ks with novel pharmacological compounds constitutes a promising area of drug development. In particular, inhibitors of PI3Ks have the potential to reduce blood pressure, restrain the development of atherosclerosis and/or stabilize atherosclerotic plaques, blunt platelet aggregation, prevent left ventricular remodeling and preserve myocardial contractility in heart failure. This review summarizes the rationale of PI3K inhibition in the most prevalent cardiovascular diseases, and the available data on the therapeutic effects of PI3K inhibitors in their preclinical models. Implications for future drug development and human therapy are also discussed.
Collapse
|
18
|
Rolland-Turner M, Goretti E, Bousquenaud M, Léonard F, Nicolas C, Zhang L, Maskali F, Marie PY, Devaux Y, Wagner D. Adenosine stimulates the migration of human endothelial progenitor cells. Role of CXCR4 and microRNA-150. PLoS One 2013; 8:e54135. [PMID: 23326587 PMCID: PMC3541240 DOI: 10.1371/journal.pone.0054135] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 12/10/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Administration of endothelial progenitor cells (EPC) represents a promising option to regenerate the heart after myocardial infarction, but is limited because of low recruitment and engraftment in the myocardium. Mobilization and migration of EPC are mainly controlled by stromal cell-derived factor 1α (SDF-1α) and its receptor CXCR4. We hypothesized that adenosine, a cardioprotective molecule, may improve the recruitment of EPC to the heart. METHODS EPC were obtained from peripheral blood mononuclear cells of healthy volunteers. Expression of chemokines and their receptors was evaluated using microarrays, quantitative PCR, and flow cytometry. A Boyden chamber assay was used to assess chemotaxis. Recruitment of EPC to the infarcted heart was evaluated in rats after permanent occlusion of the left anterior descending coronary artery. RESULTS Microarray analysis revealed that adenosine modulates the expression of several members of the chemokine family in EPC. Among these, CXCR4 was up-regulated by adenosine, and this result was confirmed by quantitative PCR (3-fold increase, P<0.001). CXCR4 expression at the cell surface was also increased. This effect involved the A(2B) receptor. Pretreatment of EPC with adenosine amplified their migration towards recombinant SDF-1α or conditioned medium from cardiac fibroblasts. Both effects were abolished by CXCR4 blocking antibodies. Adenosine also increased CXCR4 under ischemic conditions, and decreased miR-150 expression. Binding of miR-150 to the 3' untranslated region of CXCR4 was verified by luciferase assay. Addition of pre-miR-150 blunted the effect of adenosine on CXCR4. Administration of adenosine to rats after induction of myocardial infarction stimulated EPC recruitment to the heart and enhanced angiogenesis. CONCLUSION Adenosine increases the migration of EPC. The mechanism involves A(2B) receptor activation, decreased expression of miR-150 and increased expression of CXCR4. These results suggest that adenosine may be used to enhance the capacity of EPC to revascularize the ischemic heart.
Collapse
Affiliation(s)
- Magali Rolland-Turner
- Laboratory of Cardiovascular Research, Public Research Centre – Health (CRP – Santé), Luxembourg, Luxembourg
| | - Emeline Goretti
- Laboratory of Cardiovascular Research, Public Research Centre – Health (CRP – Santé), Luxembourg, Luxembourg
| | - Mélanie Bousquenaud
- Laboratory of Cardiovascular Research, Public Research Centre – Health (CRP – Santé), Luxembourg, Luxembourg
| | - Frédérique Léonard
- Laboratory of Cardiovascular Research, Public Research Centre – Health (CRP – Santé), Luxembourg, Luxembourg
| | - Christelle Nicolas
- Laboratory of Cardiovascular Research, Public Research Centre – Health (CRP – Santé), Luxembourg, Luxembourg
| | - Lu Zhang
- Laboratory of Cardiovascular Research, Public Research Centre – Health (CRP – Santé), Luxembourg, Luxembourg
| | - Fatiha Maskali
- Nancyclotep Experimental Imaging Platform, Vandoeuvre-les-Nancy, France
| | - Pierre-Yves Marie
- Nancyclotep Experimental Imaging Platform, Vandoeuvre-les-Nancy, France
| | - Yvan Devaux
- Laboratory of Cardiovascular Research, Public Research Centre – Health (CRP – Santé), Luxembourg, Luxembourg
| | - Daniel Wagner
- Laboratory of Cardiovascular Research, Public Research Centre – Health (CRP – Santé), Luxembourg, Luxembourg
- Division of Cardiology, Centre Hospitalier, Luxembourg, Luxembourg
| |
Collapse
|
19
|
Role of phosphatidylinositol 3,4,5-trisphosphate in cell signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 991:105-39. [PMID: 23775693 DOI: 10.1007/978-94-007-6331-9_7] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Many lipids present in cellular membranes are phosphorylated as part of signaling cascades and participate in the recruitment, localization, and activation of downstream protein effectors. Phosphatidylinositol (3,4,5)-trisphosphate (PtdIns(3,4,5)P3) is one of the most important second messengers and is capable of interacting with a variety of proteins through specific PtdIns(3,4,5)P3 binding domains. Localization and activation of these effector proteins controls a myriad of cellular functions including cell survival, proliferation, cytoskeletal rearrangement, and gene expression. Aberrations in the production and metabolism of PtdIns(3,4,5)P3 have been implicated in many human diseases including cancer, diabetes, inflammation, and heart disease. This chapter provides an overview of the role of PtdIns(3,4,5)P3 in cellular regulation and the implications of PtdIns(3,4,5)P3 dysregulation in human diseases. Additionally, recent attempts at targeting PtdIns(3,4,5)P3 signaling via small molecule inhibitors are summarized.
Collapse
|
20
|
Bousquenaud M, Schwartz C, Léonard F, Rolland-Turner M, Wagner D, Devaux Y. Monocyte chemotactic protein 3 is a homing factor for circulating angiogenic cells. Cardiovasc Res 2012; 94:519-25. [DOI: 10.1093/cvr/cvs140] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
21
|
Progenitor cell mobilization and recruitment: SDF-1, CXCR4, α4-integrin, and c-kit. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2012; 111:243-64. [PMID: 22917234 DOI: 10.1016/b978-0-12-398459-3.00011-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Progenitor cell retention and release are largely governed by the binding of stromal-cell-derived factor 1 (SDF-1) to CXC chemokine receptor 4 (CXCR4) and by α4-integrin signaling. Both of these pathways are dependent on c-kit activity: the mobilization of progenitor cells in response to either CXCR4 antagonism or α4-integrin blockade is impaired by the loss of c-kit kinase activity; and c-kit-kinase inactivation blocks the retention of CXCR4-positive progenitor cells in the bone marrow. SDF-1/CXCR4 and α4-integrin signaling are also crucial for the retention of progenitor cells in the ischemic region, which may explain, at least in part, why clinical trials of progenitor cell therapy have failed to display the efficacy observed in preclinical investigations. The lack of effectiveness is often attributed to poor retention of the transplanted cells and, to date, most of the trial protocols have mobilized cells with injections of granulocyte colony-stimulating factor (G-CSF), which activates extracellular proteases that irreversibly cleave cell-surface adhesion molecules, including α4-integrin and CXCR4. Thus, the retention of G-CSF-mobilized cells in the ischemic region may be impaired, and the mobilization of agents that reversibly disrupt SDF-1/CXCR4 binding, such as AMD3100, may improve patient response. Efforts to supplement SDF-1 levels in the ischemic region may also improve progenitor cell recruitment and the effectiveness of stem cell therapy.
