151
|
Endothelial progenitors as tools to study vascular disease. Stem Cells Int 2012; 2012:346735. [PMID: 22550504 PMCID: PMC3329655 DOI: 10.1155/2012/346735] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Accepted: 01/09/2012] [Indexed: 12/23/2022] Open
Abstract
Endothelial progenitor cells (EPCs) have great clinical value because they can be used as diagnostic biomarkers and as a cellular therapy for promoting vascular repair of ischaemic tissues. However, EPCs also have an additional research value in vascular disease modelling to interrogate human disease mechanisms. The term EPC is used to describe a diverse variety of cells, and we have identified a specific EPC subtype called outgrowth endothelial cell (OEC) as the best candidate for vascular disease modelling because of its high-proliferative potential and unambiguous endothelial commitment. OECs are isolated from human blood and can be exposed to pathologic conditions (forward approach) or be isolated from patients (reverse approach) in order to study vascular human disease. The use of OECs for modelling vascular disease will contribute greatly to improving our understanding of endothelial pathogenesis, which will potentially lead to the discovery of novel therapeutic strategies for vascular diseases.
Collapse
|
152
|
Tuder RM, Davis LA, Graham BB. Targeting energetic metabolism: a new frontier in the pathogenesis and treatment of pulmonary hypertension. Am J Respir Crit Care Med 2012; 185:260-6. [PMID: 22077069 PMCID: PMC3297113 DOI: 10.1164/rccm.201108-1536pp] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 10/26/2011] [Indexed: 01/01/2023] Open
Abstract
This perspective highlights advances in the understanding of the role of cellular metabolism in the pathogenesis of pulmonary hypertension. Insights gained in the past 20 years have revealed several similarities between the cellular processes underlying the pulmonary vascular remodeling in pulmonary hypertension and those seen in cancer processes. In line with these insights, there is increasing recognition that abnormal cellular metabolism, notably of aerobic glycolysis (the "Warburg effect"), the potential involvement of hypoxia-inducible factor in this process, and alterations in mitochondrial function, are key elements in the pathogenesis of this disease. The glycolytic shift may underlie the resistance to apoptosis and increased vascular cell proliferation, which are hallmarks of pulmonary hypertension. These investigations have led to novel approaches in the diagnosis and therapy of pulmonary hypertension.
Collapse
Affiliation(s)
- Rubin M Tuder
- Program in Translational Lung Research, Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, USA.
| | | | | |
Collapse
|
153
|
Suen C, Mei SH, Stewart DJ. Cell Therapy for Pulmonary Arterial Hypertension: Potential Efficacy of Endothelial Progenitor Cells and Mesenchymal Stem Cells. ACTA ACUST UNITED AC 2012. [DOI: 10.21693/1933-088x-11.1.33] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Pulmonary arterial hypertension (PAH) presents a challenging problem for health care providers, as effective long-term therapies have been elusive. An emerging paradigm for the pathogenesis of PAH is that endothelial cell injury and apoptosis at the level of the precapillary arteriole could be the initiating event in the pathogenesis of this disease. This hypothesis has spurred research on novel regenerative approaches using stem and progenitor cells. In this review, we compare findings from the latest preclinical and clinical studies using endothelial progenitor cell (EPC) and mesenchymal stem cell (MSC) therapy to treat PAH. Additionally, we highlight recent advances in gene-enhanced cell therapy, an approach that promises to augment the therapeutic potential of EPCs and MSCs especially for the reversal of established PAH. These new regenerative approaches have shown great promise in preclinical studies; however, large, rigorously designed clinical studies will be necessary to establish clinical efficacy.
Collapse
Affiliation(s)
- Colin Suen
- Sprott Centre for Stem Cell Research, The Ottawa Hospital, Research Institute & University of Ottawa, Ottawa, Ontario, Canada
| | - Shirley H.J. Mei
- Sprott Centre for Stem Cell Research, The Ottawa Hospital, Research Institute & University of Ottawa, Ottawa, Ontario, Canada
| | - Duncan J. Stewart
- Sprott Centre for Stem Cell Research, The Ottawa Hospital, Research Institute & University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
154
|
Liu X, Xie C. Human endothelial progenitor cells isolated from COPD patients are dysfunctional. Mol Cell Biochem 2011; 363:53-63. [PMID: 22139347 DOI: 10.1007/s11010-011-1157-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Accepted: 11/23/2011] [Indexed: 11/25/2022]
Abstract
Cardiovascular disease is the leading cause of morbidity and mortality in patients with moderate-to-severe chronic obstructive pulmonary disease (COPD). More than 44% of these patients present with generalized atherosclerosis at autopsy. It is accepted that endothelial progenitor cells (EPCs) participate in the repair of dysfunctional endothelium and thus protects against atherosclerosis. However, whether COPD affects the repairing capacity of EPCs is unknown. Therefore, the objective of this study was to determine whether and how EPCs are involved in the vascular repair process in patients with COPD. In our study, EPCs from 25 COPD and 16 control patients were isolated by Ficoll density-gradient centrifugation and identified using fluorescence activated cell sorting. Transwell Migratory Assay was performed to determine the number of EPC colony-forming units and the adherent capacity late-EPCs to human umbilical vein endothelial cells. Following arterial damage in NOD/SCID mice, the number of EPCs incorporated at the injured vascular site was determined using a fluorescence microscope. We found that the number of EPC clusters and cell migration, as well as the expression of CXCR4, was significantly decreased in patients with COPD. Additionally, the number of late-EPCs adherent to HUVEC tubules was significantly reduced, and fewer VEGFR2(+)-staining cells were incorporated into the injured site in COPD patients. Our study demonstrates that EPC capacity of repair was affected in COPD patients, which may contribute to altered vascular endothelium in this patient population.
Collapse
Affiliation(s)
- Xiaoran Liu
- Respiratory Department, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road, Guangzhou 51008, Guangdong Province, People's Republic of China.
| | | |
Collapse
|
155
|
Dahal BK, Heuchel R, Pullamsetti SS, Wilhelm J, Ghofrani HA, Weissmann N, Seeger W, Grimminger F, Schermuly RT. Hypoxic pulmonary hypertension in mice with constitutively active platelet-derived growth factor receptor-β. Pulm Circ 2011; 1:259-68. [PMID: 22034611 PMCID: PMC3198653 DOI: 10.4103/2045-8932.83448] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Platelet-derived growth factor (PDGF) has been implicated in the pathobiology of vascular remodeling. The multikinase inhibitor imatinib that targets PDGF receptor (PDGFR), c-kit and Abl kinases, shows therapeutic efficacy against experimental pulmonary hypertension (PH); however, the role of PDGFR-b in experimental PH has not been examined by genetic approach. We investigated the chronic hypoxia-induced PH in mice carrying an activating point mutation of PDGFR-β (D849N) and evaluated the therapeutic efficacy of imatinib. In addition, we studied pulmonary global gene expression and confirmed the expression of identified genes by immunohistochemistry. Chronically hypoxic D849N mice developed PH and strong pulmonary vascular remodeling that was improved by imatinib (100 mg/kg/day) as evident from the significantly reduced right ventricular systolic pressure, right ventricular hypertrophy and muscularization of peripheral pulmonary arteries. Global gene expression analysis revealed that stromal cell derived factor SDF)-1α was significantly upregulated, which was confirmed by immunohistochemistry. Moreover, an enhanced immunoreactivity for SDF-1α, PDGFR-β and CXCR4, the receptor for SDF-1α was localized to the α-smooth muscle cell (SMC) actin positive pulmonary vascular cells in hypoxic mice and patients with idiopathic pulmonary arterial hypertension (IPAH). In conclusion, our findings substantiate the major role of PDGFR activation in pulmonary vascular remodeling by a genetic approach. Immunohistochemistry findings suggest a role for SDF-1α/CXCR4 axis in pulmonary vascular remodeling and point to a potential interaction between the chemokine SDF-1 and the growth factor PDGF signaling. Future studies designed to elucidate an interaction between the chemokine SDF-1 and the PDGF system may uncover novel therapeutic targets.
