1
|
Castillo-Galán S, Parra V, Cuenca J. Unraveling the pathogenesis of viral-induced pulmonary arterial hypertension: Possible new therapeutic avenues with mesenchymal stromal cells and their derivatives. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167519. [PMID: 39332781 DOI: 10.1016/j.bbadis.2024.167519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/16/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024]
Abstract
Pulmonary hypertension (PH) is a severe condition characterized by elevated pressure in the pulmonary artery, where metabolic and mitochondrial dysfunction may contribute to its progression. Within the PH spectrum, pulmonary arterial hypertension (PAH) stands out with its primary pulmonary vasculopathy. PAH's prevalence varies from 0.4 to 1.4 per 100,000 individuals and is associated with diverse conditions, including viral infections such as HIV. Notably, recent observations highlight an increased occurrence of PAH among COVID-19 patients, even in the absence of pre-existing cardiopulmonary disorders. While current treatments offer partial relief, there's a pressing need for innovative therapeutic strategies, among which mesenchymal stromal cells (MSCs) and their derivatives hold promise. This review critically evaluates recent investigations into viral-induced PAH, encompassing pathogens like human immunodeficiency virus, herpesvirus, Cytomegalovirus, Hepatitis B and C viruses, SARS-CoV-2, and Human endogenous retrovirus K (HERKV), with a specific emphasis on mitochondrial dysfunction. Furthermore, we explore the underlying rationale driving novel therapeutic modalities, including MSCs, extracellular vesicles, and mitochondrial interventions, within the framework of PAH management.
Collapse
Affiliation(s)
- Sebastián Castillo-Galán
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of Medicine, Universidad de los Andes, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
| | - Valentina Parra
- Laboratory of Differentiation and Cell Metabolism (D&M), Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile; Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile; SYSTEMIX Center for Systems Biology, O'Higgins University, Rancagua, Chile
| | - Jimena Cuenca
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of Medicine, Universidad de los Andes, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile; Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile; Cells for Cells, Santiago, Chile.
| |
Collapse
|
2
|
Liu Y, He C, Zhong Q, Shi X, Li H, Fu G, Guo L, Zhao C, Tian L, Li X, Jiao X, Shan L. Tadalafil Enhances the Therapeutic Efficacy of Mesenchymal Stem Cells-Derived Exosomes in Pulmonary Hypertension by Upregulating miR-29a-3p. Int J Nanomedicine 2024; 19:13525-13546. [PMID: 39720214 PMCID: PMC11668336 DOI: 10.2147/ijn.s493047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 12/10/2024] [Indexed: 12/26/2024] Open
Abstract
Introduction Pulmonary hypertension (PH) is a progressive and life-threatening condition. Recent research has demonstrated that exosomes derived from mesenchymal stem cells (MSC) exhibit significant therapeutic potential in the treatment of PH. The composition of these exosomes is often substantially influenced by the characteristics of their parental cells. This study aimed to identify an intervention strategy to enhance the efficacy of mesenchymal stem cell exosomes in treating PH. Methods Exosomes were isolated from control MSC and tadalafil-pretreated MSCs. In vitro and in vivo studies were conducted. Results MSCTAD-Exo attenuated macrophage inflammation and improved endothelial cell (EC) apoptosis while also reducing pulmonary arterial pressure in a hypoxia-induced rat model. Furthermore, MSC exosomes can mitigate hypoxia-induced proliferation and migration of smooth muscle cells (SMC) by influencing the secretion of endothelial exosomes. MiR-29a-3p has been identified as a crucial mediator in this process, with its expression regulated by cAMP responsive element binding protein 1 (CREB1). MiR-29a-3p exerts anti-inflammatory effects by modulating the expression of ectonucleotide pyrophosphatase/phosphodiesterase 2 (ENPP2). Notably, the anti-inflammatory and anti-vascular remodeling activities of exosomes were diminished following the depletion of MiR-29a-3p. Discussion MSC treated with tadalafil can secrete better exosomes. MSCTAD-Exo may enhance anti-inflammatory and anti-vascular remodeling properties by upregulating mir-29a-3p expression, providing a novel idea for PH therapy. Future studies could explore the clinical application of this finding.
Collapse
Affiliation(s)
- Yi Liu
- Department of Respiratory Disease, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, 121000, People’s Republic of China
- Department of Critical Care Medicine, Panzhihua Central Hospital, Panzhihua, 61700, People’s Republic of China
| | - Changqing He
- Department of Respiratory Disease, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, 121000, People’s Republic of China
| | - Quanhai Zhong
- Clinical Drug Laboratory, People’s Hospital of Yichun City, Yichun, Jiangxi, 336000, People’s Republic of China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, People’s Republic of China
| | - Hongyan Li
- Department of Respiratory Disease, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, 121000, People’s Republic of China
| | - Gaoge Fu
- Department of Respiratory Disease, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, 121000, People’s Republic of China
| | - Lixuan Guo
- Department of Respiratory Disease, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, 121000, People’s Republic of China
| | - Churong Zhao
- Department of Respiratory Disease, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, 121000, People’s Republic of China
| | - Lei Tian
- Department of Respiratory Disease, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, 121000, People’s Republic of China
| | - Xin Li
- Department of Respiratory Disease, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, 121000, People’s Republic of China
| | - Xue Jiao
- Department of Respiratory Disease, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, 121000, People’s Republic of China
| | - Lina Shan
- Department of Respiratory Disease, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, 121000, People’s Republic of China
| |
Collapse
|
3
|
Singh N, Al-Naamani N, Brown MB, Long GM, Thenappan T, Umar S, Ventetuolo CE, Lahm T. Extrapulmonary manifestations of pulmonary arterial hypertension. Expert Rev Respir Med 2024; 18:189-205. [PMID: 38801029 PMCID: PMC11713041 DOI: 10.1080/17476348.2024.2361037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/24/2024] [Indexed: 05/29/2024]
Abstract
INTRODUCTION Extrapulmonary manifestations of pulmonary arterial hypertension (PAH) may play a critical pathobiological role and a deeper understanding will advance insight into mechanisms and novel therapeutic targets. This manuscript reviews our understanding of extrapulmonary manifestations of PAH. AREAS COVERED A group of experts was assembled and a complimentary PubMed search performed (October 2023 - March 2024). Inflammation is observed throughout the central nervous system and attempts at manipulation are an encouraging step toward novel therapeutics. Retinal vascular imaging holds promise as a noninvasive method of detecting early disease and monitoring treatment responses. PAH patients have gut flora alterations and dysbiosis likely plays a role in systemic inflammation. Despite inconsistent observations, the roles of obesity, insulin resistance and dysregulated metabolism may be illuminated by deep phenotyping of body composition. Skeletal muscle dysfunction is perpetuated by metabolic dysfunction, inflammation, and hypoperfusion, but exercise training shows benefit. Renal, hepatic, and bone marrow abnormalities are observed in PAH and may represent both end-organ damage and disease modifiers. EXPERT OPINION Insights into systemic manifestations of PAH will illuminate disease mechanisms and novel therapeutic targets. Additional study is needed to understand whether extrapulmonary manifestations are a cause or effect of PAH and how manipulation may affect outcomes.
Collapse
Affiliation(s)
- Navneet Singh
- Department of Medicine, Warren Alpert School of Medicine at Brown University, Providence, RI
| | - Nadine Al-Naamani
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Mary Beth Brown
- Department of Rehabilitation Medicine, University of Washington School of Medicine, Seattle, WA
| | - Gary Marshall Long
- Department of Kinesiology, Health and Sport Sciences, University of Indianapolis, Indianapolis, IN
| | - Thenappan Thenappan
- Section of Advanced Heart Failure and Pulmonary Hypertension, Cardiovascular Division, University of Minnesota, Minneapolis, MN
| | - Soban Umar
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Corey E. Ventetuolo
- Department of Medicine, Warren Alpert School of Medicine at Brown University, Providence, RI
- Department of Health Services, Policy and Practice, Brown University, Providence, RI
| | - Tim Lahm
- Department of Medicine, National Jewish Health, Denver, CO
- Department of Medicine, University of Colorado, Aurora, CO
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO
| |
Collapse
|
4
|
Schrodt MV, Behan-Bush RM, Liszewski JN, Humpal-Pash ME, Boland LK, Scroggins SM, Santillan DA, Ankrum JA. Efferocytosis of viable versus heat-inactivated MSC induces human monocytes to distinct immunosuppressive phenotypes. Stem Cell Res Ther 2023; 14:206. [PMID: 37592321 PMCID: PMC10433682 DOI: 10.1186/s13287-023-03443-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 08/07/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND Immunomodulation by mesenchymal stromal cells (MSCs) can occur through trophic factor mechanisms, however, intravenously infused MSCs are rapidly cleared from the body yet a potent immunotherapeutic response is still observed. Recent work suggests that monocytes contribute to the clearance of MSCs via efferocytosis, the body's natural mechanism for clearing dead and dying cells in a non-inflammatory manner. This begs the questions of how variations in MSC quality affect monocyte phenotype and if viable MSCs are even needed to elicit an immunosuppressive response. METHODS Herein, we sought to dissect MSC's trophic mechanism from their efferocytic mechanisms and determine if the viability of MSCs prior to efferocytosis influences the resultant phenotype of monocytes. We cultured viable or heat-inactivated human umbilical cord MSCs with human peripheral blood mononuclear cells for 24 h and observed changes in monocyte surface marker expression and secretion profile. To isolate the effect of efferocytosis from MSC trophic factors, we used cell separation techniques to remove non-efferocytosed MSCs before challenging monocytes to suppress T-cells or respond to inflammatory stimuli. For all experiments, viable and heat-inactivated efferocytic-licensing of monocytes were compared to non-efferocytic-licensing control. RESULTS We found that monocytes efferocytose viable and heat-inactivated MSCs equally, but only viable MSC-licensed monocytes suppress activated T-cells and suppression occurred even after depletion of residual MSCs. This provides direct evidence that monocytes that efferocytose viable MSCs are immunosuppressive. Further characterization of monocytes after efferocytosis showed that uptake of viable-but not heat inactivated-MSC resulted in monocytes secreting IL-10 and producing kynurenine. When monocytes were challenged with LPS, IL-2, and IFN-γ to simulate sepsis, monocytes that had efferocytosed viable MSC had higher levels of IDO while monocytes that efferocytosed heat inactivated-MSCs produced the lowest levels of TNF-α. CONCLUSION Collectively, these studies show that the quality of MSCs efferocytosed by monocytes polarize monocytes toward distinctive immunosuppressive phenotypes and highlights the need to tailor MSC therapies for specific indications.
Collapse
Affiliation(s)
- Michael V Schrodt
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52245, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52245, USA
| | - Riley M Behan-Bush
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52245, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52245, USA
| | - Jesse N Liszewski
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52245, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52245, USA
| | - Madeleine E Humpal-Pash
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52245, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52245, USA
| | - Lauren K Boland
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52245, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52245, USA
| | - Sabrina M Scroggins
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Center for Immunology and Immune Based Diseases, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Biomedical Sciences, Center for Immunology, Center for Clinical and Translational Science, University of Minnesota School of Medicine, Duluth, MN, USA
| | - Donna A Santillan
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Center for Immunology and Immune Based Diseases, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Biomedical Sciences, Center for Immunology, Center for Clinical and Translational Science, University of Minnesota School of Medicine, Duluth, MN, USA
| | - James A Ankrum
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52245, USA.
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52245, USA.
- , 103 S. Capitol St., 5621 SC, Iowa City, IA, 52242, USA.
| |
Collapse
|
5
|
Sun QW, Sun Z. Stem Cell Therapy for Pulmonary Arterial Hypertension: An Update. J Heart Lung Transplant 2022; 41:692-703. [DOI: 10.1016/j.healun.2022.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 02/04/2022] [Accepted: 02/27/2022] [Indexed: 10/18/2022] Open
|
6
|
Oh S, Jung JH, Ahn KJ, Jang AY, Byun K, Yang PC, Chung WJ. Stem Cell and Exosome Therapy in Pulmonary Hypertension. Korean Circ J 2022; 52:110-122. [PMID: 35128849 PMCID: PMC8819574 DOI: 10.4070/kcj.2021.0191] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 11/30/2021] [Accepted: 12/13/2021] [Indexed: 11/25/2022] Open
Abstract
Pulmonary hypertension (PH) is a disease that eventually causes right heart failure by remodeling pulmonary blood vessels. Based on the histopathological characteristics, PH is categorized into five subgroups. Rarely, a severe clinical entity is pulmonary arterial hypertension (PAH), subgroup 1. This disease process results in pulmonary vascular alterations through dysfunction of the pulmonary endothelium and disturbance of immune responses. Although medical treatments based on these pathophysiologic concepts have been applied for more than 30 years, PAH still cannot be cured. This review addresses the feasibility of and perspectives on stem cell therapy, including the role of exosomes in PAH. Pulmonary hypertension is a rare and progressive illness with a devastating prognosis. Promising research efforts have advanced the understanding and recognition of the pathobiology of pulmonary hypertension. Despite remarkable achievements in terms of improving the survival rate, reducing disease progression, and enhancing quality of life, pulmonary arterial hypertension (PAH) is not completely curable. Therefore, an effective treatment strategy is still needed. Recently, many studies of the underlying molecular mechanisms and technological developments have led to new approaches and paradigms for PAH treatment. Management based on stem cells and related paracrine effects, epigenetic drugs and gene therapies has yielded prospective results for PAH treatment in preclinical research. Further trials are ongoing to optimize these important insights into clinical circumstances.
