1
|
Feng J, Lu Y, Wu H, Ma W, Zhang Y, Guo N. Knockdown of TSP-4 alleviates MI/RI-induced myocardial injury and improves brain inflammation by enhancing blood-brain barrier stability. Hum Mol Genet 2025:ddaf037. [PMID: 40156906 DOI: 10.1093/hmg/ddaf037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 04/01/2025] Open
Abstract
Myocardial ischemia-reperfusion injury (MI/RI) not only affects cardiac function but also has significant implications for neurological health, potentially leading to cognitive and behavioral impairments. At present, the regulatory role of thrombospondin-4 (TSP-4) in MI/RI has not been reported. A MI/RI mouse model was constructed, and primary cardiomyocytes were isolated. An MI/RI in vitro cell model was constructed using hypoxia/reoxygenation (H/R)-induced H9c2 cells. Haematoxylin and eosin and Masson staining were performed to observe morphological differentiation and fibrosis in myocardial tissues. Evans blue staining was used to analyse blood-brain barrier (BBB) permeability. Behavioural experiments were conducted to assess the learning and cognitive functions of mice. The results showed that the expression of TSP-4 was significantly increased in the blood of patients with ischemic cardiomyopathy and in the myocardial tissue of MI/RI mice. Functional studies showed that TSP knockdown alleviated H/R-induced H9c2 cell injury, including inflammation and oxidative stress. Importantly, interference with TSP-4 alleviated myocardial dysfunction in MI/RI mice. Mechanistically, by improving BBB stability, TSP-4 knockdown alleviated neuronal injury and the inflammatory response in mice induced by MI/RI. Further research found that silencing TSP-4 alleviated cognitive impairment and improved learning in MI/RI mice. Knockdown of TSP-4 improved MI/RI-induced functional cardiomyocyte injury. In addition, by enhancing BBB stability, TSP-4 silencing alleviated MI/RI-induced neurological injury and cognitive impairment in mice.
Collapse
Affiliation(s)
- Jiahao Feng
- The First Affiliated Hospital of Xi'an Jiaotong University, Department of Cardiology, No. 277, Yanta West Road, Yanta District, Xi'an, Shaanxi 710061, China
| | - Yanfeng Lu
- The First Affiliated Hospital of Xi'an Jiaotong University, Department of Cardiology, No. 277, Yanta West Road, Yanta District, Xi'an, Shaanxi 710061, China
| | - Haoyu Wu
- The First Affiliated Hospital of Xi'an Jiaotong University, Department of Cardiology, No. 277, Yanta West Road, Yanta District, Xi'an, Shaanxi 710061, China
| | - Wangge Ma
- The First Affiliated Hospital of Xi'an Jiaotong University, Department of Cardiology, No. 277, Yanta West Road, Yanta District, Xi'an, Shaanxi 710061, China
| | - Yong Zhang
- The First Affiliated Hospital of Xi'an Jiaotong University, Department of Cardiology, No. 277, Yanta West Road, Yanta District, Xi'an, Shaanxi 710061, China
| | - Ning Guo
- The First Affiliated Hospital of Xi'an Jiaotong University, Department of Cardiology, No. 277, Yanta West Road, Yanta District, Xi'an, Shaanxi 710061, China
| |
Collapse
|
2
|
Park G, Hwang DY, Kim DY, Han JY, Lee E, Hwang H, Park JS, Kim DW, Hong S, Yim SV, Hong HS, Son Y. Identification of CD141 +vasculogenic precursor cells from human bone marrow and their endothelial engagement in the arteriogenesis by co-transplantation with mesenchymal stem cells. Stem Cell Res Ther 2024; 15:388. [PMID: 39482744 PMCID: PMC11526567 DOI: 10.1186/s13287-024-03994-9] [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: 07/09/2024] [Accepted: 10/10/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Critical limb ischemia (CLI) is a condition characterized by insufficient blood flow to the lower limbs, resulting in severe ischemia and potentially leading to amputation. This study aims to identify novel vasculogenic precursor cells (VPCs) in human bone marrow and evaluate their efficacy in combination with bone marrow-derived mesenchymal stem cells (BM-MSCs) for the treatment of CLI. METHODS Ex vivo cultured VPCs and BM-MSCs from bone marrow were characterized and their effects on neovascularization and long-term tissue regeneration were tested in a mouse CLI model. RESULTS VPCs, expressing high levels of hepatocyte growth factor and c-MET, were identified from human bone marrow aspirates. These cells exhibited strong vasculogenic capacity in vitro but possessed a cellular phenotype distinct from those of previously reported endothelial precursor cells in circulation or cord blood. They also expressed most surface markers of BM-MSCs and demonstrated multipotent differentiation ability. Screening of 376 surface markers revealed that VPCs uniquely display CD141 (thrombomodulin). CD141+VPCs are present in BM aspirates as a rare population and can be expanded ex vivo with a population doubling time of approximately 20 h, generating an elaborate vascular network even under angiogenic factor-deficient conditions and recruiting BM-MSCs to the network as pericyte-like cells. Intramuscular transplantation of a combination of human CD141+VPCs and BM-MSCs at a ratio of 2:1 resulted in limb salvage, blood flow recovery, and regeneration of large vessels in the femoral artery-removed CLI model, with an efficacy superior to that of singular transplantation. Importantly, large arteries and arterioles in dual cell transplantation expressed human CD31 in the intima and human α-smooth muscle actin in media layer at 4 and 12 weeks, likely indicating their lineage commitment to endothelial cells and vascular smooth muscle, respectively, in vivo. CONCLUSION Dual-cell therapy using BM-derived CD141+ VPCs and BM-MSCs holds potential for further development in clinical trials to treat peripheral artery disease and diabetic ulcers.
Collapse
Affiliation(s)
- Gabee Park
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
| | - Dae Yeon Hwang
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
| | - Do Young Kim
- Department of Biomedical Science and Technology, Graduated School, Kyung Hee University, Seoul, Korea
| | - Ji Young Han
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
| | - Euiseon Lee
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
| | - Hwakyung Hwang
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
| | - Jeong Seop Park
- Department of Biomedical Science and Technology, Graduated School, Kyung Hee University, Seoul, Korea
| | - Dae Wook Kim
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
- Department of Genetic Engineering, Graduate School of Biotechnology, Kyung Hee University, Yong In, Korea
| | - Seonmin Hong
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
| | - Sung Vin Yim
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Hyun Sook Hong
- Department of Biomedical Science and Technology, Graduated School, Kyung Hee University, Seoul, Korea.
- East-West Medical Research Institute, Kyung Hee University, Seoul, Korea.
| | - Youngsook Son
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea.
- Department of Genetic Engineering, Graduate School of Biotechnology, Kyung Hee University, Yong In, Korea.
| |
Collapse
|
3
|
Flores-Opazo M, Kopinke D, Helmbacher F, Fernández-Verdejo R, Tuñón-Suárez M, Lynch GS, Contreras O. Fibro-adipogenic progenitors in physiological adipogenesis and intermuscular adipose tissue remodeling. Mol Aspects Med 2024; 97:101277. [PMID: 38788527 PMCID: PMC11692456 DOI: 10.1016/j.mam.2024.101277] [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: 02/01/2024] [Revised: 04/27/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024]
Abstract
Excessive accumulation of intermuscular adipose tissue (IMAT) is a common pathological feature in various metabolic and health conditions and can cause muscle atrophy, reduced function, inflammation, insulin resistance, cardiovascular issues, and unhealthy aging. Although IMAT results from fat accumulation in muscle, the mechanisms underlying its onset, development, cellular components, and functions remain unclear. IMAT levels are influenced by several factors, such as changes in the tissue environment, muscle type and origin, extent and duration of trauma, and persistent activation of fibro-adipogenic progenitors (FAPs). FAPs are a diverse and transcriptionally heterogeneous population of stromal cells essential for tissue maintenance, neuromuscular stability, and tissue regeneration. However, in cases of chronic inflammation and pathological conditions, FAPs expand and differentiate into adipocytes, resulting in the development of abnormal and ectopic IMAT. This review discusses the role of FAPs in adipogenesis and how they remodel IMAT. It highlights evidence supporting FAPs and FAP-derived adipocytes as constituents of IMAT, emphasizing their significance in adipose tissue maintenance and development, as well as their involvement in metabolic disorders, chronic pathologies and diseases. We also investigated the intricate molecular pathways and cell interactions governing FAP behavior, adipogenesis, and IMAT accumulation in chronic diseases and muscle deconditioning. Finally, we hypothesize that impaired cellular metabolic flexibility in dysfunctional muscles impacts FAPs, leading to IMAT. A deeper understanding of the biology of IMAT accumulation and the mechanisms regulating FAP behavior and fate are essential for the development of new therapeutic strategies for several debilitating conditions.
Collapse
Affiliation(s)
| | - Daniel Kopinke
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, 32610, FL, USA; Myology Institute, University of Florida College of Medicine, Gainesville, FL, USA.
| | | | - Rodrigo Fernández-Verdejo
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA; Laboratorio de Fisiología Del Ejercicio y Metabolismo (LABFEM), Escuela de Kinesiología, Facultad de Medicina, Universidad Finis Terrae, Chile.
| | - Mauro Tuñón-Suárez
- Laboratorio de Fisiología Del Ejercicio y Metabolismo (LABFEM), Escuela de Kinesiología, Facultad de Medicina, Universidad Finis Terrae, Chile.
| | - Gordon S Lynch
- Centre for Muscle Research, Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Parkville 3010, Australia.
| | - Osvaldo Contreras
- Developmental and Regenerative Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, 2010, Australia; School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia.
| |
Collapse
|
4
|
Athamneh M, Daya N, Hentschel A, Gangfuss A, Ruck T, Marina AD, Schara‐Schmidt U, Sickmann A, Güttsches A, Deschauer M, Preusse C, Vorgerd M, Roos A. Proteomic studies in VWA1-related neuromyopathy allowed new pathophysiological insights and the definition of blood biomarkers. J Cell Mol Med 2024; 28:e18122. [PMID: 38652110 PMCID: PMC11037410 DOI: 10.1111/jcmm.18122] [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: 09/23/2023] [Revised: 12/23/2023] [Accepted: 01/02/2024] [Indexed: 04/25/2024] Open
Abstract
Bi-allelic variants in VWA1, encoding Von Willebrand Factor A domain containing 1 protein localized to the extracellular matrix (ECM), were linked to a neuromuscular disorder with manifestation in child- or adulthood. Clinical findings indicate a neuromyopathy presenting with muscle weakness. Given that pathophysiological processes are still incompletely understood, and biomarkers are still missing, we aimed to identify blood biomarkers of pathophysiological relevance: white blood cells (WBC) and plasma derived from six VWA1-patients were investigated by proteomics. Four proteins, BET1, HNRNPDL, NEFM and PHGDH, known to be involved in neurological diseases and dysregulated in WBC were further validated by muscle-immunostainings unravelling HNRNPDL as a protein showing differences between VWA1-patients, healthy controls and patients suffering from neurogenic muscular atrophy and BICD2-related neuromyopathy. Immunostaining studies of PHGDH indicate its involvement in apoptotic processes via co-localisation with caspase-3. NEFM showed an increase in cells within the ECM in biopsies of all patients studied. Plasma proteomics unravelled dysregulation of 15 proteins serving as biomarker candidates among which a profound proportion of increased ones (6/11) are mostly related to antioxidative processes and have even partially been described as blood biomarkers for other entities of neuromuscular disorders before. CRP elevated in plasma also showed an increase in the extracellular space of VWA1-mutant muscle. Results of our combined studies for the first time describe pathophysiologically relevant biomarkers for VWA1-related neuromyopathy and suggest that VWA1-patient derived blood might hold the potential to study disease processes of clinical relevance, an important aspect for further preclinical studies.
Collapse
Affiliation(s)
- Mohammed Athamneh
- Department of Neurology, Heimer Institute for Muscle ResearchUniversity Hospital Bergmannsheil, Ruhr‐University BochumBochumGermany
- Department of Clinical Science, Faculty of MedicineYarmouk UniversityIrbidJordan
| | - Nassam Daya
- Department of Neurology, Heimer Institute for Muscle ResearchUniversity Hospital Bergmannsheil, Ruhr‐University BochumBochumGermany
| | - Andreas Hentschel
- Leibniz‐Institut für Analytische Wissenschaften‐ISAS‐e.V.DortmundGermany
| | - Andrea Gangfuss
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro‐ and Behavioral SciencesUniversity Duisburg‐EssenEssenGermany
| | - Tobias Ruck
- Department of Neurology, Medical FacultyHeinrich Heine University DüsseldorfDüsseldorfGermany
| | - Adela Della Marina
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro‐ and Behavioral SciencesUniversity Duisburg‐EssenEssenGermany
| | - Ulrike Schara‐Schmidt
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro‐ and Behavioral SciencesUniversity Duisburg‐EssenEssenGermany
| | - Albert Sickmann
- Leibniz‐Institut für Analytische Wissenschaften‐ISAS‐e.V.DortmundGermany
| | - Anne‐Katrin Güttsches
- Department of Neurology, Heimer Institute for Muscle ResearchUniversity Hospital Bergmannsheil, Ruhr‐University BochumBochumGermany
| | - Marcus Deschauer
- Department of NeurologyTechnical University of Munich, School of MedicineMunichGermany
| | - Corinna Preusse
- Institute of Neuropathology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität BerlinHumboldt Universität zu Berlin, Berlin Institute of HealthBerlinGermany
| | - Matthias Vorgerd
- Department of Neurology, Heimer Institute for Muscle ResearchUniversity Hospital Bergmannsheil, Ruhr‐University BochumBochumGermany
| | - Andreas Roos
- Department of Neurology, Heimer Institute for Muscle ResearchUniversity Hospital Bergmannsheil, Ruhr‐University BochumBochumGermany
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro‐ and Behavioral SciencesUniversity Duisburg‐EssenEssenGermany
- Children's Hospital of Eastern Ontario Research InstituteUniversity of OttawaOttawaCanada
| |
Collapse
|
5
|
Pan H, Lu X, Ye D, Feng Y, Wan J, Ye J. The molecular mechanism of thrombospondin family members in cardiovascular diseases. Front Cardiovasc Med 2024; 11:1337586. [PMID: 38516004 PMCID: PMC10954798 DOI: 10.3389/fcvm.2024.1337586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/14/2024] [Indexed: 03/23/2024] Open
Abstract
Cardiovascular diseases have been identified as vital factors in global morbidity and mortality in recent years. The available evidence suggests that various cytokines and pathological proteins participate in these complicated and changeable diseases. The thrombospondin (TSP) family is a series of conserved, multidomain calcium-binding glycoproteins that cause cell-matrix and cell-cell effects via interactions with other extracellular matrix components and cell surface receptors. The TSP family has five members that can be divided into two groups (Group A and Group B) based on their different structures. TSP-1, TSP-2, and TSP-4 are the most studied proteins. Among recent studies and findings, we investigated the functions of several family members, especially TSP-5. We review the basic concepts of TSPs and summarize the relevant molecular mechanisms and cell interactions in the cardiovascular system. Targeting TSPs in CVD and other diseases has a remarkable therapeutic benefit.