Collapse
Affiliation(s)
- Bhola K Dahal
- University of Giessen Lung Centre (UGLC), Giessen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
156
|
Smadja DM, Duong-van-Huyen JP, Dal Cortivo L, Blanchard A, Bruneval P, Emmerich J, Gaussem P. Early endothelial progenitor cells in bone marrow are a biomarker of cell therapy success in patients with critical limb ischemia. Cytotherapy 2011; 14:232-9. [PMID: 22040109 DOI: 10.3109/14653249.2011.627917] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND AIMS Endothelial progenitor cells (EPC) have been proposed for autologous angiogenic therapy. The objectives of this study were to quantify EPC in the peripheral blood and bone marrow mononuclear cells (BM-MNC) of patients with critical limb ischemia that had received BM-MNC as a cell therapy product, and to study the putative relationship between the presence of EPC and the process of neovascularization in toe or transmetatarsal amputation specimens. METHODS Early and late endothelial progenitor cells (CFU-EC and ECFC) were cultivated and quantified according to published methods in peripheral blood and BM-MNC from patients with critical limb ischemia (CLI; n = 11) enrolled in the OPTIPEC trial ( http://clinicaltrials.gov/ct2/show/NCT00377897 ) to receive BM-MNC as a cell therapy product. RESULTS Eight out of the 11 patients had undergone amputations. Three of the patients displayed a neoangiogenic process that was associated with a higher number of CFU-EC in BM-MNC, while CD3+ , CFU-GM and CD34+ in BM-MNC, and EPC in peripheral blood, did not correlate with the appearance of newly formed vessels. As expected, circulating CFU-EC and ECFC counts were significantly lower in CLI patients compared with age-matched controls. CONCLUSIONS In patients with critical limb ischemia, EPC in peripheral blood were decreased compared with healthy individuals. However, in BM-MNC we found that relative numbers of CFU-EC could be used as an indicator to discriminate patients with neoangiogenic processes. These results need to be confirmed in a randomized study.
Collapse
Affiliation(s)
- David M Smadja
- Université Paris Descartes, Paris, France Sorbonne Paris Cite, France.
| | | | | | | | | | | | | |
Collapse
|
157
|
Courboulin A, Tremblay VL, Barrier M, Meloche J, Jacob MH, Chapolard M, Bisserier M, Paulin R, Lambert C, Provencher S, Bonnet S. Krüppel-like factor 5 contributes to pulmonary artery smooth muscle proliferation and resistance to apoptosis in human pulmonary arterial hypertension. Respir Res 2011; 12:128. [PMID: 21951574 PMCID: PMC3193170 DOI: 10.1186/1465-9921-12-128] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 09/27/2011] [Indexed: 12/31/2022] Open
Affiliation(s)
- Audrey Courboulin
- Department of Medicine, Faculty of Medicine, Laval University, Quebec QC, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
158
|
Schwarz J. Emerging role of c-kit+ progenitor cells in pulmonary hypertension. Am J Respir Crit Care Med 2011; 184:5-7. [PMID: 21737590 DOI: 10.1164/rccm.201104-0664ed] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
|
159
|
Jones CP, Rankin SM. Bone marrow-derived stem cells and respiratory disease. Chest 2011; 140:205-211. [PMID: 21729891 DOI: 10.1378/chest.10-2348] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Adult bone marrow contains a number of discrete populations of progenitor cells, including endothelial, mesenchymal, and epithelial progenitor cells and fibrocytes. In the context of a range of diseases, endothelial progenitor cells have been reported to promote angiogenesis, mesenchymal stem cells are potent immunosuppressors but can also contribute directly to tissue regeneration, and fibrocytes have been shown to induce tissue fibrosis. This article provides an overview of the basic biology of these different subsets of progenitor cells, reporting their distinct phenotypes and functional activities. The differences in their secretomes are highlighted, and the relative role of cellular differentiation vs paracrine effects of progenitor cells is considered. The article reviews the literature examining the contribution of progenitor cells to the pathogenesis of respiratory disease, and discusses recent studies using bone marrow progenitor cells as stem cell therapies in the context of pulmonary hypertension, COPD, and asthma.
Collapse
Affiliation(s)
- Carla P Jones
- Leukocyte Biology Section, National Heart and Lung Institute, Imperial College London, London, England
| | - Sara M Rankin
- Leukocyte Biology Section, National Heart and Lung Institute, Imperial College London, London, England.
| |
Collapse
|
160
|
Duong HT, Erzurum SC, Asosingh K. Pro-angiogenic hematopoietic progenitor cells and endothelial colony-forming cells in pathological angiogenesis of bronchial and pulmonary circulation. Angiogenesis 2011; 14:411-22. [PMID: 21796417 DOI: 10.1007/s10456-011-9228-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2011] [Accepted: 07/13/2011] [Indexed: 12/20/2022]
Abstract
Dysregulation of angiogenesis is a common feature of many disease processes. Vascular remodeling is believed to depend on the participation of endothelial progenitor cells, but the identification of endothelial progenitors in postnatal neovascularization remains elusive. Current understanding posits a role for circulating pro-angiogenic hematopoietic cells that interact with local endothelial cells to establish an environment that favors angiogenesis in physiologic and pathophysiologic responses. In the lung, increased and dysregulated angiogenesis is a hallmark of diseases of the bronchial and pulmonary circulations, manifested by asthma and pulmonary arterial hypertension (PAH), respectively. In asthma, T(Helper)-2 immune cells produce angiogenic factors that mobilize and recruit pro-inflammatory and pro-angiogenic precursors from the bone marrow into the airway wall where they induce angiogenesis and fuel inflammation. In contrast, in PAH, upregulation of hypoxia-inducible factor (HIF) in vascular cells leads to the production of bone marrow-mobilizing factors that recruit pro-angiogenic progenitor cells to the pulmonary circulation where they contribute to angiogenic remodeling of the vessel wall. This review focuses on current knowledge of pro-angiogenic progenitor cells in the pathogenesis of asthma and PAH.
Collapse
Affiliation(s)
- Heng T Duong
- Department of Pathobiology, NC22, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | | | | |
Collapse
|
161
|
Hansmann G, Plouffe BD, Hatch A, von Gise A, Sallmon H, Zamanian RT, Murthy SK. Design and validation of an endothelial progenitor cell capture chip and its application in patients with pulmonary arterial hypertension. J Mol Med (Berl) 2011; 89:971-83. [PMID: 21735044 DOI: 10.1007/s00109-011-0779-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 06/02/2011] [Accepted: 06/03/2011] [Indexed: 12/27/2022]
Abstract
The number of circulating endothelial progenitor cells (EPCs) inversely correlates with cardiovascular risk and clinical outcome, and thus has been proposed as a valuable biomarker for risk assessment, disease progression, and response to therapy. However, current strategies for isolation of these rare cells are limited to complex, laborious approaches. The goal of this study was the design and validation of a disposable microfluidic platform capable of selectively capturing and enumerating EPCs directly from human whole blood in healthy and diseased subjects, eliminating sample preprocessing. We then applied the "EPC capture chip" clinically and determined EPC numbers in blood from patients with pulmonary arterial hypertension (PAH). Blood was collected in tubes and injected into polymeric microfluidic chips containing microcolumns pre-coated with anti-CD34 antibody. Captured cells were immunofluorescently stained for the expression of stem and endothelial antigens, identified and counted. The EPC capture chip was validated with conventional flow cytometry counts (r = 0.83). The inter- and intra-day reliability of the microfluidic devices was confirmed at different time points in triplicates over 1-5 months. In a cohort of 43 patients with three forms of PAH (idiopathic/heritable, drug-induced, and connective tissue disease), EPC numbers are ≈50% lower in PAH subjects vs. matched controls and inversely related to two potential disease modifiers: body mass index and postmenopausal status. The EPC capture chip (5 × 30 × 0.05 mm(3)) requires only 200 μL of human blood and has the strong potential to serve as a rapid bedside test for the screening and monitoring of patients with PAH and other proliferative cardiovascular, pulmonary, malignant, and neurodegenerative diseases.
