1
|
Smadja DM, Mauge L, Rancic J, Gaussem P, Feraud O, Oudrhiri N, Bennaceur-Griscelli A. Comparative Evaluation of Endothelial Colony-Forming Cells from Cord and Adult Blood vs. Human Embryonic Stem Cell-Derived Endothelial Cells: Insights into Therapeutic Angiogenesis Potential. Stem Cell Rev Rep 2024:10.1007/s12015-024-10830-3. [PMID: 39612122 DOI: 10.1007/s12015-024-10830-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
The discovery of endothelial progenitor cells has revolutionized our understanding of postnatal blood vessel formation, with endothelial colony-forming cells (ECFCs) emerging as key players in vasculogenesis. Among various ECFC sources, cord blood-derived ECFCs (CB-ECFCs) are of particular interest due to their superior proliferative and clonogenic potential and their ability to promote vascular network formation. Human embryonic stem cell-derived endothelial cells (hESC-ECs) have also shown potential in regenerative medicine, though their vasculogenic efficacy remains unclear compared to CB- and adult blood-derived ECFCs (AB-ECFCs). This study aimed to directly compare the angiogenic and vasculogenic capabilities of CB-ECFCs, AB-ECFCs, and hESC-ECs in vitro and in vivo. Our results demonstrated that CB-ECFCs had a significantly higher proliferation rate than both AB-ECFCs and hESC-ECs (p < 0.01). In tube formation assays, CB-ECFCs exhibited superior ability to form capillary-like structures compared to hESC-ECs (p < 0.0001) and AB-ECFCs (p < 0.01). In vivo, CB-ECFCs significantly improved blood flow recovery in ischemic tissue (p < 0.01), outperforming both AB-ECFCs and hESC-ECs, with no significant recovery observed in the latter two groups. These findings suggest that CB-ECFCs represent a more effective cell source for therapeutic angiogenesis, while further optimization is needed to enhance the efficacy of hESC-ECs for clinical applications. Future research should explore the molecular mechanisms underlying the superior regenerative potential of CB-ECFCs and focus on improving the stability and functionality of stem cell-derived ECs for therapeutic use.
Collapse
Affiliation(s)
- David M Smadja
- Université Paris Cité, INSERM, Innovative Therapies in Hemostasis, Paris, F-75006, France.
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Paris, F-75015, France.
| | - Laetitia Mauge
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Paris, F-75015, France
- Université Paris Cité, INSERM, PARCC, Paris, F-75015, France
| | - Jeanne Rancic
- Université Paris Cité, INSERM, Innovative Therapies in Hemostasis, Paris, F-75006, France
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Paris, F-75015, France
| | - Pascale Gaussem
- Université Paris Cité, INSERM, Innovative Therapies in Hemostasis, Paris, F-75006, France
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Paris, F-75015, France
| | - Olivier Feraud
- INSERM U935/U1310 ESTeam Paris Sud Human Pluripotent Stem Cell Core Facility, Villejuif, France
| | - Noufissa Oudrhiri
- INSERM U935/U1310 ESTeam Paris Sud Human Pluripotent Stem Cell Core Facility, Villejuif, France
- CITHERA UMS45, Infrastructure-INGESTEM, INSERM, Université Paris-Saclay, Faculté de Médecine, Kremlin Bicêtre, France
- Hematology Department, AP-HP, Hôpital Universitaire Paris Sud-Kremlin Bicêtre, Kremlin-Bicêtre, France
| | - Annelise Bennaceur-Griscelli
- INSERM U935/U1310 ESTeam Paris Sud Human Pluripotent Stem Cell Core Facility, Villejuif, France.
- CITHERA UMS45, Infrastructure-INGESTEM, INSERM, Université Paris-Saclay, Faculté de Médecine, Kremlin Bicêtre, France.
- Hematology Department, AP-HP, Hôpital Universitaire Paris Sud-Kremlin Bicêtre, Kremlin-Bicêtre, France.
| |
Collapse
|
2
|
Smadja DM. Extracellular Microvesicles vs. Mitochondria: Competing for the Top Spot in Cardiovascular Regenerative Medicine. Stem Cell Rev Rep 2024; 20:1813-1818. [PMID: 38976143 DOI: 10.1007/s12015-024-10758-8] [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] [Accepted: 06/25/2024] [Indexed: 07/09/2024]
Abstract
Regenerative medicine aims to restore, replace, and regenerate human cells, tissues, and organs. Despite significant advancements, many cell therapy trials for cardiovascular diseases face challenges like cell survival and immune compatibility, with benefits largely stemming from paracrine effects. Two promising therapeutic tools have been recently emerged in cardiovascular diseases: extracellular vesicles (EVs) and mitochondrial transfer. Concerning EVs, the first pivotal study with EV-enriched secretome derived from cardiovascular progenitor cells has been done treating heart failure. This first in man demonstrated the safety and feasibility of repeated intravenous infusions and highlighted significant clinical improvements, including enhanced cardiac function and reduced symptoms in heart failure patients. The second study uncovered a novel mechanism of endothelial regeneration through mitochondrial transfer via tunneling nanotubes (TNTs). This research showed that mesenchymal stromal cells (MSCs) transfer mitochondria to endothelial cells, significantly enhancing their bioenergetics and vessel-forming capabilities. This mitochondrial transfer was crucial for endothelial cell engraftment and function, offering a new strategy for vascular regeneration without the need for additional cell types. Combining EV and mitochondrial strategies presents new clinical opportunities. These approaches could revolutionize regenerative medicine, offering new hope for treating cardiovascular and other degenerative diseases. Continued research and clinical trials will be crucial in optimizing these therapies, potentially leading to personalized medicine approaches that enhance patient outcomes.