Collapse
|
22
|
Chilian WM, Penn MS, Pung YF, Dong F, Mayorga M, Ohanyan V, Logan S, Yin L. Coronary collateral growth--back to the future. J Mol Cell Cardiol 2011; 52:905-11. [PMID: 22210280 DOI: 10.1016/j.yjmcc.2011.12.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Revised: 12/09/2011] [Accepted: 12/10/2011] [Indexed: 01/17/2023]
Abstract
The coronary collateral circulation is critically important as an adaptation of the heart to prevent the damage from ischemic insults. In their native state, collaterals in the heart would be classified as part of the microcirculation, existing as arterial-arterial anastomotic connections in the range of 30 to 100 μM in diameter. However, these vessels also show a propensity to remodel into components of the macrocirculation and can become arteries larger than 1000 μM in diameter. This process of outward remodeling is critically important in the adaptation of the heart to ischemia because the resistance to blood flow is inversely related to the fourth power of the diameter of the vessel. Thus, an expansion of a vessel from 100 to 1000 μM would reduce resistance (in this part of the circuit) to a negligible amount and enable delivery of flow to the region at risk. Our goal in this review is to highlight the voids in understanding this adaptation to ischemia-the growth of the coronary collateral circulation. In doing so we discuss the controversies and unknown aspects of the causal factors that stimulate growth of the collateral circulation, the role of genetics, and the role of endogenous stem and progenitor cells in the context of the normal, physiological situation and under more pathological conditions of ischemic heart disease or with some of the underlying risk factors, e.g., diabetes. The major conclusion of this review is that there are many gaps in our knowledge of coronary collateral growth and this knowledge is critical before the potential of stimulating collateralization in the hearts of patients can be realized. This article is part of a Special Issue entitled "Coronary Blood Flow".
Collapse
Affiliation(s)
- William M Chilian
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio 44272, USA.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Directing migration of endothelial progenitor cells with applied DC electric fields. Stem Cell Res 2011; 8:38-48. [PMID: 22099019 PMCID: PMC3238468 DOI: 10.1016/j.scr.2011.08.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Revised: 07/11/2011] [Accepted: 08/05/2011] [Indexed: 12/22/2022] Open
Abstract
Naturally-occurring, endogenous electric fields (EFs) have been detected at skin wounds, damaged tissue sites and vasculature. Applied EFs guide migration of many types of cells, including endothelial cells to migrate directionally. Homing of endothelial progenitor cells (EPCs) to an injury site is important for repair of vasculature and also for angiogenesis. However, it has not been reported whether EPCs respond to applied EFs. Aiming to explore the possibility to use electric stimulation to regulate the progenitor cells and angiogenesis, we tested the effects of direct-current (DC) EFs on EPCs. We first used immunofluorescence to confirm the expression of endothelial progenitor markers in three lines of EPCs. We then cultured the progenitor cells in EFs. Using time-lapse video microscopy, we demonstrated that an applied DC EF directs migration of the EPCs toward the cathode. The progenitor cells also align and elongate in an EF. Inhibition of vascular endothelial growth factor (VEGF) receptor signaling completely abolished the EF-induced directional migration of the progenitor cells. We conclude that EFs are an effective signal that guides EPC migration through VEGF receptor signaling in vitro. Applied EFs may be used to control behaviors of EPCs in tissue engineering, in homing of EPCs to wounds and to an injury site in the vasculature.
Collapse
|
24
|
Abstract
Progenitor cells mobilized from the bone marrow are recruited to ischemic tissues and increase neovascularization. Cell therapy is a promising new therapeutic option for treating patients with ischemic disorders. The efficiency of cell therapy to augment recovery after ischemia depends on the sufficient recruitment and engraftment of the cells to the target tissue. Homing to sites of active neovascularization is a complex process depending on a timely and spatially orchestrated interplay between chemokines, chemokine receptors, adhesion molecules (selectins and integrins), and intracellular signaling cascades, including also oxidative signaling. This review will focus on the homing mechanisms of progenitor and stem cells to ischemic tissues. Specifically, we discuss the role of chemokines and adhesion molecules such as selectins and integrins and the crosstalk between chemokines and integrins in progenitor cell homing.
Collapse
|
25
|
Foxc2 overexpression enhances benefit of endothelial progenitor cells for inhibiting neointimal formation by promoting CXCR4-dependent homing. J Vasc Surg 2011; 53:1668-78. [PMID: 21514778 DOI: 10.1016/j.jvs.2011.01.044] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2010] [Revised: 01/10/2011] [Accepted: 01/16/2011] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Endothelial progenitor cells (EPCs) are capable of enhancing re-endothelialization and attenuating neointimal formation. However, inefficient homing limits the therapeutic efficacy of EPCs transplantation. CXCR4 plays a critical role in regulating EPCs homing. Here, we studied the effect of Foxc2 overexpression on CXCR4 expression and the homing capacity of EPCs as well as the EPCs-mediated therapeutic benefit after artery injury. METHODS Bone marrow-derived EPCs were transfected with Foxc2 expression vector (Foxc2-EPCs) or empty control vector (Ctrl-EPCs) and examined 48 hours later. CXCR4 expression of EPCs was detected by flow cytometry and quantitative reverse transcriptase-polymerase chain reaction. The migration of EPCs toward SDF-1α was evaluated in a transwell migration assay, and the adhesion to fibronectin was determined using a static adhesion assay. For in vivo studies, EPCs were injected intravenously into the mice subjected to carotid injury. At 3 days after green fluorescent protein (GFP)/EPCs delivery, the recruited cells to the injury sites were detected by fluorescent microscopy. Re-endothelialization and neointimal formation were, respectively, assessed by Evans blue dye at 7 days and by the morphometric analysis for neointima and media area ratio (N/M) at 28 days after EPCs transfusion. RESULTS Foxc2 overexpression significantly increased the surface expression of CXCR4 on EPCs (about 1.9-fold of Ctrl-EPCs, P < .05). Foxc2-EPCs showed an increased migration toward SDF-1α (P < .05); Foxc2 overexpression increased also the adhesion capacity of EPCs (P < .05). In vivo, the number of recruited GFP cells was significantly higher in the mice transfused with Foxc2-GFP/EPCs compared with Ctrl-GFP/EPCs (about 2-fold of Ctrl-GFP/EPCs). The degree of re-endothelialization was higher in mice transfused with Foxc2-EPCs compared with Ctrl-EPCs (90.3% ± 1.6% vs 57.2% ± 1.3%; P < .05). Foxc2-EPCs delivery resulted in a greater inhibition of neointimal hyperplasia than Ctrl-EPCs administration (N/M: 0.38 ± 0.03 vs 0.67 ± 0.05, P < .05). Preincubation with CXCR4-Ab, AMD3100, or LY294002 significantly attenuated the enhanced in vitro and in vivo effects of Foxc2-EPCs. CONCLUSIONS Our findings indicate that Foxc2 overexpression increases CXCR4 expression of EPCs and efficiently enhances the homing potential of EPCs, thereby improving EPCs-mediated therapeutic benefit after endothelial injury. Foxc2 may be a novel molecular target for improving the therapeutic efficacy of EPCs transplantation.