Collapse
Affiliation(s)
- Seyeon Oh
- Gachon Cardiovascular Research Institute, Gachon University, Incheon, Korea
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
| | - Ji-Hye Jung
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Kyung-Jin Ahn
- Gachon Cardiovascular Research Institute, Gachon University, Incheon, Korea
- Division of Pediatric Cardiology, Department of Pediatrics, Gachon University Gil Medical Center, Incheon, Korea
| | - Albert Youngwoo Jang
- Gachon Cardiovascular Research Institute, Gachon University, Incheon, Korea
- Department of Cardiovascular Medicine, Gachon University Gil Medical Center, Incheon, Korea
| | - Kyunghee Byun
- Gachon Cardiovascular Research Institute, Gachon University, Incheon, Korea
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
- Department of Anatomy and Cell Biology, College of Medicine, Gachon University, Incheon, Korea
| | - Phillip C. Yang
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Stanford University School of Medicine, Palo Alto, CA, USA
| | - Wook-Jin Chung
- Gachon Cardiovascular Research Institute, Gachon University, Incheon, Korea
| |
Collapse
|
7
|
Marciano BE, Olivier KN, Folio LR, Zerbe CS, Hsu AP, Freeman AF, Filie AC, Spinner MA, Sanchez LA, Lovell JP, Parta M, Cuellar-Rodriguez JM, Hickstein DD, Holland SM. Pulmonary Manifestations of GATA2 Deficiency. Chest 2021; 160:1350-1359. [PMID: 34089740 PMCID: PMC8546236 DOI: 10.1016/j.chest.2021.05.046] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 05/10/2021] [Accepted: 05/15/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND GATA2 deficiency is a genetic disorder of hematopoiesis, lymphatics, and immunity caused by autosomal dominant or sporadic mutations in GATA2. The disease has a broad phenotype encompassing immunodeficiency, myelodysplasia, leukemia, and vascular or lymphatic dysfunction as well as prominent pulmonary manifestations. RESEARCH QUESTION What are the pulmonary manifestations of GATA2 deficiency? STUDY DESIGN AND METHODS A retrospective review was conducted of clinical medical records, diagnostic imaging, pulmonary pathologic specimens, and tests of pulmonary function. RESULTS Of 124 patients (95 probands and 29 ascertained), the lung was affected in 56%. In addition to chronic infections, pulmonary alveolar proteinosis (11 probands) and pulmonary arterial hypertension (nine probands) were present. Thoracic CT imaging found small nodules in 54% (54 probands and 12 relatives), reticular infiltrates in 40% (45 probands and four relatives), paraseptal emphysema in 25% (30 probands and one relative), ground-glass opacities in 35% (41 probands and two relatives), consolidation in 21% (23 probands and two relatives), and a typical crazy-paving pattern in 7% (eight probands and no relatives). Nontuberculous mycobacteria were the most frequent organisms associated with chronic infection. Allogeneic hematopoietic stem cell transplantation successfully reversed myelodysplasia and immune deficiency and also improved pulmonary hypertension and pulmonary alveolar proteinosis in most patients. INTERPRETATION GATA2 deficiency has prominent pulmonary manifestations. These clinical observations confirm the essential role of hematopoietic cells in many aspects of pulmonary function, including infections, alveolar proteinosis, and pulmonary hypertension, many of which precede the formal diagnosis, and many of which respond to stem cell transplantation.
Collapse
Affiliation(s)
- Beatriz E Marciano
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Kenneth N Olivier
- Pulmonary Branch, National Heart, Lung and Blood Institute, National Cancer Institute, National Institutes of Health, Bethesda, MD.
| | - Les R Folio
- Department of Radiology and Imaging Sciences, Clinical Center, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Christa S Zerbe
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Amy P Hsu
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Alexandra F Freeman
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Armando C Filie
- Cytology Services Laboratory Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Michael A Spinner
- Division of Oncology, Department of Medicine, Stanford University, Stanford
| | - Lauren A Sanchez
- Division of Allergy, Immunology, and Bone Marrow Transplantation, Department of Pediatrics, University of California, San Francisco, CA
| | - Jana P Lovell
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Mark Parta
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jennifer M Cuellar-Rodriguez
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Dennis D Hickstein
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Steven M Holland
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
8
|
Ambade AS, Hassoun PM, Damico RL. Basement Membrane Extracellular Matrix Proteins in Pulmonary Vascular and Right Ventricular Remodeling in Pulmonary Hypertension. Am J Respir Cell Mol Biol 2021; 65:245-258. [PMID: 34129804 PMCID: PMC8485997 DOI: 10.1165/rcmb.2021-0091tr] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/14/2021] [Indexed: 12/13/2022] Open
Abstract
The extracellular matrix (ECM), a highly organized network of structural and nonstructural proteins, plays a pivotal role in cellular and tissue homeostasis. Changes in the ECM are critical for normal tissue repair, whereas dysregulation contributes to aberrant tissue remodeling. Pulmonary arterial hypertension is a severe disorder of the pulmonary vasculature characterized by pathologic remodeling of the pulmonary vasculature and right ventricle, increased production and deposition of structural and nonstructural proteins, and altered expression of ECM growth factors and proteases. Furthermore, ECM remodeling plays a significant role in disease progression, as several dynamic changes in its composition, quantity, and organization are documented in both humans and animal models of disease. These ECM changes impact vascular cell biology and affect proliferation of resident cells. Furthermore, ECM components determine the tissue architecture of the pulmonary and myocardial vasculature as well as the myocardium itself and provide mechanical stability crucial for tissue homeostasis. However, little is known about the basement membrane (BM), a specialized, self-assembled conglomerate of ECM proteins, during remodeling. In the vasculature, the BM is in close physical association with the vascular endothelium and smooth muscle cells. While in the myocardium, each cardiomyocyte is enclosed by a BM that serves as the interface between cardiomyocytes and the surrounding interstitial matrix. In this review, we provide a brief overview on the current state of knowledge of the BM and its ECM composition and their impact on pulmonary vascular remodeling and right ventricle dysfunction and failure in pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Anjira S Ambade
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Paul M Hassoun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Rachel L Damico
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
9
|
Comparative analysis on the anti-inflammatory/immune effect of mesenchymal stem cell therapy for the treatment of pulmonary arterial hypertension. Sci Rep 2021; 11:2012. [PMID: 33479312 PMCID: PMC7820276 DOI: 10.1038/s41598-021-81244-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 12/30/2020] [Indexed: 02/06/2023] Open
Abstract
Despite the advancement of targeted therapy for pulmonary arterial hypertension (PAH), poor prognosis remains a reality. Mesenchymal stem cells (MSCs) are one of the most clinically feasible alternative treatment options. We compared the treatment effects of adipose tissue (AD)-, bone marrow (BD)-, and umbilical cord blood (UCB)-derived MSCs in the rat monocrotaline-induced pulmonary hypertension (PH) model. The greatest improvement in the right ventricular function was observed in the UCB-MSCs treated group. The UCB-MSCs treated group also exhibited the greatest improvement in terms of the largest decrease in the medial wall thickness, perivascular fibrosis, and vascular cell proliferation, as well as the lowest levels of recruitment of innate and adaptive immune cells and associated inflammatory cytokines. Gene expression profiling of lung tissue confirmed that the UCB-MSCs treated group had the most notably attenuated immune and inflammatory profiles. Network analysis further revealed that the UCB-MSCs group had the greatest therapeutic effect in terms of the normalization of all three classical PAH pathways. The intravenous injection of the UCB-MSCs, compared with those of other MSCs, showed superior therapeutic effects in the PH model for the (1) right ventricular function, (2) vascular remodeling, (3) immune/inflammatory profiles, and (4) classical PAH pathways.
Collapse
|
10
|
Zawia A, Arnold ND, West L, Pickworth JA, Turton H, Iremonger J, Braithwaite AT, Cañedo J, Johnston SA, Thompson AAR, Miller G, Lawrie A. Altered Macrophage Polarization Induces Experimental Pulmonary Hypertension and Is Observed in Patients With Pulmonary Arterial Hypertension. Arterioscler Thromb Vasc Biol 2020; 41:430-445. [PMID: 33147993 PMCID: PMC7752239 DOI: 10.1161/atvbaha.120.314639] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Supplemental Digital Content is available in the text. To determine whether global reduction of CD68 (cluster of differentiation) macrophages impacts the development of experimental pulmonary arterial hypertension (PAH) and whether this reduction affects the balance of pro- and anti-inflammatory macrophages within the lung. Additionally, to determine whether there is evidence of an altered macrophage polarization in patients with PAH.
Collapse
Affiliation(s)
- Amira Zawia
- Department of Infection, Immunity and Cardiovascular Disease (A.Z., N.D.A., L.W., J.A.P., H.T., J.I., A.T.B., J.C., S.A.J., A.A.R.T., A.L.), University of Sheffield, United Kingdom
| | - Nadine D Arnold
- Department of Infection, Immunity and Cardiovascular Disease (A.Z., N.D.A., L.W., J.A.P., H.T., J.I., A.T.B., J.C., S.A.J., A.A.R.T., A.L.), University of Sheffield, United Kingdom
| | - Laura West
- Department of Infection, Immunity and Cardiovascular Disease (A.Z., N.D.A., L.W., J.A.P., H.T., J.I., A.T.B., J.C., S.A.J., A.A.R.T., A.L.), University of Sheffield, United Kingdom
| | - Josephine A Pickworth
- Department of Infection, Immunity and Cardiovascular Disease (A.Z., N.D.A., L.W., J.A.P., H.T., J.I., A.T.B., J.C., S.A.J., A.A.R.T., A.L.), University of Sheffield, United Kingdom
| | - Helena Turton
- Department of Infection, Immunity and Cardiovascular Disease (A.Z., N.D.A., L.W., J.A.P., H.T., J.I., A.T.B., J.C., S.A.J., A.A.R.T., A.L.), University of Sheffield, United Kingdom
| | - James Iremonger
- Department of Infection, Immunity and Cardiovascular Disease (A.Z., N.D.A., L.W., J.A.P., H.T., J.I., A.T.B., J.C., S.A.J., A.A.R.T., A.L.), University of Sheffield, United Kingdom
| | - Adam T Braithwaite
- Department of Infection, Immunity and Cardiovascular Disease (A.Z., N.D.A., L.W., J.A.P., H.T., J.I., A.T.B., J.C., S.A.J., A.A.R.T., A.L.), University of Sheffield, United Kingdom
| | - Jaime Cañedo
- Department of Infection, Immunity and Cardiovascular Disease (A.Z., N.D.A., L.W., J.A.P., H.T., J.I., A.T.B., J.C., S.A.J., A.A.R.T., A.L.), University of Sheffield, United Kingdom
| | - Simon A Johnston
- Department of Infection, Immunity and Cardiovascular Disease (A.Z., N.D.A., L.W., J.A.P., H.T., J.I., A.T.B., J.C., S.A.J., A.A.R.T., A.L.), University of Sheffield, United Kingdom
| | - A A Roger Thompson
- Department of Infection, Immunity and Cardiovascular Disease (A.Z., N.D.A., L.W., J.A.P., H.T., J.I., A.T.B., J.C., S.A.J., A.A.R.T., A.L.), University of Sheffield, United Kingdom
| | - Gaynor Miller
- Department of Oncology and Metabolism (G.M.), University of Sheffield, United Kingdom.,College of Medical and Dental Science, University of Birmingham, United Kingdom (G.M.)
| | - Allan Lawrie
- Department of Infection, Immunity and Cardiovascular Disease (A.Z., N.D.A., L.W., J.A.P., H.T., J.I., A.T.B., J.C., S.A.J., A.A.R.T., A.L.), University of Sheffield, United Kingdom
| |
Collapse
|
11
|
Bisserier M, Pradhan N, Hadri L. Current and emerging therapeutic approaches to pulmonary hypertension. Rev Cardiovasc Med 2020; 21:163-179. [PMID: 32706206 PMCID: PMC7389678 DOI: 10.31083/j.rcm.2020.02.597] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 05/25/2020] [Indexed: 12/15/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive and fatal lung disease of multifactorial etiology. Most of the available drugs and FDA-approved therapies for treating pulmonary hypertension attempt to overcome the imbalance between vasoactive and vasodilator mediators, and restore the endothelial cell function. Traditional medications for treating PAH include the prostacyclin analogs and receptor agonists, phosphodiesterase 5 inhibitors, endothelin-receptor antagonists, and cGMP activators. While the current FDA-approved drugs showed improvements in quality of life and hemodynamic parameters, they have shown only very limited beneficial effects on survival and disease progression. None of them offers a cure against PAH, and the median survival rate remains less than three years from diagnosis. Extensive research efforts have led to the emergence of innovative therapeutic approaches in the area of PAH. In this review, we provide an overview of the current FDA-approved therapies in PAH and discuss the associated clinical trials and reported-side effects. As recent studies have led to the emergence of innovative therapeutic approaches in the area of PAH, we also focus on the latest promising therapies in preclinical studies such as stem cell-based therapies, gene transfer, and epigenetic therapies.