Collapse
Affiliation(s)
- Georg Hansmann
- Department of Cardiology, Children's Hospital Boston, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA.
| | | | | | | | | | | | | |
Collapse
|
162
|
Montani D, Perros F, Gambaryan N, Girerd B, Dorfmuller P, Price LC, Huertas A, Hammad H, Lambrecht B, Simonneau G, Launay JM, Cohen-Kaminsky S, Humbert M. C-Kit–Positive Cells Accumulate in Remodeled Vessels of Idiopathic Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 2011; 184:116-23. [DOI: 10.1164/rccm.201006-0905oc] [Citation(s) in RCA: 149] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
|
163
|
Yeager ME, Nguyen CM, Belchenko DD, Colvin KL, Takatsuki S, Ivy DD, Stenmark KR. Circulating fibrocytes are increased in children and young adults with pulmonary hypertension. Eur Respir J 2011; 39:104-11. [PMID: 21700605 DOI: 10.1183/09031936.00072311] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Chronic inflammation is an important component of the fibroproliferative changes that characterise pulmonary hypertensive vasculopathy. Fibrocytes contribute to tissue remodelling in settings of chronic inflammation, including animal models of pulmonary hypertension (PH). We sought to determine whether circulating fibrocytes were increased in children and young adults with PH. 26 individuals with PH and 10 with normal cardiac anatomy were studied. Fresh blood was analysed by flow cytometry for fibrocytes expressing CD45 and procollagen. Fibrocyte numbers were correlated to clinical and haemodynamic parameters, and circulating CC chemokine ligand (CCL)2 and CXC chemokine ligand (CXCL)12 levels. We found an enrichment of circulating fibrocytes among those with PH. No differences in fibrocytes were observed among those with idiopathic versus secondary PH. Higher fibrocytes correlated to increasing mean pulmonary artery pressure and age, but not to length or type of treatment. Immunofluorescence analysis confirmed flow sorting specificity. Differences in plasma levels of CCL2 or CXCL12, which could mobilise fibrocytes from the bone marrow, were not found. We conclude that circulating fibrocytes are significantly increased in individuals with PH compared with controls. We speculate that these cells might play important roles in vascular remodelling in children and young adults with pulmonary hypertension.
Collapse
Affiliation(s)
- M E Yeager
- Dept of Paediatrics, Division of Pulmonary and Critical Care Medicine, University of Colorado Denver, Aurora, CO 80045, USA.
| | | | | | | | | | | | | |
Collapse
|
164
|
Abstract
Our understanding of, and approach to, pulmonary arterial hypertension has undergone a paradigm shift in the past decade. Once a condition thought to be dominated by increased vasoconstrictor tone and thrombosis, pulmonary arterial hypertension is now seen as a vasculopathy in which structural changes driven by excessive vascular cell growth and inflammation, with recruitment and infiltration of circulating cells, play a major role. Perturbations of a number of molecular mechanisms have been described, including pathways involving growth factors, cytokines, metabolic signaling, elastases, and proteases, that may underlie the pathogenesis of the disease. Elucidating their contribution to the pathophysiology of pulmonary arterial hypertension could offer new drug targets. The role of progenitor cells in vascular repair is also under active investigation. The right ventricular response to increased pressure load is recognized as critical to survival and the molecular mechanisms involved are attracting increasing interest. The challenge now is to integrate this new knowledge and explore how it can be used to categorize patients by molecular phenotype and tailor treatment more effectively.
Collapse
Affiliation(s)
- Ralph T. Schermuly
- Max-Planck-Institute for Heart and Lung Research, Parkstrasse 1, Bad Nauheim, 61231 Germany
| | - Hossein A. Ghofrani
- University Hospital Giessen and Marburg, University of Giessen Lung Center, Klinikstrasse 36, Giessen, 35392 Germany
| | - Martin R. Wilkins
- Division of Experimental Medicine, Centre for Pharmacology and Therapeutics, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN UK
| | - Friedrich Grimminger
- University Hospital Giessen and Marburg, University of Giessen Lung Center, Klinikstrasse 36, Giessen, 35392 Germany
| |
Collapse
|
165
|
Bradford CN, Ely DR, Raizada MK. Targeting the vasoprotective axis of the renin-angiotensin system: a novel strategic approach to pulmonary hypertensive therapy. Curr Hypertens Rep 2011; 12:212-9. [PMID: 20556668 PMCID: PMC2957877 DOI: 10.1007/s11906-010-0122-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A decade has passed since the discovery of angiotensin-converting enzyme 2 (ACE2), a component of the ACE2–angiotensin (Ang)-(1-7)–Mas counterregulatory axis of the renin angiotensin system (RAS). ACE2 is considered an endogenous regulator of the vasoconstrictive, proliferative, fibrotic, and proinflammatory effects of the ACE–Ang II–angiotensin II type 1 receptor (AT1R) axis. Both animal and clinical studies have emerged to define a role for ACE2 in pulmonary arterial hypertension (PAH). There is scientific evidence supporting the concept that ACE2 maintains the RAS balance and plays a protective role in PAH. The activation of pulmonary ACE2 could influence the pathogenesis of PAH and serve as a novel therapeutic target in PAH. Current therapeutic strategies and interventions have limited success, and PAH remains a fatal disease. Thus, more research that establishes the novel therapeutic potential and defines the mechanism of the ACE2–Ang-(1-7)–Mas counterregulatory axis in PAH is needed.
Collapse
Affiliation(s)
- Chastity N Bradford
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, PO Box 100274, Gainesville, FL 32610, USA.
| | | | | |
Collapse
|
166
|
Weiss DJ, Bertoncello I, Borok Z, Kim C, Panoskaltsis-Mortari A, Reynolds S, Rojas M, Stripp B, Warburton D, Prockop DJ. Stem cells and cell therapies in lung biology and lung diseases. PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY 2011; 8:223-72. [PMID: 21653527 PMCID: PMC3132784 DOI: 10.1513/pats.201012-071dw] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Accepted: 02/03/2011] [Indexed: 11/20/2022]
Abstract
The University of Vermont College of Medicine and the Vermont Lung Center, with support of the National Heart, Lung, and Blood Institute (NHLBI), the Alpha-1 Foundation, the American Thoracic Society, the Emory Center for Respiratory Health,the Lymphangioleiomyomatosis (LAM) Treatment Alliance,and the Pulmonary Fibrosis Foundation, convened a workshop,‘‘Stem Cells and Cell Therapies in Lung Biology and Lung Diseases,’’ held July 26-29, 2009 at the University of Vermont,to review the current understanding of the role of stem and progenitor cells in lung repair after injury and to review the current status of cell therapy approaches for lung diseases. These are rapidly expanding areas of study that provide further insight into and challenge traditional views of the mechanisms of lung repair after injury and pathogenesis of several lung diseases. The goals of the conference were to summarize the current state of the field, discuss and debate current controversies, and identify future research directions and opportunities for both basic and translational research in cell-based therapies for lung diseases.
