1
|
Jing Jia, Ma B, Zhao X. Fetal endothelial colony-forming cells: Possible targets for prevention of the fetal origins of adult diseases. Placenta 2024; 145:80-88. [PMID: 38100962 DOI: 10.1016/j.placenta.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 11/20/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023]
Abstract
Endothelial colony-forming cells (ECFCs), a subset of circulating and resident endothelial progenitor cells, are capable of self-renewal and de novo vessel formation, and are known key regulators of vascular integrity and homeostasis. Numerous studies have found that exposure to hostile environment during the fetal development exerts a profound influence on the level and function of ECFCs, which may be the underlying factor linking endothelial dysfunction to cardiovascular disease of the offspring in later life. Herein, we focus on the latest findings regarding the effects of pregnancy-related disorders on the frequency and function of fetal ECFCs. Subsequently, we discuss about placental ECFCs and put forward some details that should be paid attention to in the process of ECFC isolation and culture. Overall, the information presented in this review highlight the potential of ECFCs as a future biomarker or even therapeutic targets for the pregnancy-related adverse maternal and fetal outcomes.
Collapse
Affiliation(s)
- Jing Jia
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Baitao Ma
- Department of Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xianlan Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
2
|
Dudley AC, Griffioen AW. Pathological angiogenesis: mechanisms and therapeutic strategies. Angiogenesis 2023; 26:313-347. [PMID: 37060495 PMCID: PMC10105163 DOI: 10.1007/s10456-023-09876-7] [Citation(s) in RCA: 164] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/26/2023] [Indexed: 04/16/2023]
Abstract
In multicellular organisms, angiogenesis, the formation of new blood vessels from pre-existing ones, is an essential process for growth and development. Different mechanisms such as vasculogenesis, sprouting, intussusceptive, and coalescent angiogenesis, as well as vessel co-option, vasculogenic mimicry and lymphangiogenesis, underlie the formation of new vasculature. In many pathological conditions, such as cancer, atherosclerosis, arthritis, psoriasis, endometriosis, obesity and SARS-CoV-2(COVID-19), developmental angiogenic processes are recapitulated, but are often done so without the normal feedback mechanisms that regulate the ordinary spatial and temporal patterns of blood vessel formation. Thus, pathological angiogenesis presents new challenges yet new opportunities for the design of vascular-directed therapies. Here, we provide an overview of recent insights into blood vessel development and highlight novel therapeutic strategies that promote or inhibit the process of angiogenesis to stabilize, reverse, or even halt disease progression. In our review, we will also explore several additional aspects (the angiogenic switch, hypoxia, angiocrine signals, endothelial plasticity, vessel normalization, and endothelial cell anergy) that operate in parallel to canonical angiogenesis mechanisms and speculate how these processes may also be targeted with anti-angiogenic or vascular-directed therapies.
Collapse
Affiliation(s)
- Andrew C Dudley
- Department of Microbiology, Immunology and Cancer Biology, The University of Virginia, Charlottesville, VA, 22908, USA.
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
3
|
Giubelan A, Stancu MI, Honţaru SO, Mălăescu GD, Badea-Voiculescu O, Firoiu C, Mogoantă SŞ. Tumor angiogenesis in gastric cancer. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY = REVUE ROUMAINE DE MORPHOLOGIE ET EMBRYOLOGIE 2023; 64:311-318. [PMID: 37867349 PMCID: PMC10720935 DOI: 10.47162/rjme.64.3.03] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/05/2023] [Indexed: 10/24/2023]
Abstract
Gastric cancer (GC) is still a major health problem, being one of the leading causes of cancer-related death in the world. Although the overall incidence of GC is decreasing in the United States and Western Europe, it is still high in many countries from Asia, South America, and Eastern Europe. The process of angiogenesis or the formation of new blood vessels plays an important role in cancer progression, as it allows oxygen supply, nutrients, and factors to grow tumor cells. In our study, we found that gastric neoplasms have high vascularity, with anarchic distribution, uneven in tumor stroma, sometimes with congestion vessels and microhemorrhages. Most vessels were capillaries, with a discontinuous endothelium, poorly structured basement membrane, without junctions between endothelial cells, hyperpermeable, creating the possibility of local edema in the tumor microenvironment (TME), and also extravasation of the plasma, leukocytes and even red blood cells. Angiogenesis vessels showed a low number of pericytes, which shows that they are young vessels, both morphologically and functionally immature. Tumor cells can synthesize biochemical factors [vascular endothelial growth factor-A (VEGF-A)] that stimulate the formation of new vessels (angiogenesis) to ensure their growth and metastasis. Some connective cells (tumor-associated mast cells, tumor-associated fibroblasts) are also involved in the angiogenesis process, which stimulate the progression of tumor cells and remodel the TME.
Collapse
Affiliation(s)
- Alexandru Giubelan
- PhD Student, Doctoral School, Faculty of Medicine, Titu Maiorescu University, Bucharest, Romania
| | - Marius Ionuţ Stancu
- PhD Student, Department of Histology, University of Medicine and Pharmacy of Craiova, Romania
| | - Sorina Octavia Honţaru
- Department of Health Care and Physiotherapy, Faculty of Sciences, Physical Education and Informatics, University of Piteşti, Romania
| | - Gheorghe Dan Mălăescu
- Department of Anatomy, Faculty of Nursing, Târgu Jiu Subsidiary, Titu Maiorescu University, Bucharest, Romania
| | - Oana Badea-Voiculescu
- Department of Modern Languages, University of Medicine and Pharmacy of Craiova, Romania
| | - Camelia Firoiu
- Department of Pathology, Emergency County Hospital, Târgu Jiu, Romania
| | - Stelian Ştefăniţă Mogoantă
- Department of Surgery, University of Medicine and Pharmacy of Craiova, Romania
- 3rd General Surgery Clinic, Emergency County Hospital, Craiova, Romania
| |
Collapse
|
4
|
Robertson JO, Erzurum SC, Asosingh K. Pathological Roles for Endothelial Colony-Forming Cells in Neonatal and Adult Lung Disease. Am J Respir Cell Mol Biol 2023; 68:13-22. [PMID: 36215049 PMCID: PMC9817912 DOI: 10.1165/rcmb.2022-0318ps] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 10/10/2022] [Indexed: 02/05/2023] Open
Abstract
Endothelial colony-forming cells (ECFCs) are vascular resident and circulating endothelial cell subtypes with potent angiogenic capacity, a hierarchy of single-cell clonogenic potentials, and the ability to participate in de novo blood vessel formation and endothelial repair. Existing literature regarding ECFCs in neonatal and adult pulmonary diseases is confounded by the study of ambiguously defined "endothelial progenitor cells," which are often not true ECFCs. This review contrasts adult and fetal ECFCs, discusses the effect of prematurity on ECFCs, and examines their different pathological roles in neonatal and adult pulmonary diseases, such as bronchopulmonary dysplasia, congenital diaphragmatic hernia, pulmonary artery hypertension, pulmonary fibrosis, and chronic obstructive pulmonary disease. Therapeutic potential is also discussed in light of available preclinical data.