Collapse
Affiliation(s)
- Heng Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xiyi Lu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yongqi Feng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jing Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
6
|
Lin HF, Wu MN, Chen CY, Lim K, Juo SHH, Chen CS. Thrombospondin-1 associated with carotid intima-media thickness among individuals with hypertension. J Investig Med 2024; 72:279-286. [PMID: 38217383 DOI: 10.1177/10815589241228589] [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] [Indexed: 01/15/2024]
Abstract
In vivo and in vitro studies have demonstrated that thrombospondin-1 (TSP-1) is involved in atherosclerotic pathogenesis. However, the role of TSP-1 in clinical atherosclerosis remains unknown. This cross-sectional study investigated the relationship between TSP-1 and carotid intima-media thickness (IMT) and examined whether it interacts with conventional cardiovascular risk factors. A total of 587 participants were enrolled from February 2018 to December 2021. TSP-1 was dichotomized based on median value. Carotid IMT was measured bilaterally in each segment, and the average value was taken as the overall IMT variable. Analysis of covariance models were used to ascertain the main and interaction effects of cardiovascular risk factors and circulating TSP-1 levels on carotid IMT. Those with high TSP-1 (n = 294) had significantly higher carotid IMT than did those with low TSP-1 (n = 293; 0.74 ± 0.12 vs 0.72 ± 0.11 mm; p = 0.011). After the combined effects of TSP-1 and vascular risk factors on carotid IMT were evaluated, an interaction effect on IMT was observed between TSP-1 and hypertension (adjusted F = 8.760; p = 0.003). Stratification analysis revealed that individuals with hypertension and high TSP-1 had significantly higher IMT than did those with low TSP-1 (adjusted p = 0.007). However, this difference was not observed in normotensive individuals (adjusted p = 0.636). In conclusion, this is the first study to provide clinical data supporting the correlation between TSP-1 and atherosclerosis. TSP-1 may be a crucial marker of increased susceptibility to atherosclerosis in individuals with hypertension.
Collapse
Affiliation(s)
- Hsiu-Fen Lin
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Neurology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Meng-Ni Wu
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Neurology, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chien-Yuan Chen
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kelly Lim
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Suh-Hang Hank Juo
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- Institute of Translational Medicine and New Drug Development, China Medical University, Taichung, Taiwan
- Drug Development Center, China Medical University, Taichung, Taiwan
| | - Cheng-Sheng Chen
- Department of Psychiatry, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Psychiatry, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
7
|
Cervia-Hasler C, Brüningk SC, Hoch T, Fan B, Muzio G, Thompson RC, Ceglarek L, Meledin R, Westermann P, Emmenegger M, Taeschler P, Zurbuchen Y, Pons M, Menges D, Ballouz T, Cervia-Hasler S, Adamo S, Merad M, Charney AW, Puhan M, Brodin P, Nilsson J, Aguzzi A, Raeber ME, Messner CB, Beckmann ND, Borgwardt K, Boyman O. Persistent complement dysregulation with signs of thromboinflammation in active Long Covid. Science 2024; 383:eadg7942. [PMID: 38236961 DOI: 10.1126/science.adg7942] [Citation(s) in RCA: 152] [Impact Index Per Article: 152.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 11/24/2023] [Indexed: 01/23/2024]
Abstract
Long Covid is a debilitating condition of unknown etiology. We performed multimodal proteomics analyses of blood serum from COVID-19 patients followed up to 12 months after confirmed severe acute respiratory syndrome coronavirus 2 infection. Analysis of >6500 proteins in 268 longitudinal samples revealed dysregulated activation of the complement system, an innate immune protection and homeostasis mechanism, in individuals experiencing Long Covid. Thus, active Long Covid was characterized by terminal complement system dysregulation and ongoing activation of the alternative and classical complement pathways, the latter associated with increased antibody titers against several herpesviruses possibly stimulating this pathway. Moreover, markers of hemolysis, tissue injury, platelet activation, and monocyte-platelet aggregates were increased in Long Covid. Machine learning confirmed complement and thromboinflammatory proteins as top biomarkers, warranting diagnostic and therapeutic interrogation of these systems.
Collapse
Affiliation(s)
- Carlo Cervia-Hasler
- Department of Immunology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Sarah C Brüningk
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Tobias Hoch
- Department of Immunology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Bowen Fan
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Giulia Muzio
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Ryan C Thompson
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Laura Ceglarek
- Department of Immunology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Roman Meledin
- Department of Immunology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Patrick Westermann
- Precision Proteomics Center, Swiss Institute of Allergy and Asthma Research, University of Zurich, 7265 Davos, Switzerland
| | - Marc Emmenegger
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Patrick Taeschler
- Department of Immunology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Yves Zurbuchen
- Department of Immunology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Michele Pons
- Department of Immunology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Dominik Menges
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, 8001 Zurich, Switzerland
| | - Tala Ballouz
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, 8001 Zurich, Switzerland
| | - Sara Cervia-Hasler
- Department of Immunology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Sarah Adamo
- Department of Immunology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Miriam Merad
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alexander W Charney
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Milo Puhan
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, 8001 Zurich, Switzerland
| | - Petter Brodin
- Unit for Clinical Pediatrics, Department of Women's and Children's Health, Karolinska Institute, 17165 Solna, Sweden
- Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK
| | - Jakob Nilsson
- Department of Immunology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Miro E Raeber
- Department of Immunology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Christoph B Messner
- Precision Proteomics Center, Swiss Institute of Allergy and Asthma Research, University of Zurich, 7265 Davos, Switzerland
| | - Noam D Beckmann
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Division of Data Driven and Digital Medicine (D3M), Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Karsten Borgwardt
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Onur Boyman
- Department of Immunology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
- Faculty of Medicine and Faculty of Science, University of Zurich, 8006 Zurich, Switzerland
| |
Collapse
|
8
|
Tang Y, Wang Y, Wang S, Wang R, Xu J, Peng Y, Ding L, Zhao J, Zhou G, Sun S, Zhang Z. Methylation and transcriptomic expression profiles of HUVEC in the oxygen and glucose deprivation model and its clinical implications in AMI patients. Front Genet 2023; 14:1293393. [PMID: 38145212 PMCID: PMC10740152 DOI: 10.3389/fgene.2023.1293393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/15/2023] [Indexed: 12/26/2023] Open
Abstract
The obstructed coronary artery undergoes a series of pathological changes due to ischemic-hypoxic shocks during acute myocardial infarction (AMI). However, the altered DNA methylation levels in endothelial cells under these conditions and their implication for the etiopathology of AMI have not been investigated in detail. This study aimed to explore the relationship between DNA methylation and pathologically altered gene expression profile in human umbilical vein endothelial cells (HUVECs) subjected to oxygen-glucose deprivation (OGD), and its clinical implications in AMI patients. The Illumina Infinium MethylationEPIC BeadChip assay was used to explore the genome-wide DNA methylation profile using the Novaseq6000 platform for mRNA sequencing in 3 pairs of HUVEC-OGD and control samples. GO and KEGG pathway enrichment analyses, as well as correlation, causal inference test (CIT), and protein-protein interaction (PPI) analyses identified 22 hub genes that were validated by MethylTarget sequencing as well as qRT-PCR. ELISA was used to detect four target molecules associated with the progression of AMI. A total of 2,524 differentially expressed genes (DEGs) and 22,148 differentially methylated positions (DMPs) corresponding to 6,642 differentially methylated genes (DMGs) were screened (|Δβ|>0.1 and detection p < 0.05). After GO, KEGG, correlation, CIT, and PPI analyses, 441 genes were filtered. qRT-PCR confirmed the overexpression of VEGFA, CCL2, TSP-1, SQSTM1, BCL2L11, and TIMP3 genes, and downregulation of MYC, CD44, BDNF, GNAQ, RUNX1, ETS1, NGFR, MME, SEMA6A, GNAI1, IFIT1, and MEIS1. DNA fragments BDNF_1_ (r = 0.931, p < 0.0001) and SQSTM1_2_NEW (r = 0.758, p = 0.0043) were positively correlated with the expressions of corresponding genes, and MYC_1_ (r = -0.8245, p = 0.001) was negatively correlated. Furthermore, ELISA confirmed TNFSF10 and BDNF were elevated in the peripheral blood of AMI patients (p = 0.0284 and p = 0.0142, respectively). Combined sequencing from in vitro cellular assays with clinical samples, aiming to establish the potential causal chain of the causal factor (DNA methylation) - mediator (mRNA)-cell outcome (endothelial cell ischemic-hypoxic injury)-clinical outcome (AMI), our study identified promising OGD-specific genes, which provided a solid basis for screening fundamental diagnostic and prognostic biomarkers of coronary endothelial cell injury of AMI. Moreover, it furnished the first evidence that during ischemia and hypoxia, the expression of BNDF was regulated by DNA methylation in endothelial cells and elevated in peripheral blood.
Collapse
Affiliation(s)
- Yuning Tang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- Gansu Key Laboratory of Cardiovascular Diseases, The First Hospital of Lanzhou University, Lanzhou, China
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou, China
- Cardiovascular Clinical Research Center of Gansu Province, Lanzhou, China
| | - Yongxiang Wang
- Gansu Key Laboratory of Cardiovascular Diseases, The First Hospital of Lanzhou University, Lanzhou, China
- Cardiovascular Clinical Research Center of Gansu Province, Lanzhou, China
- Heart Center, The First Hospital of Lanzhou University, Lanzhou, China
| | - Shengxiang Wang
- School of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Runqing Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- Gansu Key Laboratory of Cardiovascular Diseases, The First Hospital of Lanzhou University, Lanzhou, China
- Cardiovascular Clinical Research Center of Gansu Province, Lanzhou, China
| | - Jin Xu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- Gansu Key Laboratory of Cardiovascular Diseases, The First Hospital of Lanzhou University, Lanzhou, China
- Cardiovascular Clinical Research Center of Gansu Province, Lanzhou, China
| | - Yu Peng
- Gansu Key Laboratory of Cardiovascular Diseases, The First Hospital of Lanzhou University, Lanzhou, China
- Cardiovascular Clinical Research Center of Gansu Province, Lanzhou, China
- Heart Center, The First Hospital of Lanzhou University, Lanzhou, China
| | - Liqiong Ding
- Gansu Key Laboratory of Cardiovascular Diseases, The First Hospital of Lanzhou University, Lanzhou, China
- Cardiovascular Clinical Research Center of Gansu Province, Lanzhou, China
- Heart Center, The First Hospital of Lanzhou University, Lanzhou, China
| | - Jing Zhao
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- Gansu Key Laboratory of Cardiovascular Diseases, The First Hospital of Lanzhou University, Lanzhou, China
- Cardiovascular Clinical Research Center of Gansu Province, Lanzhou, China
- Heart Center, The First Hospital of Lanzhou University, Lanzhou, China
| | - Gang Zhou
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Shougang Sun
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou, China
| | - Zheng Zhang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- Gansu Key Laboratory of Cardiovascular Diseases, The First Hospital of Lanzhou University, Lanzhou, China
- Cardiovascular Clinical Research Center of Gansu Province, Lanzhou, China
- Heart Center, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
9
|
Sánchez Marrero G, Villa-Roel N, Li F, Park C, Kang DW, Hekman KE, Jo H, Brewster LP. Single-Cell RNA sequencing investigation of female-male differences under PAD conditions. Front Cardiovasc Med 2023; 10:1251141. [PMID: 37745110 PMCID: PMC10512722 DOI: 10.3389/fcvm.2023.1251141] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/15/2023] [Indexed: 09/26/2023] Open
Abstract
Peripheral arterial disease (PAD) is an age-related medical condition affecting mostly muscular arteries of the limb. It is the 3rd leading cause of atherosclerotic morbidity. The mechanical environment of endothelial cells (ECs) in PAD is characterized by disturbed blood flow (d-flow) and stiff extracellular matrices. In PAD, the stiffness of arteries is due to decreased elastin function and increased collagen content. These flow and stiffness parameters are largely missing from current models of PAD. It has been previously proven that ECs exposed to d-flow or stiff substrates lead to proatherogenic pathways, but the effect of both, d-flow and stiffness, on EC phenotype has not been fully investigated. In this study, we sought to explore the effect of sex on proatherogenic pathways that could result from exposing endothelial cells to a d-flow and stiff environment. We utilized the scRNA-seq tool to analyze the gene expression of ECs exposed to the different mechanical conditions both in vitro and in vivo. We found that male ECs exposed to different mechanical stimuli presented higher expression of genes related to fibrosis and d-flow in vitro. We validated our findings in vivo by exposing murine carotid arteries to d-flow via partial carotid artery ligation. Since women have delayed onset of arterial stiffening and subsequent PAD, this work may provide a framework for some of the pathways in which biological sex interacts with sex-based differences in PAD.
Collapse
Affiliation(s)
- Gloriani Sánchez Marrero
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Nicolas Villa-Roel
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Feifei Li
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States
| | - Christian Park
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Dong-Won Kang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Katherine E. Hekman
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States
- Surgical and Research Services, Atlanta Veteran Affairs Medical Center, Decatur, GA, United States
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Luke P. Brewster
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States
- Surgical and Research Services, Atlanta Veteran Affairs Medical Center, Decatur, GA, United States
| |
Collapse
|
10
|
WNK1 collaborates with TGF-β in endothelial cell junction turnover and angiogenesis. Proc Natl Acad Sci U S A 2022; 119:e2203743119. [PMID: 35867836 PMCID: PMC9335306 DOI: 10.1073/pnas.2203743119] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Angiogenesis is essential for growth of new blood vessels, remodeling existing vessels, and repair of damaged vessels, and these require reorganization of endothelial cell-cell junctions through a partial endothelial-mesenchymal transition. Homozygous disruption of the gene encoding the protein kinase WNK1 results in lethality in mice near embryonic day (E) 12 due to impaired angiogenesis. This angiogenesis defect can be rescued by endothelial-specific expression of an activated form of the WNK1 substrate kinase OSR1. We show that inhibition of WNK1 kinase activity not only prevents sprouting of endothelial cells from aortic slices but also vessel extension in inhibitor-treated embryos ex vivo. Mutations affecting TGF-β signaling also result in abnormal vascular development beginning by E10 and, ultimately, embryonic lethality. Previously, we demonstrated cross-talk of WNK1 with TGF-β-regulated SMAD signaling, and OSR1 was identified as a component of the TGF-β interactome. However, molecular events jointly regulated by TGF-β and WNK1/OSR1 have not been delineated. Here, we show that inhibition of WNK1 promotes TGF-β-dependent degradation of the tyrosine kinase receptor AXL, which is involved in TGF-β-mediated cell migration and angiogenesis. We also show that interaction between OSR1 and occludin, a protein associated with endothelial tight junctions, is an essential step to enable tight junction turnover. Furthermore, we show that these phenomena are WNK1 dependent, and sensitive to TGF-β. These findings demonstrate intimate connections between WNK1/OSR1 and multiple TGF-β-sensitive molecules controlling angiogenesis and suggest that WNK1 may modulate many TGF-β-regulated functions.