Collapse
Affiliation(s)
- Daniel J Weiss
- Vermont Lung Center, University of Vermont College of Medicine, Burlington, Vermont 05405, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
167
|
Plexiform lesions in pulmonary arterial hypertension composition, architecture, and microenvironment. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:167-79. [PMID: 21703400 DOI: 10.1016/j.ajpath.2011.03.040] [Citation(s) in RCA: 132] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Revised: 03/02/2011] [Accepted: 03/29/2011] [Indexed: 12/12/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a debilitating disease with a high mortality rate. A hallmark of PAH is plexiform lesions (PLs), complex vascular formations originating from remodeled pulmonary arteries. The development and significance of these lesions have been debated and are not yet fully understood. Some features of PLs resemble neoplastic disorders, and there is a striking resemblance to glomeruloid-like lesions (GLLs) in glioblastomas. To further elucidate PLs, we used in situ methods, such as (fluorescent) IHC staining, three-dimensional reconstruction, and laser microdissection, followed by mRNA expression analysis. We generated compartment-specific expression patterns in the lungs of 25 patients (11 with PAH associated with systemic shunts, 6 with idiopathic PAH, and 8 controls) and GLLs from 5 glioblastomas. PLs consisted of vascular channels lined by a continuously proliferating endothelium and backed by a uniform myogenic interstitium. They also showed up-regulation of remodeling-associated genes, such as HIF1a, TGF-β1, VEGF-α, VEGFR-1/-2, Ang-1, Tie-2, and THBS1, but also of cKIT and sprouting-associated markers, such as NOTCH and matrix metalloproteinases. The cellular composition and signaling seen in GLLs in neural neoplasms differed significantly from those in PLs. In conclusion, PLs show a distinct cellular composition and microenvironment, which contribute to the plexiform phenotype and set them apart from other processes of vascular remodeling in patients with PAH. Neoplastic models of angiogenesis seem to be of limited use in further study of plexiform vasculopathy.
Collapse
|
168
|
Baliga RS, MacAllister RJ, Hobbs AJ. New perspectives for the treatment of pulmonary hypertension. Br J Pharmacol 2011; 163:125-40. [PMID: 21175577 PMCID: PMC3085874 DOI: 10.1111/j.1476-5381.2010.01164.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 11/12/2010] [Accepted: 11/16/2010] [Indexed: 11/30/2022] Open
Abstract
Pulmonary hypertension (PH) is a debilitating disease with a poor prognosis. Therapeutic options remain limited despite the introduction of prostacyclin analogues, endothelin receptor antagonists and phosphodiesterase 5 inhibitors within the last 15 years; these interventions address predominantly the endothelial and vascular dysfunctionS associated with the condition, but simply delay progression of the disease rather than offer a cure. In an attempt to improve efficacy, emerging approaches have focused on targeting the pro-proliferative phenotype that underpins the pulmonary vascular remodelling in the lung and contributes to the impaired circulation and right heart failure. Many novel targets have been investigated and validated in animal models of PH, including modulation of guanylate cyclases, phosphodiesterases, tyrosine kinases, Rho kinase, bone morphogenetic proteins signalling, 5-HT, peroxisome proliferator activator receptors and ion channels. In addition, there is hope that combinations of such treatments, harnessing and optimizing vasodilator and anti-proliferative properties, will provide a further, possibly synergistic, increase in efficacy; therapies directed at the right heart may also offer an additional benefit. This overview highlights current therapeutic options, promising new therapies, and provides the rationale for a combination approach to treat the disease.
Collapse
|
169
|
Toya SP, Malik AB. Role of endothelial injury in disease mechanisms and contribution of progenitor cells in mediating endothelial repair. Immunobiology 2011; 217:569-80. [PMID: 21513999 DOI: 10.1016/j.imbio.2011.03.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Accepted: 03/09/2011] [Indexed: 12/30/2022]
Abstract
Recent research on the endothelium demonstrates complex interactions of endothelial cells with circulating immune cells, mediators such as cytokines, hormones and growth factors, and with the underlying parenchymal cells. These disparate interactions are involved in promotion of vascular development; maintenance of tissue homeostasis; and regulation of vascular repair. Injury to the endothelial monolayer is the sine qua non of organ dysfunction with endothelial repair the necessary first step needed for recovery. Thus, the capacity of the endothelium to regenerate itself is a key determinant of organ repair and survival after injury. Using the example of the lung, we will review the current state of knowledge regarding the importance of endothelium in the above mentioned processes with a focus on the role of stem cells, both endogenous (i.e., localized within the vessel wall) as well as exogenous (i.e., arriving in the vessel wall from distant sites such as the bone marrow) in promoting endothelial repair and regeneration. The subject of endothelial regeneration and the ways in which stem and progenitor cells contribute to this process has promise in treating vascular diseases. As we will highlight in this review, some questions have been addressed but many more remain and need to be addressed before cell-based therapies become a viable option.
Collapse
Affiliation(s)
- Sophie P Toya
- Department of Pharmacology and the Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, IL 60612, USA
| | | |
Collapse
|
170
|
Wang W, Wang YL, Chen XY, Li YT, Hao W, Jin YP, Han B. Dexamethasone attenuates development of monocrotaline-induced pulmonary arterial hypertension. Mol Biol Rep 2011; 38:3277-84. [PMID: 21431360 DOI: 10.1007/s11033-010-0390-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Accepted: 11/08/2010] [Indexed: 01/26/2023]
Abstract
Immunity and inflammation are well established factors in the pathogenesis of pulmonary arterial hypertension (PAH). We aimed to investigate whether dexamethasone (Dex), a potent immunosuppressant, could prevent the development of monocrotaline (MCT)-induced PAH in rats as compared with pyrrolidine dithiocarbamate (PDTC) and its effect on the immune mechanism. PAH in rats (n = 66) was induced by MCT (50 mg/kg) injected intraperitoneally. Two days after MCT treatment, Dex (1.0 mg/kg) and PDTC (100 mg/kg) were administered once daily for 21 days. Samples were collected at 7, 14, and 21 days. Dex effectively inhibited MCT-induced PAH and reduced the T-helper (Th) 1 dominant cytokine response (interferon-γ) but up-regulated the Th2 one (interleukin 4). It increased the number of CD4+ T cells and decreased the number of CD8+ T cells around pulmonary arteries, upregulated the mRNA expression of fractalkine and downregulated that of CX3CR1 in the lung. Serum levels of interferon γ and interleukin 4 did not significantly differ from that of controls. Dex attenuated the process of MCT-induced PAH through its immunomodulatory property. Dex could be an appropriate therapy for PAH, although more studies are needed to define the appropriate treatment regimen.
Collapse
Affiliation(s)
- Wei Wang
- Department of Pediatrics, Shandong Provincial Hospital, Shandong University, 44 Wenxi Road, Ji'nan, 250012 Shandong Province, China
| | | | | | | | | | | | | |
Collapse
|
171
|
Mirsky R, Jahn S, Koskenvuo JW, Sievers RE, Yim SM, Ritner C, Bernstein HS, Angeli FS, Boyle AJ, De Marco T, Yeghiazarians Y. Treatment of pulmonary arterial hypertension with circulating angiogenic cells. Am J Physiol Lung Cell Mol Physiol 2011; 301:L12-9. [PMID: 21398496 DOI: 10.1152/ajplung.00215.2010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Despite advances in the treatment of pulmonary arterial hypertension, a truly restorative therapy has not been achieved. Attention has been given to circulating angiogenic cells (CACs, also termed early endothelial progenitor cells) because of their ability to home to sites of vascular injury and regenerate blood vessels. We studied the efficacy of human CAC therapy in the treatment of pulmonary arterial hypertension at two different stages of disease severity. Cells were isolated from peripheral blood and administered to nude rats on day 14 ("early") or day 21 ("late") after monocrotaline injection. The control group received monocrotaline but no cell treatment. Disease progression was assessed using right heart catheterization and echocardiography at multiple time points. Survival differences, right ventricular hypertrophy (RVH), and vascular hypertrophy were analyzed at the study endpoint. Quantitative PCR was performed to evaluate cell engraftment. Treatment with human CACs either at the early or late time points did not result in increased survival, and therapy did not prevent or reduce the severity of disease compared with control. Histological analysis of RVH and vascular muscularization showed no benefit with therapy compared with control. No detectable signal was seen of human transcript in transplanted lungs at 14 or 21 days after cell transplant. In conclusion, CAC therapy was not associated with increased survival and did not result in either clinical or histological benefits. Future studies should be geared toward either earlier therapeutic time points with varying doses of unmodified CACs or genetically modified cells as a means of delivery of factors to the pulmonary arterial circulation.