Collapse
Affiliation(s)
| | - Serpil C. Erzurum
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Kewal Asosingh
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
5
|
Kourek C, Briasoulis A, Zouganeli V, Karatzanos E, Nanas S, Dimopoulos S. Exercise Training Effects on Circulating Endothelial and Progenitor Cells in Heart Failure. J Cardiovasc Dev Dis 2022; 9:222. [PMID: 35877584 PMCID: PMC9322098 DOI: 10.3390/jcdd9070222] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/29/2022] [Accepted: 07/07/2022] [Indexed: 02/06/2023] Open
Abstract
Heart failure (HF) is a major public health issue worldwide with increased prevalence and a high number of hospitalizations. Patients with chronic HF and either reduced ejection fraction (HFrEF) or mildly reduced ejection fraction (HFmrEF) present vascular endothelial dysfunction and significantly decreased circulating levels of endothelial progenitor cells (EPCs). EPCs are bone marrow-derived cells involved in endothelium regeneration, homeostasis, and neovascularization. One of the unsolved issues in the field of EPCs is the lack of an established method of identification. The most widely approved method is the use of monoclonal antibodies and fluorescence-activated cell sorting (FACS) analysis via flow cytometry. The most frequently used markers are CD34, VEGFR-2, CD45, CD31, CD144, and CD146. Exercise training has demonstrated beneficial effects on EPCs by increasing their number in peripheral circulation and improving their functional capacities in patients with HFrEF or HFmrEF. There are two potential mechanisms of EPCs mobilization: shear stress and the hypoxic/ischemic stimulus. The combination of both leads to the release of EPCs in circulation promoting their repairment properties on the vascular endothelium barrier. EPCs are important therapeutic targets and one of the most promising fields in heart failure and, therefore, individualized exercise training programs should be developed in rehabilitation centers.
Collapse
Affiliation(s)
- Christos Kourek
- Clinical Ergospirometry, Exercise & Rehabilitation Laboratory, 1st Critical Care Medicine Department, Evangelismos Hospital, National and Kapodistrian University of Athens, 10676 Athens, Greece; (C.K.); (E.K.); (S.N.)
- Department of Cardiology, 417 Army Share Fund Hospital of Athens (NIMTS), 11521 Athens, Greece
| | - Alexandros Briasoulis
- Department of Clinical Therapeutics, Alexandra Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece;
- Division of Cardiovascular Medicine, Section of Heart Failure and Transplantation, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Virginia Zouganeli
- Second Cardiology Department, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Eleftherios Karatzanos
- Clinical Ergospirometry, Exercise & Rehabilitation Laboratory, 1st Critical Care Medicine Department, Evangelismos Hospital, National and Kapodistrian University of Athens, 10676 Athens, Greece; (C.K.); (E.K.); (S.N.)
| | - Serafim Nanas
- Clinical Ergospirometry, Exercise & Rehabilitation Laboratory, 1st Critical Care Medicine Department, Evangelismos Hospital, National and Kapodistrian University of Athens, 10676 Athens, Greece; (C.K.); (E.K.); (S.N.)
| | - Stavros Dimopoulos
- Clinical Ergospirometry, Exercise & Rehabilitation Laboratory, 1st Critical Care Medicine Department, Evangelismos Hospital, National and Kapodistrian University of Athens, 10676 Athens, Greece; (C.K.); (E.K.); (S.N.)
- Cardiac Surgery Intensive Care Unit, Onassis Cardiac Surgery Center, 17674 Athens, Greece
| |
Collapse
|
6
|
Rossi E, Kauskot A, Saller F, Frezza E, Poirault-Chassac S, Lokajczyk A, Bourdoncle P, Saubaméa B, Gaussem P, Pericacho M, Bobe R, Bachelot-Loza C, Pasquali S, Bernabeu C, Smadja DM. Endoglin Is an Endothelial Housekeeper against Inflammation: Insight in ECFC-Related Permeability through LIMK/Cofilin Pathway. Int J Mol Sci 2021; 22:ijms22168837. [PMID: 34445542 PMCID: PMC8396367 DOI: 10.3390/ijms22168837] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/04/2021] [Accepted: 08/12/2021] [Indexed: 11/16/2022] Open
Abstract
Endoglin (Eng) is an endothelial cell (EC) transmembrane glycoprotein involved in adhesion and angiogenesis. Eng mutations result in vessel abnormalities as observed in hereditary hemorrhagic telangiectasia of type 1. The role of Eng was investigated in endothelial functions and permeability under inflammatory conditions, focusing on the actin dynamic signaling pathway. Endothelial Colony-Forming Cells (ECFC) from human cord blood and mouse lung/aortic EC (MLEC, MAEC) from Eng+/+ and Eng+/- mice were used. ECFC silenced for Eng with Eng-siRNA and ctr-siRNA were used to test tubulogenesis and permeability +/- TNFα and +/- LIM kinase inhibitors (LIMKi). In silico modeling of TNFα-Eng interactions was carried out from PDB IDs 5HZW and 5HZV. Calcium ions (Ca2+) flux was studied by Oregon Green 488 in epifluorescence microscopy. Levels of cofilin phosphorylation and tubulin post-translational modifications were evaluated by Western blot. F-actin and actin-tubulin distribution/co-localization were evaluated in cells by confocal microscopy. Eng silencing in ECFCs resulted in a decrease of cell sprouting by 50 ± 15% (p < 0.05) and an increase in pseudo-tube width (41 ± 4.5%; p < 0.001) compared to control. Upon TNFα stimulation, ECFC Eng-siRNA displayed a significant higher permeability compared to ctr-siRNA (p < 0.01), which is associated to a higher Ca2+ mobilization (p < 0.01). Computational analysis suggested that Eng mitigated TNFα activity. F-actin polymerization was significantly increased in ECFC Eng-siRNA, MAEC+/-, and MLEC+/- compared to controls (p < 0.001, p < 0.01, and p < 0.01, respectively) as well as actin/tubulin distribution (p < 0.01). Furthermore, the inactive form of cofilin (P-cofilin at Ser3) was significantly decreased by 36.7 ± 4.8% in ECFC Eng-siRNA compared to ctr-siRNA (p < 0.001). Interestingly, LIMKi reproduced the absence of Eng on TNFα-induced ECFC-increased permeability. Our data suggest that Eng plays a critical role in the homeostasis regulation of endothelial cells under inflammatory conditions (TNFα), and loss of Eng influences ECFC-related permeability through the LIMK/cofilin/actin rearrangement-signaling pathway.
Collapse
Affiliation(s)
- Elisa Rossi
- Faculty of Pharmacy, University of Paris, F-75006 Paris, France; (E.F.); (S.P.-C.); (A.L.); (B.S.); (P.G.); (C.B.-L.); (S.P.); (D.M.S.)
- IThEM, Inserm UMR-S 1140, F-75006 Paris, France
- Correspondence:
| | - Alexandre Kauskot
- HITh, UMR-S 1176, INSERM—Faculty of Medicine, University Paris-Saclay, F-94270 Le Kremlin-Bicêtre, France; (A.K.); (F.S.); (R.B.)
| | - François Saller
- HITh, UMR-S 1176, INSERM—Faculty of Medicine, University Paris-Saclay, F-94270 Le Kremlin-Bicêtre, France; (A.K.); (F.S.); (R.B.)
| | - Elisa Frezza
- Faculty of Pharmacy, University of Paris, F-75006 Paris, France; (E.F.); (S.P.-C.); (A.L.); (B.S.); (P.G.); (C.B.-L.); (S.P.); (D.M.S.)
- CiTCoM, CNRS, Université de Paris, F-75006 Paris, France
| | - Sonia Poirault-Chassac
- Faculty of Pharmacy, University of Paris, F-75006 Paris, France; (E.F.); (S.P.-C.); (A.L.); (B.S.); (P.G.); (C.B.-L.); (S.P.); (D.M.S.)
- IThEM, Inserm UMR-S 1140, F-75006 Paris, France
| | - Anna Lokajczyk
- Faculty of Pharmacy, University of Paris, F-75006 Paris, France; (E.F.); (S.P.-C.); (A.L.); (B.S.); (P.G.); (C.B.-L.); (S.P.); (D.M.S.)