Collapse
|
11
|
Bui L, Edwards S, Hall E, Alderfer L, Round K, Owen M, Sainaghi P, Zhang S, Nallathamby PD, Haneline LS, Hanjaya-Putra D. Engineering bioactive nanoparticles to rejuvenate vascular progenitor cells. Commun Biol 2022; 5:635. [PMID: 35768543 PMCID: PMC9243106 DOI: 10.1038/s42003-022-03578-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 06/08/2022] [Indexed: 11/29/2022] Open
Abstract
Fetal exposure to gestational diabetes mellitus (GDM) predisposes children to future health complications including type-2 diabetes mellitus, hypertension, and cardiovascular disease. A key mechanism by which these complications occur is through stress-induced dysfunction of endothelial progenitor cells (EPCs), including endothelial colony-forming cells (ECFCs). Although several approaches have been previously explored to restore endothelial function, their widespread adoption remains tampered by systemic side effects of adjuvant drugs and unintended immune response of gene therapies. Here, we report a strategy to rejuvenate circulating vascular progenitor cells by conjugation of drug-loaded liposomal nanoparticles directly to the surface of GDM-exposed ECFCs (GDM-ECFCs). Bioactive nanoparticles can be robustly conjugated to the surface of ECFCs without altering cell viability and key progenitor phenotypes. Moreover, controlled delivery of therapeutic drugs to GDM-ECFCs is able to normalize transgelin (TAGLN) expression and improve cell migration, which is a critical key step in establishing functional vascular networks. More importantly, sustained pseudo-autocrine stimulation with bioactive nanoparticles is able to improve in vitro and in vivo vasculogenesis of GDM-ECFCs. Collectively, these findings highlight a simple, yet promising strategy to rejuvenate GDM-ECFCs and improve their therapeutic potential. Promising results from this study warrant future investigations on the prospect of the proposed strategy to improve dysfunctional vascular progenitor cells in the context of other chronic diseases, which has broad implications for addressing various cardiovascular complications, as well as advancing tissue repair and regenerative medicine. Drug-loaded liposomal nanoparticles conjugated to endothelial colony-forming cells can improve the vasculogenic potential of vascular progenitor cells exposed to gestational diabetes mellitus.
Collapse
Affiliation(s)
- Loan Bui
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Shanique Edwards
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Riley Hospital for Children at Indiana University Health, Indianapolis, IN, 46202, USA
| | - Eva Hall
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Laura Alderfer
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Kellen Round
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Madeline Owen
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Pietro Sainaghi
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Siyuan Zhang
- Department of Biological Science, University of Notre Dame, Notre Dame, IN, 46556, USA.,Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Prakash D Nallathamby
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Laura S Haneline
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Riley Hospital for Children at Indiana University Health, Indianapolis, IN, 46202, USA.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Donny Hanjaya-Putra
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, 46556, USA. .,Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, 46556, USA. .,Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA. .,Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, 46556, USA.
| |
Collapse
|
12
|
Weiss L, Keaney J, Szklanna PB, Prendiville T, Uhrig W, Wynne K, Kelliher S, Ewins K, Comer SP, Egan K, O'Rourke E, Moran E, Petrov G, Patel A, Lennon Á, Blanco A, Kevane B, Murphy S, Ní Áinle F, Maguire PB. Nonvalvular atrial fibrillation patients anticoagulated with rivaroxaban compared with warfarin exhibit reduced circulating extracellular vesicles with attenuated pro-inflammatory protein signatures. J Thromb Haemost 2021; 19:2583-2595. [PMID: 34161660 DOI: 10.1111/jth.15434] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 06/02/2021] [Accepted: 06/22/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Rivaroxaban, a direct oral factor Xa inhibitor, mediates anti-inflammatory and cardiovascular-protective effects besides its well-established anticoagulant properties; however, these remain poorly characterized. Extracellular vesicles (EVs) are important circulating messengers regulating a myriad of biological and pathological processes and may be highly relevant to the pathophysiology of atrial fibrillation as they reflect alterations in platelet and endothelial biology. However, the effects of rivaroxaban on circulating pro-inflammatory EVs remain unknown. OBJECTIVES We hypothesized that rivaroxaban's anti-inflammatory properties are reflected upon differential molecular profiles of circulating EVs. METHODS Differences in circulating EV profiles were assessed using a combination of single vesicle analysis by Nanoparticle Tracking Analysis and flow cytometry, and proteomics. RESULTS We demonstrate, for the first time, that rivaroxaban-treated non-valvular atrial fibrillation (NVAF) patients (n=8) exhibit attenuated inflammation compared with matched warfarin controls (n=15). Circulating EV profiles were fundamentally altered. Moreover, quantitative proteomic analysis of enriched plasma EVs from six pooled biological donors per treatment group revealed a profound decrease in highly pro-inflammatory protein expression and complement factors, together with increased expression of negative regulators of inflammatory pathways. Crucially, a reduction in circulating levels of soluble P-selectin was observed in rivaroxaban-treated patients (compared with warfarin controls), which negatively correlated with the patient's time on treatment. CONCLUSION Collectively, these data demonstrate that NVAF patients anticoagulated with rivaroxaban (compared with warfarin) exhibit both a reduced pro-inflammatory state and evidence of reduced endothelial activation. These findings are of translational relevance toward characterizing the anti-inflammatory and cardiovascular-protective mechanisms associated with rivaroxaban therapy.
Collapse
Affiliation(s)
- Luisa Weiss
- UCD Conway SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - John Keaney
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Cardiology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Paulina B Szklanna
- UCD Conway SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Tadhg Prendiville
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Wido Uhrig
- UCD Conway SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
| | - Kieran Wynne
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
| | - Sarah Kelliher
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Karl Ewins
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Shane P Comer
- UCD Conway SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Karl Egan
- UCD Conway SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Ellen O'Rourke
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Eric Moran
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Georgi Petrov
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Ashish Patel
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Áine Lennon
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Alfonso Blanco
- Flow Cytometry Core, Conway Institute, University College Dublin, Dublin, Ireland
| | - Barry Kevane
- UCD Conway SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Sean Murphy
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Stroke Medicine, Mater Misericordiae University Hospital, Dublin, Ireland
- School of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Fionnuala Ní Áinle
- UCD Conway SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Patricia B Maguire
- UCD Conway SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
- UCD Institute for Discovery, O'Brien Centre for Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
13
|
Valproic Acid Decreases Endothelial Colony Forming Cells Differentiation and Induces Endothelial-to-Mesenchymal Transition-like Process. Stem Cell Rev Rep 2021; 16:357-368. [PMID: 31898801 DOI: 10.1007/s12015-019-09950-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Valproic acid (VPA), a histone deacetylase (HDAC) inhibitor is a widely used anticonvulsant drug. VPA is also under clinical evaluation to be employed in anticancer therapy, as an antithrombotic agent or a molecule to be used in the stem cells expansion protocols. Since endothelial colony forming cells (ECFC) has been identified as the human postnatal vasculogenic cells involved in thrombotic disorders and serve as a promising source of immature cell for vascular repair, objectives of the present study were to determine how VPA contributes to ECFC commitment and their angiogenic properties. We examined the effect of VPA on ECFC obtained from cord blood by evaluating colony number, proliferation, migration and their sprouting ability in vitro, as well as their in vivo vasculogenic properties. VPA inhibited endothelial differentiation potential from of cord blood derived stem cells associated with decreased proliferation and sprouting activity of cultured ECFC. VPA treatment significantly decreased the vessel-forming ability of ECFC transplanted together with mesenchymal stem cells (MSC) in Matrigel implants in nude mice model. Surprisingly, a microscopic evaluation revealed that VPA induces marked morphological changes from a cobblestone-like EC morphology to enlarged spindle shaped morphology of ECFC. RT-qPCR and a CD31/CD90 flow cytometry analysis confirmed a phenotypic switch of VPA-treated ECFC to mesenchymal-like phenotype. In conclusion, the pan-HDAC inhibitor VPA described for expansion of hematopoietic stem cells and very small embryonic like stem cells cannot be successfully employed for differentiation of endothelial lineage committed ECFC into functional endothelial cells. Our data also suggest that VPA based therapeutics may induce endothelial dysfunction associated with fibrosis that might induce thrombosis recurrence or venous insufficiency.
Collapse
|
14
|
Helle E, Ampuja M, Antola L, Kivelä R. Flow-Induced Transcriptomic Remodeling of Endothelial Cells Derived From Human Induced Pluripotent Stem Cells. Front Physiol 2020; 11:591450. [PMID: 33178051 PMCID: PMC7593792 DOI: 10.3389/fphys.2020.591450] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 09/16/2020] [Indexed: 12/31/2022] Open
Abstract
The vascular system is essential for the development and function of all organs and tissues in our body. The molecular signature and phenotype of endothelial cells (EC) are greatly affected by blood flow-induced shear stress, which is a vital component of vascular development and homeostasis. Recent advances in differentiation of ECs from human induced pluripotent stem cells (hiPSC) have enabled development of in vitro experimental models of the vasculature containing cells from healthy individuals or from patients harboring genetic variants or diseases of interest. Here we have used hiPSC-derived ECs and bulk- and single-cell RNA sequencing to study the effect of flow on the transcriptomic landscape of hiPSC-ECs and their heterogeneity. We demonstrate that hiPS-ECs are plastic and they adapt to flow by expressing known flow-induced genes. Single-cell RNA sequencing showed that flow induced a more homogenous and homeostatically more stable EC population compared to static cultures, as genes related to cell polarization, barrier formation and glucose and fatty acid transport were induced. The hiPS-ECs increased both arterial and venous markers when exposed to flow. Interestingly, while in general there was a greater increase in the venous markers, one cluster with more arterial-like hiPS-ECs was detected. Single-cell RNA sequencing revealed that not all hiPS-ECs are similar even after sorting, but exposing them to flow increases their homogeneity. Since hiPS-ECs resemble immature ECs and demonstrate high plasticity in response to flow, they provide an excellent model to study vascular development.
Collapse
Affiliation(s)
- Emmi Helle
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- New Children’s Hospital, and Pediatric Research Center Helsinki University Hospital, Helsinki, Finland
| | - Minna Ampuja
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Laura Antola
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Riikka Kivelä
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Wihuri Research Institute, Helsinki, Finland
| |
Collapse
|
15
|
Therapeutic Potential of Endothelial Colony-Forming Cells in Ischemic Disease: Strategies to Improve their Regenerative Efficacy. Int J Mol Sci 2020; 21:ijms21197406. [PMID: 33036489 PMCID: PMC7582994 DOI: 10.3390/ijms21197406] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/02/2020] [Accepted: 10/02/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular disease (CVD) comprises a range of major clinical cardiac and circulatory diseases, which produce immense health and economic burdens worldwide. Currently, vascular regenerative surgery represents the most employed therapeutic option to treat ischemic disorders, even though not all the patients are amenable to surgical revascularization. Therefore, more efficient therapeutic approaches are urgently required to promote neovascularization. Therapeutic angiogenesis represents an emerging strategy that aims at reconstructing the damaged vascular network by stimulating local angiogenesis and/or promoting de novo blood vessel formation according to a process known as vasculogenesis. In turn, circulating endothelial colony-forming cells (ECFCs) represent truly endothelial precursors, which display high clonogenic potential and have the documented ability to originate de novo blood vessels in vivo. Therefore, ECFCs are regarded as the most promising cellular candidate to promote therapeutic angiogenesis in patients suffering from CVD. The current briefly summarizes the available information about the origin and characterization of ECFCs and then widely illustrates the preclinical studies that assessed their regenerative efficacy in a variety of ischemic disorders, including acute myocardial infarction, peripheral artery disease, ischemic brain disease, and retinopathy. Then, we describe the most common pharmacological, genetic, and epigenetic strategies employed to enhance the vasoreparative potential of autologous ECFCs by manipulating crucial pro-angiogenic signaling pathways, e.g., extracellular-signal regulated kinase/Akt, phosphoinositide 3-kinase, and Ca2+ signaling. We conclude by discussing the possibility of targeting circulating ECFCs to rescue their dysfunctional phenotype and promote neovascularization in the presence of CVD.
Collapse
|
16
|
Alehagen U, Shamoun L, Wågsäter D. Increased cardiovascular mortality in females with the a/a genotype of the SNPs rs1478604 and rs2228262 of thrombospondin-1. BMC MEDICAL GENETICS 2020; 21:179. [PMID: 32917134 PMCID: PMC7488716 DOI: 10.1186/s12881-020-01118-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 09/02/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Cardiovascular diseases are still the major cause of death in the Western world, with different outcomes between the two genders. Efforts to identify those at risk are therefore given priority in the handling of health resources. Thrombospondins (TSP) are extracellular matrix proteins associated with cardiovascular diseases. The aim of this study was to investigate variations in single nucleotide polymorphisms (SNPs) of TSP-1 and plasma expression, and associations with mortality from a gender perspective. METHODS A population of 470 community-living persons were invited to participate. The participants were followed for 7.9 years and underwent a clinical examination and blood sampling. SNP analyses of TSP-1 rs1478604 and rs2228262 using allelic discrimination and plasma measurement of TSP-1 using ELISA were performed, RESULTS: During the follow-up period, 135 (28.7%) all-cause and 83 (17.7%) cardiovascular deaths were registered. In the female population, the A/A genotype of rs2228262 and the T/T genotype of rs1478604 exhibited significantly more cardiovascular deaths compared with the A/G and G/G, or the T/C and C/C genotypes amalgamated (rs2228262: 13.7% vs 2.0%; Χ2:5.29; P = 0.02; rs1478604:17.7% vs 4.7%; Χ2:9.50; P = 0.002). Applied in a risk evaluation, the A/A, or T/T genotypes exhibited an increased risk of cardiovascular mortality (rs2228262: HR: 7.1; 95%CI 1.11-45.8; P = 0.04; rs1478604: HR: 3.18; 95%CI 1.35-7.50; p = 0.008). No differences among the three genotypes could be seen in the male group. CONCLUSION In this study the female group having the A/A genotype of rs2228262, or the T/T genotype of rs1478604 of TSP-1 exhibited higher cardiovascular mortality after a follow-up of almost 8 years. No corresponding genotype differences could be found in the male group. Genotype evaluations should be considered as one of the options to identify individuals at risk. However, this study should be regarded as hypothesis-generating, and more research in the field is needed.
Collapse
Affiliation(s)
- Urban Alehagen
- Institution of Medical and Health Sciences, Division of Cardiovascular Medicine, Department of Medicine and Health Sciences, Faculty of Health Sciences, Linköping University, SE-581 85, Linköping, Sweden.
| | - Levar Shamoun
- Division of Medical Diagnostics, Department of Laboratory Medicine, Jönköping County, Jönköping, Sweden.,Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Dick Wågsäter
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
17
|
Julovi SM, Sanganeria B, Minhas N, Ghimire K, Nankivell B, Rogers NM. Blocking thrombospondin-1 signaling via CD47 mitigates renal interstitial fibrosis. J Transl Med 2020; 100:1184-1196. [PMID: 32366943 DOI: 10.1038/s41374-020-0434-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 03/31/2020] [Accepted: 04/11/2020] [Indexed: 11/10/2022] Open
Abstract
Acute kidney injury triggers a complex cascade of molecular responses that can culminate in maladaptive repair and fibrosis. We have previously reported that the matrix protein thrombospondin-1 (TSP1), binding its high affinity its receptor CD47, promotes acute kidney injury. However, the role of this pathway in promoting fibrosis is less clear. Hypothesizing that limiting TSP1-CD47 signaling is protective against fibrosis, we interrogated this pathway in a mouse model of chronic ischemic kidney injury. Plasma and renal parenchymal expression of TSP1 in patients with chronic kidney disease was also assessed. We found that CD47-/- mice or wild-type mice treated with a CD47 blocking antibody showed clear amelioration of fibrotic histological changes compared to control animals. Wild-type mice showed upregulated TSP1 and pro-fibrotic markers which were significantly abrogated in CD47-/- and antibody-treated cohorts. Renal tubular epithelial cells isolated from WT mice showed robust upregulation of pro-fibrotic markers following hypoxic stress or exogenous TSP1, which was mitigated in CD47-/- cells. Patient sera showed a proportionate correlation between TSP1 levels and worsening glomerular filtration rate. Immunohistochemistry of human kidney tissue demonstrated tubular and glomerular matrix localization of TSP1 expression in patients with CKD. These data suggest that renal tubular epithelial cells contribute to fibrosis by activating TSP1-CD47 signaling, and point to CD47 as a potential target to limit fibrosis following ischemic injury.