Collapse
Affiliation(s)
- Rachel Mirsky
- Department of Medicine, University of California, San Francisco, San Francisco, California 94143, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
172
|
Paulin R, Courboulin A, Meloche J, Mainguy V, Dumas de la Roque E, Saksouk N, Côté J, Provencher S, Sussman MA, Bonnet S. Signal transducers and activators of transcription-3/pim1 axis plays a critical role in the pathogenesis of human pulmonary arterial hypertension. Circulation 2011; 123:1205-15. [PMID: 21382889 DOI: 10.1161/circulationaha.110.963314] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Pulmonary artery hypertension (PAH) is a proliferative disorder associated with enhanced pulmonary artery smooth muscle cell proliferation and suppressed apoptosis. The sustainability of this phenotype required the activation of a prosurvival transcription factor like signal transducers and activators of transcription-3 (STAT3) and nuclear factor of activated T cell (NFAT). Because these factors are implicated in several physiological processes, their inhibition in PAH patients could be associated with detrimental effects. Therefore, a better understanding of the mechanism accounting for their expression/activation in PAH pulmonary artery smooth muscle cells is of great therapeutic interest. METHODS AND RESULTS Using multidisciplinary and translational approaches, we demonstrated that STAT3 activation in both human and experimental models of PAH accounts for the expression of both NFATc2 and the oncoprotein kinase Pim1, which trigger NFATc2 activation. Because Pim1 expression correlates with the severity of PAH in humans and is confined to the PAH pulmonary artery smooth muscle cell, Pim1 was identified as an attractive therapeutic target for PAH. Indeed, specific Pim1 inhibition in vitro decreases pulmonary artery smooth muscle cell proliferation and promotes apoptosis, all of which are sustained by NFATc2 inhibition. In vivo, tissue-specific inhibition of Pim1 by nebulized siRNA reverses monocrotaline-induced PAH in rats, whereas Pim1 knockout mice are resistant to PAH development. CONCLUSION We demonstrated for the first time that inhibition of the inappropriate activation of STAT3/Pim1 axis is a novel, specific, and attractive therapeutic strategy to reverse PAH.
Collapse
Affiliation(s)
- Roxane Paulin
- Centre de recherche de L'Hôtel-Dieu de Québec, 10 Rue McMahon, Québec, QC G1R 2J6, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
173
|
Yu L, Hales CA. Effect of chemokine receptor CXCR4 on hypoxia-induced pulmonary hypertension and vascular remodeling in rats. Respir Res 2011; 12:21. [PMID: 21294880 PMCID: PMC3042398 DOI: 10.1186/1465-9921-12-21] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Accepted: 02/04/2011] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND CXCR4 is the receptor for chemokine CXCL12 and reportedly plays an important role in systemic vascular repair and remodeling, but the role of CXCR4 in development of pulmonary hypertension and vascular remodeling has not been fully understood. METHODS In this study we investigated the role of CXCR4 in the development of pulmonary hypertension and vascular remodeling by using a CXCR4 inhibitor AMD3100 and by electroporation of CXCR4 shRNA into bone marrow cells and then transplantation of the bone marrow cells into rats. RESULTS We found that the CXCR4 inhibitor significantly decreased chronic hypoxia-induced pulmonary hypertension and vascular remodeling in rats and, most importantly, we found that the rats that were transplanted with the bone marrow cells electroporated with CXCR4 shRNA had significantly lower mean pulmonary pressure (mPAP), ratio of right ventricular weight to left ventricular plus septal weight (RV/(LV+S)) and wall thickness of pulmonary artery induced by chronic hypoxia as compared with control rats. CONCLUSIONS The hypothesis that CXCR4 is critical in hypoxic pulmonary hypertension in rats has been demonstrated. The present study not only has shown an inhibitory effect caused by systemic inhibition of CXCR4 activity on pulmonary hypertension, but more importantly also has revealed that specific inhibition of the CXCR4 in bone marrow cells can reduce pulmonary hypertension and vascular remodeling via decreasing bone marrow derived cell recruitment to the lung in hypoxia. This study suggests a novel therapeutic approach for pulmonary hypertension by inhibiting bone marrow derived cell recruitment.
Collapse
Affiliation(s)
- Lunyin Yu
- Pulmonary and Critical Care Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | | |
Collapse
|
174
|
Hypoxia-inducible factors in human pulmonary arterial hypertension: a link to the intrinsic myeloid abnormalities. Blood 2011; 117:3485-93. [PMID: 21258008 DOI: 10.1182/blood-2010-09-306357] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a proliferative vasculopathy characterized by high circulating CD34(+)CD133(+) proangiogenic progenitors, and endothelial cells that have pathologic expression of hypoxia-inducible factor 1 α (HIF-1α). Here, CD34(+)CD133(+) progenitor cell numbers are shown to be higher in PAH bone marrow, blood, and pulmonary arteries than in healthy controls. The HIF-inducible myeloid-activating factors erythropoietin, stem cell factor (SCF), and hepatocyte growth factor (HGF) are also present at higher than normal levels in PAH blood, and related to disease severity. Primary endothelial cells harvested from human PAH lungs produce greater HGF and progenitor recruitment factor stromal-derived factor 1 α (SDF-1α) than control lung endothelial cells, and thus may contribute to bone marrow activation. Even though PAH patients had normal numbers of circulating blood elements, hematopoietic alterations in myeloid and erythroid lineages and reticulin fibrosis identified a subclinical myeloproliferative process. Unexpectedly, evaluation of bone marrow progenitors and reticulin in nonaffected family members of patients with familial PAH revealed similar myeloid abnormalities. Altogether, the results show that PAH is linked to myeloid abnormalities, some of which may be related to increased production of HIF-inducible factors by diseased pulmonary vasculature, but findings in nonaffected family suggest myeloid abnormalities may be intrinsic to the disease process.
Collapse
|
175
|
Lung. Regen Med 2011. [DOI: 10.1007/978-90-481-9075-1_30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
176
|
Abstract
Pulmonary hypertension is characterized by cellular and structural changes in the walls of pulmonary arteries. Intimal thickening and fibrosis, medial hypertrophy and fibroproliferative changes in the adventitia are commonly observed, as is the extension of smooth muscle into the previously non-muscularized vessels. A majority of these changes are associated with the enhanced presence of α-SM-actin+ cells and inflammatory cells. Atypical abundances of functionally distinct endothelial cells, particularly in the intima (plexiform lesions), and also in the perivascular regions, are also described. At present, neither the origin(s) of these cells nor the molecular mechanisms responsible for their accumulation, in any of the three compartments of the vessel wall, have been fully elucidated. The possibility that they arise from either resident vascular progenitors or bone marrow-derived progenitor cells is now well established. Resident vascular progenitor cells have been demonstrated to exist within the vessel wall, and in response to certain stimuli, to expand and express myofibroblastic, endothelial or even hematopoietic markers. Bone marrow-derived or circulating progenitor cells have also been shown to be recruited to sites of vascular injury and to assume both endothelial and SM-like phenotypes. Here, we review the data supporting the contributory role of vascular progenitors (including endothelial progenitor cells, smooth muscle progenitor cells, pericytes, and fibrocytes) in vascular remodeling. A more complete understanding of the processes by which progenitor cells modulate pulmonary vascular remodeling will undoubtedly herald a renaissance of therapies extending beyond the control of vascular tonicity and reduction of pulmonary artery pressure.