- IThEM, Inserm UMR-S 1140, F-75006 Paris, France
| | - Pierre Bourdoncle
- Plate-Forme IMAG’IC Institut Cochin Inserm U1016-CNRS UMR8104, Université Paris Descartes, F-75006 Paris, France;
| | - Bruno Saubaméa
- Faculty of Pharmacy, University of Paris, F-75006 Paris, France; (E.F.); (S.P.-C.); (A.L.); (B.S.); (P.G.); (C.B.-L.); (S.P.); (D.M.S.)
- UMR-S 1144, F-75006 Paris, France
| | - Pascale Gaussem
- Faculty of Pharmacy, University of Paris, F-75006 Paris, France; (E.F.); (S.P.-C.); (A.L.); (B.S.); (P.G.); (C.B.-L.); (S.P.); (D.M.S.)
- IThEM, Inserm UMR-S 1140, F-75006 Paris, France
- AP-HP, Hematology Department, Hôpital Européen Georges Pompidou, F-75015 Paris, France
| | - Miguel Pericacho
- Department of Physiology and Pharmacology, Universidad de Salamanca, 37008 Salamanca, Spain;
| | - Regis Bobe
- HITh, UMR-S 1176, INSERM—Faculty of Medicine, University Paris-Saclay, F-94270 Le Kremlin-Bicêtre, France; (A.K.); (F.S.); (R.B.)
| | - Christilla Bachelot-Loza
- Faculty of Pharmacy, University of Paris, F-75006 Paris, France; (E.F.); (S.P.-C.); (A.L.); (B.S.); (P.G.); (C.B.-L.); (S.P.); (D.M.S.)
- IThEM, Inserm UMR-S 1140, F-75006 Paris, France
| | - Samuela Pasquali
- Faculty of Pharmacy, University of Paris, F-75006 Paris, France; (E.F.); (S.P.-C.); (A.L.); (B.S.); (P.G.); (C.B.-L.); (S.P.); (D.M.S.)
- CiTCoM, CNRS, Université de Paris, F-75006 Paris, France
| | - Carmelo Bernabeu
- Centro de Investigaciones Biológicas Margarita Salas, 28040 Madrid, Spain;
- Consejo Superior de Investigaciones Científicas (CSIC) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28040 Madrid, Spain
| | - David M. Smadja
- Faculty of Pharmacy, University of Paris, F-75006 Paris, France; (E.F.); (S.P.-C.); (A.L.); (B.S.); (P.G.); (C.B.-L.); (S.P.); (D.M.S.)
- IThEM, Inserm UMR-S 1140, F-75006 Paris, France
- AP-HP, Hematology Department, Hôpital Européen Georges Pompidou, F-75015 Paris, France
- Biosurgical Research Lab (Carpentier Foundation), F-75000 Paris, France
| |
Collapse
|
7
|
Díaz del Moral S, Barrena S, Muñoz-Chápuli R, Carmona R. Embryonic circulating endothelial progenitor cells. Angiogenesis 2020; 23:531-541. [DOI: 10.1007/s10456-020-09732-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 06/22/2020] [Indexed: 12/26/2022]
|
8
|
Proust R, Ponsen AC, Rouffiac V, Schenowitz C, Montespan F, Ser-Le Roux K, De Leeuw F, Laplace-Builhé C, Mauduit P, Carosella ED, Banzet S, Lataillade JJ, Rouas-Freiss N, Uzan G, Peltzer J. Cord blood-endothelial colony forming cells are immunotolerated and participate at post-ischemic angiogenesis in an original dorsal chamber immunocompetent mouse model. Stem Cell Res Ther 2020; 11:172. [PMID: 32381102 PMCID: PMC7206734 DOI: 10.1186/s13287-020-01687-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 03/30/2020] [Accepted: 04/22/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Cardiovascular diseases are the main cause of morbidity and mortality worldwide. Restoring blood supply to ischemic tissues is an essential goal for the successful treatment of these diseases. Growth factor or gene therapy efficacy remains controversial, but stem cell transplantation is emerging as an interesting approach to stimulate angiogenesis. Among the different stem cell populations, cord blood-endothelial progenitor cells (CB-EPCs) and more particularly cord blood-endothelial progenitor cell-derived endothelial colony forming cells (CB-ECFCs) have a great proliferative potential without exhibiting signs of senescence. Even if it was already described that CB-ECFCs were able to restore blood perfusion in hind-limb ischemia in an immunodeficient mouse model, until now, the immunogenic potential of allogenic CB-ECFCs remains controversial. Therefore, our objectives were to evaluate the immune tolerance potency of CB-ECFCs and their capacity to restore a functional vascular network under ischemic condition in immunocompetent mice. METHODS In vitro, the expression and secretion of immunoregulatory markers (HLA-G, IL-10, and TGF-β1) were evaluated on CB-ECFCs. Moreover, CB-ECFCs were co-cultured with activated peripheral blood mononuclear cells (PBMCs) for 6 days. PBMC proliferation was evaluated by [3H]-thymidine incorporation on the last 18 h. In vivo, CB-ECFCs were administered in the spleen and muscle of immunocompetent mice. Tissues were collected at day 14 after surgery. Finally, CB-ECFCs were injected intradermally in C57BL/6JRj mice close to ischemic macrovessel induced by thermal cauterization. Mice recovered until day 5 and were imaged, twice a week until day 30. RESULTS Firstly, we demonstrated that CB-ECFCs expressed HLA-G, IL-10, and TGF-β1 and secreted IL-10 and TGF-β1 and that they could display immunosuppressive properties in vitro. Secondly, we showed that CB-ECFCs could be tolerated until 14 days in immunocompetent mice. Thirdly, we revealed in an original ischemic model of dorsal chamber that CB-ECFCs were integrated in a new functional vascular network. CONCLUSION These results open up new perspectives about using CB-ECFCs as an allogeneic cell therapy product and gives new impulse to the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Richard Proust
- INSERM UMR-S-MD 1197/Ministry of the Armed Forces, Biomedical Research Institut of the Armed Forces (IRBA), Paul-Brousse Hospital Villejuif and CTSA Clamart, Clamart, France
| | - Anne-Charlotte Ponsen
- INSERM UMR-S-MD 1197/Ministry of the Armed Forces, Biomedical Research Institut of the Armed Forces (IRBA), Paul-Brousse Hospital Villejuif and CTSA Clamart, Clamart, France
| | - Valérie Rouffiac
- Paris-Saclay University, Paris-Sud University, Gustave Roussy Institute, INSERM, CNRS, Molecular Analysis, Modeling and Imaging of Cancer Disease, Villejuif, France
| | - Chantal Schenowitz
- CEA, DRF-IBFJ, Hemato-Immunology Research Unit, INSERM UMR-S 976, IRSL - Paris University, Saint-Louis Hospital, Paris, France
| | - Florent Montespan
- INSERM UMR-S-MD 1197/Ministry of the Armed Forces, Biomedical Research Institut of the Armed Forces (IRBA), Paul-Brousse Hospital Villejuif and CTSA Clamart, Clamart, France
| | - Karine Ser-Le Roux
- Paris-Saclay University, Paris-Sud University, Gustave Roussy Institute, INSERM, CNRS, Molecular Analysis, Modeling and Imaging of Cancer Disease, Villejuif, France
| | - Frédéric De Leeuw
- Paris-Saclay University, Paris-Sud University, Gustave Roussy Institute, INSERM, CNRS, Molecular Analysis, Modeling and Imaging of Cancer Disease, Villejuif, France
| | - Corinne Laplace-Builhé
- Paris-Saclay University, Paris-Sud University, Gustave Roussy Institute, INSERM, CNRS, Molecular Analysis, Modeling and Imaging of Cancer Disease, Villejuif, France
| | - Philippe Mauduit
- INSERM UMR-S-MD 1197/Ministry of the Armed Forces, Biomedical Research Institut of the Armed Forces (IRBA), Paul-Brousse Hospital Villejuif and CTSA Clamart, Clamart, France
| | - Edgardo D Carosella
- CEA, DRF-IBFJ, Hemato-Immunology Research Unit, INSERM UMR-S 976, IRSL - Paris University, Saint-Louis Hospital, Paris, France
| | - Sébastien Banzet
- INSERM UMR-S-MD 1197/Ministry of the Armed Forces, Biomedical Research Institut of the Armed Forces (IRBA), Paul-Brousse Hospital Villejuif and CTSA Clamart, Clamart, France
| | - Jean-Jacques Lataillade
- INSERM UMR-S-MD 1197/Ministry of the Armed Forces, Biomedical Research Institut of the Armed Forces (IRBA), Paul-Brousse Hospital Villejuif and CTSA Clamart, Clamart, France
| | - Nathalie Rouas-Freiss
- CEA, DRF-IBFJ, Hemato-Immunology Research Unit, INSERM UMR-S 976, IRSL - Paris University, Saint-Louis Hospital, Paris, France
| | - Georges Uzan
- INSERM UMR-S-MD 1197/Ministry of the Armed Forces, Biomedical Research Institut of the Armed Forces (IRBA), Paul-Brousse Hospital Villejuif and CTSA Clamart, Clamart, France
| | - Juliette Peltzer
- INSERM UMR-S-MD 1197/Ministry of the Armed Forces, Biomedical Research Institut of the Armed Forces (IRBA), Paul-Brousse Hospital Villejuif and CTSA Clamart, Clamart, France.