Collapse
Affiliation(s)
- Sohel M Julovi
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Camperdown, NSW, Australia
| | - Barkha Sanganeria
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Camperdown, NSW, Australia
| | - Nikita Minhas
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Camperdown, NSW, Australia
| | - Kedar Ghimire
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Camperdown, NSW, Australia
| | - Brian Nankivell
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Camperdown, NSW, Australia.,Westmead Clinical Medical School, University of Sydney, Camperdown, NSW, Australia.,Renal Division, Westmead Hospital, Camperdown, NSW, Australia
| | - Natasha M Rogers
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Camperdown, NSW, Australia. .,Westmead Clinical Medical School, University of Sydney, Camperdown, NSW, Australia. .,Renal Division, Westmead Hospital, Camperdown, NSW, Australia. .,Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
18
|
Rossi E, Poirault-Chassac S, Bieche I, Chocron R, Schnitzler A, Lokajczyk A, Bourdoncle P, Dizier B, Bacha NC, Gendron N, Blandinieres A, Guerin CL, Gaussem P, Smadja DM. Human Endothelial Colony Forming Cells Express Intracellular CD133 that Modulates their Vasculogenic Properties. Stem Cell Rev Rep 2020; 15:590-600. [PMID: 30879244 DOI: 10.1007/s12015-019-09881-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Stem cells at the origin of endothelial progenitor cells and in particular endothelial colony forming cells (ECFCs) subtype have been largely supposed to be positive for the CD133 antigen, even though no clear correlation has been established between its expression and function in ECFCs. We postulated that CD133 in ECFCs might be expressed intracellularly, and could participate to vasculogenic properties. ECFCs extracted from cord blood were used either fresh (n = 4) or frozen (n = 4), at culture days <30, to investigate the intracellular presence of CD133 by flow cytometry and confocal analysis. Comparison with HUVEC and HAEC mature endothelial cells was carried out. Then, CD133 was silenced in ECFCs using specific siRNA (siCD133-ECFCs) or scramble siRNA (siCtrl-ECFCs). siCD133-ECFCs (n = 12), siCtrl-ECFCs (n = 12) or PBS (n = 12) were injected in a hind-limb ischemia nude mouse model and vascularization was quantified at day 14 with H&E staining and immunohistochemistry for CD31. Results of flow cytometry and confocal microscopy evidenced the positivity of CD133 in ECFCs after permeabilization compared with not permeabilized ECFCs (p < 0.001) and mature endothelial cells (p < 0.03). In the model of mouse hind-limb ischemia, silencing of CD133 in ECFCs significantly abolished post-ischemic revascularization induced by siCtrl-ECFCs; indeed, a significant reduction in cutaneous blood flows (p = 0.03), capillary density (CD31) (p = 0.01) and myofiber regeneration (p = 0.04) was observed. Also, a significant necrosis (p = 0.02) was observed in mice receiving siCD133-ECFCs compared to those treated with siCtrl-ECFCs. In conclusion, our work describes for the first time the intracellular expression of the stemness marker CD133 in ECFCs. This feature could resume the discrepancies found in the literature concerning CD133 positivity and ontogeny in endothelial progenitors.
Collapse
Affiliation(s)
- Elisa Rossi
- Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Inserm UMR-S1140, Paris, France
| | - Sonia Poirault-Chassac
- Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Inserm UMR-S1140, Paris, France
| | - Ivan Bieche
- Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Department of genetics, Pharmacogenomics Unit, Institut Curie, Paris, France
| | - Richard Chocron
- Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Inserm UMR-S970, Paris, France.,AP-HP, Emergency Medicine Department, Hôpital Européen Georges Pompidou, Paris, France
| | - Anne Schnitzler
- Department of genetics, Pharmacogenomics Unit, Institut Curie, Paris, France
| | - Anna Lokajczyk
- Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Inserm UMR-S1140, Paris, France
| | - Pierre Bourdoncle
- Plate-forme IMAG'IC Institut Cochin Inserm U1016-CNRS UMR8104, Université Paris Descartes, Paris, France
| | - Blandine Dizier
- Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Inserm UMR-S1140, Paris, France
| | - Nour C Bacha
- Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Inserm UMR-S1140, Paris, France
| | - Nicolas Gendron
- Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Inserm UMR-S1140, Paris, France.,AP-HP, Hematology Department, Hôpital Européen Georges Pompidou, Paris, France
| | - Adeline Blandinieres
- Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Inserm UMR-S1140, Paris, France.,AP-HP, Hematology Department, Hôpital Européen Georges Pompidou, Paris, France
| | - Coralie L Guerin
- Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Inserm UMR-S1140, Paris, France.,Cytometry Unit, Institut Curie, Paris, France
| | - Pascale Gaussem
- Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Inserm UMR-S1140, Paris, France.,AP-HP, Hematology Department, Hôpital Européen Georges Pompidou, Paris, France
| | - David M Smadja
- Sorbonne Paris Cité, Université Paris Descartes, Paris, France. .,Inserm UMR-S1140, Paris, France. .,AP-HP, Hematology Department, Hôpital Européen Georges Pompidou, Paris, France. .,Laboratory of Biosurgical Research, Carpentier Foundation, Hôpital Européen Georges Pompidou, Paris, France.
| |
Collapse
|
19
|
Patsouras M, Tsiki E, Karagianni P, Vlachoyiannopoulos PG. The role of thrombospondin-1 in the pathogenesis of antiphospholipid syndrome. J Autoimmun 2020; 115:102527. [PMID: 32709480 DOI: 10.1016/j.jaut.2020.102527] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/12/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Antiphospholipid syndrome (APS) is an acquired thrombophilia characterized by recurrent thrombosis and/or pregnancy morbidity, in the presence of antibodies to β2 glycoprotein-I (β2GPI), prothrombin or Lupus anticoagulant (LA). Anti-β2GPI antibodies recognize complexes of β2GPI dimers with CXCL4 chemokine and activate platelets. Thrombospondin 1 (TSP-1) is secreted by platelets and exhibits prothrombotic and proinflammatory properties. Therefore, we investigated its implication in APS. METHODS Plasma from APS patients (n = 100), Systemic Lupus Erythematosus (SLE) (n = 27) and healthy donors (HD) (n = 50) was analyzed for TSP-1, IL-1β, IL-17A and free active TGF-β1 by ELISA. Human Umbilical Vein Endothelial Cells (HUVECs) and HD monocytes were treated with total HD-IgG or anti-β2GPI, β2GPI and CXCL4 and CD4+ T-cells were stimulated by monocyte supernatants. TSP-1, IL-1β, IL-17A TGF-β1 levels were quantified by ELISA and Real-Time PCR. RESULTS Higher plasma levels of TSP-1 and TGF-β1, which positively correlated each other, were observed in APS but not HDs or SLE patients. Patients with arterial thrombotic events or those undergoing a clinical event had the highest TSP-1 levels. These patients also had detectable IL-1β, IL-17A in their plasma. HD-derived monocytes and HUVECs stimulated with anti-β2GPI-IgG-β2GPI-CXCL4 secreted the highest TSP-1 and IL-1β levels. Supernatants from anti-β2GPI-β2GPI-CXCL4 treated monocytes induced IL-17A expression from CD4+ T-cells. Transcript levels followed a similar pattern. CONCLUSIONS TSP-1 is probably implicated in the pathogenesis of APS. In vitro cell treatments along with high TSP-1 levels in plasma of APS patients suggest that high TSP-1 levels could mark a prothrombotic state and an underlying inflammatory process.
Collapse
Affiliation(s)
- M Patsouras
- Department of Pathophysiology, Medical School, National and Kapodistrian University of Athens, Greece
| | - E Tsiki
- Department of Pathophysiology, Medical School, National and Kapodistrian University of Athens, Greece
| | - P Karagianni
- Department of Pathophysiology, Medical School, National and Kapodistrian University of Athens, Greece
| | - P G Vlachoyiannopoulos
- Department of Pathophysiology, Medical School, National and Kapodistrian University of Athens, Greece.
| |
Collapse
|
20
|
Recent Advancements in CD47 Signal Transduction Pathways Involved in Vascular Diseases. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4749135. [PMID: 32733941 PMCID: PMC7378613 DOI: 10.1155/2020/4749135] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 04/09/2020] [Indexed: 12/18/2022]
Abstract
Cardiovascular and cerebrovascular diseases caused by atherosclerosis have a high disability rate and reduce the quality of life of the population. Therefore, understanding the mechanism of atherosclerosis and its control may interfere with the progression of atherosclerosis and thus control the occurrence of diseases closely related to atherosclerosis. TSP-1 is a factor that has been found to have an antiangiogenic effect, and CD47, as the receptor of TSP-1, can participate in the regulation of antiangiogenesis of atherosclerosis. VEGF is an important regulator of angiogenesis, and TSP-1/CD47 can cause VEGF and its downstream expression. Therefore, the TSP-1/CD47/VEGF/VEGFR2 signal may have an important influence on atherosclerosis. In addition, some inflammatory factors, such as IL-1 and NLRP3, can also affect atherosclerosis. This review will be expounded focusing on the pathogenesis and influencing factors of atherosclerosis.
Collapse
|
21
|
He Y, Sun X, Rong W, Yang R, Liang H, Qi Y, Li L, Zen K. CD47 is a negative regulator of intestinal epithelial cell self-renewal following DSS-induced experimental colitis. Sci Rep 2020; 10:10180. [PMID: 32576895 PMCID: PMC7311394 DOI: 10.1038/s41598-020-67152-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 06/04/2020] [Indexed: 02/06/2023] Open
Abstract
CD47 deficient mice are resistant to dextran sulfate sodium (DSS)-induced experimental colitis. The underlying mechanism, however, remains incompletely understood. In this study, we characterized the role of CD47 in modulating homeostasis of gastrointestinal tract. We found that CD47 expression in both human and mouse intestinal epithelium was upregulated in colitic condition compared to that under normal condition. In line with this, CD47 deficiency protected mice from DSS-induced colitis. Analysis based on both intestinal organoid and cultured cell assays showed that CD47 deficiency accelerated intestinal epithelial cell proliferation and migration. Mechanistically, western blot and functional assays indicated that CD47 deficiency promoting mouse intestinal epithelial cell proliferation and migration follow cell injury is likely through upregulating expression of four Yamanaka transcriptional factors Oct4, Sox2, Klf4 and c-Myc (OSKM in abbreviation). Our studies thus reveal CD47 as a negative regulator in intestinal epithelial cell renewal during colitis through downregulating OSKM transcriptional factors.
Collapse
Affiliation(s)
- Yueqin He
- Nanjing University Advanced Institute of Life Sciences, Nanjing, China
| | - Xinlei Sun
- Nanjing University Advanced Institute of Life Sciences, Nanjing, China
| | - Weiwei Rong
- Nanjing University Advanced Institute of Life Sciences, Nanjing, China
| | - Rong Yang
- Nanjing University Advanced Institute of Life Sciences, Nanjing, China
| | - Hongwei Liang
- Nanjing University Advanced Institute of Life Sciences, Nanjing, China
| | - Ying Qi
- Department of Gastroenterology and Hepatology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Limin Li
- Nanjing University Advanced Institute of Life Sciences, Nanjing, China.
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, Nanjing University, Nanjing, Jiangsu, 210093, China.
| | - Ke Zen
- Nanjing University Advanced Institute of Life Sciences, Nanjing, China.
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, Nanjing University, Nanjing, Jiangsu, 210093, China.
| |
Collapse
|
22
|
Isenberg JS, Roberts DD. Thrombospondin-1 in maladaptive aging responses: a concept whose time has come. Am J Physiol Cell Physiol 2020; 319:C45-C63. [PMID: 32374675 DOI: 10.1152/ajpcell.00089.2020] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Numerous age-dependent alterations at the molecular, cellular, tissue and organ systems levels underlie the pathophysiology of aging. Herein, the focus is upon the secreted protein thrombospondin-1 (TSP1) as a promoter of aging and age-related diseases. TSP1 has several physiological functions in youth, including promoting neural synapse formation, mediating responses to ischemic and genotoxic stress, minimizing hemorrhage, limiting angiogenesis, and supporting wound healing. These acute functions of TSP1 generally require only transient expression of the protein. However, accumulating basic and clinical data reinforce the view that chronic diseases of aging are associated with accumulation of TSP1 in the extracellular matrix, which is a significant maladaptive contributor to the aging process. Identification of the relevant cell types that chronically produce and respond to TSP1 and the molecular mechanisms that mediate the resulting maladaptive responses could direct the development of therapeutic agents to delay or revert age-associated maladies.
Collapse
Affiliation(s)
| | - David D Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
23
|
Wieczór R, Rość D, Wieczór AM, Kulwas A. VASCULAR-1 and VASCULAR-2 as a New Potential Angiogenesis and Endothelial Dysfunction Markers in Peripheral Arterial Disease. Clin Appl Thromb Hemost 2020; 25:1076029619877440. [PMID: 31564130 PMCID: PMC6829630 DOI: 10.1177/1076029619877440] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The quotient of concentrations concerning the key proangiogenic factor, that is, the vascular endothelial growth factor (VEGF-A) and the angiogenesis inhibitor, namely, its soluble receptors (sVEGFR-1 or sVEGFR-2), seems to reflect increased hypoxia and intensity of compensation angiogenesis. Therefore, it can be an ischemic and endothelial dysfunction marker reflected in intermittent claudication (IC) or critical limb ischemia (CLI) in patients with symptomatic peripheral arterial disease (PAD). The main objective of this study was to evaluate the levels of VEGF-A/sVEGFR-1 and VEGF-A/sVEGFR-2—presented using a novelty acronym VASCULAR-1 and VASCULAR-2—in patients with IC and CLI, as well as displayed in 4 classes of severity of PAD. VASCULAR-1 and VASCULAR-2 were calculated using the plasma of venous blood sampled from 80 patients with IC (n = 65) and CLI (n = 15) and the control group (n = 30). Patients with CLI were reported to have a slightly higher index of VASCULAR-1 and double VASCULAR-2 levels as compared to patients with IC (P = nonsignificant), and these markers were significantly higher than controls (P < .01 and P < .01, respectively). VASCULAR-2 levels were observed to have an increasing tendency in the subsequent degrees of PAD severity according to the Fontaine classification (P = .02). In view of the need to consider the role of the proangiogenic and antiangiogenic factor in the assessment of the so-called “angiogenic potential,” VASCULAR-1 ratio and VASCULAR-2 ratio may be a new useful biomarker of limb ischemia in patients with IC and CLI. However, this requires further studies and evidence on a very large group of patients with PAD.