Collapse
Affiliation(s)
- Malik Bisserier
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Natasha Pradhan
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Lahouaria Hadri
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
12
|
Ouyang S, Chen W, Zeng G, Lei C. Aquaporin-2 expression in the kidney and urine is elevated in rats with monocrotaline-induced pulmonary heart disease. J Int Med Res 2020; 48:300060519894448. [PMID: 32000538 PMCID: PMC7114290 DOI: 10.1177/0300060519894448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Objective Little is known about how renal aquaporin-2 (AQP2) expression is affected by right heart failure caused by pulmonary heart disease (PHD). Therefore, we examined the expression of AQP2 in a rat model of PHD induced by monocrotaline (MCT). Methods After 4 weeks of treatment, urine and blood samples were collected from sham-treated and MCT-treated rats. Plasma arginine vasopressin (AVP) levels were measured by radioimmunoassay, and kidney Aqp2 mRNA expression was detected by reverse transcription (RT)-PCR. Kidney AQP2 protein expression was quantified by immunohistochemistry and western blotting assays. The concentration of urine AQP2 was determined by indirect enzyme-linked immunosorbent assay. Results We successfully established an animal model of MCT-induced PHD in rats. MCT-treated rats had significantly higher mRNA and protein levels of AQP2 in their kidney tissue. Following MCT treatment, rats also had markedly increased concentrations of both urine AQP2 and plasma AVP. Conclusions AQP2 expression was significantly increased in the kidney tissues and urine of rats with PHD induced by MCT. Our findings suggest that the evaluation of AQP2 expression contributes to an early diagnosis of PHD, and may also be an important reference to improve PHD therapeutics.
Collapse
Affiliation(s)
- Shao Ouyang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Wei Chen
- Department of Respiratory Medicine, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Gaofeng Zeng
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Changcheng Lei
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
| |
Collapse
|
13
|
Muhammad SA, Abbas AY, Saidu Y, Fakurazi S, Bilbis LS. Therapeutic efficacy of mesenchymal stromal cells and secretome in pulmonary arterial hypertension: A systematic review and meta-analysis. Biochimie 2019; 168:156-168. [PMID: 31678635 DOI: 10.1016/j.biochi.2019.10.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 10/28/2019] [Indexed: 12/12/2022]
Abstract
Mesenchymal stromal cells (MSCs) and secretome are promising therapies for pulmonary arterial hypertension (PAH). This meta-analysis aimed to provide a precise estimate and compare the therapeutic efficacy of MSC and secretome in PAH. We searched six databases (CINAHL, Cochrane, Ovid Medline, PubMed, Science Direct and Scopus) until December 2018 using search terms related to MSCs, secretome and PAH. Twenty-three studies were included for the meta-analysis. The effect size of pulmonary hemodynamics and right ventricular hypertrophy markers was estimated using random effects model. MSCs and secretome significantly improved pulmonary hemodynamics and right ventricular hypertrophy compared to control. Comparison between MSCs and secretome indicate no significant difference in reducing right ventricular systolic pressure (RVSP) and medial wall thickening (MWT). However, treatment of PAH with secretome significantly improved mean pulmonary arterial pressure (mPAP) (p = 0.018) and right ventricular/left ventricular + septum (RV/LV+S) (p = 0.017) better than MSCs. Meta-regression shows that cell type (p = 0.034) is a predictor of MSCs to reduce RVSP in PAH. Similarly, the effect of secretome on MWT was significantly (p = 0.011) better at 4 weeks compared to 2 weeks of intervention. The overall risk of bias ranges from low to moderate; however, some of the essential elements required in reports of animal trials were not reported. There was evidence of publication bias for RV/LV+S and MWT, but not RVSP. This meta-analysis provides evidence of the therapeutic benefits of MSCs and secretome in PAH and the effect of secretome was similar or superior to MSCs.
Collapse
Affiliation(s)
| | | | - Yusuf Saidu
- Department of Biochemistry, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Sharida Fakurazi
- Department of Human Anatomy, Universiti Putra Malaysia, Malaysia
| | | |
Collapse
|
14
|
Abstract
Introduction: Mesenchymal stem/stromal cells (MSCs) have been shown to improve lung function and survival in chronic inflammatory lung diseases, including asthma, chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), pulmonary arterial hypertension (PAH), and silicosis.Areas covered: This review covers rationale for the use of MSC therapy, along with preclinical studies and clinical trials with MSC therapy in chronic lung diseases.Expert opinion: MSC therapy holds promise for the treatment of chronic lung diseases, mainly when administered at early stages. In clinical trials, MSC administration was safe, but associated with limited effects on clinical outcomes. Further studies are required to elucidate unresolved issues, including optimal MSC source and dose, route of administration, and frequency (single vs. multiple-dose regimens). A better understanding of the mechanisms of MSC action, local microenvironment of each disease, and development of strategies to potentiate the beneficial effects of MSCs may improve outcomes.
Collapse
|
15
|
Fukumitsu M, Suzuki K. Mesenchymal stem/stromal cell therapy for pulmonary arterial hypertension: Comprehensive review of preclinical studies. J Cardiol 2019; 74:304-312. [PMID: 31109735 DOI: 10.1016/j.jjcc.2019.04.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 03/12/2019] [Accepted: 04/06/2019] [Indexed: 12/21/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a disease characterized by progressive pulmonary vascular remodeling, resulting in right-sided heart failure and premature death. Current available therapies for PAH have limited efficacy, and new therapeutic strategies need to be developed. Mesenchymal stem/stromal cells (MSCs) may offer a novel therapeutic approach to PAH. Since the first report in 2006, a number of preclinical studies have demonstrated a potential therapeutic effect of this approach, with attenuated hemodynamic and histological progression of PAH, in animal models of PAH. However, there remain several issues that should be addressed for this approach to be clinically successful. With the aim to highlight such issues, this review clarifies existing knowledge on MSC therapy for PAH in preclinical studies, including types of PAH animal models used for MSC therapy, MSC sources, and administration protocol (route, cell dose, and timing of administration). This review thereafter summarizes thoroughly and discusses the mechanism underpinning MSC therapy for PAH. For clinical success of MSC therapy, insufficient evidence of safety (e.g. critical risk of pulmonary embolism) and therapeutic efficacy of MSCs on established PAH with severe vascular remodeling, as well as further optimization of the MSC administration protocol, are considered as remaining issues to be addressed. In terms of the efficacy, it is controversial whether angiogenic cytokines, which are considered as one of the therapeutic mechanisms of MSC, have beneficial effect for human PAH. To address these issues, further preclinical data using more clinically-relevant animal models of PAH, such as SU5416 model, should be accumulated, whereas most preclinical studies have been conducted using monocrotaline-induced PAH model. While MSC therapy has a great potential to become a novel therapy in PAH, continuing careful preclinical research is warranted for clinical success in PAH.
Collapse
Affiliation(s)
- Masafumi Fukumitsu
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Ken Suzuki
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.
| |
Collapse
|
16
|
Suen CM, Stewart DJ, Montroy J, Welsh C, Levac B, Wesch N, Zhai A, Fergusson D, McIntyre L, Lalu MM. Regenerative cell therapy for pulmonary arterial hypertension in animal models: a systematic review. Stem Cell Res Ther 2019; 10:75. [PMID: 30841915 PMCID: PMC6404277 DOI: 10.1186/s13287-019-1172-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/30/2019] [Accepted: 02/11/2019] [Indexed: 12/21/2022] Open
Abstract
Background Pulmonary arterial hypertension (PAH) is a rare disease characterized by widespread loss of the pulmonary microcirculation and elevated pulmonary arterial pressures leading to pathological right ventricular remodeling and ultimately right heart failure. Regenerative cell therapies could potentially restore the effective lung microcirculation and provide a curative therapy for PAH. The objective of this systematic review was to compare the efficacy of regenerative cell therapies in preclinical models of PAH. Methods A systematic search strategy was developed and executed. We included preclinical animal studies using regenerative cell therapy in experimental models of PAH. Primary outcomes were right ventricular systolic pressure (RVSP) and mean pulmonary arterial pressure (mPAP). The secondary outcome was right ventricle/left ventricle + septum weight ratio (RV/LV+S). Pooled effect sizes were undertaken using random effects inverse variance models. Risk of bias and publication bias were assessed. Results The systematic search yielded 1285 studies, of which 44 met eligibility criteria. Treatment with regenerative cell therapy was associated with decreased RVSP (SMD − 2.10; 95% CI − 2.59 to − 1.60), mPAP (SMD − 2.16; 95% CI − 2.97 to − 1.35), and RV/LV+S (SMD − 1.31, 95% CI − 1.64 to − 0.97). Subgroup analysis demonstrated that cell modification resulted in greater reduction in RVSP. The effects on RVSP and mPAP remained statistically significant even after adjustment for publication bias. The majority of studies had an unclear risk of bias. Conclusions Preclinical studies of regenerative cell therapy demonstrated efficacy in animal models of PAH; however, future studies should consider incorporating design elements to reduce the risk of bias. Systematic review registration Suen CM, Zhai A, Lalu MM, Welsh C, Levac BM, Fergusson D, McIntyre L and Stewart DJ. Efficacy and safety of regenerative cell therapy for pulmonary arterial hypertension in animal models: a preclinical systematic review protocol. Syst Rev. 2016;5:89. Trial registration CAMARADES-NC3Rs Preclinical Systematic Review & Meta-analysis Facility (SyRF). http://syrf.org.uk/protocols/. Syst Rev 5:89, 2016 Electronic supplementary material The online version of this article (10.1186/s13287-019-1172-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Colin M Suen
- Regenerative Medicine Program, The Ottawa Hospital Research Institute, 501 Smyth Road, PO Box 201B, Ottawa, ON, K1H 8L6, Canada.,Department of Cell and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Duncan J Stewart
- Regenerative Medicine Program, The Ottawa Hospital Research Institute, 501 Smyth Road, PO Box 201B, Ottawa, ON, K1H 8L6, Canada.,Department of Cell and Molecular Medicine, University of Ottawa, Ottawa, Canada.,Department of Medicine, University of Ottawa, Ottawa, Canada
| | - Joshua Montroy
- Clinical Epidemiology Program, Ottawa, Canada.,Blueprint Translational Research Group, The Ottawa Hospital Research Institute, Ottawa, Canada
| | | | - Brendan Levac
- Department of Medicine, University of Ottawa, Ottawa, Canada
| | - Neil Wesch
- Clinical Epidemiology Program, Ottawa, Canada.,Blueprint Translational Research Group, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Alexander Zhai
- Regenerative Medicine Program, The Ottawa Hospital Research Institute, 501 Smyth Road, PO Box 201B, Ottawa, ON, K1H 8L6, Canada
| | - Dean Fergusson
- Clinical Epidemiology Program, Ottawa, Canada.,Blueprint Translational Research Group, The Ottawa Hospital Research Institute, Ottawa, Canada.,Department of Medicine, University of Ottawa, Ottawa, Canada.,Depatrment of Surgery, University of Ottawa, Ottawa, Canada.,Department of Epidemiology and Community Medicine, University of Ottawa, Ottawa, Canada
| | - Lauralyn McIntyre
- Clinical Epidemiology Program, Ottawa, Canada.,Blueprint Translational Research Group, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Manoj M Lalu
- Regenerative Medicine Program, The Ottawa Hospital Research Institute, 501 Smyth Road, PO Box 201B, Ottawa, ON, K1H 8L6, Canada. .,Department of Cell and Molecular Medicine, University of Ottawa, Ottawa, Canada. .,Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, The Ottawa Hospital Research Institute, Ottawa, Canada. .,Clinical Epidemiology Program, Ottawa, Canada. .,Blueprint Translational Research Group, The Ottawa Hospital Research Institute, Ottawa, Canada.
| |
Collapse
|
17
|
Schmuck EG, Hacker TA, Schreier DA, Chesler NC, Wang Z. Beneficial effects of mesenchymal stem cell delivery via a novel cardiac bioscaffold on right ventricles of pulmonary arterial hypertensive rats. Am J Physiol Heart Circ Physiol 2019; 316:H1005-H1013. [PMID: 30822119 DOI: 10.1152/ajpheart.00091.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Right ventricular failure (RVF) is a common cause of death in patients suffering from pulmonary arterial hypertension (PAH). The current treatment for PAH only moderately improves symptoms, and RVF ultimately occurs. Therefore, it is necessary to develop new treatment strategies to protect against right ventricle (RV) maladaptation despite PAH progression. In this study, we hypothesize that local mesenchymal stem cell (MSC) delivery via a novel bioscaffold can improve RV function despite persistent PAH. To test our hypothesis, we induced PAH in adult rats with SU5416 and chronic hypoxia exposure; treated with rat MSCs delivered by intravenous injection, intramyocardial injection, or epicardial placement of a bioscaffold; and then examined treatment effectiveness by in vivo pressure-volume measurement, echocardiography, histology, and immunohistochemistry. Our results showed that compared with other treatment groups, only the MSC-seeded bioscaffold group resulted in RV functional improvement, including restored stroke volume, cardiac output, and improved stroke work. Diastolic function indicated by end-diastolic pressure-volume relationship was improved by the local MSC treatments or bioscaffold alone. Cardiomyocyte hypertrophy and RV fibrosis were both reduced, and von Willebrand factor expression was restored by the MSC-seeded bioscaffold treatment. Overall, our study suggests a potential new regenerative therapy to rescue the pressure-overload failing RV with persistent pulmonary vascular disease, which may improve quality of life and/or survival of PAH patients. NEW & NOTEWORTHY We explored the effects of mesenchymal stem cell-seeded bioscaffold on right ventricles (RVs) of rats with established pulmonary arterial hypertension (PAH). Some beneficial effects were observed despite persistent PAH, suggesting that this may be a new therapy for RV to improve quality of life and/or survival of PAH patients.