Collapse
|
26
|
Baragli A, Ghè C, Arnoletti E, Granata R, Ghigo E, Muccioli G. Acylated and unacylated ghrelin attenuate isoproterenol-induced lipolysis in isolated rat visceral adipocytes through activation of phosphoinositide 3-kinase γ and phosphodiesterase 3B. Biochim Biophys Acta Mol Cell Biol Lipids 2011; 1811:386-96. [PMID: 21435395 DOI: 10.1016/j.bbalip.2011.03.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Revised: 03/02/2011] [Accepted: 03/04/2011] [Indexed: 11/30/2022]
Abstract
The acylated peptide ghrelin (AG) and its endogenous non-acylated isoform (UAG) protect cardiomyocytes, pancreatic β-cells, and preadipocytes from apoptosis, and induce preadipocytes differentiation into adipocytes. These events are mediated by AG and UAG binding to a still unidentified receptor, which determines the activation of phosphoinositide 3-kinase (PI3K), protein kinase B (AKT), and mitogen-activated protein kinase (MAPK) ERK1/2. AG and UAG also possess antilipolytic activity in vitro, but the underlying mechanism remains unknown. Thus, the objective of the current study was to characterize the molecular events involved in AG/UAG receptor signaling cascade. We treated rat primary visceral adipocytes with isoproterenol (ISO) and forskolin (FSK) to stimulate lipolysis, simultaneously incubating them with or without AG or UAG. Both peptides blocked ISO- and FSK-induced lipolysis. By direct measurement of cAMP intracellular content, we demonstrated that AG/UAG effect was associated to a reduction of ISO-induced cAMP accumulation. Moreover, the cAMP analog 8Br-cAMP abolished AG/UAG effect. As AG and UAG were ineffective against lipolysis induced by db-cAMP, another poorly hydrolyzable cAMP analog, phosphodiesterase (PDE) involvement was hypothesized. Indeed, cilostamide, a specific PDE3B inhibitor, blocked AG/UAG effect on ISO-induced lipolysis. Furthermore, the PI3K inhibitor wortmannin and AKT inhibitor 1,3-dihydro-1-(1-((4-(6-phenyl-1H-imidazo(4,5-g)quinoxalin-7-yl)phenyl)methyl)-4piperidinyl)-2H-benzimidazol-2-one trifluoroacetate also blocked AG/UAG action, suggesting a role in PDE3B activation. In particular, PI3K isoenzyme gamma (PI3Kγ) selective inhibition through the compound AS605240 prevented AG/UAG effect on ISO-stimulated lipolysis, hampering AKT phosphorylation on Ser(473). Taken together, these data demonstrate for the first time that AG/UAG attenuation of ISO-induced lipolysis involves PI3Kγ/AKT and PDE3B.
Collapse
Affiliation(s)
- Alessandra Baragli
- Department of Anatomy, Pharmacology, and Forensic Medicine, Division of Medical Pharmacology, University of Torino, Via P. Giuria 13, 10125 Torino, Italy
| | | | | | | | | | | |
Collapse
|
27
|
Wilson LS, Baillie GS, Pritchard LM, Umana B, Terrin A, Zaccolo M, Houslay MD, Maurice DH. A phosphodiesterase 3B-based signaling complex integrates exchange protein activated by cAMP 1 and phosphatidylinositol 3-kinase signals in human arterial endothelial cells. J Biol Chem 2011; 286:16285-96. [PMID: 21393242 DOI: 10.1074/jbc.m110.217026] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Enzymes of the phosphodiesterase 3 (PDE3) and PDE4 families each regulate the activities of both protein kinases A (PKAs) and exchange proteins activated by cAMP (EPACs) in cells of the cardiovascular system. At present, the mechanisms that allow selected PDEs to individually regulate the activities of these two effectors are ill understood. The objective of this study was to determine how a specific PDE3 variant, namely PDE3B, interacts with and regulates EPAC1-based signaling in human arterial endothelial cells (HAECs). Using several biochemical approaches, we show that PDE3B and EPAC1 bind directly through protein-protein interactions. By knocking down PDE3B expression or by antagonizing EPAC1 binding with PDE3B, we show that PDE3B regulates cAMP binding by its tethered EPAC1. Interestingly, we also show that PDE3B binds directly to p84, a PI3Kγ regulatory subunit, and that this interaction allows PI3Kγ recruitment to the PDE3B-EPAC1 complex. Of potential cardiovascular importance, we demonstrate that PDE3B-tethered EPAC1 regulates HAEC PI3Kγ activity and that this allows dynamic cAMP-dependent regulation of HAEC adhesion, spreading, and tubule formation. We identify and molecularly characterize a PDE3B-based "signalosome" that integrates cAMP- and PI3Kγ-encoded signals and show how this signal integration regulates HAEC functions of importance in angiogenesis.
Collapse
Affiliation(s)
- Lindsay S Wilson
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Schmidt S, Nakchbandi I, Ruppert R, Kawelke N, Hess MW, Pfaller K, Jurdic P, Fässler R, Moser M. Kindlin-3-mediated signaling from multiple integrin classes is required for osteoclast-mediated bone resorption. ACTA ACUST UNITED AC 2011; 192:883-97. [PMID: 21357746 PMCID: PMC3051823 DOI: 10.1083/jcb.201007141] [Citation(s) in RCA: 133] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Loss of kindlin-3 impairs activation of β1, β2, and β3 integrin classes, resulting in osteopetrotic defects in osteoclast adhesion and spreading. The blood cell–specific kindlin-3 protein is required to activate leukocyte and platelet integrins. In line with this function, mutations in the KINDLIN-3 gene in man cause immunodeficiency and severe bleeding. Some patients also suffer from osteopetrosis, but the underlying mechanism leading to abnormal bone turnover is unknown. Here we show that kindlin-3–deficient mice develop severe osteopetrosis because of profound adhesion and spreading defects in bone-resorbing osteoclasts. Mechanistically, loss of kindlin-3 impairs the activation of β1, β2, and β3 integrin classes expressed on osteoclasts, which in turn abrogates the formation of podosomes and sealing zones required for bone resorption. In agreement with these findings, genetic ablation of all integrin classes abolishes the development of podosomes, mimicking kindlin-3 deficiency. Although loss of single integrin classes gives rise to podosomes, their resorptive activity is impaired. These findings show that osteoclasts require their entire integrin repertoire to be regulated by kindlin-3 to orchestrate bone homeostasis.