Collapse
Affiliation(s)
- Radosław Wieczór
- Department of Pathophysiology, Faculty of Pharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland.,Dr Jan Biziel University Hospital No. 2, Bydgoszcz, Poland
| | - Danuta Rość
- Department of Pathophysiology, Faculty of Pharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Anna Maria Wieczór
- Department of Pathophysiology, Faculty of Pharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Arleta Kulwas
- Department of Pathophysiology, Faculty of Pharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| |
Collapse
|
24
|
Do HS, Park SW, Im I, Seo D, Yoo HW, Go H, Kim YH, Koh GY, Lee BH, Han YM. Enhanced thrombospondin-1 causes dysfunction of vascular endothelial cells derived from Fabry disease-induced pluripotent stem cells. EBioMedicine 2020; 52:102633. [PMID: 31981984 PMCID: PMC6992938 DOI: 10.1016/j.ebiom.2020.102633] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 01/04/2020] [Accepted: 01/07/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Fabry disease (FD) is a recessive X-linked lysosomal storage disorder caused by α-galactosidase A (GLA) deficiency. Although the mechanism is unclear, GLA deficiency causes an accumulation of globotriaosylceramide (Gb3), leading to vasculopathy. METHODS To explore the relationship between the accumulation of Gb3 and vasculopathy, induced pluripotent stem cells generated from four Fabry patients (FD-iPSCs) were differentiated into vascular endothelial cells (VECs). Genome editing using CRISPR-Cas9 system was carried out to correct the GLA mutation or to delete Thrombospondin-1 (TSP-1). Global transcriptomes were compared between wild-type (WT)- and FD-VECs by RNA-sequencing analysis. FINDINGS Here, we report that overexpression of TSP-1 contributes to the dysfunction of VECs in FD. VECs originating from FD-iPSCs (FD-VECs) showed aberrant angiogenic functionality even upon treatment with recombinant α-galactosidase. Intriguingly, FD-VECs produced more p-SMAD2 and TSP-1 than WT-VECs. We also found elevated TSP-1 in the peritubular capillaries of renal tissues biopsied from FD patients. Inhibition of SMAD2 signaling or knock out of TSP-1 (TSP-1-/-) rescues normal vascular functionality in FD-VECs, like in gene-corrected FD-VECs. In addition, the enhanced oxygen consumption rate is reduced in TSP-1-/- FD-VECs. INTERPRETATION The overexpression of TSP-1 secondary to Gb3 accumulation is primarily responsible for the observed FD-VEC dysfunction. Our findings implicate dysfunctional VEC angiogenesis in the peritubular capillaries in some of the complications of Fabry disease. FUNDING This study was supported by grant 2018M3A9H1078330 from the National Research Foundation of the Republic of Korea.
Collapse
Affiliation(s)
- Hyo-Sang Do
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Sang-Wook Park
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea; New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Ilkyun Im
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34141, Republic of Korea
| | - Donghyuk Seo
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Han-Wook Yoo
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Heounjeong Go
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Yoo Hyung Kim
- College of Natural Sciences, KAIST, Daejeon 34141, Republic of Korea; Center for Vascular Research, Institute for Basic Sciences, Daejeon 34141, Republic of Korea
| | - Gou Young Koh
- Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea; Center for Vascular Research, Institute for Basic Sciences, Daejeon 34141, Republic of Korea
| | - Beom-Hee Lee
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea.
| | - Yong-Mahn Han
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea.
| |
Collapse
|
25
|
Lee NG, Jeung IC, Heo SC, Song J, Kim W, Hwang B, Kwon MG, Kim YG, Lee J, Park JG, Shin MG, Cho YL, Son MY, Bae KH, Lee SH, Kim JH, Min JK. Ischemia-induced Netrin-4 promotes neovascularization through endothelial progenitor cell activation via Unc-5 Netrin receptor B. FASEB J 2019; 34:1231-1246. [PMID: 31914695 DOI: 10.1096/fj.201900866rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 10/31/2019] [Accepted: 11/11/2019] [Indexed: 11/11/2022]
Abstract
Endothelial progenitor cells (EPCs) promote neovascularization and tissue repair by migrating to vascular injury sites; therefore, factors that enhance EPC homing to damaged tissues are of interest. Here, we provide evidence of the prominent role of the Netrin-4 (NTN4)-Unc-5 Netrin receptor B (UNC5B) axis in EPC-specific promotion of ischemic neovascularization. Our results showed that NTN4 promoted the proliferation, chemotactic migration, and paracrine effects of small EPCs (SEPCs) and significantly increased the incorporation of large EPCs (LEPCs) into tubule networks. Additionally, NTN4 prominently augmented neovascularization in mice with hindlimb ischemia by increasing the homing of exogenously transplanted EPCs to the ischemic limb and incorporating EPCs into vessels. Moreover, silencing of UNC5B, an NTN4 receptor, abrogated the NTN4-induced cellular activities of SEPCs in vitro and blood-flow recovery and neovascularization in vivo in ischemic muscle by reducing EPC homing and incorporation. These findings suggest NTN4 as an EPC-based therapy for treating angiogenesis-dependent diseases.
Collapse
Affiliation(s)
- Na Geum Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, South Korea
| | - In Cheul Jeung
- Department of Obstetrics and Gynecology, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Soon Chul Heo
- Department of Physiology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Jinhoi Song
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Wooil Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, South Korea
| | - Byungtae Hwang
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Min-Gi Kwon
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, South Korea
| | - Yeon-Gu Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Jangwook Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Jong-Gil Park
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Min-Gyeong Shin
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Young-Lai Cho
- Research Center for Metabolic Regulation, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Mi-Young Son
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Kwang-Hee Bae
- Research Center for Metabolic Regulation, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Sang-Hyun Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Jae Ho Kim
- Department of Physiology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Jeong-Ki Min
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, South Korea
| |
Collapse
|
26
|
Zierfuss B, Höbaus C, Herz CT, Pesau G, Koppensteiner R, Schernthaner GH. Thrombospondin-4 increases with the severity of peripheral arterial disease and is associated with diabetes. Heart Vessels 2019; 35:52-58. [PMID: 31227875 PMCID: PMC6942020 DOI: 10.1007/s00380-019-01453-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 06/14/2019] [Indexed: 02/07/2023]
Abstract
Thrombospondin-4 (TSP-4) is an extracellular matrix protein of the vessel wall. Despite bench evidence, its significance in the clinical setting of atherosclerosis is missing. TSP-4 (ng/ml) was measured in 365 PAD patientsusing a commercially available ELISA. PAD was diagnosed by the ankle–brachial index (ABI) and clinically graded using the Fontaine classification. TSP-4 levels were significantly higher in Fontaine II vs. Fontaine I (4.78 ± 0. 42, 4.69 ± 0.42, p = 0.043). TSP-4 significantly correlated with ABI (r = − 0.141, p = 0.023, n = 259) after the exclusion of mediasclerotic patients. Binary logistic regression analysis for Fontaine I vs. II showed an OR of 1.70 (1.02–2.82) in a multivariable model adjusted for traditional risk factors. Interestingly, TSP-4 levels were higher in patients with type 2 diabetes mellitus or prediabetes (DGT) compared with normal glucose tolerance (NGT) (4.76 ± 0.42 vs. 4.66 ± 0.41, p = 0.035). ANOVA for PAD and diabetes subgroups showed a linear increase with disease burden with the highest difference between Fontaine I-NGT and Fontaine II-DGT (4.59 ± 0.40, 4.79 ± 0.43, p = 0.015). TSP-4 levels increased with PAD severity and showed a former unknown association with diabetes. Thus, TSP-4 could be a novel marker of atherosclerotic activity, especially in the major subgroup of patients with concomitant diabetes.
Collapse
Affiliation(s)
- Bernhard Zierfuss
- Division of Angiology, Department of Internal Medicine 2, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria.
| | - Clemens Höbaus
- Division of Angiology, Department of Internal Medicine 2, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Carsten T Herz
- Division of Endocrinology and Metabolism, Department of Internal Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Gerfried Pesau
- Division of Angiology, Department of Internal Medicine 2, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Renate Koppensteiner
- Division of Angiology, Department of Internal Medicine 2, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Gerit-Holger Schernthaner
- Division of Angiology, Department of Internal Medicine 2, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| |
Collapse
|
27
|
Novelli EM, Little-Ihrig L, Knupp HE, Rogers NM, Yao M, Baust JJ, Meijles D, St Croix CM, Ross MA, Pagano PJ, DeVallance ER, Miles G, Potoka KP, Isenberg JS, Gladwin MT. Vascular TSP1-CD47 signaling promotes sickle cell-associated arterial vasculopathy and pulmonary hypertension in mice. Am J Physiol Lung Cell Mol Physiol 2019; 316:L1150-L1164. [PMID: 30892078 PMCID: PMC6620668 DOI: 10.1152/ajplung.00302.2018] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 03/07/2019] [Accepted: 03/14/2019] [Indexed: 02/08/2023] Open
Abstract
Pulmonary hypertension (PH) is a leading cause of death in sickle cell disease (SCD) patients. Hemolysis and oxidative stress contribute to SCD-associated PH. We have reported that the protein thrombospondin-1 (TSP1) is elevated in the plasma of patients with SCD and, by interacting with its receptor CD47, limits vasodilation of distal pulmonary arteries ex vivo. We hypothesized that the TSP1-CD47 interaction may promote PH in SCD. We found that TSP1 and CD47 are upregulated in the lungs of Berkeley (BERK) sickling (Sickle) mice and patients with SCD-associated PH. We then generated chimeric animals by transplanting BERK bone marrow into C57BL/6J (n = 24) and CD47 knockout (CD47KO, n = 27) mice. Right ventricular (RV) pressure was lower in fully engrafted Sickle-to-CD47KO than Sickle-to-C57BL/6J chimeras, as shown by the reduced maximum RV pressure (P = 0.013) and mean pulmonary artery pressure (P = 0.020). The afterload of the sickle-to-CD47KO chimeras was also lower, as shown by the diminished pulmonary vascular resistance (P = 0.024) and RV effective arterial elastance (P = 0.052). On myography, aortic segments from Sickle-to-CD47KO chimeras showed improved relaxation to acetylcholine. We hypothesized that, in SCD, TSP1-CD47 signaling promotes PH, in part, by increasing reactive oxygen species (ROS) generation. In human pulmonary artery endothelial cells, treatment with TSP1 stimulated ROS generation, which was abrogated by CD47 blockade. Explanted lungs of CD47KO chimeras had less vascular congestion and a smaller oxidative footprint. Our results show that genetic absence of CD47 ameliorates SCD-associated PH, which may be due to decreased ROS levels. Modulation of TSP1-CD47 may provide a new molecular approach to the treatment of SCD-associated PH.
Collapse
Affiliation(s)
- Enrico M Novelli
- Heart, Lung, Blood, and Vascular Medicine Institute and Division of Hematology/Oncology, UPMC Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Lynda Little-Ihrig
- Heart, Lung, Blood, and Vascular Medicine Institute and Division of Hematology/Oncology, UPMC Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Heather E Knupp
- UPMC Children's Hospital of Pittsburgh , Pittsburgh, Pennsylvania
| | - Natasha M Rogers
- Department of Medicine, Westmead Clinical School, University of Sydney , Sydney, New South Wales , Australia
| | - Mingyi Yao
- Department of Pharmaceutical Science, Midwestern University , Glendale, Arizona
| | - Jeffrey J Baust
- Heart, Lung, Blood, and Vascular Medicine Institute and Division of Hematology/Oncology, UPMC Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Daniel Meijles
- School of Biological Sciences, University of Reading , Reading , United Kingdom
| | - Claudette M St Croix
- Center for Biologic Imaging, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Mark A Ross
- Center for Biologic Imaging, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Patrick J Pagano
- Heart, Lung, Blood, and Vascular Medicine Institute and Division of Hematology/Oncology, UPMC Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Evan R DeVallance
- Heart, Lung, Blood, and Vascular Medicine Institute and Division of Hematology/Oncology, UPMC Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - George Miles
- Department of Molecular and Human Genetics, Baylor College of Medicine , Houston, Texas
| | - Karin P Potoka
- Heart, Lung, Blood, and Vascular Medicine Institute and Division of Hematology/Oncology, UPMC Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
- UPMC Children's Hospital of Pittsburgh , Pittsburgh, Pennsylvania
| | - Jeffrey S Isenberg
- Heart, Lung, Blood, and Vascular Medicine Institute and Division of Hematology/Oncology, UPMC Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Mark T Gladwin
- Heart, Lung, Blood, and Vascular Medicine Institute and Division of Hematology/Oncology, UPMC Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| |
Collapse
|
28
|
Matsumoto T, Tanaka M, Ikeji T, Maeshige N, Sakai Y, Akisue T, Kondo H, Ishihara A, Fujino H. Application of transcutaneous carbon dioxide improves capillary regression of skeletal muscle in hyperglycemia. J Physiol Sci 2019; 69:317-326. [PMID: 30478742 PMCID: PMC10717691 DOI: 10.1007/s12576-018-0648-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 11/14/2018] [Indexed: 12/17/2022]
Abstract
The purpose of the present study was to determine the effects of transcutaneous CO2 application on the blood flow and capillary architecture of the soleus muscle in rats with streptozotocin (STZ)-induced hyperglycemia. Wistar rats were randomly divided into four groups: control, control + CO2-treated, STZ-induced hyperglycemia, and STZ-induced hyperglycemia + CO2-treated groups. Blood flow in soleus muscle increased during the transcutaneous CO2 exposure, and continued to increase for 30 min after the treatment. In addition, the transcutaneous CO2 attenuated a decrease in capillary and the expression level of eNOS and VEGF protein, and an increase in the expression level of MDM-2 and TSP-1 protein of soleus muscle due to STZ-induced hyperglycemia. These results indicate that the application of transcutaneous CO2 could improve capillary regression via the change of pro- and anti-angiogenesis factors, which might be induced by an increase in blood flow.