Collapse
Affiliation(s)
- Eric G Schmuck
- Department of Medicine, University of Wisconsin , Madison, Wisconsin
| | - Timothy A Hacker
- Department of Medicine, University of Wisconsin , Madison, Wisconsin
| | - David A Schreier
- Department of Biomedical Engineering, University of Wisconsin , Madison, Wisconsin
| | - Naomi C Chesler
- Department of Medicine, University of Wisconsin , Madison, Wisconsin.,Department of Biomedical Engineering, University of Wisconsin , Madison, Wisconsin
| | - Zhijie Wang
- Department of Biomedical Engineering, University of Wisconsin , Madison, Wisconsin.,Department of Mechanical Engineering, Colorado State University , Fort Collins, Colorado
| |
Collapse
|
18
|
Ding XF, Liang HY, Yuan B, Li LF, Wang T, Kan QC, Wang LX, Sun TW. Efficacy of stem cell therapy for pulmonary arterial hypertension: a systematic review and meta-analysis of preclinical studies. Stem Cell Res Ther 2019; 10:55. [PMID: 30760312 PMCID: PMC6374914 DOI: 10.1186/s13287-019-1162-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 01/06/2019] [Accepted: 02/04/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Despite significant progress in drug treatment, the prognosis of patients with advanced pulmonary arterial hypertension (PAH) remains extremely poor. Many preclinical studies have reported the efficacy of stem cell (SC) therapy for PAH; however, this approach remains controversial. The aim of this systematic review and meta-analysis is to assess the potential efficacy of SC therapy for PAH. METHODS The Medline, EMBASE, Cochrane Library, and Web of Science databases were searched from inception to August 12, 2018. Preclinical studies that evaluated the use of SC therapy for PAH were included. The primary outcome was pulmonary haemodynamics, as assessed by measurement of the right ventricular systolic pressure (RVSP), mean pulmonary arterial pressure (mPAP), and/or mean right ventricle pressure (mRVP). The secondary outcomes included the weight ratio of the right ventricle to the left ventricle plus septum (RV/LV+S), the right ventricle to body weight ratio (RV/BW), the percentage of pulmonary arteriole area index (WA), and/or the percentage of medial wall thickness of the pulmonary arteriole (WT). The quality of outcomes was evaluated using the SYstematic Review Centre for Laboratory animal Experimentation (SYRCLE) bias risk tool. The inverse-variance method with random-effects modelling was used to calculate pooled weighted mean differences (WMDs) and 95% CIs. Statistical analysis was performed with STATA 14.0. RESULTS Twenty-eight eligible articles (722 animals) were included. SC therapy reduced the pooled WMDs (95% CIs) of RVSP, mPAP, mRVP, RV/LV+S, RV/BW, WA, and WT for animals with PAH, with values of - 14.12 (- 14.63, - 13.61), - 11.86 (- 12.35, - 11.36), - 17.33 (- 18.10, - 16.56), - 0.10 (- 0.10, - 0.09), 0.23 (0.21, 0.24), - 13.66 (- 15.71, - 11.62), and - 7.96 (- 7.99, - 7.93), respectively. CONCLUSIONS SC therapy is effective for PAH in preclinical studies. These results may help to standardise preclinical animal studies and provide a theoretical basis for clinical trial design in the future. SYSTEMATIC REVIEW REGISTRATION PROSPERO ( http://www.crd.york.ac.uk/PROSPERO ).
Collapse
Affiliation(s)
- Xian-Fei Ding
- General ICU, Henan Key Laboratory of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
| | - Huo-Yan Liang
- General ICU, Henan Key Laboratory of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
| | - Bo Yuan
- General ICU, Henan Key Laboratory of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
| | - Li-Feng Li
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
| | - Tian Wang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
| | - Quan-Cheng Kan
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
| | - Le-Xin Wang
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW 2650 Australia
| | - Tong-Wen Sun
- General ICU, Henan Key Laboratory of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
| |
Collapse
|
19
|
Burgess JK, Heijink IH. Paving the Road for Mesenchymal Stem Cell-Derived Exosome Therapy in Bronchopulmonary Dysplasia and Pulmonary Hypertension. STEM CELL-BASED THERAPY FOR LUNG DISEASE 2019. [PMCID: PMC7122497 DOI: 10.1007/978-3-030-29403-8_8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic neonatal lung disease characterized by inflammation and arrest of alveolarization. Its common sequela, pulmonary hypertension (PH), presents with elevated pulmonary vascular resistance associated with remodeling of the pulmonary arterioles. Despite notable advancements in neonatal medicine, there is a severe lack of curative treatments to help manage the progressive nature of these diseases. Numerous studies in preclinical models of BPD and PH have demonstrated that therapies based on mesenchymal stem/stromal cells (MSCs) can resolve pulmonary inflammation and ameliorate the severity of disease. Recent evidence suggests that novel, cell-free approaches based on MSC-derived exosomes (MEx) might represent a compelling therapeutic alternative offering major advantages over treatments based on MSC transplantation. Here, we will discuss the development of MSC-based therapies, stressing the centrality of paracrine action as the actual vector of MSC therapeutic functionality, focusing on MEx. We will briefly present our current understanding of the biogenesis and secretion of MEx, and discuss potential mechanisms by which they afford such beneficial effects, including immunomodulation and restoration of homeostasis in diseased states. We will also review ongoing clinical trials using MSCs as treatment for BPD that pave the way for bringing cell-free, MEx-based therapeutics from the bench to the NICU setting.
Collapse
Affiliation(s)
- Janette K. Burgess
- The University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| | - Irene H. Heijink
- The University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| |
Collapse
|
20
|
Willis GR, Fernandez-Gonzalez A, Reis M, Mitsialis SA, Kourembanas S. Macrophage Immunomodulation: The Gatekeeper for Mesenchymal Stem Cell Derived-Exosomes in Pulmonary Arterial Hypertension? Int J Mol Sci 2018; 19:ijms19092534. [PMID: 30150544 PMCID: PMC6164282 DOI: 10.3390/ijms19092534] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 08/21/2018] [Accepted: 08/22/2018] [Indexed: 12/13/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease characterized by remodeling of the pulmonary arteries, increased pulmonary infiltrates, loss of vascular cross-sectional area, and elevated pulmonary vascular resistance. Despite recent advances in the management of PAH, there is a pressing need for the development of new tools to effectively treat and reduce the risk of further complications. Dysregulated immunity underlies the development of PAH, and macrophages orchestrate both the initiation and resolution of pulmonary inflammation, thus, manipulation of lung macrophage function represents an attractive target for emerging immunomodulatory therapies, including cell-based approaches. Indeed, mesenchymal stem cell (MSC)-based therapies have shown promise, effectively modulating the macrophage fulcrum to favor an anti-inflammatory, pro-resolving phenotype, which is associated with both histological and functional benefits in preclinical models of pulmonary hypertension (PH). The complex interplay between immune system homeostasis and MSCs remains incompletely understood. Here, we highlight the importance of macrophage function in models of PH and summarize the development of MSC-based therapies, focusing on the significance of MSC exosomes (MEx) and the immunomodulatory and homeostatic mechanisms by which such therapies may afford their beneficial effects.
Collapse
Affiliation(s)
- Gareth R Willis
- Division of Newborn Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| | - Angeles Fernandez-Gonzalez
- Division of Newborn Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| | - Monica Reis
- Division of Newborn Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| | - S Alex Mitsialis
- Division of Newborn Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| | - Stella Kourembanas
- Division of Newborn Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
21
|
Myeloid-Derived Suppressor Cells and Pulmonary Hypertension. Int J Mol Sci 2018; 19:ijms19082277. [PMID: 30081463 PMCID: PMC6121540 DOI: 10.3390/ijms19082277] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 07/31/2018] [Accepted: 08/01/2018] [Indexed: 01/04/2023] Open
Abstract
Myeloid–derived suppressor cells (MDSCs) comprised a heterogeneous subset of bone marrow–derived myeloid cells, best studied in cancer research, that are increasingly implicated in the pathogenesis of pulmonary vascular remodeling and the development of pulmonary hypertension. Stem cell transplantation represents one extreme interventional strategy for ablating the myeloid compartment but poses a number of translational challenges. There remains an outstanding need for additional therapeutic targets to impact MDSC function, including the potential to alter interactions with innate and adaptive immune subsets, or alternatively, alter trafficking receptors, metabolic pathways, and transcription factor signaling with readily available and safe drugs. In this review, we summarize the current literature on the role of myeloid cells in the development of pulmonary hypertension, first in pulmonary circulation changes associated with myelodysplastic syndromes, and then by examining intrinsic myeloid cell changes that contribute to disease progression in pulmonary hypertension. We then outline several tractable targets and pathways relevant to pulmonary hypertension via MDSC regulation. Identifying these MDSC-regulated effectors is part of an ongoing effort to impact the field of pulmonary hypertension research through identification of myeloid compartment-specific therapeutic applications in the treatment of pulmonary vasculopathies.
Collapse
|
22
|
Chaubey S, Thueson S, Ponnalagu D, Alam MA, Gheorghe CP, Aghai Z, Singh H, Bhandari V. Early gestational mesenchymal stem cell secretome attenuates experimental bronchopulmonary dysplasia in part via exosome-associated factor TSG-6. Stem Cell Res Ther 2018; 9:173. [PMID: 29941022 PMCID: PMC6019224 DOI: 10.1186/s13287-018-0903-4] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 04/25/2018] [Accepted: 05/13/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are promising tools for the treatment of human lung disease and other pathologies relevant to newborn medicine. Recent studies have established MSC exosomes (EXO), as one of the main therapeutic vectors of MSCs in mouse models of multifactorial chronic lung disease of preterm infants, bronchopulmonary dysplasia (BPD). However, the mechanisms underlying MSC-EXO therapeutic action are not completely understood. Using a neonatal mouse model of human BPD, we evaluated the therapeutic efficiency of early gestational age (GA) human umbilical cord (hUC)-derived MSC EXO fraction and its exosomal factor, tumor necrosis factor alpha-stimulated gene-6 (TSG-6). METHODS Conditioned media (CM) and EXO fractions were isolated from 25 and 30 weeks GA hUC-MSC cultures grown in serum-free media (SFM) for 24 h. Newborn mice were exposed to hyperoxia (> 95% oxygen) and were given intraperitoneal injections of MSC-CM or MSC-CM EXO fractions at postnatal (PN) day 2 and PN4. They were then returned to room air until PN14 (in a mouse model of severe BPD). The treatment regime was followed with (rh)TSG-6, TSG-6-neutralizing antibody (NAb), TSG-6 (si)RNA-transfected MSC-CM EXO and their appropriate controls. Echocardiography was done at PN14 followed by harvesting of lung, heart and brain for assessment of pathology parameters. RESULTS Systemic administration of CM or EXO in the neonatal BPD mouse model resulted in robust improvement in lung, cardiac and brain pathology. Hyperoxia-exposed BPD mice exhibited pulmonary inflammation accompanied by alveolar-capillary leakage, increased chord length, and alveolar simplification, which was ameliorated by MSC CM/EXO treatment. Pulmonary hypertension and right ventricular hypertrophy was also corrected. Cell death in brain was decreased and the hypomyelination reversed. Importantly, we detected TSG-6, an immunomodulatory glycoprotein, in EXO. Administration of TSG-6 attenuated BPD and its associated pathologies, in lung, heart and brain. Knockdown of TSG-6 by NAb or by siRNA in EXO abrogated the therapeutic effects of EXO, suggesting TSG-6 as an important therapeutic molecule. CONCLUSIONS Preterm hUC-derived MSC-CM EXO alleviates hyperoxia-induced BPD and its associated pathologies, in part, via exosomal factor TSG-6. The work indicates early systemic intervention with TSG-6 as a robust option for cell-free therapy, particularly for treating BPD.