| |
Collapse
|
9
|
Kutikhin AG, Tupikin AE, Matveeva VG, Shishkova DK, Antonova LV, Kabilov MR, Velikanova EA. Human Peripheral Blood-Derived Endothelial Colony-Forming Cells Are Highly Similar to Mature Vascular Endothelial Cells yet Demonstrate a Transitional Transcriptomic Signature. Cells 2020; 9:cells9040876. [PMID: 32260159 PMCID: PMC7226818 DOI: 10.3390/cells9040876] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/30/2020] [Accepted: 04/02/2020] [Indexed: 02/06/2023] Open
Abstract
Endothelial colony-forming cells (ECFC) are currently considered as a promising cell population for the pre-endothelialization or pre-vascularization of tissue-engineered constructs, including small-diameter biodegradable vascular grafts. However, the extent of heterogeneity between ECFC and mature vascular endothelial cells (EC) is unclear. Here, we performed a transcriptome-wide study to compare gene expression profiles of ECFC, human coronary artery endothelial cells (HCAEC), and human umbilical vein endothelial cells (HUVEC). Characterization of the abovementioned cell populations was carried out by immunophenotyping, tube formation assay, and evaluation of proliferation capability while global gene expression profiling was conducted by means of RNA-seq. ECFC were similar to HUVEC in terms of immunophenotype (CD31+vWF+KDR+CD146+CD34-CD133-CD45-CD90-) and tube formation activity yet had expectedly higher proliferative potential. HCAEC and HUVEC were generally similar to ECFC with regards to their global gene expression profile; nevertheless, ECFC overexpressed specific markers of all endothelial lineages (NRP2, NOTCH4, LYVE1), in particular lymphatic EC (LYVE1), and had upregulated extracellular matrix and basement membrane genes (COL1A1, COL1A2, COL4A1, COL4A2). Proteomic profiling for endothelial lineage markers and angiogenic molecules generally confirmed RNA-seq results, indicating ECFC as an intermediate population between HCAEC and HUVEC. Therefore, gene expression profile and behavior of ECFC suggest their potential to be applied for a pre-endothelialization of bioartificial vascular grafts, whereas in terms of endothelial hierarchy they differ from HCAEC and HUVEC, having a transitional phenotype.
Collapse
Affiliation(s)
- Anton G. Kutikhin
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia; (V.G.M.); (D.K.S.); (L.V.A.); (E.A.V.)
- Correspondence: ; Tel.: +7-960-907-70-67
| | - Alexey E. Tupikin
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 8 Lavrentiev Avenue, Novosibirsk 630090, Russia; (A.E.T.); (M.R.K.)
| | - Vera G. Matveeva
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia; (V.G.M.); (D.K.S.); (L.V.A.); (E.A.V.)
| | - Daria K. Shishkova
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia; (V.G.M.); (D.K.S.); (L.V.A.); (E.A.V.)
| | - Larisa V. Antonova
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia; (V.G.M.); (D.K.S.); (L.V.A.); (E.A.V.)
| | - Marsel R. Kabilov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 8 Lavrentiev Avenue, Novosibirsk 630090, Russia; (A.E.T.); (M.R.K.)
| | - Elena A. Velikanova
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia; (V.G.M.); (D.K.S.); (L.V.A.); (E.A.V.)
| |
Collapse
|
10
|
Gao K, He S, Kumar P, Farmer D, Zhou J, Wang A. Clonal isolation of endothelial colony-forming cells from early gestation chorionic villi of human placenta for fetal tissue regeneration. World J Stem Cells 2020; 12:123-138. [PMID: 32184937 PMCID: PMC7062038 DOI: 10.4252/wjsc.v12.i2.123] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 12/03/2019] [Accepted: 12/23/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Endothelial colony-forming cells (ECFCs) have been implicated in the process of vascularization, which includes vasculogenesis and angiogenesis. Vasculogenesis is a de novo formation of blood vessels, and is an essential physiological process that occurs during embryonic development and tissue regeneration. Angiogenesis is the growth of new capillaries from pre-existing blood vessels, which is observed both prenatally and postnatally. The placenta is an organ composed of a variety of fetal-derived cells, including ECFCs, and therefore has significant potential as a source of fetal ECFCs for tissue engineering. AIM To investigate the possibility of isolating clonal ECFCs from human early gestation chorionic villi (CV-ECFCs) of the placenta, and assess their potential for tissue engineering. METHODS The early gestation chorionic villus tissue was dissociated by enzyme digestion. Cells expressing CD31 were selected using magnetic-activated cell sorting, and plated in endothelial-specific growth medium. After 2-3 wks in culture, colonies displaying cobblestone-like morphology were manually picked using cloning cylinders. We characterized CV-ECFCs by flow cytometry, immunophenotyping, tube formation assay, and Dil-Ac-LDL uptake assay. Viral transduction of CV-ECFCs was performed using a Luciferase/tdTomato-containing lentiviral vector, and transduction efficiency was tested by fluorescent microscopy and flow cytometry. Compatibility of CV-ECFCs with a delivery vehicle was determined using an FDA approved, small intestinal submucosa extracellular matrix scaffold. RESULTS After four passages in 6-8 wks of culture, we obtained a total number of 1.8 × 107 CV-ECFCs using 100 mg of early gestational chorionic villus tissue. Immunophenotypic analyses by flow cytometry demonstrated that CV-ECFCs highly expressed the endothelial markers CD31, CD144, CD146, CD105, CD309, only partially expressed CD34, and did not express CD45 and CD90. CV-ECFCs were capable of acetylated low-density lipoprotein uptake and tube formation, similar to cord blood-derived ECFCs (CB-ECFCs). CV-ECFCs can be transduced with a Luciferase/tdTomato-containing lentiviral vector at a transduction efficiency of 85.1%. Seeding CV-ECFCs on a small intestinal submucosa extracellular matrix scaffold confirmed that CV-ECFCs were compatible with the biomaterial scaffold. CONCLUSION In summary, we established a magnetic sorting-assisted clonal isolation approach to derive CV-ECFCs. A substantial number of CV-ECFCs can be obtained within a short time frame, representing a promising novel source of ECFCs for fetal treatments.