Collapse
Affiliation(s)
- Sarah Schmidt
- Max Planck Institute of Biochemistry, D-82152 Martinsried, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Spinetti G, Fortunato O, Cordella D, Portararo P, Kränkel N, Katare R, Sala-Newby GB, Richer C, Vincent MP, Alhenc-Gelas F, Tonolo G, Cherchi S, Emanueli C, Madeddu P. Tissue kallikrein is essential for invasive capacity of circulating proangiogenic cells. Circ Res 2011; 108:284-93. [PMID: 21164105 PMCID: PMC3596779 DOI: 10.1161/circresaha.110.236786] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Accepted: 12/06/2010] [Indexed: 01/20/2023]
Abstract
RATIONALE Homing of proangiogenic cells (PACs) is guided by chemoattractants and requires proteases to disrupt the extracellular matrix. The possibility that PAC recruitment involves an interaction between proteases and chemotactic factor receptors remains largely unexplored. OBJECTIVE To determine the role of human tissue kallikrein (hK1) in PAC invasion and its dependency on kinin receptor signaling. METHODS AND RESULTS Human mononuclear cells (MNCs) and culture-selected PACs express and release mature hK1 protein. HK1 gene (KLK1) silencing reduced PACs migratory, invasive, and proangiogenic activities. KLK1-knockout mouse bone marrow-derived MNCs showed similar impairments and were unable to support reparative angiogenesis in a mouse model of peripheral ischemia. Conversely, adenovirus-mediated KLK1 (Ad.KLK1) gene transfer enhanced PAC-associated functions, whereas the catalytically inactive variant R53H-KLK1 was ineffective. HK1-induced effects are mediated by a kinin B(2) receptor (B(2)R)-dependent mechanism involving inducible nitric oxide synthase and metalloproteinase-2 (MMP2). Lower hK1 protein levels were observed in PACs from type 2 diabetic (T2D) patients, whereas KLK1 mRNA levels were similar to those of healthy subjects, suggesting a post-transcriptional defect. Furthermore, B(2)R is normally expressed on T2D-PACs but remains uncoupled from downstream signaling. Importantly, whereas Ad.KLK1 alone could not restore T2D-PAC invasion capacity, combined KLK1 and B(2)R expression rescued the diabetic phenotype. CONCLUSIONS This study reveals new interactive components of the PACs invasive machinery, acting via protease- and kinin receptor-dependent mechanisms.
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Case-Control Studies
- Cell Movement/physiology
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Down-Regulation
- Female
- Hindlimb/blood supply
- Humans
- Ischemia/metabolism
- Kallikreins/genetics
- Kallikreins/metabolism
- Kinins/metabolism
- Leukocytes, Mononuclear/cytology
- Leukocytes, Mononuclear/metabolism
- Male
- Matrix Metalloproteinase 2/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Middle Aged
- Models, Animal
- Neovascularization, Physiologic/physiology
- Nitric Oxide Synthase/metabolism
- RNA, Messenger/metabolism
- Receptor, Bradykinin B2/metabolism
- Signal Transduction/physiology
Collapse
Affiliation(s)
- Gaia Spinetti
- Chair of Experimental Cardiovascular Medicine, University of Bristol, Bristol BS28HW, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
|
31
|
Siragusa M, Katare R, Hirsch E, Madeddu P. Response to the Letter by Seropian et al. Circ Res 2010. [DOI: 10.1161/circresaha.110.225185] [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: 11/16/2022]
Affiliation(s)
- Mauro Siragusa
- Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, United Kingdom
| | - Rajesh Katare
- Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, United Kingdom
| | - Emilio Hirsch
- Department of Genetics, Biology, and Biochemistry, University of Turin, Italy
| | - Paolo Madeddu
- Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, United Kingdom,
| |
Collapse
|
32
|
Stellos K, Langer H, Gnerlich S, Panagiota V, Paul A, Schönberger T, Ninci E, Menzel D, Mueller I, Bigalke B, Geisler T, Bültmann A, Lindemann S, Gawaz M. Junctional Adhesion Molecule A Expressed on Human CD34
+
Cells Promotes Adhesion on Vascular Wall and Differentiation Into Endothelial Progenitor Cells. Arterioscler Thromb Vasc Biol 2010; 30:1127-36. [DOI: 10.1161/atvbaha.110.204370] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Objective—
To investigate the role of junctional adhesion molecule A (JAM-A) on adhesion and differentiation of human CD34
+
cells into endothelial progenitor cells.
Methods and Results—
Tissue healing and vascular regeneration is a multistep process requiring firm adhesion of circulating progenitor cells to the vascular wall and their further differentiation into endothelial cells. The role of JAM-A in platelet-mediated adhesion of progenitor cells was investigated by adhesion assays in vitro and with the help of intravital fluorescence microscopy in mice. Preincubation of human CD34
+
progenitor cells with soluble JAM-A-Fc (sJAM-A-Fc) resulted in significantly decreased adhesion over immobilized platelets or inflammatory endothelium under high shear stress in vitro and after carotid ligation in vivo or ischemia/reperfusion injury in the microcirculation of mice. Human CD34
+
cells express JAM-A, as defined by flow cytometry and Western blot analysis. JAM-A mediates differentiation of CD34
+
cells to endothelial progenitor cells and facilitates CD34
+
cell-induced reendothelialization in vitro. Pretreatment of human CD34
+
cells with sJAM-A-Fc resulted in increased neointima formation 3 weeks after endothelial denudation in the carotid arteries of nonobese diabetic/severe combined immunodeficient mice.
Conclusion—
These results indicate that the expression of JAM-A on CD34
+
cells mediates adhesion to the vascular wall after injury and differentiation into endothelial progenitor cells, a mechanism potentially involved in vascular regeneration. Human CD34
+
cells express JAM-A, mediating their interaction with platelets and endothelial cells. Specifically, JAM-A expressed on human CD34
+
progenitor cells regulates their adhesion over immobilized platelets or inflammatory endothelium under high shear stress in vitro and after carotid ligation in vivo or ischemia/reperfusion injury in the microcirculation of mice. Moreover, it mediates differentiation of CD34
+
cells to endothelial progenitor cells and facilitates reendothelialization.