Collapse
Affiliation(s)
- Sushma Chaubey
- Section of Neonatal-Perinatal Medicine, Department of Pediatrics, Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA, 19102, USA
| | - Sam Thueson
- Section of Neonatal-Perinatal Medicine, Department of Pediatrics, Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA, 19102, USA
| | - Devasena Ponnalagu
- Department of Pharmacology & Physiology, Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA, 19102, USA
| | - Mohammad Afaque Alam
- Section of Neonatal-Perinatal Medicine, Department of Pediatrics, Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA, 19102, USA
| | - Ciprian P Gheorghe
- Department of Obstetrics and Gynecology, Loma Linda University School of Medicine, 11370 Anderson Street, Loma Linda, CA, 92354, USA
| | - Zubair Aghai
- Divison of Neonatology, Department of Pediatrics, Thomas Jefferson University Hospital, 132S, 10th Street, Philadelphia, PA, 19107, USA
| | - Harpreet Singh
- Department of Pharmacology & Physiology, Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA, 19102, USA.,Department of Medicine, Division of Cardiology, Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA, 19102, USA
| | - Vineet Bhandari
- Section of Neonatal-Perinatal Medicine, Department of Pediatrics, Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA, 19102, USA.
| |
Collapse
|
23
|
Loisel F, Provost B, Haddad F, Guihaire J, Amsallem M, Vrtovec B, Fadel E, Uzan G, Mercier O. Stem cell therapy targeting the right ventricle in pulmonary arterial hypertension: is it a potential avenue of therapy? Pulm Circ 2018; 8:2045893218755979. [PMID: 29480154 PMCID: PMC5844533 DOI: 10.1177/2045893218755979] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is an incurable disease characterized by an increase in pulmonary arterial pressure due to pathological changes to the pulmonary vascular bed. As a result, the right ventricle (RV) is subject to an increased afterload and undergoes multiple changes, including a decrease in capillary density. All of these dysfunctions lead to RV failure. A number of studies have shown that RV function is one of the main prognostic factors for PAH patients. Many stem cell therapies targeting the left ventricle are currently undergoing development. The promising results observed in animal models have led to clinical trials that have shown an improvement of cardiac function. In contrast to left heart disease, stem cell therapy applied to the RV has remained poorly studied, even though it too may provide a therapeutic benefit. In this review, we discuss stem cell therapy as a treatment for RV failure in PAH. We provide an overview of the results of preclinical and clinical studies for RV cell therapies. Although a large number of studies have targeted the pulmonary circulation rather than the RV directly, there are nonetheless encouraging results in the literature that indicate that cell therapies may have a direct beneficial effect on RV function. This cell therapy strategy may therefore hold great promise and warrants further studies in PAH patients.
Collapse
Affiliation(s)
- Fanny Loisel
- 1 36705 Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Universite Paris Sud, Paris-Saclay University, Le Plessis Robinson, France.,2 Inserm 1197 Research Unit, Universite Paris Sud, Paris-Saclay University, Villejuif, France
| | - Bastien Provost
- 1 36705 Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Universite Paris Sud, Paris-Saclay University, Le Plessis Robinson, France
| | - François Haddad
- 3 Cardiovascular Medicine, Stanford Hospital, Stanford University, CA, USA
| | - Julien Guihaire
- 1 36705 Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Universite Paris Sud, Paris-Saclay University, Le Plessis Robinson, France
| | - Myriam Amsallem
- 1 36705 Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Universite Paris Sud, Paris-Saclay University, Le Plessis Robinson, France
| | - Bojan Vrtovec
- 4 Department of Cardiology, Advanced Heart Failure and Transplantation Center, University Medical Center Ljubljana, Ljubljana, Slovenia
| | - Elie Fadel
- 1 36705 Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Universite Paris Sud, Paris-Saclay University, Le Plessis Robinson, France.,5 Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Marie Lannelongue Hospital, Universite Paris Sud, Paris-Saclay University, Le Plessis Robinson, France
| | - Georges Uzan
- 2 Inserm 1197 Research Unit, Universite Paris Sud, Paris-Saclay University, Villejuif, France
| | - Olaf Mercier
- 1 36705 Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Universite Paris Sud, Paris-Saclay University, Le Plessis Robinson, France.,5 Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Marie Lannelongue Hospital, Universite Paris Sud, Paris-Saclay University, Le Plessis Robinson, France
| |
Collapse
|
24
|
Mittal R, Jhaveri VM, McMurry HS, Kay SIS, Sutherland KJ, Nicole L, Mittal J, Jayant RD. Recent treatment modalities for cardiovascular diseases with a focus on stem cells, aptamers, exosomes and nanomedicine. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:831-840. [PMID: 29447002 DOI: 10.1080/21691401.2018.1436555] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide. Due to the significant impact of CVD on humans, there is a need to develop novel treatment modalities tailored to major classes of cardiac diseases including hypertension, coronary artery disease, cardiomyopathies, arrhythmias, valvular disease and inflammatory diseases. In this article, we discuss recent advancements regarding development of therapeutic strategies based on stem cells, aptamers, exosomes, drug-eluting and dissolvable stents, immunotherapy and nanomedicine for the treatment of CVD. We summarize current research and clinical advances in cardiovascular therapeutics, with a focus on therapies that move beyond current oral- or sublingual-based regimens. This review article provides insight into current research and future treatment strategies that hold a great relevance for future clinical practice in pursuit of improving quality of life of patients suffering from CVD.
Collapse
Affiliation(s)
- Rahul Mittal
- a Department of Otolaryngology , University of Miami Miller School of Medicine , Miami , FL , USA
| | - Vasanti M Jhaveri
- a Department of Otolaryngology , University of Miami Miller School of Medicine , Miami , FL , USA
| | - Hannah S McMurry
- a Department of Otolaryngology , University of Miami Miller School of Medicine , Miami , FL , USA
| | - Sae-In Samantha Kay
- b Dr. Kiran C. Patel College of Osteopathic Medicine , Nova Southeastern University , Fort Lauderdale , FL , USA
| | - Kyle J Sutherland
- a Department of Otolaryngology , University of Miami Miller School of Medicine , Miami , FL , USA
| | - Lin Nicole
- a Department of Otolaryngology , University of Miami Miller School of Medicine , Miami , FL , USA
| | - Jeenu Mittal
- a Department of Otolaryngology , University of Miami Miller School of Medicine , Miami , FL , USA
| | - Rahul Dev Jayant
- c Department of Immunology , Center for Personalized Nanomedicine, Herbert Wertheim College of Medicine, Florida International University , Miami , FL , USA
| |
Collapse
|
25
|
Cheng Y, Gong Y, Qian S, Mou Y, Li H, Chen X, Kong H, Xie W, Wang H, Zhang Y, Huang Z. Identification of a Novel Hybridization from Isosorbide 5-Mononitrate and Bardoxolone Methyl with Dual Activities of Pulmonary Vasodilation and Vascular Remodeling Inhibition on Pulmonary Arterial Hypertension Rats. J Med Chem 2018; 61:1474-1482. [PMID: 29377691 DOI: 10.1021/acs.jmedchem.7b01153] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Yusheng Cheng
- Department
of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029, P. R. China
| | | | | | | | | | | | - Hui Kong
- Department
of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029, P. R. China
| | - Weiping Xie
- Department
of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029, P. R. China
| | - Hong Wang
- Department
of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029, P. R. China
| | | | | |
Collapse
|
26
|
Intratracheal Administration of Autologous Bone Marrow-Derived Cells Ameliorates Monocrotaline-Induced Pulmonary Vessel Remodeling and Lung Inflammation in Rats. Lung 2017; 196:147-155. [PMID: 29264652 DOI: 10.1007/s00408-017-0075-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 11/27/2017] [Indexed: 12/28/2022]
Abstract
PURPOSE Inflammation is a feature of lung injury and plays a critical role in pulmonary vascular remodeling. Bone marrow-derived cells (BMCs) have anti-inflammatory properties and favor macrophage differentiation into an alternatively activated regulatory M2 profile. We investigated the effect of autologous BMCs on monocrotaline-induced pulmonary vessel remodeling and lung inflammation in rats, by direct administration into lungs via the airway. METHODS BMCs were isolated and plastic-adherent cells were cultured for 3 weeks. 1 week following monocrotaline (60 mg/kg) treatment, fluorescently labeled autologous BMCs (1 × 106 cells) or vehicle were administered intratracheally to male Sprague-Dawley rats. 4 weeks following monocrotaline treatment, lung pathology was evaluated. RESULTS Monocrotaline increased pulmonary vessel wall thickness, perivascular infiltration, alveolar septal thickening, and inflammatory cell infiltration including T lymphocytes and monocytes/macrophages in alveolar areas, and also increased mRNA expression of inflammatory-related cytokines including IL-10 in the lung. Intratracheal administration of autologous BMCs prevented pulmonary vessel wall thickening and perivascular infiltration, and increased CD163-positive M2-like macrophages in perivascular areas. BMC administration inhibited the thickening of alveolar septa and reduced monocrotaline-induced inflammatory cell infiltration in lung parenchyma compared with monocrotaline-vehicle-treated-rats. Furthermore, BMCs administration increased expression of CD163-positive cells in perivascular areas and maintained the increased mRNA expression of IL-10. CONCLUSIONS Intratracheal administration of autologous BMCs prevented monocrotaline-induced pulmonary vessel remodeling and lung inflammation, at least in part, through induction of alternatively activated macrophages and regulation of the local lung environment toward resolving inflammation.
Collapse
|
27
|
de Mendonça L, Felix NS, Blanco NG, Da Silva JS, Ferreira TP, Abreu SC, Cruz FF, Rocha N, Silva PM, Martins V, Capelozzi VL, Zapata-Sudo G, Rocco PRM, Silva PL. Mesenchymal stromal cell therapy reduces lung inflammation and vascular remodeling and improves hemodynamics in experimental pulmonary arterial hypertension. Stem Cell Res Ther 2017; 8:220. [PMID: 28974252 PMCID: PMC5627397 DOI: 10.1186/s13287-017-0669-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/29/2017] [Accepted: 09/12/2017] [Indexed: 12/30/2022] Open
Abstract
Background Experimental research has reported beneficial effects of mesenchymal stromal cell (MSC) therapy in pulmonary arterial hypertension (PAH). However, these studies either were based on prophylactic protocols or assessed basic remodeling features without evaluating possible mechanisms. We analyzed the effects of MSC therapy on lung vascular remodeling and hemodynamics and its possible mechanisms of action in monocrotaline (MCT)-induced PAH. Methods Twenty-eight Wistar rats were randomly divided into two groups. In the PAH group, animals received MCT 60 mg/kg intraperitoneally, while a control group received saline (SAL) instead. On day 14, both groups were further randomized to receive 105 adipose-derived MSCs or SAL intravenously (n = 7/group). On day 28, right ventricular systolic pressure (RVSP) and the gene expression of mediators associated with apoptosis, inflammation, fibrosis, Smad-1 levels, cell proliferation, and endothelial–mesenchymal transition were determined. In addition, lung histology (smooth muscle cell proliferation and plexiform-like injuries), CD68+ and CD163+ macrophages, and plasma levels of vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) were evaluated. Results In the PAH group, adipose-derived MSCs, compared to SAL, reduced mean RVSP (29 ± 1 vs 39 ± 2 mmHg, p < 0.001), lung tissue collagen fiber content, smooth muscle cell proliferation, CD68+ macrophages, interleukin-6 expression, and the antiapoptotic mediators Bcl-2 and survivin. Conversely, expression of the proapoptotic mediator procaspase-3 and plasma VEGF increased, with no changes in PDGF. In the pulmonary artery, MSCs dampened the endothelial–mesenchymal transition. Conclusion In MCT-induced PAH, MSC therapy reduced lung vascular remodeling, thus improving hemodynamics. These beneficial effects were associated with increased levels of proapoptotic markers, mesenchymal-to-endothelial transition, reduced cell proliferation markers, and inflammation due to a shift away from the M1 phenotype. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0669-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lucas de Mendonça
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil
| | - Nathane S Felix
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil
| | - Natália G Blanco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil
| | - Jaqueline S Da Silva
- Laboratory of Cardiovascular Pharmacology, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Tatiana P Ferreira
- Laboratory of Inflammation, Oswaldo Cruz Institute-Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Soraia C Abreu
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil
| | - Fernanda F Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil
| | - Nazareth Rocha
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,Department of Physiology, Fluminense Federal University, Niterói, RJ, Brazil
| | - Patrícia M Silva
- Laboratory of Inflammation, Oswaldo Cruz Institute-Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Vanessa Martins
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,Laboratory of Histomorphometry and Lung Genomics, University of São Paulo Faculty of Medicine, São Paulo, SP, Brazil
| | - Vera L Capelozzi
- Laboratory of Histomorphometry and Lung Genomics, University of São Paulo Faculty of Medicine, São Paulo, SP, Brazil
| | - Gizele Zapata-Sudo
- Laboratory of Cardiovascular Pharmacology, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil
| | - Pedro L Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil. .,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
28
|
Involvement of Ca 2+-activated K + channel 3.1 in hypoxia-induced pulmonary arterial hypertension and therapeutic effects of TRAM-34 in rats. Biosci Rep 2017; 37:BSR20170763. [PMID: 28679649 PMCID: PMC5529208 DOI: 10.1042/bsr20170763] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 07/02/2017] [Accepted: 07/04/2017] [Indexed: 02/05/2023] Open
Abstract
Pulmonary artery hypertension (PAH) is an incurable disease associated with the proliferation of pulmonary artery smooth muscle cells (PASMCs) and vascular remodeling. The present study examined whether TRAM-34, a highly selective blocker of calcium-activated potassium channel 3.1 (Kca3.1), can help prevent such hypertension by reducing proliferation in PASMCs. Rats were exposed to hypoxia (10% O2) for 3 weeks and treated daily with TRAM-34 intraperitoneally from the first day of hypoxia. Animals were killed and examined for vascular hypertrophy, Kca3.1 expression, and downstream signaling pathways. In addition, primary cultures of rat PASMCs were exposed to hypoxia (3% O2) or normoxia (21% O2) for 24 h in the presence of TRAM-34 or siRNA against Kca3.1. Activation of cell signaling pathways was examined using Western blot analysis. In animal experiments, hypoxia triggered significant medial hypertrophy of pulmonary arterioles and right ventricular hypertrophy, and it significantly increased pulmonary artery pressure, Kca3.1 mRNA levels and ERK/p38 MAP kinase signaling. These effects were attenuated in the presence of TRAM-34. In cell culture experiments, blocking Kca3.1 using TRAM-34 or siRNA inhibited hypoxia-induced ERK/p38 signaling. Kca3.1 may play a role in the development of PAH by activating ERK/p38 MAP kinase signaling, which may then contribute to hypoxia-induced pulmonary vascular remodeling. TRAM-34 may protect against hypoxia-induced PAH.