Collapse
Affiliation(s)
- Kewa Gao
- Department of Burns and Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan Province, China
- Surgical Bioengineering Laboratory, Department of Surgery, University of California Davis, Sacramento, CA 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, United States
| | - Siqi He
- Department of Burns and Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan Province, China
- Surgical Bioengineering Laboratory, Department of Surgery, University of California Davis, Sacramento, CA 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, United States
| | - Priyadarsini Kumar
- Surgical Bioengineering Laboratory, Department of Surgery, University of California Davis, Sacramento, CA 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, United States
| | - Diana Farmer
- Surgical Bioengineering Laboratory, Department of Surgery, University of California Davis, Sacramento, CA 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, United States
| | - Jianda Zhou
- Department of Burns and Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan Province, China
| | - Aijun Wang
- Surgical Bioengineering Laboratory, Department of Surgery, University of California Davis, Sacramento, CA 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, United States
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95817, United States.
| |
Collapse
|
11
|
Use of Human Umbilical Vein Endothelial Cells (HUVEC) as a Model to Study Cardiovascular Disease: A Review. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10030938] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide, and extensive research has been performed to understand this disease better, using various experimental models. The endothelium plays a crucial role in the development of CVD, since it is an interface between bloodstream components, such as monocytes and platelets, and other arterial wall components. Human umbilical vein endothelial cell (HUVEC) isolation from umbilical cord was first described in 1973. To date, this model is still widely used because of the high HUVEC isolation success rate, and because HUVEC are an excellent model to study a broad array of diseases, including cardiovascular and metabolic diseases. We here review the history of HUVEC isolation, the HUVEC model over time, HUVEC culture characteristics and conditions, advantages and disadvantages of this model and finally, its applications in the area of cardiovascular diseases.
Collapse
|
12
|
Moxibustion therapy improving delayed memory deficits via promoting neurogenesis and angiogenesis of hippocampus in a vascular dementia rat model. JOURNAL OF ACUPUNCTURE AND TUINA SCIENCE 2019. [DOI: 10.1007/s11726-019-1140-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
13
|
High Proliferative Placenta-Derived Multipotent Cells Express Cytokeratin 7 at Low Level. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2098749. [PMID: 31392209 PMCID: PMC6662495 DOI: 10.1155/2019/2098749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 05/30/2019] [Accepted: 06/25/2019] [Indexed: 12/11/2022]
Abstract
The purpose of this study was to investigate the immunophenotypes and gene expression profile of high proliferative placenta-derived multipotent cells (PDMCs) population at different stages of culture. We demonstrated that the colonies resulting from single cells were either positive or negative for CK7, whereas only PDMC clones with weak CK7 expression (CK7low-clones) were highly proliferative. Interestingly, vimentin positive (Vim+) placental stromal mesenchymal cells did not express CK7 in situ, but double CK7+Vim+ cells detection in tissue explants and explants outgrowth indicated CK7 inducible expression in vitro. PCNA presence in CK7+Vim+ cells during placental explants culturing confirmed belonging of these cells to proliferative subpopulation. Transcription factors CDX2 and EOMES were expressed in both CK7low-clones and subset of stromal mesenchymal cells of first-trimester placental tissue in situ. Meanwhile, CK7low -clones and stromal mesenchymal cells of full-term placental tissue in situ expressed ERG heterogeneously. SPP1, COL2A1, and PPARG2 mesodermal-related genes expression by CK7low-clones additionally confirms their mesenchymal origin. Inherent stem cell-related gene expression (IFTM3, POU5F1, and VASA) in CK7low-clones might indicate their enrichment for progenitors. Finally, in CK7low-clones we observed expression of such trophoblast-associated genes as CGB types I and II, fusogenic ERVW-1, GCM1, and GATA3. Thus, our results indicate that PDMCs acquired the representative immunophenotype signature under culture conditions.
Collapse
|
14
|
Taglauer E, Abman SH, Keller RL. Recent advances in antenatal factors predisposing to bronchopulmonary dysplasia. Semin Perinatol 2018; 42:413-424. [PMID: 30389227 PMCID: PMC6286866 DOI: 10.1053/j.semperi.2018.09.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Bronchopulmonary dysplasia (BPD) remains a major cause of late morbidities and death after preterm birth. BPD is characterized by an arrest of vascular and alveolar growth and high risk for pulmonary hypertension; yet mechanisms contributing to its pathogenesis and early strategies to prevent BPD are poorly understood. Strong epidemiologic studies have shown that the "new BPD" reflects the long-lasting impact of antenatal factors on lung development, partly due to placental dysfunction, as reflected in recent data from animal models. Improved understanding of mechanisms through which antenatal stress alters placental function and contributes to BPD may lead to preventive therapies.
Collapse
Affiliation(s)
| | - Steven H. Abman
- Pediatric Heart Lung Center, Department of Pediatrics, University of Colorado Anschutz School of Medicine, Aurora CO USA
| | - Roberta L. Keller
- Division of Neonatology, Department of Pediatrics, University of California San Francisco, San Francisco, CA USA
| |
Collapse
|
15
|
Qiu Y, Zhang C, Zhang G, Tao J. Endothelial progenitor cells in cardiovascular diseases. Aging Med (Milton) 2018; 1:204-208. [PMID: 31942498 PMCID: PMC6880702 DOI: 10.1002/agm2.12041] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 08/21/2018] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death in both developed and developing countries. Endothelial progenitor cells (EPCs) are derived from hematopoietic stem cells with powerful function of angiogenesis. There are many studies on the relation between coronary heart disease and circulating EPCs. In this review, we discuss biological characteristics of endothelial progenitor cells, some influencing factors of the number and function of EPCs, and the role of EPCs in the treatment of cardiovascular disease. At last, we bring some perspectives on the future of endothelial progenitor cell therapy.
Collapse
Affiliation(s)
- Yumin Qiu
- Department of Hypertension and Vascular DiseaseThe First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouChina
| | - Chanjuan Zhang
- Department of Hypertension and Vascular DiseaseThe First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouChina
| | - Gaoxing Zhang
- Department of Cardiovascular DiseaseThe Jiangmen Central HospitalJiangmenChina
| | - Jun Tao
- Department of Hypertension and Vascular DiseaseThe First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouChina
| |
Collapse
|
16
|
Lu W, Li X. PDGFs and their receptors in vascular stem/progenitor cells: Functions and therapeutic potential in retinal vasculopathy. Mol Aspects Med 2018; 62:22-32. [DOI: 10.1016/j.mam.2017.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/04/2017] [Indexed: 02/07/2023]
|
17
|
Bollini S, Silini AR, Banerjee A, Wolbank S, Balbi C, Parolini O. Cardiac Restoration Stemming From the Placenta Tree: Insights From Fetal and Perinatal Cell Biology. Front Physiol 2018; 9:385. [PMID: 29695981 PMCID: PMC5904405 DOI: 10.3389/fphys.2018.00385] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 03/28/2018] [Indexed: 12/27/2022] Open
Abstract
Efficient cardiac repair and ultimate regeneration still represents one of the main challenges of modern medicine. Indeed, cardiovascular disease can derive from independent conditions upsetting heart structure and performance: myocardial ischemia and infarction (MI), pharmacological cardiotoxicity, and congenital heart defects, just to name a few. All these disorders have profound consequences on cardiac tissue, inducing the onset of heart failure over time. Since the cure is currently represented by heart transplantation, which is extremely difficult due to the shortage of donors, much effort is being dedicated to developing innovative therapeutic strategies based on stem cell exploitation. Among the broad scenario of stem/progenitor cell subpopulations, fetal and perinatal sources, namely amniotic fluid and term placenta, have gained interest due to their peculiar regenerative capacity, high self-renewal capability, and ease of collection from clinical waste material. In this review, we will provide the state-of-the-art on fetal perinatal stem cells for cardiac repair and regeneration. We will discuss different pathological conditions and the main therapeutic strategies proposed, including cell transplantation, putative paracrine therapy, reprogramming, and tissue engineering approaches.