Collapse
Affiliation(s)
- Konstantinos Stellos
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (K.S., H.L., S.G., V.P., A.P., T.S., E.N., I.M., B.B., T.G., A.B., S.L., M.G.), Eberhard Karls-Universität Tübingen, Tübingen, Germany; and Zentrum für Klinische Transfusionsmedizin GmbH (D.M.), Tübingen, Germany
| | - Harald Langer
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (K.S., H.L., S.G., V.P., A.P., T.S., E.N., I.M., B.B., T.G., A.B., S.L., M.G.), Eberhard Karls-Universität Tübingen, Tübingen, Germany; and Zentrum für Klinische Transfusionsmedizin GmbH (D.M.), Tübingen, Germany
| | - Stephan Gnerlich
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (K.S., H.L., S.G., V.P., A.P., T.S., E.N., I.M., B.B., T.G., A.B., S.L., M.G.), Eberhard Karls-Universität Tübingen, Tübingen, Germany; and Zentrum für Klinische Transfusionsmedizin GmbH (D.M.), Tübingen, Germany
| | - Victoria Panagiota
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (K.S., H.L., S.G., V.P., A.P., T.S., E.N., I.M., B.B., T.G., A.B., S.L., M.G.), Eberhard Karls-Universität Tübingen, Tübingen, Germany; and Zentrum für Klinische Transfusionsmedizin GmbH (D.M.), Tübingen, Germany
| | - Angela Paul
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (K.S., H.L., S.G., V.P., A.P., T.S., E.N., I.M., B.B., T.G., A.B., S.L., M.G.), Eberhard Karls-Universität Tübingen, Tübingen, Germany; and Zentrum für Klinische Transfusionsmedizin GmbH (D.M.), Tübingen, Germany
| | - Tanja Schönberger
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (K.S., H.L., S.G., V.P., A.P., T.S., E.N., I.M., B.B., T.G., A.B., S.L., M.G.), Eberhard Karls-Universität Tübingen, Tübingen, Germany; and Zentrum für Klinische Transfusionsmedizin GmbH (D.M.), Tübingen, Germany
| | - Elena Ninci
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (K.S., H.L., S.G., V.P., A.P., T.S., E.N., I.M., B.B., T.G., A.B., S.L., M.G.), Eberhard Karls-Universität Tübingen, Tübingen, Germany; and Zentrum für Klinische Transfusionsmedizin GmbH (D.M.), Tübingen, Germany
| | - Dagmar Menzel
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (K.S., H.L., S.G., V.P., A.P., T.S., E.N., I.M., B.B., T.G., A.B., S.L., M.G.), Eberhard Karls-Universität Tübingen, Tübingen, Germany; and Zentrum für Klinische Transfusionsmedizin GmbH (D.M.), Tübingen, Germany
| | - Iris Mueller
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (K.S., H.L., S.G., V.P., A.P., T.S., E.N., I.M., B.B., T.G., A.B., S.L., M.G.), Eberhard Karls-Universität Tübingen, Tübingen, Germany; and Zentrum für Klinische Transfusionsmedizin GmbH (D.M.), Tübingen, Germany
| | - Boris Bigalke
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (K.S., H.L., S.G., V.P., A.P., T.S., E.N., I.M., B.B., T.G., A.B., S.L., M.G.), Eberhard Karls-Universität Tübingen, Tübingen, Germany; and Zentrum für Klinische Transfusionsmedizin GmbH (D.M.), Tübingen, Germany
| | - Tobias Geisler
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (K.S., H.L., S.G., V.P., A.P., T.S., E.N., I.M., B.B., T.G., A.B., S.L., M.G.), Eberhard Karls-Universität Tübingen, Tübingen, Germany; and Zentrum für Klinische Transfusionsmedizin GmbH (D.M.), Tübingen, Germany
| | - Andreas Bültmann
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (K.S., H.L., S.G., V.P., A.P., T.S., E.N., I.M., B.B., T.G., A.B., S.L., M.G.), Eberhard Karls-Universität Tübingen, Tübingen, Germany; and Zentrum für Klinische Transfusionsmedizin GmbH (D.M.), Tübingen, Germany
| | - Stephan Lindemann
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (K.S., H.L., S.G., V.P., A.P., T.S., E.N., I.M., B.B., T.G., A.B., S.L., M.G.), Eberhard Karls-Universität Tübingen, Tübingen, Germany; and Zentrum für Klinische Transfusionsmedizin GmbH (D.M.), Tübingen, Germany
| | - Meinrad Gawaz
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (K.S., H.L., S.G., V.P., A.P., T.S., E.N., I.M., B.B., T.G., A.B., S.L., M.G.), Eberhard Karls-Universität Tübingen, Tübingen, Germany; and Zentrum für Klinische Transfusionsmedizin GmbH (D.M.), Tübingen, Germany
| |
Collapse
|
33
|
Bouvard C, Gafsou B, Dizier B, Galy-Fauroux I, Lokajczyk A, Boisson-Vidal C, Fischer AM, Helley D. alpha6-integrin subunit plays a major role in the proangiogenic properties of endothelial progenitor cells. Arterioscler Thromb Vasc Biol 2010; 30:1569-75. [PMID: 20508204 DOI: 10.1161/atvbaha.110.209163] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Alpha6 integrin subunit (alpha6) expression is increased by proangiogenic growth factors such as vascular endothelial growth factor (VEGF) and fibroblast growth factor. This increase correlates with enhanced in vitro tube formation by endothelial cells and their progenitors called endothelial colony-forming cells (ECFCs). We thus studied the role of alpha6 in vasculogenesis induced by human ECFCs, in a mouse model of hindlimb ischemia. METHODS AND RESULTS We used small interfering RNA (siRNA) to inhibit alpha6 expression on the surface of ECFCs. For in vivo studies, human ECFCs were injected intravenously into a nude mouse model of unilateral hind limb ischemia. Transfection with siRNA alpha6 abrogated neovessel formation and reperfusion of the ischemic hind limb induced by ECFCs (P<0.01 and P<0.001, respectively). It also inhibited ECFC incorporation into the vasculature of the ischemic muscle (P<0.001). In vitro, siRNA alpha6 inhibited ECFC adhesion (P<0.01), pseudotube formation on Matrigel, migration, and AKT phosphorylation (P<0.0001), with no effect on cell proliferation or apoptosis. CONCLUSIONS alpha6 Expression is required for ECFC migration, adhesion, recruitment at the site of ischemia, and the promotion of the postischemic vascular repair. Thus, we have demonstrated a major role of alpha6 in the proangiogenic properties of ECFCs.
Collapse
|
34
|
Assmus B, Tonn T, Seeger FH, Yoon CH, Leistner D, Klotsche J, Schächinger V, Seifried E, Zeiher AM, Dimmeler S. Red blood cell contamination of the final cell product impairs the efficacy of autologous bone marrow mononuclear cell therapy. J Am Coll Cardiol 2010; 55:1385-94. [PMID: 20338501 DOI: 10.1016/j.jacc.2009.10.059] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2009] [Revised: 09/30/2009] [Accepted: 10/14/2009] [Indexed: 12/26/2022]
Abstract
OBJECTIVES The aim of this study was to identify an association between the quality and functional activity of bone marrow-derived progenitor cells (BMCs) used for cardiovascular regenerative therapies and contractile recovery in patients with acute myocardial infarction included in the placebo-controlled REPAIR-AMI (Reinfusion of Enriched Progenitor cells And Infarct Remodeling in Acute Myocardial Infarction) trial. BACKGROUND Isolation procedures of autologous BMCs might affect cell functionality and therapeutic efficacy. METHODS Quality of cell isolation was assessed by measuring the total number of isolated BMCs, CD34+ and CD133+ cells, their colony-forming unit (CFU) and invasion capacity, cell viability, and contamination of the final BMC preparation with thrombocytes and red blood cells (RBCs). RESULTS The number of RBCs contaminating the final cell product significantly correlated with reduced recovery of left ventricular ejection fraction 4 months after BMC therapy (p = 0.007). Higher numbers of RBCs in the BMC preparation were associated with reduced BMC viability (r = -0.23, p = 0.001), CFU capacity (r = -0.16, p = 0.03), and invasion capacity (r = -0.27, p < 0.001). To assess a causal role for RBC contamination, we coincubated isolated BMCs with RBCs for 24 h in vitro. The addition of RBCs dose-dependently abrogated migratory capacity (p = 0.003) and reduced CFU capacity (p < 0.05) of isolated BMCs. Neovascularization capacity was significantly impaired after infusion of BMCs contaminated with RBCs, compared with BMCs alone (p < 0.05). Mechanistically, the addition of RBCs was associated with a profound reduction in mitochondrial membrane potential of BMCs. CONCLUSIONS Contaminating RBCs affects the functionality of isolated BMCs and determines the extent of left ventricular ejection fraction recovery after intracoronary BMC infusion in patients with acute myocardial infarction. These results suggest a bioactivity response relationship very much like a dose-response relationship in drug trials. (Reinfusion of Enriched Progenitor cells and Infarct Remodeling in Acute Myocardial Infarction [REPAIR-AMI]; NCT00279175).