Collapse
Affiliation(s)
- Tomohiro Matsumoto
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, Hyogo, 654-0142, Japan
| | - Masayuki Tanaka
- Department of Physical Therapy, Faculty of Human Sciences, Osaka University of Human Sciences, 1-4-1 Shojaku, Settsu, Osaka, 566-8501, Japan
| | - Takuya Ikeji
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, Hyogo, 654-0142, Japan
| | - Noriaki Maeshige
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, Hyogo, 654-0142, Japan
| | - Yoshitada Sakai
- Division of Rehabilitation Medicine, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Toshihiro Akisue
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, Hyogo, 654-0142, Japan
| | - Hiroyo Kondo
- Department of Food Science and Nutrition, Nagoya Women's University, 4-21 Shioji-cho, Mizuho-ku, Nagoya, Aichi, 467-8611, Japan
| | - Akihiko Ishihara
- Laboratory of Cell Biology and Life Science, Graduate School of Human and Environmental Studies, Kyoto University, Yoshida-nihonmatsu-cho, Sakyo-ku, Kyoto-shi, Kyoto, 606-8501, Japan
| | - Hidemi Fujino
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, Hyogo, 654-0142, Japan.
| |
Collapse
|
29
|
Bitar MS. Diabetes Impairs Angiogenesis and Induces Endothelial Cell Senescence by Up-Regulating Thrombospondin-CD47-Dependent Signaling. Int J Mol Sci 2019; 20:ijms20030673. [PMID: 30720765 PMCID: PMC6386981 DOI: 10.3390/ijms20030673] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/03/2019] [Accepted: 01/06/2019] [Indexed: 02/07/2023] Open
Abstract
Endothelial dysfunction, impaired angiogenesis and cellular senescence in type 2 diabetes constitute dominant risk factors for chronic non-healing wounds and other cardiovascular disorders. Studying these phenomena in the context of diabetes and the TSP1-CD-47 signaling dictated the use of the in vitro wound endothelial cultured system and an in vivo PVA sponge model of angiogenesis. Herein we report that diabetes impaired the in vivo sponge angiogenic capacity by decreasing cell proliferation, fibrovascular invasion and capillary density. In contrast, a heightened state of oxidative stress and elevated expression of TSP1 and CD47 both at the mRNA and protein levels were evident in this diabetic sponge model of wound healing. An in vitro culturing system involving wound endothelial cells confirmed the increase in ROS generation and the up-regulation of TSP1-CD47 signaling as a function of diabetes. We also provided evidence that diabetic wound endothelial cells (W-ECs) exhibited a characteristic feature that is consistent with cellular senescence. Indeed, enhanced SA-β-gal activity, cell cycle arrest, increased cell cycle inhibitors (CKIs) p53, p21 and p16 and decreased cell cycle promoters including Cyclin D1 and CDK4/6 were all demonstrated in these cells. The functional consequence of this cascade of events was illustrated by a marked reduction in diabetic endothelial cell proliferation, migration and tube formation. A genetic-based strategy in diabetic W-ECs using CD47 siRNA significantly ameliorated in these cells the excessiveness in oxidative stress, attenuation in angiogenic potential and more importantly the inhibition in cell cycle progression and its companion cellular senescence. To this end, the current data provide evidence linking the overexpression of TSP1-CD47 signaling in diabetes to a number of parameters associated with endothelial dysfunction including impaired angiogenesis, cellular senescence and a heightened state of oxidative stress. Moreover, it may also point to TSP1-CD47 as a potential therapeutic target in the treatment of the aforementioned pathologies.
Collapse
Affiliation(s)
- Milad S Bitar
- Department of Pharmacology& Toxicology, Faculty of Medicine, Kuwait University, P.O. Box 24923, Safat 13110, Kuwait.
| |
Collapse
|
30
|
Vasculogenic Stem and Progenitor Cells in Human: Future Cell Therapy Product or Liquid Biopsy for Vascular Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1201:215-237. [PMID: 31898789 DOI: 10.1007/978-3-030-31206-0_11] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
New blood vessel formation in adults was considered to result exclusively from sprouting of preexisting endothelial cells, a process referred to angiogenesis. Vasculogenesis, the formation of new blood vessels from endothelial progenitor cells, was thought to occur only during embryonic life. Discovery of adult endothelial progenitor cells (EPCs) in 1997 opened the door for cell therapy in vascular disease. Endothelial progenitor cells contribute to vascular repair and are now well established as postnatal vasculogenic cells in humans. It is now admitted that endothelial colony-forming cells (ECFCs) are the vasculogenic subtype. ECFCs could be used as a cell therapy product and also as a liquid biopsy in several vascular diseases or as vector for gene therapy. However, despite a huge interest in these cells, their tissue and molecular origin is still unclear. We recently proposed that endothelial progenitor could come from very small embryonic-like stem cells (VSELs) isolated in human from CD133 positive cells. VSELs are small dormant stem cells related to migratory primordial germ cells. They have been described in bone marrow and other organs. This chapter discusses the reported findings from in vitro data and also preclinical studies that aimed to explore stem cells at the origin of vasculogenesis in human and then explore the potential use of ECFCs to promote newly formed vessels or serve as liquid biopsy to understand vascular pathophysiology and in particular pulmonary disease and haemostasis disorders.
Collapse
|
31
|
Mandler WK, Nurkiewicz TR, Porter DW, Kelley EE, Olfert IM. Microvascular Dysfunction Following Multiwalled Carbon Nanotube Exposure Is Mediated by Thrombospondin-1 Receptor CD47. Toxicol Sci 2018; 165:90-99. [PMID: 29788500 PMCID: PMC6111784 DOI: 10.1093/toxsci/kfy120] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pulmonary exposure to multiwalled carbon nanotubes (MWCNTs) disrupts peripheral microvascular function. Thrombospondin-1 (TSP-1) is highly expressed during lung injury and has been shown to alter microvascular reactivity. It is unclear exactly how TSP-1 exerts effects on vascular function, but we hypothesized that the TSP-1 receptor CD47 may mediate changes in vasodilation. Wildtype (WT) or CD47 knockout (CD47 KO) C57B6/J-background animals were exposed to 50 µg of MWCNT or saline control via pharyngeal aspiration. Twenty-four hours postexposure, intravital microscopy was performed to assess arteriolar dilation and venular leukocyte adhesion and rolling. To assess tissue redox status, electron paramagnetic resonance and NOx measurements were performed, while inflammatory biomarkers were measured via multiplex assay.Vasodilation was impaired in the WT + MWCNT group compared with control (57 ± 9 vs 90 ± 2% relaxation), while CD47 KO animals showed no impairment (108 ± 8% relaxation). Venular leukocyte adhesion and rolling increased by >2-fold, while the CD47 KO group showed no change. Application of the antioxidant apocynin rescued normal leukocyte activity in the WT + MWCNT group. Lung and plasma NOx were reduced in the WT + MWCNT group by 47% and 32%, respectively, while the CD47 KO groups were unchanged from control. Some inflammatory cytokines were increased in the CD47 + MWCNT group only. In conclusion, TSP-1 is an important ligand mediating MWCNT-induced microvascular dysfunction, and CD47 is a component of this dysregulation. CD47 activation likely disrupts nitric oxide (•NO) signaling and promotes leukocyte-endothelial interactions. Impaired •NO production, signaling, and bioavailability is linked to a variety of cardiovascular diseases in which TSP-1/CD47 may play an important role.
Collapse
Affiliation(s)
- William Kyle Mandler
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV 26506
- Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Timothy R Nurkiewicz
- Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV 26506
- Department of Physiology, Pharmacology and Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
- West Virginia Clinical and Translational Science Institute, Robert C. Byrd Health Sciences Center, Morgantown, WV 26506
| | - Dale W Porter
- Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV 26506
- National Institute for Occupational Safety and Health, Morgantown, WV 26505
| | - Eric E Kelley
- Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV 26506
- Department of Physiology, Pharmacology and Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
- West Virginia Clinical and Translational Science Institute, Robert C. Byrd Health Sciences Center, Morgantown, WV 26506
| | - Ivan Mark Olfert
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV 26506
- Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV 26506
- Department of Physiology, Pharmacology and Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
- West Virginia Clinical and Translational Science Institute, Robert C. Byrd Health Sciences Center, Morgantown, WV 26506
| |
Collapse
|
32
|
Rogers NM, Ghimire K, Calzada MJ, Isenberg JS. Matricellular protein thrombospondin-1 in pulmonary hypertension: multiple pathways to disease. Cardiovasc Res 2018; 113:858-868. [PMID: 28472457 DOI: 10.1093/cvr/cvx094] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 05/03/2017] [Indexed: 12/24/2022] Open
Abstract
Matricellular proteins are secreted molecules that have affinities for both extracellular matrix and cell surface receptors. Through interaction with structural proteins and the cells that maintain the matrix these proteins can alter matrix strength. Matricellular proteins exert control on cell activity primarily through engagement of membrane receptors that mediate outside-in signaling. An example of this group is thrombospondin-1 (TSP1), first identified as a component of the secreted product of activated platelets. As a result, TSP1 was initially studied in relation to coagulation, growth factor signaling and angiogenesis. More recently, TSP1 has been found to alter the effects of the gaseous transmitter nitric oxide (NO). This latter capacity has provided motivation to study TSP1 in diseases associated with loss of NO signaling as observed in cardiovascular disease and pulmonary hypertension (PH). PH is characterized by progressive changes in the pulmonary vasculature leading to increased resistance to blood flow and subsequent right heart failure. Studies have linked TSP1 to pre-clinical animal models of PH and more recently to clinical PH. This review will provide analysis of the vascular and non-vascular effects of TSP1 that contribute to PH, the experimental and translational studies that support a role for TSP1 in disease promotion and frame the relevance of these findings to therapeutic strategies.
Collapse
Affiliation(s)
- Natasha M Rogers
- Medicine, Westmead Clinical School, University of Sydney, Sydney, New South Wales 2145, Australia
| | - Kedar Ghimire
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Maria J Calzada
- Department of Medicine, Universidad Autónoma of Madrid, Diego de León, Hospital Universitario of the Princesa, 62?28006 Madrid, Spain
| | - Jeffrey S Isenberg
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.,Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| |
Collapse
|
33
|
Bazzazi H, Zhang Y, Jafarnejad M, Isenberg JS, Annex BH, Popel AS. Computer Simulation of TSP1 Inhibition of VEGF-Akt-eNOS: An Angiogenesis Triple Threat. Front Physiol 2018; 9:644. [PMID: 29899706 PMCID: PMC5988849 DOI: 10.3389/fphys.2018.00644] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/11/2018] [Indexed: 01/08/2023] Open
Abstract
The matricellular protein thrombospondin-1 (TSP1) is a potent inhibitor of angiogenesis. Specifically, TSP1 has been experimentally shown to inhibit signaling downstream of vascular endothelial growth factor (VEGF). The molecular mechanism of this inhibition is not entirely clear. We developed a detailed computational model of VEGF signaling to Akt-endothelial nitric oxide synthase (eNOS) to investigate the quantitative molecular mechanism of TSP1 inhibition. The model demonstrated that TSP1 acceleration of VEGFR2 degradation is sufficient to explain the inhibition of VEGFR2 and eNOS phosphorylation. However, Akt inhibition requires TSP1-induced phosphatase recruitment to VEGFR2. The model was then utilized to test various strategies for the rescue of VEGF signaling to Akt and eNOS. Inhibiting TSP1 was predicted to be not as effective as CD47 depletion in rescuing signaling to Akt. The model further predicts that combination strategy involving depletion of CD47 and inhibition of TSP1 binding to CD47 is necessary for effective recovery of signaling to eNOS. In all, computational modeling offers insight to molecular mechanisms involving TSP1 interaction with VEGF signaling and provides strategies for rescuing angiogenesis by targeting TSP1-CD47 axis.
Collapse
Affiliation(s)
- Hojjat Bazzazi
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Yu Zhang
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Mohammad Jafarnejad
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Jeffrey S Isenberg
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States.,Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Brian H Annex
- Division of Cardiovascular Medicine, Department of Medicine, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Aleksander S Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
34
|
Zhao C, Isenberg JS, Popel AS. Human expression patterns: qualitative and quantitative analysis of thrombospondin-1 under physiological and pathological conditions. J Cell Mol Med 2018; 22:2086-2097. [PMID: 29441713 PMCID: PMC5867078 DOI: 10.1111/jcmm.13565] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 01/07/2018] [Indexed: 12/12/2022] Open
Abstract
Thrombospondin-1 (TSP-1), a matricellular protein and one of the first endogenous anti-angiogenic molecules identified, has long been considered a potent modulator of human diseases. While the therapeutic effect of TSP-1 to suppress cancer was investigated in both research and clinical settings, the mechanisms of how TSP-1 is regulated in cancer remain elusive, and the scientific answers to the question of whether TSP-1 expressions can be utilized as diagnostic or prognostic marker for patients with cancer are largely inconsistent. Moreover, TSP-1 plays crucial functions in angiogenesis, inflammation and tissue remodelling, which are essential biological processes in the progression of many cardiovascular diseases, and therefore, its dysregulated expressions in such conditions may have therapeutic significance. Herein, we critically analysed the literature pertaining to TSP-1 expression in circulating blood and pathological tissues in various types of cancer as well as cardiovascular and inflammation-related diseases in humans. We compare the secretion rates of TSP-1 by different cancer and non-cancer cells and discuss the potential connection between the expression changes of TSP-1 and vascular endothelial growth factor (VEGF) observed in patients with cancer. Moreover, the pattern and emerging significance of TSP-1 profiles in cardiovascular disease, such as peripheral arterial disease, diabetes and other related non-cancer disorders, are highlighted. The analysis of published TSP-1 data presented in this review may have implications for the future exploration of novel TSP-1-based treatment strategies for cancer and cardiovascular-related diseases.
Collapse
Affiliation(s)
- Chen Zhao
- Department of Biomedical EngineeringSchool of MedicineJohns Hopkins UniversityBaltimoreMDUSA
| | - Jeffrey S. Isenberg
- Division of Pulmonary, Allergy and Critical CareDepartment of MedicineHeart, Lung, Blood and Vascular Medicine InstituteUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Aleksander S. Popel
- Department of Biomedical EngineeringSchool of MedicineJohns Hopkins UniversityBaltimoreMDUSA
| |
Collapse
|
35
|
Roberts DD, Kaur S, Isenberg JS. Regulation of Cellular Redox Signaling by Matricellular Proteins in Vascular Biology, Immunology, and Cancer. Antioxid Redox Signal 2017; 27:874-911. [PMID: 28712304 PMCID: PMC5653149 DOI: 10.1089/ars.2017.7140] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 07/11/2017] [Accepted: 07/13/2017] [Indexed: 12/15/2022]
Abstract
SIGNIFICANCE In contrast to structural elements of the extracellular matrix, matricellular proteins appear transiently during development and injury responses, but their sustained expression can contribute to chronic disease. Through interactions with other matrix components and specific cell surface receptors, matricellular proteins regulate multiple signaling pathways, including those mediated by reactive oxygen and nitrogen species and H2S. Dysregulation of matricellular proteins contributes to the pathogenesis of vascular diseases and cancer. Defining the molecular mechanisms and receptors involved is revealing new therapeutic opportunities. Recent Advances: Thrombospondin-1 (TSP1) regulates NO, H2S, and superoxide production and signaling in several cell types. The TSP1 receptor CD47 plays a central role in inhibition of NO signaling, but other TSP1 receptors also modulate redox signaling. The matricellular protein CCN1 engages some of the same receptors to regulate redox signaling, and ADAMTS1 regulates NO signaling in Marfan syndrome. In addition to mediating matricellular protein signaling, redox signaling is emerging as an important pathway that controls the expression of several matricellular proteins. CRITICAL ISSUES Redox signaling remains unexplored for many matricellular proteins. Their interactions with multiple cellular receptors remains an obstacle to defining signaling mechanisms, but improved transgenic models could overcome this barrier. FUTURE DIRECTIONS Therapeutics targeting the TSP1 receptor CD47 may have beneficial effects for treating cardiovascular disease and cancer and have recently entered clinical trials. Biomarkers are needed to assess their effects on redox signaling in patients and to evaluate how these contribute to their therapeutic efficacy and potential side effects. Antioxid. Redox Signal. 27, 874-911.