Collapse
|
29
|
Lee H, Kim KC, Choi SJ, Hong YM. Optimal Dose and Timing of Umbilical Stem Cells Treatment in Pulmonary Arterial Hypertensive Rats. Yonsei Med J 2017; 58:570-580. [PMID: 28332363 PMCID: PMC5368143 DOI: 10.3349/ymj.2017.58.3.570] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 12/02/2016] [Accepted: 12/08/2016] [Indexed: 01/18/2023] Open
Abstract
PURPOSE Pulmonary arterial hypertension (PAH) is a fatal disease which is characterized by an increase in pulmonary arterial pressure leading to increases in right ventricular afterload. Human umbilical cord blood derived-mesenchymal stem cells (hUCB-MSCs) administered via the jugular vein have been previously shown to improve PAH by reversal treatment. However, the effect of low dosage and transfusion timing of hUCB-MSCs on PAH has not yet been clearly established. Obviously, low dosage treatment can lead to a reduction in costs. This is the first study on early transfusion effect. MATERIALS AND METHODS This study was divided into two parts. The first part is an investigation of dose-dependent effect. hUCB-MSCs were administered into 3 groups of rats (UA: 3×10⁶ cells, UB: 1.5×10⁶ cells, UC: 3×10⁵ cells) via the external jugular vein at week 1 after monocrotaline (MCT) injection. The second part is a search for optimal treatment timing in 3×10⁵ cells dose of hUCB-MSCs administered at day 1 for UD group (low dose of hUCB-MSCs at day 1), at day 1 and week 1 for the UE group (dual transfusion of low dose of hUCB-MSCs at day 1 and week 1) and at 1 week for the UF group (reversal treatment of low dose hUCB-MSC at week 1) after MCT injection. RESULTS The administration of 3×10⁵ hUCB-MSCs was as effective as the 3×10⁶ dose in decreasing mean right ventricle (RV) pressure and pulmonary pathological changes. Early treatment with hUCB-MSCs improved mean RV pressure, pulmonary pathological changes and heart collagen 3 protein expression levels in PAH. CONCLUSION Low-dose early treatment of hUCB-MSCs is as effective as a high dose treatment of hUCB-MSCs in improving PAH although dual or reversal treatment is still more effective.
Collapse
Affiliation(s)
- Hyeryon Lee
- Department of Pediatrics, Ewha Womans University School of Medicine, Seoul, Korea
| | - Kwan Chang Kim
- Department of Thoracic and Cardiovascular Surgery, Ewha Womans University School of Medicine, Seoul, Korea
| | - Soo Jin Choi
- Biomedical Research Institute, MEDIPOST, Co., Seoul, Korea
| | - Young Mi Hong
- Department of Pediatrics, Ewha Womans University School of Medicine, Seoul, Korea.
| |
Collapse
|
30
|
Zhang X, Hou HT, Wang J, Liu XC, Yang Q, He GW. Plasma Proteomic Study in Pulmonary Arterial Hypertension Associated with Congenital Heart Diseases. Sci Rep 2016; 6:36541. [PMID: 27886187 PMCID: PMC5122864 DOI: 10.1038/srep36541] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 10/17/2016] [Indexed: 02/06/2023] Open
Abstract
Pulmonary arterial hypertension associated with congenital heart disease (CHD-PAH) has serious consequence and plasma protein profiles in CHD-PAH are unknown. We aimed to reveal the differential plasma proteins in 272 CHD patients with or without PAH. Various types of CHD-PAH were studied. Differential plasma proteins were first detected by iTRAQ proteomic technology and those with significant clinical relevance were selected for further ELISA validation in new cohort of patients. Among the 190 differential plasma proteins detected by iTRAQ, carbamoyl-phosphate synthetase I (CPSI, related to urea cycle and endogenous nitric oxide production) and complement factor H-related protein 2 (CFHR2, related to complement system and coagulant mechanism) were selected for further ELISA validation in new cohort of 152 patients. Both CPSI and CFHR2 were down-regulated with decreased plasma levels (p < 0.01). Thus, we for the first time in CHD-PAH patients identified a large number of differential plasma proteins. The decreased CPSI expression in CHD-PAH patients may reveal a mechanism related to endogenous nitric oxide and the decrease of CFHR2 protein may demonstrate the deficiency of the immune system and coagulation mechanism. The findings may open a new direction for translational medicine in CHD-PAH with regard to the diagnosis and progress of the disease.
Collapse
Affiliation(s)
- Xi Zhang
- Department of Surgery & Center for Basic Medical Research, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Tianjin & The Affiliated Hospital of Hangzhou Normal University & Zhejiang University, Hangzhou, China
| | - Hai-Tao Hou
- TEDA International Cardiovascular Hospital, CAMS & PUMC, Tianjin, China
| | - Jun Wang
- TEDA International Cardiovascular Hospital, CAMS & PUMC, Tianjin, China
| | - Xiao-Cheng Liu
- TEDA International Cardiovascular Hospital, CAMS & PUMC, Tianjin, China
| | - Qin Yang
- TEDA International Cardiovascular Hospital, CAMS & PUMC, Tianjin, China
- The Chinese University of Hong Kong, Shatin, China
| | - Guo-Wei He
- Department of Surgery & Center for Basic Medical Research, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Tianjin & The Affiliated Hospital of Hangzhou Normal University & Zhejiang University, Hangzhou, China
- Department of Surgery, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
31
|
Varshney R, Ali Q, Wu C, Sun Z. Monocrotaline-Induced Pulmonary Hypertension Involves Downregulation of Antiaging Protein Klotho and eNOS Activity. Hypertension 2016; 68:1255-1263. [PMID: 27672025 DOI: 10.1161/hypertensionaha.116.08184] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 08/15/2016] [Indexed: 12/20/2022]
Abstract
The objective of this study is to investigate whether stem cell delivery of secreted Klotho (SKL), an aging-suppressor protein, attenuates monocrotaline-induced pulmonary vascular dysfunction and remodeling. Overexpression of SKL in mesenchymal stem cells (MSCs) was achieved by transfecting MSCs with lentiviral vectors expressing SKL-green fluorescent protein (GFP). Four groups of rats were treated with monocrotaline, whereas an additional group was given saline (control). Three days later, 4 monocrotaline-treated groups received intravenous delivery of nontransfected MSCs, MSC-GFP, MSC-SKL-GFP, and PBS, respectively. Ex vivo vascular relaxing responses to acetylcholine were diminished in small pulmonary arteries (PAs) in monocrotaline-treated rats, indicating pulmonary vascular endothelial dysfunction. Interestingly, delivery of MSCs overexpressing SKL (MSC-SKL-GFP) abolished monocrotaline-induced pulmonary vascular endothelial dysfunction and PA remodeling. Monocrotaline significantly increased right ventricular systolic blood pressure, which was attenuated significantly by MSC-SKL-GFP, indicating improved PA hypertension. MSC-SKL-GFP also attenuated right ventricular hypertrophy. Nontransfected MSCs slightly, but not significantly, improved PA hypertension and pulmonary vascular endothelial dysfunction. MSC-SKL-GFP attenuated monocrotaline-induced inflammation, as evidenced by decreased macrophage infiltration around PAs. MSC-SKL-GFP increased SKL levels, which rescued the downregulation of SIRT1 (Sirtuin 1) expression and endothelial NO synthase (eNOS) phosphorylation in the lungs of monocrotaline-treated rats. In cultured endothelial cells, SKL abolished monocrotaline-induced downregulation of eNOS activity and NO levels and enhanced cell viability. Therefore, stem cell delivery of SKL is an effective therapeutic strategy for pulmonary vascular endothelial dysfunction and PA remodeling. SKL attenuates monocrotaline-induced PA remodeling and PA smooth muscle cell proliferation, likely by reducing inflammation and restoring SIRT1 levels and eNOS activity.
Collapse
Affiliation(s)
- Rohan Varshney
- From the Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Quaisar Ali
- From the Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Chengxiang Wu
- From the Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Zhongjie Sun
- From the Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City.
| |
Collapse
|
32
|
Yan L, Chen X, Talati M, Nunley BW, Gladson S, Blackwell T, Cogan J, Austin E, Wheeler F, Loyd J, West J, Hamid R. Bone Marrow-derived Cells Contribute to the Pathogenesis of Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 2016; 193:898-909. [PMID: 26651104 DOI: 10.1164/rccm.201502-0407oc] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
RATIONALE Pulmonary arterial hypertension (PAH) is a progressive lung disease of the pulmonary microvasculature. Studies suggest that bone marrow (BM)-derived circulating cells may play an important role in its pathogenesis. OBJECTIVES We used a genetic model of PAH, the Bmpr2 mutant mouse, to study the role of BM-derived circulating cells in its pathogenesis. METHODS Recipient mice, either Bmpr2(R899X) mutant or controls, were lethally irradiated and transplanted with either control or Bmpr2(R899X) BM cells. Donor cells were traced in female recipient mice by Y chromosome painting. Molecular and function insights were provided by expression and cytokine arrays combined with flow cytometry, colony-forming assays, and competitive transplant assays. MEASUREMENTS AND MAIN RESULTS We found that mutant BM cells caused PAH with remodeling and inflammation when transplanted into control mice, whereas control BM cells had a protective effect against the development of disease, when transplanted into mutant mice. Donor BM-derived cells were present in the lungs of recipient mice. Functional and molecular analysis identified mutant BM cell dysfunction suggestive of a PAH phenotype soon after activation of the transgene and long before the development of lung pathology. CONCLUSIONS Our data show that BM cells played a key role in PAH pathogenesis and that the transplanted BM cells were able to drive the lung phenotype in a myeloablative transplant model. Furthermore, the specific cell types involved were derived from hematopoietic stem cells and exhibit dysfunction long before the development of lung pathology.
Collapse
Affiliation(s)
- Ling Yan
- 1 Division of Medical Genetics and Genomic Medicine, Department of Pediatrics
| | - Xinping Chen
- 2 Division of Respiratory and Critical Care, Department of Medicine
| | - Megha Talati
- 2 Division of Respiratory and Critical Care, Department of Medicine
| | | | - Santhi Gladson
- 2 Division of Respiratory and Critical Care, Department of Medicine
| | - Tom Blackwell
- 2 Division of Respiratory and Critical Care, Department of Medicine
| | - Joy Cogan
- 1 Division of Medical Genetics and Genomic Medicine, Department of Pediatrics
| | - Eric Austin
- 3 Division of Allergy, Immunology and Pulmonary Medicine, Department of Pediatrics, and
| | - Ferrin Wheeler
- 4 Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - James Loyd
- 2 Division of Respiratory and Critical Care, Department of Medicine
| | - James West
- 2 Division of Respiratory and Critical Care, Department of Medicine
| | - Rizwan Hamid
- 1 Division of Medical Genetics and Genomic Medicine, Department of Pediatrics
| |
Collapse
|
33
|
Zhu Z, Fang Z, Hu X, Zhou S. MicroRNAs and mesenchymal stem cells: hope for pulmonary hypertension. Braz J Cardiovasc Surg 2016; 30:380-5. [PMID: 26313730 PMCID: PMC4541786 DOI: 10.5935/1678-9741.20150033] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 05/12/2015] [Indexed: 12/21/2022] Open
Abstract
Pulmonary hypertension is a devastating and refractory disease and there is no cure
for this disease. Recently, microRNAs and mesenchymal stem cells emerged as novel
methods to treat pulmonary hypertension. More than 20 kinds of microRNAs may
participate in the process of pulmonary hypertension. It seems microRNAs or
mesenchymal stem cells can ameliorate some symptoms of pulmonary hypertension in
animals and even improve heart and lung function during pulmonary hypertension.
Nevertheless, the relationship between mesenchymal stem cells, microRNAs and
pulmonary hypertension is not clear. And the mechanisms underlying their function
still need to be investigated. In this study we review the recent findings in
mesenchymal stem cells - and microRNAs-based pulmonary hypertension treatment,
focusing on the potential role of microRNAs regulated mesenchymal stem cells in
pulmonary hypertension and the role of exosomes between mesenchymal stem cells and
pulmonary hypertension.