Collapse
Affiliation(s)
- Birgit Assmus
- Cardiology, Department of Medicine III, Goethe University, Frankfurt, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Carretero-Ortega J, Walsh CT, Hernández-García R, Reyes-Cruz G, Brown JH, Vázquez-Prado J. Phosphatidylinositol 3,4,5-triphosphate-dependent Rac exchanger 1 (P-Rex-1), a guanine nucleotide exchange factor for Rac, mediates angiogenic responses to stromal cell-derived factor-1/chemokine stromal cell derived factor-1 (SDF-1/CXCL-12) linked to Rac activation, endothelial cell migration, and in vitro angiogenesis. Mol Pharmacol 2010; 77:435-42. [PMID: 20018810 PMCID: PMC3202486 DOI: 10.1124/mol.109.060400] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2009] [Accepted: 12/17/2009] [Indexed: 01/15/2023] Open
Abstract
Stromal cell-derived factor-1 (SDF-1/CXCL-12) and vascular endothelial growth factor (VEGF), which can be secreted by hypoxic tumors, promote the generation of new blood vessels. These potent angiogenic factors stimulate endothelial cell migration via the activation of Rho GTPases and the phosphatidylinositol 3-kinase (PI3K)/AKT signaling pathway. Thus, characterization of guanine nucleotide exchange factors critical in the angiogenic signaling cascades offers the possibility of identifying novel molecular targets. We demonstrated previously that mammalian target of rapamycin, an important effector and regulator of PI3K/AKT, activates phosphatidylinositol 3,4,5-triphosphate-dependent Rac exchanger 1 (P-Rex1), a Rac guanine nucleotide exchange factor identified as a target of G betagamma and PI3K, via direct interactions. In this study, we tested the hypothesis that P-Rex1 is involved in the angiogenic responses elicited by SDF-1 and VEGF. Using a knockdown approach, we demonstrate that P-Rex1 is indeed required for SDF-1 promoted signaling pathway, because there is decreased Rac activation, cell migration, and in vitro angiogenesis in P-Rex1 knockdown cells stimulated with SDF-1. In contrast, P-Rex1 knockdown does not affect responses to VEGF, and signaling to extracellular signal-regulated kinase in response to either angiogenic factor is not sensitive to P-Rex1 knockdown. We also demonstrate that in endothelial cells, VEGF promotes an increase in the expression of endogenous P-Rex1 and the SDF-1 receptor CXCR4, In addition, VEGF-pretreated cells show an increased migratory and angiogenic response to SDF-1, suggesting that VEGF stimulation can complement SDF-1/CXCR4 signaling to induce angiogenesis. We conclude that P-Rex1 is a key element in SDF-1-induced angiogenic responses and a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Jorge Carretero-Ortega
- Departments of Pharmacology (J.C.-O., R.H.-G., J.V.-P.) and Cell Biology (G.R.-C.), Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico, D.F., Mexico; and the Department of Pharmacology (C.T.W., J.H.B.), University of California, San Diego, California
| | - Colin T. Walsh
- Departments of Pharmacology (J.C.-O., R.H.-G., J.V.-P.) and Cell Biology (G.R.-C.), Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico, D.F., Mexico; and the Department of Pharmacology (C.T.W., J.H.B.), University of California, San Diego, California
| | - Ricardo Hernández-García
- Departments of Pharmacology (J.C.-O., R.H.-G., J.V.-P.) and Cell Biology (G.R.-C.), Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico, D.F., Mexico; and the Department of Pharmacology (C.T.W., J.H.B.), University of California, San Diego, California
| | - Guadalupe Reyes-Cruz
- Departments of Pharmacology (J.C.-O., R.H.-G., J.V.-P.) and Cell Biology (G.R.-C.), Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico, D.F., Mexico; and the Department of Pharmacology (C.T.W., J.H.B.), University of California, San Diego, California
| | - Joan Heller Brown
- Departments of Pharmacology (J.C.-O., R.H.-G., J.V.-P.) and Cell Biology (G.R.-C.), Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico, D.F., Mexico; and the Department of Pharmacology (C.T.W., J.H.B.), University of California, San Diego, California
| | - José Vázquez-Prado
- Departments of Pharmacology (J.C.-O., R.H.-G., J.V.-P.) and Cell Biology (G.R.-C.), Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico, D.F., Mexico; and the Department of Pharmacology (C.T.W., J.H.B.), University of California, San Diego, California
| |
Collapse
|
36
|
Siragusa M, Katare R, Meloni M, Damilano F, Hirsch E, Emanueli C, Madeddu P. Involvement of phosphoinositide 3-kinase gamma in angiogenesis and healing of experimental myocardial infarction in mice. Circ Res 2010; 106:757-68. [PMID: 20056919 DOI: 10.1161/circresaha.109.207449] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
RATIONALE Phosphoinositide 3-kinase (PI3K)gamma is expressed in hematopoietic cells, endothelial cells (ECs), and cardiomyocytes and regulates different cellular functions relevant to inflammation, tissue remodeling and cicatrization. Recently, PI3Kgamma inhibitors have been indicated for the treatment of chronic inflammatory/autoimmune diseases and atherosclerosis. OBJECTIVE We aimed to determine PI3Kgamma contribution to the angiogenic capacity of ECs and the effect of PI3Kgamma inhibition on healing of myocardial infarction (MI). METHODS AND RESULTS Human umbilical ECs were treated with a selective PI3Kgamma inhibitor, AS605240, or a pan-phosphoinositide 3-kinases inhibitor, LY294002. Both inhibitory treatments and small interfering RNA-mediated PI3Kgamma knockdown strongly impaired ECs angiogenic capacity, because of suppression of the PI3K/Akt and mitogen-activated protein kinase pathways. Constitutive activation of Akt rescued the angiogenic defect. Reparative angiogenesis was studied in vivo in a model of MI. AS605240 did not affect MI-induced PI3Kgamma upregulation, whereas it suppressed Akt activation and downstream signaling. AS605240 strongly reduced inflammation, enhanced cardiomyocyte apoptosis, and impaired survival and proliferation of ECs in peri-infarct zone, which resulted in defective reparative neovascularization. As a consequence, AS605240-treated MI hearts showed increased infarct size and impaired recovery of left ventricular function. Similarly, PI3Kgamma-deficient mice showed impaired reparative neovascularization, enhanced cardiomyocyte apoptosis and marked deterioration of cardiac function following MI. Mice expressing catalytically inactive PI3Kgamma also failed to mount a proper neovascularization, although cardiac dysfunction was similar to wild-type controls. CONCLUSIONS PI3Kgamma expression and catalytic activity are involved at different levels in reparative neovascularization and healing of MI.