Collapse
Affiliation(s)
- Michael E. Yeager
- Department of Pediatrics and Critical Care, University of Colorado at Denver and Health Sciences Center, Colorado, USA
| | - Maria G. Frid
- Developmental Lung Biology Laboratory, Denver, Colorado, USA
| | | |
Collapse
|
177
|
|
178
|
Sakao S, Tatsumi K, Voelkel NF. Reversible or irreversible remodeling in pulmonary arterial hypertension. Am J Respir Cell Mol Biol 2010; 43:629-34. [PMID: 20008280 PMCID: PMC2993084 DOI: 10.1165/rcmb.2009-0389tr] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Accepted: 11/21/2009] [Indexed: 12/15/2022] Open
Abstract
Vascular remodeling is an important pathological feature of pulmonary arterial hypertension (PAH), which leads to increased pulmonary vascular resistance, with marked proliferation of pulmonary artery smooth muscle cells (SMC) and/or endothelial cells (EC). Successful treatment of experimental PAH with a platelet-derived growth factor (PDGF) receptor tyrosine kinase inhibitor offers the perspective of "reverse remodeling" (i.e., the regression of established pulmonary vascular lesions). Here we ask the question: which forms of pulmonary vascular remodeling are reversible and can such remodeling caused by angiogenic proliferation of EC be reversed? It is important to emphasize that the report showing reduction of vascular remodeling by PDGF receptor tyrosine kinase inhibitor showed only a reduction of the pulmonary artery muscularization in chronic hypoxia and monocrotaline models, which lack the feature of clustered proliferated EC in the lumen of pulmonary arteries. The regression of vascular muscularization is an important manifestation, whereby proliferative adult SMC convert back to a nonproliferative state. In contrast, in vitro experiments assessing the contribution of EC to the development of PAH demonstrated that phenotypically altered EC generated as a consequence of a vascular endothelial growth factor receptor blockade did not reverse to normal EC. Whereas it is suggested that the proliferative state of SMC may be reversible, it remains unknown whether phenotypically altered EC can switch back to a normal monolayer-forming EC. This article reviews the pathogenetic concepts of severe PAH and explains the many forms in PAH with reversible or irreversible remodeling.
Collapse
Affiliation(s)
- Seiichiro Sakao
- Department of Respirology (B2), Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, Japan.
| | | | | |
Collapse
|
179
|
Toshner M, Morrell NW. Endothelial progenitor cells in pulmonary hypertension - dawn of cell-based therapy? Int J Clin Pract 2010:7-12. [PMID: 19958395 DOI: 10.1111/j.1742-1241.2009.02232.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
There is mounting interest in the concept of endothelial progenitor cell (EPC) therapy for the treatment of pulmonary arterial hypertension (PAH). Recent successful pilot studies in idiopathic PAH have raised questions about the contribution of progenitor cells circulating in the peripheral blood to pulmonary vascular homeostasis and to the process of vascular remodelling. This review will summarise the work performed to date in animal and human therapeutic trials and clarify what is known about the potential contribution of EPCs to the pathophysiology of PAH.
Collapse
|
180
|
Treprostinil increases the number and angiogenic potential of endothelial progenitor cells in children with pulmonary hypertension. Angiogenesis 2010; 14:17-27. [PMID: 21049284 PMCID: PMC3040815 DOI: 10.1007/s10456-010-9192-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Accepted: 10/19/2010] [Indexed: 02/05/2023]
Abstract
Background Pulmonary vasodilators in general and prostacyclin therapy in particular, have markedly improved the outcome of patients with pulmonary arterial hypertension (PAH). As endothelial dysfunction is a key feature of PAH, and as endothelial progenitor cells (EPC) may contribute to vascular repair in PAH, we suspected that prostacyclin therapy might enhance EPC numbers and functions. In the present study, objectives were to determine whether EPC may contribute to vasodilator treatment efficacy in PAH. Methods We quantified CD34+ cells, CFU-Hill and ECFC (endothelial colony forming cells) in peripheral blood from children with idiopathic PAH (n = 27) or PAH secondary to congenital heart disease (n = 52). CD34+ were enumerated by flow cytometry, CFU-Hill and ECFC by a culture assay. ECFC grown ex vivo were tested for their angiogenic capacities before and after prostacyclin analog therapy (subcutaneous treprostinil). Results ECFC counts were significantly enhanced in the 8 children treated with treprostinil, while no change was observed in children receiving oral therapy with endothelin antagonists and/or PDE5 inhibitors. CD34+ cell and CFU-Hill counts were unaffected. ECFC from patients treated with treprostinil had a hyperproliferative phenotype and showed enhanced angiogenic potential in a nude mouse preclinical model of limb ischemia. Conclusions ECFC may partly mediate the clinical benefits of prostanoids in pulmonary arterial hypertension.
Collapse
|
181
|
Dewachter L, Dewachter C, Naeije R. New therapies for pulmonary arterial hypertension: an update on current bench to bedside translation. Expert Opin Investig Drugs 2010; 19:469-88. [PMID: 20367190 DOI: 10.1517/13543781003727099] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
IMPORTANCE OF THE FIELD Treatments of pulmonary arterial hypertension (PAH) that have so far proven efficacious are all based on the restoration of endothelium control of pulmonary vascular tone and structure, by administration of prostacyclins, endothelin receptor antagonists and phosphodiesterase-5 inhibitors. However, results remain unsatisfactory, with persistent high mortality, insufficient clinical improvement and no convincing report of any reversal of the disease process. AREAS COVERED IN THIS REVIEW New antiproliferative approaches that aim to actively limit pulmonary vascular remodeling are being sought. Several such treatments have shown promise in experimental models and in preliminary clinical studies. Noteworthy among these are dichloroacetate, survivin antagonists, nuclear factor of activated T-cell inhibitors, PPAR-gamma agonists, tyrosine kinase inhibitors, Rho-kinase inhibitors, statins, vasoactive intestinal peptide, soluble guanylate cyclase stimulators/activators, adrenomedullin, elastase inhibitors, serotonin reuptake inhibitors, anti-inflammatory agents, and bone marrow-derived progenitor cells. WHAT THE READER WILL GAIN Update on various strategies targeting proliferative, inflammatory and regenerating processes currently under evaluation in patients with PAH. TAKE HOME MESSAGE In spite of favorable results in experimental models, none of these strategies has achieved the ultimate goal of curing PAH. Further developments will depend on progress made in our pathobiological understanding of the disease and carefully designed randomized, controlled trials.