Collapse
Affiliation(s)
- Sveva Bollini
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Antonietta R Silini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza - Istituto Ospedaliero, Brescia, Italy
| | - Asmita Banerjee
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Susanne Wolbank
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Carolina Balbi
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Ornella Parolini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza - Istituto Ospedaliero, Brescia, Italy.,Institute of Human Anatomy and Cell Biology, "A. Gemelli" Faculty of Medicine and Surgery, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
18
|
Shear stress: An essential driver of endothelial progenitor cells. J Mol Cell Cardiol 2018; 118:46-69. [PMID: 29549046 DOI: 10.1016/j.yjmcc.2018.03.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/08/2018] [Accepted: 03/09/2018] [Indexed: 02/06/2023]
Abstract
The blood flow through vessels produces a tangential, or shear, stress sensed by their innermost layer (i.e., endothelium) and representing a major hemodynamic force. In humans, endothelial repair and blood vessel formation are mainly performed by circulating endothelial progenitor cells (EPCs) characterized by a considerable expression of vascular endothelial growth factor receptor 2 (VEGFR2), CD34, and CD133, pronounced tube formation activity in vitro, and strong reendothelialization or neovascularization capacity in vivo. EPCs have been proposed as a promising agent to induce reendothelialization of injured arteries, neovascularization of ischemic tissues, and endothelialization or vascularization of bioartificial constructs. A number of preconditioning approaches have been suggested to improve the regenerative potential of EPCs, including the use of biophysical stimuli such as shear stress. However, in spite of well-defined influence of shear stress on mature endothelial cells (ECs), articles summarizing how it affects EPCs are lacking. Here we discuss the impact of shear stress on homing, paracrine effects, and differentiation of EPCs. Unidirectional laminar shear stress significantly promotes homing of circulating EPCs to endothelial injury sites, induces anti-thrombotic and anti-atherosclerotic phenotype of EPCs, increases their capability to form capillary-like tubes in vitro, and enhances differentiation of EPCs into mature ECs in a dose-dependent manner. These effects are mediated by VEGFR2, Tie2, Notch, and β1/3 integrin signaling and can be abrogated by means of complementary siRNA/shRNA or selective pharmacological inhibitors of the respective proteins. Although the testing of sheared EPCs for vascular tissue engineering or regenerative medicine applications is still an unaccomplished task, favorable effects of unidirectional laminar shear stress on EPCs suggest its usefulness for their preconditioning.
Collapse
|
19
|
Shafiee A, Patel J, Lee JS, Hutmacher DW, Fisk NM, Khosrotehrani K. Mesenchymal stem/stromal cells enhance engraftment, vasculogenic and pro-angiogenic activities of endothelial colony forming cells in immunocompetent hosts. Sci Rep 2017; 7:13558. [PMID: 29051567 PMCID: PMC5648925 DOI: 10.1038/s41598-017-13971-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 10/04/2017] [Indexed: 02/08/2023] Open
Abstract
The clinical use of endothelial colony forming cells (ECFC) is hampered by their restricted engraftment. We aimed to assess engraftment, vasculogenic and pro-angiogenic activities of ECFC in immunocompetent (C57BL/6: WT) or immunodeficient (rag1 -/- C57BL/6: Rag1) mice. In addition, the impact of host immune system was investigated where ECFC were co-implanted with mesenchymal stem/stromal cells (MSC) from adult bone marrow (AdBM-MSC), fetal bone marrow (fBM-MSC), fetal placental (fPL-MSC), or maternal placental (MPL-MSC). Transplantation of ECFCs in Matrigel plugs resulted in less cell engraftment in WT mice compared to Rag1 mice. Co-implantation with different MSCs resulted in a significant increase in cell engraftment up to 9 fold in WT mice reaching levels of engraftment observed when using ECFCs alone in Rag1 mice but well below levels of engraftment with MSC-ECFC combination in Rag1 recipients. Furthermore, MSCs did not reduce murine splenic T cell proliferation in response to ECFCs in vitro. ECFCs enhanced the murine neo-vascularization through paracrine effect, but with no difference between Rag1 and WT mice. In conclusions, the host adaptive immune system affects the engraftment of ECFCs. MSC co-implantation improves ECFC engraftment and function even in immunocompetent hosts mostly through non-immune mechanisms.
Collapse
Affiliation(s)
- Abbas Shafiee
- The University of Queensland, UQ Centre for Clinical Research, Brisbane, 4029, QLD, Australia
- Queensland University of Technology, Brisbane, 4000, QLD, Australia
| | - Jatin Patel
- The University of Queensland, UQ Centre for Clinical Research, Brisbane, 4029, QLD, Australia
- The University of Queensland, UQ Diamantina Institute, Brisbane, 4102, QLD, Australia
| | - James S Lee
- The University of Queensland, UQ Centre for Clinical Research, Brisbane, 4029, QLD, Australia
- The University of Queensland, UQ Diamantina Institute, Brisbane, 4102, QLD, Australia
| | | | - Nicholas M Fisk
- The University of Queensland, UQ Centre for Clinical Research, Brisbane, 4029, QLD, Australia
- Centre for Advanced Prenatal Care, Royal Brisbane & Women's Hospital, Brisbane, 4029, QLD, Australia
| | - Kiarash Khosrotehrani
- The University of Queensland, UQ Centre for Clinical Research, Brisbane, 4029, QLD, Australia.
- The University of Queensland, UQ Diamantina Institute, Brisbane, 4102, QLD, Australia.