Collapse
Affiliation(s)
- Mauro Siragusa
- Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Upper Maudlin Street, Bristol, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
37
|
Zhu G, Song M, Wang H, Zhao G, Yu Z, Yin Y, Zhao X, Huang L. Young environment reverses the declined activity of aged rat-derived endothelial progenitor cells: involvement of the phosphatidylinositol 3-kinase/Akt signaling pathway. Ann Vasc Surg 2009; 23:519-34. [PMID: 19540437 DOI: 10.1016/j.avsg.2008.11.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2008] [Revised: 11/25/2008] [Accepted: 11/26/2008] [Indexed: 01/13/2023]
Abstract
BACKGROUND Although age-related impairment of endothelial progenitor cells (EPCs) has been documented in recent studies, the detailed role of aging-induced environment in EPCs remains unclear. METHODS Two and 20 months old Sprague-Dawley female rats were used in the present study. EPCs isolated from young (YEPCs) and aged (AEPCs) rats were cultured with young or aged serum. EPC migration and proliferation were detected with a modified Boyden chamber and the MTT assay, respectively; EPC differentiation was detected by reverse-transcription polymerase chain reaction or fluorescence-activated cell sorting; Akt and phosphorylated-Akt protein expression was detected with Western blotting. EPC transplantation was performed in the rat carotid artery injury models. RESULTS Young serum significantly promotes AEPC migration, proliferation, and differentiation and increases phosphatidylinositol 3-kinase (PI3-K) and endothelial nitric oxide synthase activity in AEPCs compared with aged serum; total-Akt and phosphorylated-Akt protein expressions in AEPCs are also significantly upregulated by young serum. Transplanted AEPC numbers at vascular injury sites in the young rat carotid artery injury model significantly increased compared with those in aged models. Those effects could be reasonably attenuated by the PI3-K-specific blocker wortmannin. CONCLUSION A young environment partly restores the declined AEPC activity and promotes AEPC homing to vascular injury sites; activation of the PI3-K/Akt signaling pathway is at least partly responsible for this process.
Collapse
Affiliation(s)
- Guangxu Zhu
- Department of Clinical Laboratory, Kunming General Hospital, Chengdu Military Area of PLA, Kunming, Yunnan Province, PR China.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Thymosin beta4 induces endothelial progenitor cell migration via PI3K/Akt/eNOS signal transduction pathway. J Cardiovasc Pharmacol 2009; 53:209-14. [PMID: 19247195 DOI: 10.1097/fjc.0b013e318199f326] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Thymosin beta4, a G-actin-sequestering peptide, has been shown to play an important role in cell migration. However, little is known about the effect of thymosin beta4 on circulating endothelial progenitor cell (EPC) directional migration, which is essential for EPC-mediated reendothelialization and neovascularization. In our study, using a transwell migration assay, we showed that thymosin beta4 induced EPC migration in a concentration-dependent manner. Western blot analysis revealed that treatment of EPCs with thymosin beta4 resulted in a time and concentration-dependent phosphorylation of Akt, endothelial nitric oxide synthase (eNOS), and extracellular signal-regulated kinase (ERK)1/2. Functional analysis showed that thymosin beta4-induced EPC migration was blocked by phosphatidylinositol 3-kinase inhibitors (LY294002 or wortmannin) or eNOS inhibitor (Nomega-nitro-L-arginine methyl ester) but was not significantly attenuated by mitogen-activated protein kinase (MAPK)/ERK inhibitor (PD98059). These findings suggest that thymosin beta4 stimulates EPC directional migration via phosphatidylinositol 3-kinase/Akt/eNOS, rather than via MAPK/ERK signal transduction pathway.
Collapse
|
39
|
Park DH, Eve DJ, Musso J, Klasko SK, Cruz E, Borlongan CV, Sanberg PR. Inflammation and Stem Cell Migration to the Injured Brain in Higher Organisms. Stem Cells Dev 2009; 18:693-702. [DOI: 10.1089/scd.2009.0008] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Dong-Hyuk Park
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery, University of South Florida, Tampa, Florida
- Department of Neurosurgery, Korea University Medical Center, Korea University, Seoul, Korea
| | - David J. Eve
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery, University of South Florida, Tampa, Florida
| | - James Musso
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery, University of South Florida, Tampa, Florida
| | | | - Eduardo Cruz
- Cryopraxis, CellPraxis, BioRio, Pólo de Biotecnologia do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cesario V. Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery, University of South Florida, Tampa, Florida
| | - Paul R. Sanberg
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery, University of South Florida, Tampa, Florida
| |
Collapse
|
40
|
Abstract
The diverse effects mediated by PI3K/PTEN (phosphoinositide 3-kinase/phosphatase and tensin homologue deleted on chromosome 10) signalling in the heart clearly support an important biological and pathophysiological role for this signalling cascade. PI3Ks are a family of evolutionarily conserved lipid kinases that mediate many cellular responses to physiological and pathophysiological stimuli. Class I PI3K can be activated by either receptor tyrosine kinase/cytokine receptor activation (class IA) or G-protein-coupled receptors (class IB), leading to the generation of phosphatidyl inositol (3,4,5)P3 and recruitment and activation of Akt/protein kinase B, 3'-phosphoinositide-dependent kinase-1 (PDK1), or monomeric G-proteins, and phosphorylation of a wide range of downstream targets including glycogen synthase kinase 3beta (GSK3beta), mTOR (mammalian target of rapamycin), p70S6 kinase, endothelial nitric oxide synthase, and several anti-apoptotic effectors. Class IA (PI3Kalpha, beta, and delta) and class IB (PI3Kgamma) PI3Ks mediate distinct phenotypes in the heart under negative control by the 3'-lipid phosphatase PTEN, which dephosphorylates PtdIns(3,4,5)P3 to generate PtdIns(4,5)P2. PI3Kalpha, PI3Kgamma, and PTEN are expressed in cardiomyocytes, fibroblasts, endothelial cells, and vascular smooth muscle cells, where they modulate cell survival, hypertrophy, contractility, metabolism, and mechanotransduction. The PI3K/PTEN signalling pathways are involved in a wide variety of diseases including myocardial hypertrophy and contractility, heart failure, and preconditioning. In this review, we discuss the signalling pathways mediated by PI3K class I isoforms and PTEN and their roles in cardiac structure and function.