Collapse
Affiliation(s)
- David D. Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Sukhbir Kaur
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jeffrey S. Isenberg
- Division of Pulmonary, Allergy and Critical Care, Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
36
|
LeBlanc AJ, Kelm NQ. Thrombospondin-1, Free Radicals, and the Coronary Microcirculation: The Aging Conundrum. Antioxid Redox Signal 2017; 27:785-801. [PMID: 28762749 PMCID: PMC5647494 DOI: 10.1089/ars.2017.7292] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
SIGNIFICANCE Successful matching of cardiac metabolism to perfusion is accomplished primarily through vasodilation of the coronary resistance arterioles, but the mechanism that achieves this effect changes significantly as aging progresses and involves the contribution of reactive oxygen species (ROS). Recent Advances: A matricellular protein, thrombospondin-1 (Thbs-1), has been shown to be a prolific contributor to the production and modulation of ROS in large conductance vessels and in the peripheral circulation. Recently, the presence of physiologically relevant circulating Thbs-1 levels was proven to also disrupt vasodilation to nitric oxide (NO) in coronary arterioles from aged animals, negatively impacting coronary blood flow reserve. CRITICAL ISSUES This review seeks to reconcile how ROS can be successfully utilized as a substrate to mediate vasoreactivity in the coronary microcirculation as "normal" aging progresses, but will also examine how Thbs-1-induced ROS production leads to dysfunctional perfusion and eventual ischemia and why this is more of a concern in advancing age. FUTURE DIRECTIONS Current therapies that may effectively disrupt Thbs-1 and its receptor CD47 in the vascular wall and areas for future exploration will be discussed. Antioxid. Redox Signal. 27, 785-801.
Collapse
Affiliation(s)
- Amanda J LeBlanc
- Department of Physiology, Cardiovascular Innovation Institute, University of Louisville , Louisville, Kentucky
| | - Natia Q Kelm
- Department of Physiology, Cardiovascular Innovation Institute, University of Louisville , Louisville, Kentucky
| |
Collapse
|
37
|
Labrousse-Arias D, Martínez-Ruiz A, Calzada MJ. Hypoxia and Redox Signaling on Extracellular Matrix Remodeling: From Mechanisms to Pathological Implications. Antioxid Redox Signal 2017; 27:802-822. [PMID: 28715969 DOI: 10.1089/ars.2017.7275] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
SIGNIFICANCE The extracellular matrix (ECM) is an essential modulator of cell behavior that influences tissue organization. It has a strong relevance in homeostasis and translational implications for human disease. In addition to ECM structural proteins, matricellular proteins are important regulators of the ECM that are involved in a myriad of different pathologies. Recent Advances: Biochemical studies, animal models, and study of human diseases have contributed to the knowledge of molecular mechanisms involved in remodeling of the ECM, both in homeostasis and disease. Some of them might help in the development of new therapeutic strategies. This review aims to review what is known about some of the most studied matricellular proteins and their regulation by hypoxia and redox signaling, as well as the pathological implications of such regulation. CRITICAL ISSUES Matricellular proteins have complex regulatory functions and are modulated by hypoxia and redox signaling through diverse mechanisms, in some cases with controversial effects that can be cell or tissue specific and context dependent. Therefore, a better understanding of these regulatory processes would be of great benefit and will open new avenues of considerable therapeutic potential. FUTURE DIRECTIONS Characterizing the specific molecular mechanisms that modulate matricellular proteins in pathological processes that involve hypoxia and redox signaling warrants additional consideration to harness the potential therapeutic value of these regulatory proteins. Antioxid. Redox Signal. 27, 802-822.
Collapse
Affiliation(s)
- David Labrousse-Arias
- 1 Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP) , Madrid, Spain
| | - Antonio Martínez-Ruiz
- 1 Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP) , Madrid, Spain .,2 Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV) , Madrid, Spain
| | - María J Calzada
- 1 Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP) , Madrid, Spain .,3 Departmento de Medicina, Universidad Autónoma de Madrid , Madrid, Spain
| |
Collapse
|
38
|
Vitamin D supplementation lowers thrombospondin-1 levels and blood pressure in healthy adults. PLoS One 2017; 12:e0174435. [PMID: 28489857 PMCID: PMC5425007 DOI: 10.1371/journal.pone.0174435] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 03/09/2017] [Indexed: 01/21/2023] Open
Abstract
INTRODUCTION Vitamin D insufficiency, defined as 25-hydroxyvitamin D (25(OH)D) levels < 75nmol/L is associated with cardio-metabolic dysfunction. Vitamin D insufficiency is associated with inflammation and fibrosis, but it remains uncertain whether these anomalies are readily reversible. Therefore, we aimed to determine the effects of vitamin D supplementation on markers of: 1) nitric oxide (NO) signaling, 2) inflammation, and 3) fibrosis, in healthy volunteers with mild hypovitaminosis. METHODS Healthy volunteers (n = 35) (mean age: 45 ± 11 years) with 25(OH)D levels <75nmol/L, received vitamin D supplementation (Ostelin ® capsules 2000IU) for 12 weeks. Resting systolic and diastolic blood pressures (BP) were assessed. Routine biochemistry was examined. Plasma concentrations of asymmetric dimethylarginine (ADMA), thrombospondin-1 (TSP-1), plasminogen activator inhibitor-1 (PAI-1), hs-CRP, activin-A, and follistatin-like 3 (FSTL3) were quantitated. RESULTS Vitamin D administration for 12 weeks significantly increased 25-(OH)D levels (48.8 ± 16 nmol/L to 100.8 ± 23.7 nmol/L, p<0.001). There was significant lowering of systolic and diastolic BP, while there was no significant change in lipid profiles, or fasting insulin. Plasma concentrations of ADMA, hs-CRP, PAI-1, activin A, and FSTL-3 did not change with vitamin D supplementation. However, there was a marked reduction of TSP-1 (522.7 ± 379.8 ng/mL vs 206.7 ± 204.5 ng/mL, p<0.001). CONCLUSIONS Vitamin D supplementation in vitamin D insufficient, but otherwise healthy individuals markedly decreased TSP-1 levels and blood pressure. Since TSP-1 suppresses signaling of NO, it is possible that the fall in BP is engendered by restoration of NO effect.
Collapse
|
39
|
Desai P, Helkin A, Odugbesi A, Stein J, Bruch D, Lawler J, Maier KG, Gahtan V. Fluvastatin inhibits intimal hyperplasia in wild-type but not Thbs1 -null mice. J Surg Res 2017; 210:1-7. [DOI: 10.1016/j.jss.2016.10.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 09/23/2016] [Accepted: 10/05/2016] [Indexed: 10/20/2022]
|
40
|
Mandler WK, Nurkiewicz TR, Porter DW, Olfert IM. Thrombospondin-1 mediates multi-walled carbon nanotube induced impairment of arteriolar dilation. Nanotoxicology 2017; 11:112-122. [PMID: 28024456 DOI: 10.1080/17435390.2016.1277275] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pulmonary exposure to multi-walled carbon nanotubes (MWCNT) has been shown to disrupt endothelium-dependent arteriolar dilation in the peripheral microcirculation. The molecular mechanisms behind these arteriolar disruptions have yet to be fully elucidated. The secreted matricellular matrix protein thrombospondin-1 (TSP-1) is capable of moderating arteriolar vasodilation by inhibiting soluble guanylate cyclase activity. We hypothesized that TSP-1 may be a link between nanomaterial exposure and observed peripheral microvascular dysfunction. To test this hypothesis, wild-type C57B6J (WT) and TSP-1 knockout (KO) mice were exposed via lung aspiration to 50 μg MWCNT or a Sham dispersion medium control. Following exposure (24 h), arteriolar characteristics and reactivity were measured in the gluteus maximus muscle using intravital microscopy (IVM) coupled with microiontophoretic delivery of acetylcholine (ACh) or sodium nitroprusside (SNP). In WT mice exposed to MWCNT, skeletal muscle TSP-1 protein increased > fivefold compared to Sham exposed, and exhibited a 39% and 47% decrease in endothelium-dependent and -independent vasodilation, respectively. In contrast, TSP-1 protein was not increased following MWCNT exposure in KO mice and exhibited no loss in dilatory capacity. Microvascular leukocyte-endothelium interactions were measured by assessing leukocyte adhesion and rolling activity in third order venules. The WT + MWCNT group demonstrated 223% higher leukocyte rolling compared to the WT + Sham controls. TSP-1 KO animals exposed to MWCNT showed no differences from the WT + Sham control. These data provide evidence that TSP-1 is likely a central mediator of the systemic microvascular dysfunction that follows pulmonary MWCNT exposure.
Collapse
Affiliation(s)
- W Kyle Mandler
- a Division of Exercise Physiology , West Virginia University School of Medicine , Morgantown , WV , USA
| | - Timothy R Nurkiewicz
- b Department of Physiology and Pharmacology , West Virginia University School of Medicine , Morgantown , WV , USA.,c Center for Cardiovascular & Respiratory Sciences , West Virginia University, Robert C. Byrd Health Sciences Center , Morgantown , WV , USA
| | - Dale W Porter
- d National Institute for Occupational Safety and Health , Morgantown , WV , USA
| | - I Mark Olfert
- a Division of Exercise Physiology , West Virginia University School of Medicine , Morgantown , WV , USA.,c Center for Cardiovascular & Respiratory Sciences , West Virginia University, Robert C. Byrd Health Sciences Center , Morgantown , WV , USA
| |
Collapse
|
41
|
Zhao C, Isenberg JS, Popel AS. Transcriptional and Post-Transcriptional Regulation of Thrombospondin-1 Expression: A Computational Model. PLoS Comput Biol 2017; 13:e1005272. [PMID: 28045898 PMCID: PMC5207393 DOI: 10.1371/journal.pcbi.1005272] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 11/29/2016] [Indexed: 01/09/2023] Open
Abstract
Hypoxia is an important physiological stress signal that drives angiogenesis, the formation of new blood vessels. Besides an increase in the production of pro-angiogenic signals such as vascular endothelial growth factor (VEGF), hypoxia also stimulates the production of anti-angiogenic signals. Thrombospondin-1 (TSP-1) is one of the anti-angiogenic factors whose synthesis is driven by hypoxia. Cellular synthesis of TSP-1 is tightly regulated by different intermediate biomolecules including proteins that interact with hypoxia-inducible factors (HIFs), transcription factors that are activated by receptor and intracellular signaling, and microRNAs which are small non-coding RNA molecules that function in post-transcriptional modification of gene expression. Here we present a computational model that describes the mechanistic interactions between intracellular biomolecules and cooperation between signaling pathways that together make up the complex network of TSP-1 regulation both at the transcriptional and post-transcriptional level. Assisted by the model, we conduct in silico experiments to compare the efficacy of different therapeutic strategies designed to modulate TSP-1 synthesis in conditions that simulate tumor and peripheral arterial disease microenvironment. We conclude that TSP-1 production in endothelial cells depends on not only the availability of certain growth factors but also the fine-tuned signaling cascades that are initiated by hypoxia.
Collapse
Affiliation(s)
- Chen Zhao
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| | - Jeffrey S. Isenberg
- Vascular Medicine Institute, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Aleksander S. Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
42
|
Rogers NM, Sharifi-Sanjani M, Yao M, Ghimire K, Bienes-Martinez R, Mutchler SM, Knupp HE, Baust J, Novelli EM, Ross M, St Croix C, Kutten JC, Czajka CA, Sembrat JC, Rojas M, Labrousse-Arias D, Bachman TN, Vanderpool RR, Zuckerbraun BS, Champion HC, Mora AL, Straub AC, Bilonick RA, Calzada MJ, Isenberg JS. TSP1-CD47 signaling is upregulated in clinical pulmonary hypertension and contributes to pulmonary arterial vasculopathy and dysfunction. Cardiovasc Res 2017; 113:15-29. [PMID: 27742621 PMCID: PMC5220673 DOI: 10.1093/cvr/cvw218] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 08/30/2016] [Accepted: 09/22/2016] [Indexed: 12/23/2022] Open
Abstract
AIMS Thrombospondin-1 (TSP1) is a ligand for CD47 and TSP1-/- mice are protected from pulmonary hypertension (PH). We hypothesized the TSP1-CD47 axis is upregulated in human PH and promotes pulmonary arterial vasculopathy. METHODS AND RESULTS We analyzed the molecular signature and functional response of lung tissue and distal pulmonary arteries (PAs) from individuals with (n = 23) and without (n = 16) PH. Compared with controls, lungs and distal PAs from PH patients showed induction of TSP1-CD47 and endothelin-1/endothelin A receptor (ET-1/ETA) protein and mRNA. In control PAs, treatment with exogenous TSP1 inhibited vasodilation and potentiated vasoconstriction to ET-1. Treatment of diseased PAs from PH patients with a CD47 blocking antibody improved sensitivity to vasodilators. Hypoxic wild type (WT) mice developed PH and displayed upregulation of pulmonary TSP1, CD47, and ET-1/ETA concurrent with down regulation of the transcription factor cell homolog of the v-myc oncogene (cMyc). In contrast, PH was attenuated in hypoxic CD47-/- mice while pulmonary TSP1 and ET-1/ETA were unchanged and cMyc was overexpressed. In CD47-/- pulmonary endothelial cells cMyc was increased and ET-1 decreased. In CD47+/+ cells, forced induction of cMyc suppressed ET-1 transcript, whereas suppression of cMyc increased ET-1 signaling. Furthermore, disrupting TSP1-CD47 signaling in pulmonary smooth muscle cells abrogated ET-1-stimulated hypertrophy. Finally, a CD47 antibody given 2 weeks after monocrotaline challenge in rats upregulated pulmonary cMyc and improved aberrations in PH-associated cardiopulmonary parameters. CONCLUSIONS In pre-clinical models of PH CD47 targets cMyc to increase ET-1 signaling. In clinical PH TSP1-CD47 is upregulated, and in both, contributes to pulmonary arterial vasculopathy and dysfunction.
Collapse
Affiliation(s)
- Natasha M Rogers
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Division of Renal and Electrolytes, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Starzl Transplant Institute, University of Pittsburgh, PA, USA
| | - Maryam Sharifi-Sanjani
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Mingyi Yao
- Department of Pharmaceutical Science, College of Pharmacy-Glendale, Midwestern University, Glendale, AZ 85308, USA
| | - Kedar Ghimire
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Raquel Bienes-Martinez
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Stephanie M Mutchler
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Heather E Knupp
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Jeffrey Baust
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Enrico M Novelli
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Mark Ross
- Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Claudette St Croix
- Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Johannes C Kutten
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Caitlin A Czajka
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - John C Sembrat
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Dorothy P. & Richard P. Simmons Center for Interstitial Lung Disease, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mauricio Rojas
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Dorothy P. & Richard P. Simmons Center for Interstitial Lung Disease, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - David Labrousse-Arias
- Hospital of the Princesa, Department of Medicine, Universidad Autónoma, Diego de León, 62 28006 Madrid, Spain
| | - Timothy N Bachman
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Rebecca R Vanderpool
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Brian S Zuckerbraun
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hunter C Champion
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ana L Mora
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Adam C Straub
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Richard A Bilonick
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Maria J Calzada
- Hospital of the Princesa, Department of Medicine, Universidad Autónoma, Diego de León, 62 28006 Madrid, Spain
| | - Jeffrey S Isenberg
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
43
|
Maringer K, Sims-Lucas S. The multifaceted role of the renal microvasculature during acute kidney injury. Pediatr Nephrol 2016; 31:1231-40. [PMID: 26493067 PMCID: PMC4841763 DOI: 10.1007/s00467-015-3231-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 09/28/2015] [Accepted: 09/29/2015] [Indexed: 12/20/2022]
Abstract
Pediatric acute kidney injury (AKI) represents a complex disease process for clinicians as it is multifactorial in cause and only limited treatment or preventatives are available. The renal microvasculature has recently been implicated in AKI as a strong therapeutic candidate involved in both injury and recovery. Significant progress has been made in the ability to study the renal microvasculature following ischemic AKI and its role in repair. Advances have also been made in elucidating cell-cell interactions and the molecular mechanisms involved in these interactions. The ability of the kidney to repair post AKI is closely linked to alterations in hypoxia, and these studies are elucidated in this review. Injury to the microvasculature following AKI plays an integral role in mediating the inflammatory response, thereby complicating potential therapeutics. However, recent work with experimental animal models suggests that the endothelium and its cellular and molecular interactions are attractive targets to prevent injury or hasten repair following AKI. Here, we review the cellular and molecular mechanisms of the renal endothelium in AKI, as well as repair and recovery, and potential therapeutics to prevent or ameliorate injury and hasten repair.