| |
Collapse
|
20
|
Zhang H, Tao Y, Ren S, Liu H, Zhou H, Hu J, Tang Y, Zhang B, Chen H. Isolation and characterization of human umbilical cord-derived endothelial colony-forming cells. Exp Ther Med 2017; 14:4160-4166. [PMID: 29067104 PMCID: PMC5647737 DOI: 10.3892/etm.2017.5035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 03/31/2017] [Indexed: 12/30/2022] Open
Abstract
Endothelial colony-forming cells (ECFCs) are a population of endothelial progenitor cells (EPCs) that display robust proliferative potential and vessel-forming capability. Previous studies have demonstrated that a limited number of ECFCs may be obtained from adult bone marrow, peripheral blood and umbilical cord (UC) blood. The present study describes an effective method for isolating ECFCs from human UC. The ECFCs derived from human UC displayed the full properties of EPCs. Analysis of the growth kinetics, cell cycle and colony-forming ability of the isolated human UC-ECFCs indicated that the cells demonstrated properties of stem cells, including relative stability and rapid proliferation in vitro. Gene expression of Fms related tyrosine kinase 1, kinase insert domain receptor, vascular endothelial cadherin, cluster of differentiation (CD)31, CD34, epidermal growth factor homology domains-2, von Willebrand factor and endothelial nitric oxide synthase was assessed by reverse transcription-polymerase chain reaction. The cells were positive for CD34, CD31, CD73, CD105 and vascular endothelial growth factor receptor-2, and negative for CD45, CD90 and human leukocyte antigen-antigen D related protein according to flow cytometry. 1,1'-dioctadecyl-3,3,3',3'-tetra-methyl-indocarbocyanine perchlorate-labeled acetylated low-density lipoprotein and fluorescein isothiocyanate-Ulex europaeus-l were used to verify the identity of the UC-ECFCs. Matrigel was used to investigate tube formation capability. The results demonstrated that the reported technique is a valuable method for isolating human UC-ECFCs, which have potential for use in vascular regeneration.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing 100071, P.R. China.,Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Yanling Tao
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Saisai Ren
- Department of Graduate School, Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Haihui Liu
- Department of Graduate School, Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Hui Zhou
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Jiangwei Hu
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing 100071, P.R. China
| | - Yongyong Tang
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing 100071, P.R. China.,Cell and Gene Therapy Center, Academy of Military Medical Sciences, Beijing 100071, P.R. China
| | - Bin Zhang
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing 100071, P.R. China.,Cell and Gene Therapy Center, Academy of Military Medical Sciences, Beijing 100071, P.R. China
| | - Hu Chen
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing 100071, P.R. China.,Cell and Gene Therapy Center, Academy of Military Medical Sciences, Beijing 100071, P.R. China
| |
Collapse
|
21
|
Green L, Ofstein RH, Rapp B, Saadatzadeh MR, Bhavsar JR, Fajardo A, Dalsing MC, Ingram DA, Murphy MP. Adult venous endothelium is a niche for highly proliferative and vasculogenic endothelial colony-forming cells. J Vasc Surg 2017; 66:1854-1863. [PMID: 28655551 DOI: 10.1016/j.jvs.2016.11.059] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 11/30/2016] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Postnatal resident endothelium of blood vessels has been proposed to represent terminally differentiated tissue that does not replicate. We previously isolated endothelial colony-forming cells (ECFCs) from human umbilical cord blood (CB) and term placenta by using colony-forming assays and immunocytochemistry. We showed that ECFCs are highly proliferative and form functioning vessels in vivo, the defining characteristics of a true endothelial progenitor cell. This exploratory investigation was conducted to determine whether the endothelium of healthy adult blood vessels contained resident ECFCs. METHODS The endothelium of great saphenous vein (GSV) obtained from vein stripping procedures was collected with mechanical scraping, and ECFCs were isolated according to established protocols. RESULTS GSV ECFCs incorporated acetylated low-density lipoprotein, formed tubules in Matrigel (BD Biosciences, San Jose, Calif) at 24 hours, and expressed endothelial antigens cluster of differentiation (CD) 144, CD31, CD105, and kinase insert domain receptor but not hematopoietic antigen CD45. Using cumulative population doublings and single-cell assays, we demonstrated that GSV ECFCs exhibited comparable proliferative capacities compared with CB ECFCs, including similar numbers of highly proliferative cells. When injected in collagen/fibronectin gels implanted in nonobese diabetic/severe combined immune deficiency mice, GSV ECFCs formed blood vessels with circulating murine red blood cells, demonstrating their vasculogenic potential. CONCLUSIONS The ECFCs of the GSV contain a hierarchy of progenitor cells with a comparable number of highly proliferative clones as ECFCs of CB. The results of this investigation demonstrate that the adult endothelium contains resident progenitor cells that may have a critical role in vascular homeostasis and repair and could potentially be used as a source of autologous cells for cell therapies focusing on vasculogenesis.
Collapse
Affiliation(s)
- Linden Green
- Health Center for Aortic Disease, Indiana University Health Center for Aortic Disease, Indianapolis, Ind; Department of Cellular and Integrative Physiology, Indiana University Health Center for Aortic Disease, Indianapolis, Ind.
| | - Richard H Ofstein
- Department of Surgery, Indiana University Health Center for Aortic Disease, Indianapolis, Ind
| | - Brian Rapp
- Department of Surgery, Indiana University Health Center for Aortic Disease, Indianapolis, Ind
| | - M Reza Saadatzadeh
- Department of Surgery, Indiana University Health Center for Aortic Disease, Indianapolis, Ind; Indiana Center for Vascular Biology and Medicine, Indiana University Health Center for Aortic Disease, Indianapolis, Ind
| | - Janak R Bhavsar
- Department of Surgery, Indiana University Health Center for Aortic Disease, Indianapolis, Ind; Indiana Center for Vascular Biology and Medicine, Indiana University Health Center for Aortic Disease, Indianapolis, Ind
| | - Andres Fajardo
- Department of Surgery, Indiana University Health Center for Aortic Disease, Indianapolis, Ind
| | - Michael C Dalsing
- Department of Surgery, Indiana University Health Center for Aortic Disease, Indianapolis, Ind
| | - David A Ingram
- Indiana Center for Vascular Biology and Medicine, Indiana University Health Center for Aortic Disease, Indianapolis, Ind; Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University Health Center for Aortic Disease, Indianapolis, Ind; Department of Biochemistry and Molecular Biology, Indiana University Health Center for Aortic Disease, Indianapolis, Ind
| | - Michael P Murphy
- Health Center for Aortic Disease, Indiana University Health Center for Aortic Disease, Indianapolis, Ind; Department of Cellular and Integrative Physiology, Indiana University Health Center for Aortic Disease, Indianapolis, Ind; Department of Surgery, Indiana University Health Center for Aortic Disease, Indianapolis, Ind; Indiana Center for Vascular Biology and Medicine, Indiana University Health Center for Aortic Disease, Indianapolis, Ind
| |
Collapse
|
22
|
Gumina DL, Su EJ. Endothelial Progenitor Cells of the Human Placenta and Fetoplacental Circulation: A Potential Link to Fetal, Neonatal, and Long-term Health. Front Pediatr 2017; 5:41. [PMID: 28361046 PMCID: PMC5350128 DOI: 10.3389/fped.2017.00041] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 02/15/2017] [Indexed: 12/16/2022] Open
Abstract
The fetoplacental circulation plays a key role in both short- and long-term outcomes, and aberrant flow indices as manifested by abnormal fetal Doppler velocimetry within this compartment have been associated with significant adverse consequences. These include fetal growth restriction, which often coexists with preeclampsia, and long-lasting medical issues as a result of both the underlying pathology and prematurity such as bronchopulmonary dysplasia, chronic lung disease, and neurodevelopmental delay. Furthermore, it is also clear that exposure to an abnormal in utero environment increases risk for long-term, adulthood issues such as cardiovascular disease. Endothelial progenitor cells (EPCs) have been implicated in vasculogenesis and angiogenesis, and they have been isolated from both human placenta and umbilical cord blood. This review outlines the extensive nomenclature of EPCs, summarizes existing literature surrounding human placental and umbilical cord blood EPCs, explores their potential role in pregnancy complications and adverse perinatal outcome, and highlights key areas where future investigations are needed.
Collapse
Affiliation(s)
- Diane L Gumina
- Obstetrics and Gynecology, University of Colorado School of Medicine , Aurora, CO , USA
| | - Emily J Su
- Obstetrics and Gynecology, University of Colorado School of Medicine , Aurora, CO , USA
| |
Collapse
|
23
|
Shafiee A, Patel J, Wong HY, Donovan P, Hutmacher DW, Fisk NM, Khosrotehrani K. Priming of endothelial colony-forming cells in a mesenchymal niche improves engraftment and vasculogenic potential by initiating mesenchymal transition orchestrated by NOTCH signaling. FASEB J 2016; 31:610-624. [PMID: 28045376 DOI: 10.1096/fj.201600937] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 10/11/2016] [Indexed: 12/31/2022]
Abstract
The prospect of using endothelial progenitors is currently hampered by their low engraftment upon transplantation. We report that mesenchymal stem/stromal cells (MSCs), independent of source and age, improve the engraftment of endothelial colony forming cells (ECFCs). MSC coculture altered ECFC appearance to an elongated mesenchymal morphology with reduced proliferation. ECFC primed via MSC contact had reduced self-renewal potential, but improved capacity to form tube structures in vitro and engraftment in vivo Primed ECFCs displayed major differences in transcriptome compared to ECFCs never exposed to MSCs, affecting genes involved in the cell cycle, up-regulating of genes influencing mesenchymal transition, adhesion, extracellular matrix. Inhibition of NOTCH signaling, a potential upstream regulator of mesenchymal transition, in large part modulated this gene expression pattern and functionally reversed the mesenchymal morphology of ECFCs. The collective results showed that primed ECFCs survive better and undergo a mesenchymal transition that is dependent on NOTCH signaling, resulting in significantly increased vasculogenic potential.-Shafiee, A., Patel, J., Wong, H. Y., Donovan, P., Hutmacher, D. W., Fisk, N. M., Khosrotehrani, K. Priming of endothelial colony-forming cells in a mesenchymal niche improves engraftment and vasculogenic potential by initiating mesenchymal transition orchestrated by NOTCH signaling.