Collapse
Affiliation(s)
- Gavin Y Oudit
- Division of Cardiology, Department of Medicine, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada T6G 2B7.
| | | |
Collapse
|
41
|
Scharner D, Rössig L, Carmona G, Chavakis E, Urbich C, Fischer A, Kang TB, Wallach D, Chiang YJ, Deribe YL, Dikic I, Zeiher AM, Dimmeler S. Caspase-8 is involved in neovascularization-promoting progenitor cell functions. Arterioscler Thromb Vasc Biol 2009; 29:571-8. [PMID: 19122169 DOI: 10.1161/atvbaha.108.182006] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Endothelial progenitor cells (EPCs) comprise a heterogeneous population of cells, which improve therapeutic neovascularization after ischemia. The neovascularization-promoting potential of progenitor cells depends on survival and retention of the infused cells to the tissue. Caspases mediate apoptosis but are also involved in other critical biological processes. Therefore, we aimed to address the role of caspases in proangiogenic cells. METHODS AND RESULTS The caspase-8 inhibitor zIETD abrogated the ex vivo formation of EPCs, inhibited EPC adhesion and migration, and reduced their capacity to improve neovascularization in vivo. Consistently, cells isolated from caspase-8-deficient mice exhibited a reduced capacity for enhancing neovascularization when transplanted into mice after hindlimb ischemia. Because inhibition of Caspase-8 reduced the adhesion and homing functions of EPCs, we further determined the surface expression of integrins and receptors involved in cell recruitment to ischemic tissues. Pharmacological inhibition of caspase-8 and genetic depletion of caspase-8 reduced the expression of the fibronectin receptor subunits alpha5 and beta1 and the SDF-1 receptor CXCR4. Moreover, we identified the E3 ubiquitin ligase Cbl-b, which negatively regulates integrin and receptor-mediated signaling, as a potential Caspase-8 substrate. CONCLUSIONS In summary, our data demonstrate a novel apoptosis-unrelated role of caspase-8 in proangiogenic cells.
Collapse
Affiliation(s)
- Dörte Scharner
- Department of Internal Medicine III, University of Frankfurt, Frankfurt, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Morello F, Perino A, Hirsch E. Phosphoinositide 3-kinase signalling in the vascular system. Cardiovasc Res 2008; 82:261-71. [PMID: 19038971 DOI: 10.1093/cvr/cvn325] [Citation(s) in RCA: 167] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Phosphoinositide 3-kinases (PI3Ks) are protein and lipid kinases activated by different classes of membrane receptors, including G-protein coupled and tyrosine kinase receptors. Several lines of evidence have uncovered specific roles for distinct PI3K isoforms in the vascular system in both physiology and disease. The present review will summarize and discuss the most recent advances regarding PI3K-Akt signalling in endothelial cells, vascular smooth muscle cells, platelets, and inflammatory cells involved in the atherosclerotic process. Of interest, the development of novel isoform-selective PI3K inhibitor drugs offers a unique opportunity to selectively and differentially target PI3K-driven pathways in the vascular system and may give rise to new strategies for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Fulvio Morello
- Molecular Biotechnology Center, University of Torino, via Nizza 52, 10126 Torino, Italy
| | | | | |
Collapse
|
43
|
Kränkel N, Katare RG, Siragusa M, Barcelos LS, Campagnolo P, Mangialardi G, Fortunato O, Spinetti G, Tran N, Zacharowski K, Wojakowski W, Mroz I, Herman A, Manning Fox JE, MacDonald PE, Schanstra JP, Bascands JL, Ascione R, Angelini G, Emanueli C, Madeddu P. Role of kinin B2 receptor signaling in the recruitment of circulating progenitor cells with neovascularization potential. Circ Res 2008; 103:1335-43. [PMID: 18927465 PMCID: PMC2821015 DOI: 10.1161/circresaha.108.179952] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Reduced migratory function of circulating angiogenic progenitor cells (CPCs) has been associated with impaired neovascularization in patients with cardiovascular disease (CVD). Previous findings underline the role of the kallikrein-kinin system in angiogenesis. We now demonstrate the involvement of the kinin B2 receptor (B(2)R) in the recruitment of CPCs to sites of ischemia and in their proangiogenic action. In healthy subjects, B(2)R was abundantly present on CD133(+) and CD34(+) CPCs as well as cultured endothelial progenitor cells (EPCs) derived from blood mononuclear cells (MNCs), whereas kinin B1 receptor expression was barely detectable. In transwell migration assays, bradykinin (BK) exerts a potent chemoattractant activity on CD133(+) and CD34(+) CPCs and EPCs via a B(2)R/phosphoinositide 3-kinase/eNOS-mediated mechanism. Migration toward BK was able to attract an MNC subpopulation enriched in CPCs with in vitro proangiogenic activity, as assessed by Matrigel assay. CPCs from cardiovascular disease patients showed low B(2)R levels and decreased migratory capacity toward BK. When injected systemically into wild-type mice with unilateral limb ischemia, bone marrow MNCs from syngenic B(2)R-deficient mice resulted in reduced homing of sca-1(+) and cKit(+)flk1(+) progenitors to ischemic muscles, impaired reparative neovascularization, and delayed perfusion recovery as compared with wild-type MNCs. Similarly, blockade of the B(2)R by systemic administration of icatibant prevented the beneficial effect of bone marrow MNC transplantation. BK-induced migration represents a novel mechanism mediating homing of circulating angiogenic progenitors. Reduction of BK sensitivity in progenitor cells from cardiovascular disease patients might contribute to impaired neovascularization after ischemic complications.
Collapse
Affiliation(s)
- Nicolle Kränkel
- Experimental Cardiovascular Medicine, Bristol Heart Institute, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Urao N, Inomata H, Razvi M, Kim HW, Wary K, McKinney R, Fukai T, Ushio-Fukai M. Role of nox2-based NADPH oxidase in bone marrow and progenitor cell function involved in neovascularization induced by hindlimb ischemia. Circ Res 2008; 103:212-20. [PMID: 18583711 PMCID: PMC2711765 DOI: 10.1161/circresaha.108.176230] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Bone marrow (BM) is the major reservoir for endothelial progenitor cells (EPCs). Postnatal neovascularization depends on not only angiogenesis but also vasculogenesis, which is mediated through mobilization of EPCs from BM and their recruitment to the ischemic sites. Reactive oxygen species (ROS) derived from Nox2-based NADPH oxidase play an important role in postnatal neovascularization; however, their role in BM and EPC function is unknown. Here we show that hindlimb ischemia of mice significantly increases Nox2 expression and ROS production in BM-mononuclear cells (BMCs), which is associated with an increase in circulating EPC-like cells. Mice lacking Nox2 show reduction of ischemia-induced flow recovery, ROS levels in BMCs, as well as EPC mobilization from BM. Transplantation of wild-type (WT)-BM into Nox2-deficient mice rescues the defective neovascularization, whereas WT mice transplanted with Nox2-deficient BM show reduced flow recovery and capillary density compared to WT-BM transplanted control. Intravenous infusion of WT- and Nox2-deficient BMCs into WT mice reveals that neovascularization and homing capacity are impaired in Nox2-deficient BMCs in vivo. In vitro, Nox2-deficient c-kit+Lin- BM stem/progenitor cells show impaired chemotaxis and invasion as well as polarization of actins in response to stromal derived factor (SDF), which is associated with blunted SDF-1-mediated phosphorylation of Akt. In conclusion, Nox2-derived ROS in BM play a critical role in mobilization, homing, and angiogenic capacity of EPCs and BM stem/progenitor cells, thereby promoting revascularization of ischemic tissue. Thus, NADPH oxidase in BM and EPCs is potential therapeutic targets for promoting neovascularization in ischemic cardiovascular diseases.
Collapse
Affiliation(s)
- Norifumi Urao
- Department of Pharmacology, Center for Lung and Vascular Biology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
|