Collapse
Affiliation(s)
- Laurence Dewachter
- Free University of Brussels, Department of Physiology, Faculty of Medicine, Erasme Campus CP 604, Lennik Road 808, B-1070 Brussels, Belgium.
| | | | | |
Collapse
|
182
|
Yue WS, Wang M, Yan GH, Yiu KH, Yin L, Lee SWL, Siu CW, Tse HF. Smoking is associated with depletion of circulating endothelial progenitor cells and elevated pulmonary artery systolic pressure in patients with coronary artery disease. Am J Cardiol 2010; 106:1248-54. [PMID: 21029820 DOI: 10.1016/j.amjcard.2010.06.045] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Revised: 06/09/2010] [Accepted: 06/09/2010] [Indexed: 01/13/2023]
Abstract
Smoking is associated with depletion of endothelial progenitor cells (EPCs) and may subsequently contribute to the development of vascular dysfunction. The aim of this study was to investigate the relation between circulating EPCs and pulmonary artery systolic pressure (PASP) as determined by flow cytometry and echocardiography in 174 patients (mean age 69 ± 9 years, 95 smokers) with established coronary artery disease. Smokers had significantly lower circulating log CD34/KDR(+) (0.86 ± 0.03 vs 0.96 ± 0.03 × 10⁻³/ml, p = 0.032) and log CD133/KDR(+) (0.68 ± 0.03 vs 0.82 ± 0.03 × 10⁻³/ml, p = 0.002) EPCs and a higher prevalence of elevated PASP >30 mm Hg (52% vs 30%, p = 0.001) than nonsmokers. Smokers with elevated PASP also had significantly lower circulating log CD34/KDR(+) (0.74 ± 0.04 vs 0.88 ± 0.06 × 10⁻³/ml, p <0.001) and log CD133/KDR(+) (0.61 ± 0.04 vs 0.78 ± 0.05 × 10⁻³/ml, p <0.001) EPCs, higher pulmonary vascular resistance, and larger right ventricular dimensions with impaired function (all p values <0.05). Log CD34/KDR(+) and log CD133/KDR(+) EPC counts were significantly and negatively correlated with PASP (r = -0.30, p <0.001, and r = -0.34, p <0.001, respectively) and pulmonary vascular resistance (r = -0.29, p = 0.002, and r = -0.18, p = 0.013, respectively). In conclusion, this study demonstrated that in patients with coronary artery disease, smoking was associated with a reduced number of EPCs and elevated PASP. This suggests that in smokers, depletion of circulating EPCs might be linked to the occurrence of pulmonary vascular dysfunction.
Collapse
Affiliation(s)
- Wen-Sheng Yue
- Department of Medicine, Queen Mary Hospital, Hong Kong, China
| | | | | | | | | | | | | | | |
Collapse
|
183
|
Affiliation(s)
- Mark T Gladwin
- Pulmonary, Allergy and Critical Care Medicine, NW 628 Montefiore Hospital, 3459 Fifth Ave., Pittsburgh, PA 15213, USA.
| | | |
Collapse
|
184
|
Abstract
IMPORTANCE OF THE FIELD Pulmonary arterial hypertension (PAH) is a clinical syndrome characterized by structural narrowing of the small pulmonary arteries that often culminates in fatal right heart failure. AREAS COVERED IN THIS REVIEW PubMed was searched for PAH and treatment. Data from scientific meetings and pharmaceutical websites are also included. There are currently eight FDA approved drugs for PAH that fall into one of three classes: prostacyclins, endothelin-receptor antagonists and PDE-5 inhibitors. All have important limitations and morbidity and mortality remain high. Several new agents with similar mechanisms of action are in clinical development. Multiple novel therapeutic targets are being explored. New applications for PAH therapies, such as pulmonary hypertension due to left heart and lung disease, are also being investigated. WHAT THE READER WILL GAIN An understanding of currently available drugs and those in clinical development for pulmonary hypertension. TAKE HOME MESSAGE Drugs targeting the pulmonary vasculature have been an extremely active area of basic and clinical research for the past 20 years and will continue to be so for the foreseeable future. Considerable progress has been made, and yet there continues to be a great unmet medical need for developing more efficacious therapies.
Collapse
Affiliation(s)
- Reda E Girgis
- Johns Hopkins University, School of Medicine, Division of Pulmonary and Critical Care Medicine, USA.
| |
Collapse
|
185
|
Everaert BR, Van Craenenbroeck EM, Hoymans VY, Haine SE, Van Nassauw L, Conraads VM, Timmermans JP, Vrints CJ. Current perspective of pathophysiological and interventional effects on endothelial progenitor cell biology: focus on PI3K/AKT/eNOS pathway. Int J Cardiol 2010; 144:350-66. [PMID: 20444511 DOI: 10.1016/j.ijcard.2010.04.018] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2010] [Accepted: 04/04/2010] [Indexed: 12/24/2022]
Abstract
For more than a decade, endothelial progenitor cells (EPCs) have been implicated in cardiovascular homeostasis. EPCs are believed to reside within the bone marrow in close contact with surrounding stromal cells, and, under stimulation of pro-inflammatory cytokines, EPCs are mobilized out of the bone marrow. Hereafter circulating EPCs home to peripheral tissues, undergoing further proliferation and differentiation. Under certain pathophysiologic conditions this process seems to be blunted, resulting in a reduced capacity of EPCs to engage in vasculogenesis at sites of endothelial injury or tissue ischemia. In this review, we focus on the effects of traditional cardiovascular risk factors on EPC biology and we explore whether pharmacological, dietary and lifestyle interventions can favorably restore EPC mobilization, differentiation, homing and angiogenic properties. Because the PI3K/Akt/eNOS pathway plays a pivotal role in the process of EPC mobilization, migration and homing, we specifically emphasize the involvement of PI3K, Akt and eNOS in EPC biology under these different (patho)physiologic conditions. (Pre)clinically used drugs or lifestyle interventions that have been shown to ameliorate EPC biology are reviewed. These treatment strategies remain attractive targets to restore the regenerative capacity of EPCs in cardiovascular diseases.
Collapse
Affiliation(s)
- Bert R Everaert
- Laboratory of Cell Biology and Histology, University of Antwerp, Antwerp, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
186
|
Abstract
Pulmonary hypertension is an orphan disease that until recently has received limited attention within the wider medical community. This has changed distinctly in the last 10 years with the advent of new classes of therapy and a renewed interest in mechanisms of pathogenesis. This review utilized information gathered from recent conferences, and a review of the literature was conducted using MedLine and Pubmed. Accepted mechanisms of pathogenesis and currently available treatments are presented. We will discuss interesting new concepts in pathogenesis, including the importance of genetic forms of the disease and in particular the transforming growth factor receptor superfamily and the evolving evidence of the contribution of dysregulated immunity. Areas of research may yield therapeutic benefits in the not-too-distant future, including anti-proliferative therapies and stem cell therapy.