Collapse
Affiliation(s)
- Abbas Shafiee
- University of Queensland (UQ) Centre for Clinical Research, The University of Queensland, Brisbane, Queensland Australia.,Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Jatin Patel
- University of Queensland (UQ) Centre for Clinical Research, The University of Queensland, Brisbane, Queensland Australia
| | - Ho Yi Wong
- University of Queensland (UQ) Centre for Clinical Research, The University of Queensland, Brisbane, Queensland Australia
| | - Prudence Donovan
- UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, Queensland, Australia; and
| | - Dietmar W Hutmacher
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Nicholas M Fisk
- University of Queensland (UQ) Centre for Clinical Research, The University of Queensland, Brisbane, Queensland Australia.,Centre for Advanced Prenatal Care, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Kiarash Khosrotehrani
- University of Queensland (UQ) Centre for Clinical Research, The University of Queensland, Brisbane, Queensland Australia; .,UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, Queensland, Australia; and
| |
Collapse
|
24
|
Obtulowicz P, Lech W, Strojek L, Sarnowska A, Domanska-Janik K. Induction of Endothelial Phenotype from Wharton's Jelly-Derived MSCs and Comparison of Their Vasoprotective and Neuroprotective Potential with Primary WJ-MSCs in CA1 Hippocampal Region Ex Vivo. Cell Transplant 2016; 25:715-27. [DOI: 10.3727/096368915x690369] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Ischemic stroke results in violent impairment of tissue homeostasis leading to severe perturbation within the neurovascular unit (NVU) during the recovery period. The aim of this study was to assess the potential of mesenchymal stem cells (MSCs) originating from Wharton's jelly (WJ) to differentiate into functionally competent cells of endothelial lineage (WJ-EPCs). The protective effect(s) of either primary WJ-MSCs or induced WJ-EPCs was investigated and compared after oxygen–glucose deprivation (OGD) of hippocampal organotypic slices (OHC) in the indirect coculture model. WJ-MSCs, primed in EGM-2 (Lonza commercial medium) under 5% O2, acquired cobblestone endothelial-like morphology, formed capillary-like structures and actively took up DiI-Ac-LDL. Both cell types (WJ-MSCs and WJ-EPCs) were positive for CD73, CD90, CD105, VEGFR-2, and VEGF, but only endothelial-like culture expressed vWF and PECAM-1 markers at significant levels. In the presence of either WJ-MSCs or WJ-EPCs in the compartment below OGD-injured slices, cell death and vascular atrophy in the hypoxia-sensitive CA1 region were substantially decreased. This suggests that a paracrine mechanism may mediate WJ-MSC- and WJ-EPC-dependent protection. Thus, finally, we estimated secretion of the neuro/angio/immunomodulatory molecules IL-6, TGF-β1, and VEGF by these cell cultures. We have found that release of TGF-β1 and IL-6 was TLR ligand [LPS and Poly(I:C)] concentration dependent and stronger in WJ-EPC than WJ-MSC cultures. Simultaneously, the uneven pattern of TLR receptors and modulatory cytokine gene expression was confirmed also on qRT-PCR level, but no significant differences were noticed between WJ-EPC and primary WJ-MSC cultures.
Collapse
Affiliation(s)
- Patrycja Obtulowicz
- Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Wioletta Lech
- Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Lukasz Strojek
- Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Sarnowska
- Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | | |
Collapse
|
25
|
Shafiee A, Fisk NM, Hutmacher DW, Khosrotehrani K, Patel J. Fetal endothelial and mesenchymal progenitors from the human term placenta: potency and clinical potential. Stem Cells Transl Med 2015; 4:419-23. [PMID: 25769652 DOI: 10.5966/sctm.2014-0224] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 02/09/2015] [Indexed: 01/11/2023] Open
Abstract
Since the isolation of fetal stem cell populations from perinatal tissues, such as umbilical cord blood and placenta, interest has been growing in understanding their greater plasticity compared with adult stem cells and exploring their potential in regenerative medicine. The phenomenon of fetal microchimerism (FMC) naturally occurring during pregnancy through the transfer of fetal stem/progenitor cells to maternal blood and tissues has been integral in developing this dogma. Specifically, microchimeric mesenchymal stem cells and endothelial progenitors of fetal origin have now demonstrated a capacity for tissue repair in the maternal host. However, the use of similar fetal stem cells in therapy has been significantly hampered by the availability of clinically relevant cell numbers and/or contamination with cells of maternal origin, particularly when using the chorionic and decidual placenta. In the present prospective review, we highlight the importance of FMC to the field of fetal stem cell biology and issues of maternal contamination from perinatal tissues and discuss specific isolation strategies to overcome these translational obstacles.
Collapse
Affiliation(s)
- Abbas Shafiee
- Experimental Dermatology Group, UQ Centre for Clinical Research, The University of Queensland, Herston, Queensland, Australia; Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Nicholas M Fisk
- Experimental Dermatology Group, UQ Centre for Clinical Research, The University of Queensland, Herston, Queensland, Australia
| | - Dietmar W Hutmacher
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Kiarash Khosrotehrani
- Experimental Dermatology Group, UQ Centre for Clinical Research, The University of Queensland, Herston, Queensland, Australia;
| | - Jatin Patel
- Experimental Dermatology Group, UQ Centre for Clinical Research, The University of Queensland, Herston, Queensland, Australia
| |
Collapse
|
26
|
Patel J, Seppanen E, Chong MSK, Yeo JSL, Teo EYL, Chan JKY, Fisk NM, Khosrotehrani K. Prospective surface marker-based isolation and expansion of fetal endothelial colony-forming cells from human term placenta. Stem Cells Transl Med 2013; 2:839-47. [PMID: 24106336 DOI: 10.5966/sctm.2013-0092] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The term placenta is a highly vascularized tissue and is usually discarded upon birth. Our objective was to isolate clinically relevant quantities of fetal endothelial colony-forming cells (ECFCs) from human term placenta and to compare them to the well-established donor-matched umbilical cord blood (UCB)-derived ECFCs. A sorting strategy was devised to enrich for CD45-CD34+CD31Lo cells prior to primary plating to obtain pure placental ECFCs (PL-ECFCs) upon culture. UCB-ECFCs were derived using a well-described assay. PL-ECFCs were fetal in origin and expressed the same cell surface markers as UCB-ECFCs. Most importantly, a single term placenta could yield as many ECFCs as 27 UCB donors. PL-ECFCs and UCB-ECFCs had similar in vitro and in vivo vessel forming capacities and restored mouse hind limb ischemia in similar proportions. Gene expression profiles were only minimally divergent between PL-ECFCs and UCB-ECFCs, probably reflecting a vascular source versus a circulating source. Finally, PL-ECFCs and UCB-ECFCs displayed similar hierarchies between high and low proliferative colonies. We report a robust strategy to isolate ECFCs from human term placentas based on their cell surface expression. This yielded much larger quantities of ECFCs than UCB, but the cells were comparable in immunophenotype, gene expression, and in vivo functional ability. We conclude that PL-ECFCs have significant bio-banking and clinical translatability potential.
Collapse
Affiliation(s)
- Jatin Patel
- University of Queensland Centre for Clinical Research, University of Queensland, Herston, Queensland, Australia
| | | | | | | | | | | | | | | |
Collapse
|