Collapse
Affiliation(s)
- Zhaowei Zhu
- The Second Xiangya Hospital, Central South University, Huan Province, CN
| | - Zhenfei Fang
- Department of Cardiology, Second Xiangya Hospital, Central South University, Huan Province, CN
| | - Xinqun Hu
- Department of Cardiology, Second Xiangya Hospital, Central South University, Huan Province, CN
| | - Shenghua Zhou
- The Second Xiangya Hospital, Central South University, Huan Province, CN
| |
Collapse
|
34
|
Heise RL, Link PA, Farkas L. From Here to There, Progenitor Cells and Stem Cells Are Everywhere in Lung Vascular Remodeling. Front Pediatr 2016; 4:80. [PMID: 27583245 PMCID: PMC4988064 DOI: 10.3389/fped.2016.00080] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 07/20/2016] [Indexed: 01/27/2023] Open
Abstract
The field of stem cell biology, cell therapy, and regenerative medicine has expanded almost exponentially, in the last decade. Clinical trials are evaluating the potential therapeutic use of stem cells in many adult and pediatric lung diseases with vascular component, such as bronchopulmonary dysplasia (BPD), chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), or pulmonary arterial hypertension (PAH). Extensive research activity is exploring the lung resident and circulating progenitor cells and their contribution to vascular complications of chronic lung diseases, and researchers hope to use resident or circulating stem/progenitor cells to treat chronic lung diseases and their vascular complications. It is becoming more and more clear that progress in mechanobiology will help to understand the various influences of physical forces and extracellular matrix composition on the phenotype and features of the progenitor cells and stem cells. The current review provides an overview of current concepts in the field.
Collapse
Affiliation(s)
- Rebecca L Heise
- Department of Biomedical Engineering, School of Engineering, Virginia Commonwealth University , Richmond, VA , USA
| | - Patrick A Link
- Department of Biomedical Engineering, School of Engineering, Virginia Commonwealth University , Richmond, VA , USA
| | - Laszlo Farkas
- Department of Internal Medicine, Division of Pulmonary Disease and Critical Care Medicine, School of Medicine, Virginia Commonwealth University , Richmond, VA , USA
| |
Collapse
|
35
|
Lee JC, Cha CI, Kim DS, Choe SY. Therapeutic Effects of Umbilical Cord Blood Derived Mesenchymal Stem Cell-Conditioned Medium on Pulmonary Arterial Hypertension in Rats. J Pathol Transl Med 2015; 49:472-80. [PMID: 26471341 PMCID: PMC4696528 DOI: 10.4132/jptm.2015.09.11] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 09/09/2015] [Indexed: 01/07/2023] Open
Abstract
Background: Human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) may have multiple therapeutic applications for cell based therapy including the treatment of pulmonary artery hypertension (PAH). As low survival rates and potential tumorigenicity of implanted cells could undermine the mesenchymal stem cell (MSC) cell-based therapy, we chose to investigate the use of conditioned medium (CM) from a culture of MSC cells as a feasible alternative. Methods: CM was prepared by culturing hUCB-MSCs in three-dimensional spheroids. In a rat model of PAH induced by monocrotaline, we infused CM or the control unconditioned culture media via the tail-vein of 6-week-old Sprague-Dawley rats. Results: Compared with the control unconditioned media, CM infusion reduced the ventricular pressure, the right ventricle/(left ventricle+interventricular septum) ratio, and maintained respiratory function in the treated animals. Also, the number of interleukin 1α (IL-1α), chemokine (C-C motif) ligand 5 (CCL5), and tissue inhibitor of metalloproteinase 1 (TIMP-1)–positive cells increased in lung samples and the number of terminal deoxynucleotidyl transferase–mediated deoxyuridine triphosphate nick-end labeling technique (TUNEL)–positive cells decreased significantly in the CM treated animals. Conclusions: From our in vivo data in the rat model, the observed decreases in the TUNEL staining suggest a potential therapeutic benefit of the CM in ameliorating PAH-mediated lung tissue damage. Increased IL-1α, CCL5, and TIMP-1 levels may play important roles in this regard.
Collapse
Affiliation(s)
- Jae Chul Lee
- Department of Biology, School of Life Sciences, Chungbuk National University, Cheongju, Korea.,Department of Surgery, Brain Korea 21 PLUS Project for Medical Sciences and HBP Surgery and Liver Transplantation, Korea University College of Medicine, Seoul, Korea.,Department of Anatomy, Seoul National University College of Medicine, Seoul, Korea
| | - Choong Ik Cha
- Department of Anatomy, Seoul National University College of Medicine, Seoul, Korea
| | - Dong-Sik Kim
- Department of Surgery, Brain Korea 21 PLUS Project for Medical Sciences and HBP Surgery and Liver Transplantation, Korea University College of Medicine, Seoul, Korea
| | - Soo Young Choe
- Department of Biology, School of Life Sciences, Chungbuk National University, Cheongju, Korea
| |
Collapse
|
36
|
Regional differences in WT-1 and Tcf21 expression during ventricular development: implications for myocardial compaction. PLoS One 2015; 10:e0136025. [PMID: 26390289 PMCID: PMC4577115 DOI: 10.1371/journal.pone.0136025] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 07/29/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Morphological and functional differences of the right and left ventricle are apparent in the adult human heart. A differential contribution of cardiac fibroblasts and smooth muscle cells (populations of epicardium-derived cells) to each ventricle may account for part of the morphological-functional disparity. Here we studied the relation between epicardial derivatives and the development of compact ventricular myocardium. RESULTS Wildtype and Wt1CreERT2/+ reporter mice were used to study WT-1 expressing cells, and Tcf21lacZ/+ reporter mice and PDGFRα-/-;Tcf21LacZ/+ mice to study the formation of the cardiac fibroblast population. After covering the heart, intramyocardial WT-1+ cells were first observed at the inner curvature, the right ventricular postero-lateral wall and left ventricular apical wall. Later, WT-1+ cells were present in the walls of both ventricles, but significantly more pronounced in the left ventricle. Tcf21-LacZ + cells followed the same distribution pattern as WT-1+ cells but at later stages, indicating a timing difference between these cell populations. Within the right ventricle, WT-1+ and Tcf21-lacZ+ cell distribution was more pronounced in the posterior inlet part. A gradual increase in myocardial wall thickness was observed early in the left ventricle and at later stages in the right ventricle. PDGFRα-/-;Tcf21LacZ/+ mice showed deficient epicardium, diminished number of Tcf21-LacZ + cells and reduced ventricular compaction. CONCLUSIONS During normal heart development, spatio-temporal differences in contribution of WT-1 and Tcf21-LacZ + cells to right versus left ventricular myocardium occur parallel to myocardial thickening. These findings may relate to lateralized differences in ventricular (patho)morphology in humans.
Collapse
|
37
|
Naaldijk Y, Johnson AA, Ishak S, Meisel HJ, Hohaus C, Stolzing A. Migrational changes of mesenchymal stem cells in response to cytokines, growth factors, hypoxia, and aging. Exp Cell Res 2015; 338:97-104. [PMID: 26335540 DOI: 10.1016/j.yexcr.2015.08.019] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 07/23/2015] [Accepted: 08/30/2015] [Indexed: 12/17/2022]
Abstract
Mesenchymal stem cells (MSCs) are non-immunogenic, multipotent cells with at least trilineage differentiation potential. They promote wound healing, improve regeneration of injured tissue, and mediate numerous other health effects. MSCs migrate to sites of injury and stimulate repair either through direct differentiation or indirectly through the stimulation of endogenous repair mechanisms. Using the in vitro scratch assay, we show that the inflammatory cytokines, chemokines, and growth factors TNF-α, SDF-1, PDGF, and bFGF enhance migration of rat MSCs under normoxic conditions, while TNF-α, IFN-γ, PDGF, and bFGF promote MSC migration under hypoxic conditions. This indicates that the oxygen concentration affects how MSCs will migrate in response to specific factors and, consistent with this, differential expression of cytokines was observed under hypoxic versus normoxic conditions. Using the transwell migration assay, we find that TNF-α, IFN-γ, bFGF, IGF-1, PDGF, and SDF-1 significantly increase transmigration of rat MSCs compared to unstimulated medium. MSCs derived from aged rats exhibited comparable migration to MSCs derived from young rats under hypoxic and normoxic conditions, even after application with specific factors. Similarly, migration in MSCs from aged, human donors did not statistically differ compared to migration in MSCs derived from human umbilical cord tissue or younger donors.
Collapse
Affiliation(s)
- Yahaira Naaldijk
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany; Translational Centre for Regenerative Medicine (TRM), Leipzig University, Germany
| | - Adiv A Johnson
- Department of Ophthalmology, Mayo Clinic, Rochester, MN, USA
| | - Stefan Ishak
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Hans Jörg Meisel
- Department of Neurosurgery, BG Clinic Bergmannstrost, Halle, Germany
| | - Christian Hohaus
- Department of Neurosurgery, BG Clinic Bergmannstrost, Halle, Germany
| | - Alexandra Stolzing
- Translational Centre for Regenerative Medicine (TRM), Leipzig University, Germany; University of Loughborough, Centre for Biological Engineering, Wolfson School of Material and Manufacturing Engineering, Epinal Way, LE113TU Loughborough, UK.
| |
Collapse
|
38
|
van der Laarse A, Cobbaert CM, Umar S. Stem and progenitor cell therapy for pulmonary arterial hypertension: effects on the right ventricle (2013 Grover Conference Series). Pulm Circ 2015; 5:73-80. [PMID: 25992272 DOI: 10.1086/679701] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 08/25/2014] [Indexed: 12/12/2022] Open
Abstract
In experimental animals and in patients with pulmonary arterial hypertension (PAH), a wide spectrum of structural and functional conditions is known that may be responsible for the switch of a state of "compensated" right ventricular (RV) hypertrophy to a state of RV failure. In recent years, therapy with differentiated cells, endothelial progenitor cells, and mesenchymal stem cells has been shown to cause partial or complete reversal of pathological characteristics of PAH. The therapeutic effects of stem or progenitor cell therapy are considered to be (1) paracrine effects from stem or progenitor cells that had engrafted in the myocardium (or elsewhere), by compounds that have anti-inflammatory, antiapoptotic, and proangiogenic actions and (2) unloading effects on the right ventricle due to stem or progenitor cell-induced decrease in pulmonary vascular resistance and decrease in pulmonary artery pressure.
Collapse
Affiliation(s)
- Arnoud van der Laarse
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands ; Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Christa M Cobbaert
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Soban Umar
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine at University of California, Los Angeles, California, USA
| |
Collapse
|
39
|
Lee JC, Kim KC, Yang YS, Oh W, Choi SJ, Choe SY, Hong YM. Microarray analysis after umbilical cord blood derived mesenchymal stem cells injection in monocrotaline-induced pulmonary artery hypertension rats. Anat Cell Biol 2014; 47:217-26. [PMID: 25548719 PMCID: PMC4276895 DOI: 10.5115/acb.2014.47.4.217] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 11/27/2014] [Accepted: 12/01/2014] [Indexed: 02/08/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is associated with structural alterations of lung vasculature. PAH is still a devastating disease needing an aggressive therapeutic approach. Despite the therapeutic potential of human umbilical cord mesenchymal stem cells (MSCs), the molecular parameters to define the stemness remain largely unknown. Using high-density oligonucleotide microarrays, the differential gene expression profiles between a fraction of mononuclear cells of human umbilical cord blood (UCB) and its MSC subpopulation were obtained. Of particular interest was a subset of 46 genes preferentially expressed at 7-fold or higher in the group treated with human UCB-MSCs. This subset contained numerous genes involved in the inflammatory response, immune response, lipid metabolism, cell adhesion, cell migration, cell differentiation, apoptosis, cell growth, transport, cell proliferation, transcription, and signal transduction. Our results provide a foundation for a more reproducible and reliable quality control using genotypic analysis for the definition of human UCB-MSCs. Therefore, our results will provide a basis for studies on molecular mechanisms controlling the core properties of human MSCs.
Collapse
Affiliation(s)
- Jae Chul Lee
- Department of Pediatrics, Ewha Womans University School of Medicine, Seoul, Korea. ; Department of Biology, School of Life Sciences, Chungbuk National University, Cheongju, Korea
| | - Kwan Chang Kim
- Department of Thoracic and Cardiovascular Surgery, Ewha Womans University School of Medicine, Seoul, Korea
| | - Yoon Sun Yang
- Biomedical Research Institute, MEDIPOST, Co., Seoul, Korea
| | - Wonil Oh
- Biomedical Research Institute, MEDIPOST, Co., Seoul, Korea
| | - Soo Jin Choi
- Biomedical Research Institute, MEDIPOST, Co., Seoul, Korea
| | - Soo Young Choe
- Department of Biology, School of Life Sciences, Chungbuk National University, Cheongju, Korea
| | - Young Mi Hong
- Department of Pediatrics, Ewha Womans University School of Medicine, Seoul, Korea
| |
Collapse
|
40
|
Suen CM, Mei SHJ, Kugathasan L, Stewart DJ. Targeted delivery of genes to endothelial cells and cell- and gene-based therapy in pulmonary vascular diseases. Compr Physiol 2014; 3:1749-79. [PMID: 24265244 DOI: 10.1002/cphy.c120034] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a devastating disease that, despite significant advances in medical therapies over the last several decades, continues to have an extremely poor prognosis. Gene therapy is a method to deliver therapeutic genes to replace defective or mutant genes or supplement existing cellular processes to modify disease. Over the last few decades, several viral and nonviral methods of gene therapy have been developed for preclinical PAH studies with varying degrees of efficacy. However, these gene delivery methods face challenges of immunogenicity, low transduction rates, and nonspecific targeting which have limited their translation to clinical studies. More recently, the emergence of regenerative approaches using stem and progenitor cells such as endothelial progenitor cells (EPCs) and mesenchymal stem cells (MSCs) have offered a new approach to gene therapy. Cell-based gene therapy is an approach that augments the therapeutic potential of EPCs and MSCs and may deliver on the promise of reversal of established PAH. These new regenerative approaches have shown tremendous potential in preclinical studies; however, large, rigorously designed clinical studies will be necessary to evaluate clinical efficacy and safety.