Collapse
Affiliation(s)
- Katherine Maringer
- Rangos Research Center, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, USA
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sunder Sims-Lucas
- Rangos Research Center, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, USA.
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
44
|
Tengood JE, Levy RJ, Stachelek SJ. The use of CD47-modified biomaterials to mitigate the immune response. Exp Biol Med (Maywood) 2016; 241:1033-41. [PMID: 27190273 DOI: 10.1177/1535370216647130] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Addressing the aberrant interactions between immune cells and biomaterials represents an unmet need in biomaterial research. Although progress has been made in the development of bioinert coatings, identifying and targeting relevant cellular and molecular pathways can provide additional therapeutic strategies to address this major healthcare concern. To that end, we describe the immune inhibitory motif, receptor-ligand pairing of signal regulatory protein alpha and its cognate ligand CD47 as a potential signaling pathway to enhance biocompatibility. The goals of this article are to detail the known roles of CD47-signal regulatory protein alpha signal transduction pathway and to describe how immobilized CD47 can be used to mitigate the immune response to biomaterials. Current applications of CD47-modified biomaterials will also be discussed herein.
Collapse
Affiliation(s)
- Jillian E Tengood
- Division of Cardiology, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert J Levy
- Division of Cardiology, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stanley J Stachelek
- Division of Cardiology, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
45
|
Divergent modulation of normal and neoplastic stem cells by thrombospondin-1 and CD47 signaling. Int J Biochem Cell Biol 2016; 81:184-194. [PMID: 27163531 DOI: 10.1016/j.biocel.2016.05.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 04/27/2016] [Accepted: 05/04/2016] [Indexed: 01/19/2023]
Abstract
Thrombospondin-1 is a secreted matricellular protein that regulates the differentiation and function of many cell types. Thrombospondin-1 is not required for embryonic development, but studies using lineage-committed adult stem cells have identified positive and negative effects of thrombospondin-1 on stem cell differentiation and self-renewal and identified several thrombospondin-1 receptors that mediate these responses. Genetic studies in mice reveal a broad inhibitory role of thrombospondin-1 mediated by its receptor CD47. Cells and tissues lacking thrombospondin-1 or CD47 exhibit an increased capacity for self-renewal associated with increased expression of the stem cell transcription factors c-Myc, Sox2, Klf4, and Oct4. Thrombospondin-1 inhibits expression of these transcription factors in a CD47-dependent manner. However, this regulation differs in some neoplastic cells. Tumor initiating/cancer stem cells express high levels of CD47, but in contrast to nontransformed stem cells CD47 signaling supports cancer stem cells. Suppression of CD47 expression in cancer stem cells or ligation of CD47 by function blocking antibodies or thrombospondin-1 results in loss of self-renewal. Therefore, the therapeutic CD47 antagonists that are in clinical development for stimulating innate anti-tumor immunity may also inhibit tumor growth by suppressing cancer stem cells. These and other therapeutic modulators of thrombospondin-1 and CD47 signaling may also have applications in regenerative medicine to enhance the function of normal stem cells.
Collapse
|
46
|
Barclay JL, Keshvari S, Whitehead JP, Inder WJ. Development of an enzyme-linked immunosorbent assay for thrombospondin-1 and comparison of human plasma and serum concentrations. Ann Clin Biochem 2016; 53:606-10. [DOI: 10.1177/0004563216628891] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/30/2015] [Indexed: 11/16/2022]
Abstract
Background Thrombospondin-1 (TSP-1) is a circulating matricellular glycoprotein produced from many cell types including platelets. Currently TSP-1 is measured in either plasma or serum, using expensive commercial assays. Aim To develop and validate a cost effective in-house immunoassay for human TSP-1 suitable for quantitating levels from both plasma and serum. Methods An in-house enzyme-linked immunosorbent assay (ELISA) was developed for the measurement of human TSP-1. Sixteen healthy volunteers (8 male and 8 female), mean age 29 years (range 21–49), body mass index (BMI) mean 23.3 kg/m2 (range 17.3–26.7) had non-fasted venous blood sampled at 0800 h and 1600 h for both plasma and serum TSP-1. Results The assay limit of quantitation was 7.8 μg/L, inter assay CV was 17–31%, intra assay CV was 3–4% for plasma and <9% for serum. Plasma TSP-1 ranged from 133 to 478 μg/L (mean concentration 290 μg/L) in normal volunteers. Serum TSP-1 was approximately 100-fold higher, ranging from 13,700 to 44,400 μg/L (mean concentration 257,00 μg/L). There was no correlation between plasma and serum TSP-1. Conclusions TSP-1 can be readily measured in human plasma using ELISA. Serum concentrations are 100-fold higher, reflecting documented TSP-1 release by platelets, and does not provide a meaningful measure of circulating concentrations.
Collapse
Affiliation(s)
- Johanna L Barclay
- Mater Research Institute, University of Queensland, Brisbane, Australia
- Translational Research Institute, Brisbane, Australia
| | - Sahar Keshvari
- Mater Research Institute, University of Queensland, Brisbane, Australia
- Translational Research Institute, Brisbane, Australia
- School of Medicine, the University of Queensland, Brisbane, Australia
| | - Jonathan P Whitehead
- Mater Research Institute, University of Queensland, Brisbane, Australia
- Translational Research Institute, Brisbane, Australia
| | - Warrick J Inder
- Translational Research Institute, Brisbane, Australia
- School of Medicine, the University of Queensland, Brisbane, Australia
- Department of Diabetes and Endocrinology, Princess Alexandra Hospital, Brisbane, Australia
| |
Collapse
|
47
|
Yin DX, Zhao HM, Sun DJ, Yao J, Ding DY. Identification of candidate target genes for human peripheral arterial disease using weighted gene co‑expression network analysis. Mol Med Rep 2015; 12:8107-12. [PMID: 26498853 DOI: 10.3892/mmr.2015.4450] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 09/16/2015] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to identify the potential treatment targets of peripheral arterial disease (PAD) and provide further insights into the underlying mechanism of PAD, based on a weighted gene co‑expression network analysis (WGCNA) method. The mRNA expression profiles (accession. no. GSE27034), which included 19 samples from patients with PAD and 18 samples from normal control individuals were extracted from the Gene Expression Omnibus database. Subsequently, the differentially expressed genes (DEGs) were obtained using the Limma package and the co‑expression network modules were screened using the WGCNA approach. In addition, the protein‑protein interaction network for the DEGs in the most significant module was constructed using Cytoscape software. Functional enrichment analyses of the DEGs in the most significant module were also performed using the Database for Annotation, Visualization and Integrated Discovery and Kyoto Encyclopedia of Genes and Genomes (KEGG) Orthology‑Based Annotation System, respectively. A total of 148 DEGs were identified in PAD, which were used to construct the WGCN, in which two modules (gray module and turquoise module) were identified, with the gray module exhibiting a higher gene significance (GS) value than the turquoise module. In addition, a co‑expression network was constructed for 60 DEGs in the gray module. The functional enrichment results showed that the DEGs in the gray module were enriched in five Gene Ontology terms and four KEGG pathways. For example, cyclin‑dependent kinase inhibitor 1A (CDKN1A), FBJ murine osteosarcoma viral oncogene homolog (FOS) and prostaglandin‑endoperoxide synthase 2 (PTGS2) were enriched in response to glucocorticoid stimulus. The results of the present study suggested that DEGs in the gray module, including CDKN1A, FOS and PTGS2, may be associated with the pathogenesis of PAD, by modulating the cell cycle, and may offer potential for use as candidate treatment targets for PAD.
Collapse
Affiliation(s)
- De-Xin Yin
- Department of Vascular Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Hao-Min Zhao
- Department of Vascular Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Da-Jun Sun
- Department of Vascular Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Jian Yao
- Department of Thoracic Surgery, Jilin Hospital of Jilin Province People's Hospital, Changchun, Jilin 130031, P.R. China
| | - Da-Yong Ding
- Department of Gastroenterology Surgery, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
48
|
Abnormalities in three-dimensional capillary architecture and imbalance between vascular endothelial growth factor-A and thrombospondin-1 in soleus muscle of ovariectomized rat. Acta Histochem 2015; 117:605-11. [PMID: 26092525 DOI: 10.1016/j.acthis.2015.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Revised: 05/29/2015] [Accepted: 06/01/2015] [Indexed: 11/23/2022]
Abstract
Reduced ovarian hormone levels associated with menopause or ovariectomy (OVX) not only result in vascular dysfunction but also lead to structural abnormalities in capillaries. Therefore, the effect of OVX on the three-dimensional (3-D) architecture of capillary networks and the underlying molecular mechanisms were investigated in rat soleus muscle. Seven-week-old female Wistar rats were divided into the OVX and sham-treated (Sham) groups. The OVX group exhibited lower endurance exercise capacity compared to the sham group and resulted in decreased capillary diameter, number of anastomoses and capillary/anastomosis volume in soleus muscle, indicating 3-D structural abnormalities of capillary networks. Furthermore, OVX led to increased concentrations of thrombospondin-1 (TSP-1) protein and a decreased VEGF-A/TSP-1 ratio, an indicator of angio-adaptations, in soleus muscle compared with the Sham group. These results indicate OVX may induce 3-D capillary regression in soleus muscle through an imbalance between VEGF-A and TSP-1 expression, possibly associated with decreased exercise tolerance in ovariectomized rats.
Collapse
|
49
|
Smadja DM, Levy M, Huang L, Rossi E, Blandinières A, Israel-Biet D, Gaussem P, Bischoff J. Treprostinil indirectly regulates endothelial colony forming cell angiogenic properties by increasing VEGF-A produced by mesenchymal stem cells. Thromb Haemost 2015; 114:735-47. [PMID: 26062754 DOI: 10.1160/th14-11-0907] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Accepted: 04/26/2015] [Indexed: 01/05/2023]
Abstract
Pulmonary vasodilators and prostacyclin therapy in particular, have markedly improved the outcome of patients with pulmonary hypertension (PH). Endothelial dysfunction is a key feature of PH, and we previously reported that treprostinil therapy increases number and proliferative potential of endothelial colony forming cells (ECFC) isolated from PH patients' blood. In the present study, the objective was to determine how treprostinil contributes to the proangiogenic functions of ECFC. We examined the effect of treprostinil on ECFC obtained from cord blood in terms of colony numbers, proliferative and clonogenic properties in vitro, as well as in vivo vasculogenic properties. Surprisingly, treprostinil inhibited viability of cultured ECFC but did not modify their clonogenic properties or the endothelial differentiation potential from cord blood stem cells. Treprostinil treatment significantly increased the vessel-forming ability of ECFC combined with mesenchymal stem cells (MSC) in Matrigel implanted in nude mice. In vitro, ECFC proliferation was stimulated by conditioned media from treprostinil-pretreated MSC, and this effect was inhibited either by the use of VEGF-A blocking antibodies or siRNA VEGF-A in MSC. Silencing VEGF-A gene in MSC also blocked the pro-angiogenic effect of treprostinil in vivo. In conclusion, increased VEGF-A produced by MSC can account for the increased vessel formation observed during treprostinil treatment. The clinical relevance of these data was confirmed by the high level of VEGF-A detected in plasma from patients with paediatric PH who had been treated with treprostinil. Moreover, our results suggest that VEGF-A level in patients could be a surrogate biomarker of treprostinil efficacy.
Collapse
Affiliation(s)
- David M Smadja
- Prof. David Smadja, Georges Pompidou European Hospital, Hematology Department, 20 rue Leblanc, 75015 Paris, France, Tel.: +31 56093933, Fax: +31 56093393, E-mail:
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Chu LH, Vijay CG, Annex BH, Bader JS, Popel AS. PADPIN: protein-protein interaction networks of angiogenesis, arteriogenesis, and inflammation in peripheral arterial disease. Physiol Genomics 2015; 47:331-43. [PMID: 26058837 DOI: 10.1152/physiolgenomics.00125.2014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 06/04/2015] [Indexed: 11/22/2022] Open
Abstract
Peripheral arterial disease (PAD) results from an obstruction of blood flow in the arteries other than the heart, most commonly the arteries that supply the legs. The complexity of the known signaling pathways involved in PAD, including various growth factor pathways and their cross talks, suggests that analyses of high-throughput experimental data could lead to a new level of understanding of the disease as well as novel and heretofore unanticipated potential targets. Such bioinformatic analyses have not been systematically performed for PAD. We constructed global protein-protein interaction networks of angiogenesis (Angiome), immune response (Immunome), and arteriogenesis (Arteriome) using our previously developed algorithm GeneHits. The term "PADPIN" refers to the angiome, immunome, and arteriome in PAD. Here we analyze four microarray gene expression datasets from ischemic and nonischemic gastrocnemius muscles at day 3 posthindlimb ischemia (HLI) in two genetically different C57BL/6 and BALB/c mouse strains that display differential susceptibility to HLI to identify potential targets and signaling pathways in angiogenesis, immune, and arteriogenesis networks. We hypothesize that identification of the differentially expressed genes in ischemic and nonischemic muscles between the strains that recovers better (C57BL/6) vs. the strain that recovers more poorly (BALB/c) will help for the prediction of target genes in PAD. Our bioinformatics analysis identified several genes that are differentially expressed between the two mouse strains with known functions in PAD including TLR4, THBS1, and PRKAA2 and several genes with unknown functions in PAD including EphA4, TSPAN7, SLC22A4, and EIF2a.
Collapse
Affiliation(s)
- Liang-Hui Chu
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland;
| | - Chaitanya G Vijay
- Cardiovascular Medicine, Department of Medicine, and the Robert M. Berne Cardiovascular Research Center University of Virginia School of Medicine, Charlottesville, Virginia; and
| | - Brian H Annex
- Cardiovascular Medicine, Department of Medicine, and the Robert M. Berne Cardiovascular Research Center University of Virginia School of Medicine, Charlottesville, Virginia; and
| | - Joel S Bader
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland; High-Throughput Biology Center, Johns Hopkins University, Baltimore, Maryland
| | - Aleksander S Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|