Collapse
Affiliation(s)
- M Toshner
- Department of Medicine, University of Cambridge, Cambridge CB2 2QQ, UK
| | | | | |
Collapse
|
187
|
Diller GP, Thum T, Wilkins MR, Wharton J. Endothelial Progenitor Cells in Pulmonary Arterial Hypertension. Trends Cardiovasc Med 2010; 20:22-9. [DOI: 10.1016/j.tcm.2010.03.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
188
|
Toshner M, Voswinckel R, Southwood M, Al-Lamki R, Howard LSG, Marchesan D, Yang J, Suntharalingam J, Soon E, Exley A, Stewart S, Hecker M, Zhu Z, Gehling U, Seeger W, Pepke-Zaba J, Morrell NW. Evidence of dysfunction of endothelial progenitors in pulmonary arterial hypertension. Am J Respir Crit Care Med 2009; 180:780-7. [PMID: 19628780 PMCID: PMC2778151 DOI: 10.1164/rccm.200810-1662oc] [Citation(s) in RCA: 186] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2008] [Accepted: 07/22/2009] [Indexed: 02/06/2023] Open
Abstract
RATIONALE Severe pulmonary arterial hypertension (PAH) is characterized by the formation of plexiform lesions and concentric intimal fibrosis in small pulmonary arteries. The origin of cells contributing to these vascular lesions is uncertain. Endogenous endothelial progenitor cells are potential contributors to this process. OBJECTIVES To determine whether progenitors are involved in the pathobiology of PAH. METHODS We performed immunohistochemistry to determine the expression of progenitor cell markers (CD133 and c-Kit) and the major homing signal pathway stromal cell-derived factor-1 and its chemokine receptor (CXCR4) in lung tissue from patients with idiopathic PAH, familial PAH, and PAH associated with congenital heart disease. Two separate flow cytometric methods were employed to determine peripheral blood circulating numbers of angiogenic progenitors. Late-outgrowth progenitor cells were expanded ex vivo from the peripheral blood of patients with mutations in the gene encoding bone morphogenetic protein receptor type II (BMPRII), and functional assays of migration, proliferation, and angiogenesis were undertaken. measurements and main results: There was a striking up-regulation of progenitor cell markers in remodeled arteries from all patients with PAH, specifically in plexiform lesions. These lesions also displayed increased stromal cell-derived factor-1 expression. Circulating angiogenic progenitor numbers in patients with PAH were increased compared with control subjects and functional studies of late-outgrowth progenitor cells from patients with PAH with BMPRII mutations revealed a hyperproliferative phenotype with impaired ability to form vascular networks. CONCLUSIONS These findings provide evidence of the involvement of progenitor cells in the vascular remodeling associated with PAH. Dysfunction of circulating progenitors in PAH may contribute to this process.
Collapse
Affiliation(s)
- Mark Toshner
- Papworth Hospital and University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom; Department of Internal Medicine, University of Giessen Lung Center, Giessen, Germany; Hammersmith Hospital, London and Royal United Hospital, Bath, United Kingdom; Department Antibody Technology, ImClone Systems, Inc., New York; and Department of Medicine, University Hospital Eppendorf, Hamburg, Germany
| | - Robert Voswinckel
- Papworth Hospital and University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom; Department of Internal Medicine, University of Giessen Lung Center, Giessen, Germany; Hammersmith Hospital, London and Royal United Hospital, Bath, United Kingdom; Department Antibody Technology, ImClone Systems, Inc., New York; and Department of Medicine, University Hospital Eppendorf, Hamburg, Germany
| | - Mark Southwood
- Papworth Hospital and University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom; Department of Internal Medicine, University of Giessen Lung Center, Giessen, Germany; Hammersmith Hospital, London and Royal United Hospital, Bath, United Kingdom; Department Antibody Technology, ImClone Systems, Inc., New York; and Department of Medicine, University Hospital Eppendorf, Hamburg, Germany
| | - Rafia Al-Lamki
- Papworth Hospital and University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom; Department of Internal Medicine, University of Giessen Lung Center, Giessen, Germany; Hammersmith Hospital, London and Royal United Hospital, Bath, United Kingdom; Department Antibody Technology, ImClone Systems, Inc., New York; and Department of Medicine, University Hospital Eppendorf, Hamburg, Germany
| | - Luke S. G. Howard
- Papworth Hospital and University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom; Department of Internal Medicine, University of Giessen Lung Center, Giessen, Germany; Hammersmith Hospital, London and Royal United Hospital, Bath, United Kingdom; Department Antibody Technology, ImClone Systems, Inc., New York; and Department of Medicine, University Hospital Eppendorf, Hamburg, Germany
| | - Denis Marchesan
- Papworth Hospital and University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom; Department of Internal Medicine, University of Giessen Lung Center, Giessen, Germany; Hammersmith Hospital, London and Royal United Hospital, Bath, United Kingdom; Department Antibody Technology, ImClone Systems, Inc., New York; and Department of Medicine, University Hospital Eppendorf, Hamburg, Germany
| | - Jun Yang
- Papworth Hospital and University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom; Department of Internal Medicine, University of Giessen Lung Center, Giessen, Germany; Hammersmith Hospital, London and Royal United Hospital, Bath, United Kingdom; Department Antibody Technology, ImClone Systems, Inc., New York; and Department of Medicine, University Hospital Eppendorf, Hamburg, Germany
| | - Jay Suntharalingam
- Papworth Hospital and University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom; Department of Internal Medicine, University of Giessen Lung Center, Giessen, Germany; Hammersmith Hospital, London and Royal United Hospital, Bath, United Kingdom; Department Antibody Technology, ImClone Systems, Inc., New York; and Department of Medicine, University Hospital Eppendorf, Hamburg, Germany
| | - Elaine Soon
- Papworth Hospital and University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom; Department of Internal Medicine, University of Giessen Lung Center, Giessen, Germany; Hammersmith Hospital, London and Royal United Hospital, Bath, United Kingdom; Department Antibody Technology, ImClone Systems, Inc., New York; and Department of Medicine, University Hospital Eppendorf, Hamburg, Germany
| | - Andrew Exley
- Papworth Hospital and University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom; Department of Internal Medicine, University of Giessen Lung Center, Giessen, Germany; Hammersmith Hospital, London and Royal United Hospital, Bath, United Kingdom; Department Antibody Technology, ImClone Systems, Inc., New York; and Department of Medicine, University Hospital Eppendorf, Hamburg, Germany
| | - Susan Stewart
- Papworth Hospital and University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom; Department of Internal Medicine, University of Giessen Lung Center, Giessen, Germany; Hammersmith Hospital, London and Royal United Hospital, Bath, United Kingdom; Department Antibody Technology, ImClone Systems, Inc., New York; and Department of Medicine, University Hospital Eppendorf, Hamburg, Germany
| | - Markus Hecker
- Papworth Hospital and University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom; Department of Internal Medicine, University of Giessen Lung Center, Giessen, Germany; Hammersmith Hospital, London and Royal United Hospital, Bath, United Kingdom; Department Antibody Technology, ImClone Systems, Inc., New York; and Department of Medicine, University Hospital Eppendorf, Hamburg, Germany
| | - Zhenping Zhu
- Papworth Hospital and University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom; Department of Internal Medicine, University of Giessen Lung Center, Giessen, Germany; Hammersmith Hospital, London and Royal United Hospital, Bath, United Kingdom; Department Antibody Technology, ImClone Systems, Inc., New York; and Department of Medicine, University Hospital Eppendorf, Hamburg, Germany
| | - Ursula Gehling
- Papworth Hospital and University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom; Department of Internal Medicine, University of Giessen Lung Center, Giessen, Germany; Hammersmith Hospital, London and Royal United Hospital, Bath, United Kingdom; Department Antibody Technology, ImClone Systems, Inc., New York; and Department of Medicine, University Hospital Eppendorf, Hamburg, Germany
| | - Werner Seeger
- Papworth Hospital and University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom; Department of Internal Medicine, University of Giessen Lung Center, Giessen, Germany; Hammersmith Hospital, London and Royal United Hospital, Bath, United Kingdom; Department Antibody Technology, ImClone Systems, Inc., New York; and Department of Medicine, University Hospital Eppendorf, Hamburg, Germany
| | - Joanna Pepke-Zaba
- Papworth Hospital and University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom; Department of Internal Medicine, University of Giessen Lung Center, Giessen, Germany; Hammersmith Hospital, London and Royal United Hospital, Bath, United Kingdom; Department Antibody Technology, ImClone Systems, Inc., New York; and Department of Medicine, University Hospital Eppendorf, Hamburg, Germany
| | - Nicholas W. Morrell
- Papworth Hospital and University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom; Department of Internal Medicine, University of Giessen Lung Center, Giessen, Germany; Hammersmith Hospital, London and Royal United Hospital, Bath, United Kingdom; Department Antibody Technology, ImClone Systems, Inc., New York; and Department of Medicine, University Hospital Eppendorf, Hamburg, Germany
| |
Collapse
|