Collapse
Affiliation(s)
- Colin M Suen
- Sprott Centre for Stem Cell Research, The Ottawa Hospital Research Institute and University of Ottawa, Ottawa, Ontario, Canada
| | | | | | | |
Collapse
|
41
|
Foster WS, Suen CM, Stewart DJ. Regenerative Cell and Tissue-based Therapies for Pulmonary Arterial Hypertension. Can J Cardiol 2014; 30:1350-60. [DOI: 10.1016/j.cjca.2014.08.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 08/13/2014] [Accepted: 08/24/2014] [Indexed: 12/21/2022] Open
|
42
|
Luo L, Lin T, Zheng S, Xie Z, Chen M, Lian G, Xu C, Wang H, Xie L. Adipose-derived stem cells attenuate pulmonary arterial hypertension and ameliorate pulmonary arterial remodeling in monocrotaline-induced pulmonary hypertensive rats. Clin Exp Hypertens 2014; 37:241-8. [PMID: 25271670 DOI: 10.3109/10641963.2014.954710] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
We investigated the effect of adipose-derived stem cells (ADSCs) transplantation effects on structural remodeling and pulmonary artery pressure in monocrotaline (MCT)-induced pulmonary hypertensive rats. In the first experiment, 32 male Sprague-Dawley (SD) rats were randomly divided into four groups (n = 8/group): 3 ADSCs treated groups and normal control (Ctrl). ADSCs were administered through the left jugular vein at 10(5), 10(6) and 10(7) cells, respectively, and a cell density of 10(6)cells/ml was shown to be optimal. The GFP-tagged ADSCs were identified in the lungs and differentiated into endothelial-like cells. In the second experiment, 96 male SD rats were randomly divided into three groups (n = 32/group): Ctrl, MCT-induced pulmonary arterial hypertension (PAH), and PAH treated with ADSCs (ADSCs). Two weeks post-MCT administration, the ADSCs group received 1 × 10(6) ADSCs via the external jugular vein. Compared to PAH rats, mean pulmonary arterial pressure was decreased in rats at 1, 2, and 3 weeks after ADSCs-treatment (18.63 ± 2.15 mmHg versus 24.53 ± 2.90 mmHg; 23.07 ± 2.84 mmHg versus 33.18 ± 2.30 mmHg; 22.98 ± 2.34 mmHg versus 36.38 ± 3.28 mmHg, p < 0.05). Meanwhile, the right heart hypertrophy index (36.2 1 ± 4.27% versus 41.01 ± 1.29%; 39.47 ± 4.02% versus 48.75 ± 2 .13%; 41.02 ± 0.9% versus 50.52 ± 1.49%, p < 0.05, respectively), ratio of wall/lumen thickness, as well as the wall/lumen area were significantly reduced in PAH rats at these time points following ADSCs-treatment, as compared with untreated PAH rats. In summary, ADSCs may colonize the pulmonary arteries, attenuate pulmonary arterial hypertension and ameliorate pulmonary arterial remodeling.
Collapse
Affiliation(s)
- Li Luo
- Fujian Hypertension Research Institute, the First Affiliated Hospital of Fujian Medical University , Fuzhou , P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Chen JY, An R, Liu ZJ, Wang JJ, Chen SZ, Hong MM, Liu JH, Xiao MY, Chen YF. Therapeutic effects of mesenchymal stem cell-derived microvesicles on pulmonary arterial hypertension in rats. Acta Pharmacol Sin 2014; 35:1121-8. [PMID: 25088001 DOI: 10.1038/aps.2014.61] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 05/26/2014] [Indexed: 02/06/2023]
Abstract
AIM Microvesicles (MVs) are nanoscale membrane fragments released from virtually all cell types upon activation or apoptosis, and may contribute to the beneficial effects of stem cell therapy. In this study, we investigated the therapeutic effects of mesenchymal stem cell (MSC) derived MVs (MSC-MVs) on pulmonary artery hypertension (PAH) in rats. METHODS MSC-MVs were isolated from rat bone marrow MSCs that were cultured in a serum-free conditioned medium. Transmission electron microscopy (TEM), flow cytometry and nanoparticle tracking analysis (NTA) were used to characterize the MVs. Adult SD rats were injected with monocrotaline (50 mg/kg, sc) to induce PAH. Three weeks later, the rats were intravenously injected with MSCs, MSC-MVs or saline for 2 weeks. At the end of treatments, the hemodynamic parameters and pathological right ventricular and pulmonary arterial remodeling were analyzed in each group. RESULTS The MSC-MVs showed general morphologic characteristics of MVs and expressed annexin V and CD29 markers under TEM, and their size ranged from 40 to 300 nm. Intravenous injection of MSC-MVs or MSCs significantly ameliorated the mean pulmonary artery pressure (mPAP) and mean right ventricle pressure (mRVP) in PAH rats. Furthermore, intravenous injection of MSC-MVs or MSCs significantly decreased the right ventricle (RV) hypertrophy and pulmonary arteriole area index (AI) and thickness index (TI) in PAH rats. CONCLUSION Intravenous injection of MSC-MVs or MSCs produces similar beneficial effects for treating PAH, and our results provide a basis for cell-free approach in stem cell therapy.
Collapse
|
44
|
Cardiac tissue structure, properties, and performance: a materials science perspective. Ann Biomed Eng 2014; 42:2003-13. [PMID: 25081385 DOI: 10.1007/s10439-014-1071-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 07/14/2014] [Indexed: 12/24/2022]
Abstract
From an engineering perspective, many forms of heart disease can be thought of as a reduction in biomaterial performance, in which the biomaterial is the tissue comprising the ventricular wall. In materials science, the structure and properties of a material are recognized to be interconnected with performance. In addition, for most measurements of structure, properties, and performance, some processing is required. Here, we review the current state of knowledge regarding cardiac tissue structure, properties, and performance as well as the processing steps taken to acquire those measurements. Understanding the impact of these factors and their interactions may enhance our understanding of heart function and heart failure. We also review design considerations for cardiac tissue property and performance measurements because, to date, most data on cardiac tissue has been obtained under non-physiological loading conditions. Novel measurement systems that account for these design considerations may improve future experiments and lead to greater insight into cardiac tissue structure, properties, and ultimately performance.
Collapse
|
45
|
Lanzola E, Farha S, Erzurum SC, Asosingh K. Bone marrow-derived vascular modulatory cells in pulmonary arterial hypertension. Pulm Circ 2014; 3:781-91. [PMID: 25006394 DOI: 10.1086/674769] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 06/28/2013] [Indexed: 12/23/2022] Open
Abstract
Hematopoiesis and vascular homeostasis are closely linked to each other via subsets of circulating bone marrow-derived cells with potent activity to repair endothelial injury and promote angiogenesis. As a consequence, abnormalities in hematopoiesis will eventually affect vascular health. Pulmonary arterial hypertension (PAH) is a vascular disease characterized by severe remodeling of the pulmonary artery wall. Over the past decade, circulating hematopoietic cells have been assigned an increasing role in the remodeling, such that these cells have been used in new therapeutic strategies. More recently, research has been extended to the bone marrow where these cells originate to identify abnormalities in hematopoiesis that may underlie PAH. Here, we review the current literature and identify gaps in knowledge of the myeloid effects on PAH.
Collapse
Affiliation(s)
- Emily Lanzola
- Department of Pathobiology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Samar Farha
- Department of Pathobiology, Cleveland Clinic, Cleveland, Ohio, USA ; Lerner Research Institute and Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Serpil C Erzurum
- Department of Pathobiology, Cleveland Clinic, Cleveland, Ohio, USA ; Lerner Research Institute and Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Kewal Asosingh
- Department of Pathobiology, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
46
|
Stem cells, cell therapies, and bioengineering in lung biology and diseases. Comprehensive review of the recent literature 2010-2012. Ann Am Thorac Soc 2014; 10:S45-97. [PMID: 23869446 DOI: 10.1513/annalsats.201304-090aw] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
A conference, "Stem Cells and Cell Therapies in Lung Biology and Lung Diseases," was held July 25 to 28, 2011 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 and ex vivo bioengineering approaches for lung diseases. These are rapidly expanding areas of study that provide further insight into and challenge traditional views of 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, to discuss and debate current controversies, and to identify future research directions and opportunities for basic and translational research in cell-based therapies for lung diseases. The goal of this article, which accompanies the formal conference report, is to provide a comprehensive review of the published literature in lung regenerative medicine from the last conference report through December 2012.
Collapse
|
47
|
Maarman G, Lecour S, Butrous G, Thienemann F, Sliwa K. A comprehensive review: the evolution of animal models in pulmonary hypertension research; are we there yet? Pulm Circ 2013; 3:739-56. [PMID: 25006392 PMCID: PMC4070827 DOI: 10.1086/674770] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Accepted: 06/28/2013] [Indexed: 02/06/2023] Open
Abstract
Pulmonary hypertension (PH) is a disorder that develops as a result of remodeling of the pulmonary vasculature and is characterized by narrowing/obliteration of small pulmonary arteries, leading to increased mean pulmonary artery pressure and pulmonary vascular resistance. Subsequently, PH increases the right ventricular afterload, which leads to right ventricular hypertrophy and eventually right ventricular failure. The pathophysiology of PH is not fully elucidated, and current treatments have only a modest impact on patient survival and quality of life. Thus, there is an urgent need for improved treatments or a cure. The use of animal models has contributed extensively to the current understanding of PH pathophysiology and the investigation of experimental treatments. However, PH in current animal models may not fully represent current clinical observations. For example, PH in animal models appears to be curable with many therapeutic interventions, and the severity of PH in animal models is also believed to correlate poorly with that observed in humans. In this review, we discuss a variety of animal models in PH research, some of their contributions to the field, their shortcomings, and how these have been addressed. We highlight the fact that the constant development and evolution of animal models will help us to more closely model the severity and heterogeneity of PH observed in humans.
Collapse
Affiliation(s)
- Gerald Maarman
- Hatter Institute for Cardiovascular Research in Africa (HICRA), Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular Research in Africa (HICRA), Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Ghazwan Butrous
- Pulmonary Vascular Research Institute, Kent Enterprise Hub, University of Kent, Canterbury, United Kingdom
| | - Friedrich Thienemann
- Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Karen Sliwa
- Hatter Institute for Cardiovascular Research in Africa (HICRA), Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
48
|
Zhang Y, Liang X, Lian Q, Tse HF. Perspective and challenges of mesenchymal stem cells for cardiovascular regeneration. Expert Rev Cardiovasc Ther 2013; 11:505-17. [PMID: 23570363 DOI: 10.1586/erc.13.5] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Mesenchymal stem cells (MSCs) exhibit multipotent differentiation potential and can be derived from embryonic, neonatal and adult differentiation stage III tissue sources. While increasing preclinical studies and clinical trials have indicated that MSC-based therapy is a promising strategy for cardiovascular regeneration, there are major challenges to overcome before this stem-cell technology can be widely applied in clinical settings. In this review, the following important issues will be addressed. First, optimal sources of MSC derivation suitable for myocardial repair are not determined. Second, assessments for preclinical and clinical studies of MSCs require more scientific data analysis. Third, mechanisms of MSC-based therapy for cardiovascular regeneration have not been fully understood yet. Finally, the potential benefit-risk ratio of MSC therapy needs to be evaluated systematically. Additionally, future development of MSC therapy will be discussed.
Collapse
Affiliation(s)
- Yuelin Zhang
- Cardiology Division, Department of Medicine, University of Hong Kong, Hong Kong
| | | | | | | |
Collapse
|
49
|
Inamdar AC, Inamdar AA. Mesenchymal stem cell therapy in lung disorders: pathogenesis of lung diseases and mechanism of action of mesenchymal stem cell. Exp Lung Res 2013; 39:315-27. [PMID: 23992090 DOI: 10.3109/01902148.2013.816803] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Lung disorders such as asthma, acute respiratory distress syndrome (ARDS), chronic obstructive lung disease (COPD), and interstitial lung disease (ILD) show a few common threads of pathogenic mechanisms: inflammation, aberrant immune activity, infection, and fibrosis. Currently no modes of effective treatment are available for ILD or emphysema. Being anti-inflammatory, immunomodulatory, and regenerative in nature, the administration of mesenchymal stem cells (MSCs) has shown the capacity to control immune dysfunction and inflammation in the lung. The intravenous infusion of MSCs, the common mode of delivery, is followed by their entrapment in lung vasculature before MSCs reach to other organ systems thus indicating the feasible and promising approach of MSCs therapy for lung diseases. In this review, we discuss the mechanistic basis for MSCs therapy for asthma, ARDS, COPD, and ILD.
Collapse
|
50
|
Dell'Italia LJ. The forgotten left ventricle in right ventricular pressure overload. J Am Coll Cardiol 2013; 57:929-30. [PMID: 21329839 DOI: 10.1016/j.jacc.2010.08.647] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Accepted: 08/17/2010] [Indexed: 10/18/2022]
|