Collapse
Affiliation(s)
- David M Smadja
- Université Paris Cité, INSERM, Innovative Therapies in Hemostasis, Paris, F-75006, France.
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Inserm UMR-S1140, 56 rue Leblanc, Paris, F-75015, France.
| |
Collapse
|
3
|
Huang L, Ye Y, Sun Y, Zhou Z, Deng T, Liu Y, Wu R, Wang K, Yao C. LncRNA H19/miR-107 regulates endothelial progenitor cell pyroptosis and promotes flow recovery of lower extremity ischemia through targeting FADD. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167323. [PMID: 38925483 DOI: 10.1016/j.bbadis.2024.167323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 06/02/2024] [Accepted: 06/21/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Peripheral artery disease (PAD) is an ischemic disease with a rising incidence worldwide. The lncRNA H19 (H19) is enriched in endothelial progenitor cells (EPCs), and transplantation of pyroptosis-resistant H19-overexpressed EPCs (oe-H19-EPCs) may promote vasculogenesis and blood flow recovery in PAD, especially with critical limb ischemia (CLI). METHODS EPCs isolated from human peripheral blood was characterized using immunofluorescence and flow cytometry. Cell proliferation was determined with CCK8 and EdU assays. Cell migration was assessed by Transwell and wound healing assays. The angiogenic potential was evaluated using tube formation assay. The pyroptosis pathway-related protein in EPCs was detected by western blot. The binding sites of H19 and FADD on miR-107 were analyzed using Luciferase assays. In vivo, oe-H19-EPCs were transplanted into a mouse ischemic limb model, and blood flow was detected by laser Doppler imaging. The transcriptional landscape behind the therapeutic effects of oe-H19-EPCs on ischemic limbs were examined with whole transcriptome sequencing. RESULTS Overexpression of H19 in EPCs led to an increase in proliferation, migration, and tube formation abilities. These effects were mediated through pyroptosis pathway, which is regulated by the H19/miR-107/FADD axis. Transplantation of oe-H19-EPCs in a mouse ischemic limb model promoted vasculogenesis and blood flow recovery. Whole transcriptome sequencing indicated significant activation of vasculogenesis pathway in the ischemic limbs following treatment with oe-H19-EPCs. CONCLUSIONS Overexpression of H19 increases FADD level by competitively binding to miR-107, leading to enhanced proliferation, migration, vasculogenesis, and inhibition of pyroptosis in EPCs. These effects ultimately promote the recovery of blood flow in CLI.
Collapse
Affiliation(s)
- Lin Huang
- Division of Vascular Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510800, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Yanchen Ye
- Division of Vascular Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510800, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Yunhao Sun
- Division of Vascular Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510800, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhihao Zhou
- Division of Vascular Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510800, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Tang Deng
- Division of Vascular Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510800, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Yunyan Liu
- Division of Vascular Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510800, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Ridong Wu
- Division of Vascular Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510800, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China.
| | - Kangjie Wang
- Division of Vascular Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510800, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China.
| | - Chen Yao
- Division of Vascular Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510800, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
4
|
Rojas-Torres M, Beltrán-Camacho L, Martínez-Val A, Sánchez-Gomar I, Eslava-Alcón S, Rosal-Vela A, Jiménez-Palomares M, Doiz-Artázcoz E, Martínez-Torija M, Moreno-Luna R, Olsen JV, Duran-Ruiz MC. Unraveling the differential mechanisms of revascularization promoted by MSCs & ECFCs from adipose tissue or umbilical cord in a murine model of critical limb-threatening ischemia. J Biomed Sci 2024; 31:71. [PMID: 39004727 PMCID: PMC11247736 DOI: 10.1186/s12929-024-01059-w] [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: 01/08/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND Critical limb-threatening ischemia (CLTI) constitutes the most severe manifestation of peripheral artery disease, usually induced by atherosclerosis. CLTI patients suffer from high risk of amputation of the lower extremities and elevated mortality rates, while they have low options for surgical revascularization due to associated comorbidities. Alternatively, cell-based therapeutic strategies represent an effective and safe approach to promote revascularization. However, the variability seen in several factors such as cell combinations or doses applied, have limited their success in clinical trials, being necessary to reach a consensus regarding the optimal "cellular-cocktail" prior further application into the clinic. To achieve so, it is essential to understand the mechanisms by which these cells exert their regenerative properties. Herein, we have evaluated, for the first time, the regenerative and vasculogenic potential of a combination of endothelial colony forming cells (ECFCs) and mesenchymal stem cells (MSCs) isolated from adipose-tissue (AT), compared with ECFCs from umbilical cord blood (CB-ECFCs) and AT-MSCs, in a murine model of CLTI. METHODS Balb-c nude mice (n:32) were distributed in four different groups (n:8/group): control shams, and ischemic mice (after femoral ligation) that received 50 µl of physiological serum alone or a cellular combination of AT-MSCs with either CB-ECFCs or AT-ECFCs. Follow-up of blood flow reperfusion and ischemic symptoms was carried out for 21 days, when mice were sacrificed to evaluate vascular density formation. Moreover, the long-term molecular changes in response to CLTI and both cell combinations were analyzed in a proteomic quantitative approach. RESULTS AT-MSCs with either AT- or CB-ECFCs, promoted a significant recovery of blood flow in CLTI mice 21 days post-ischemia. Besides, they modulated the inflammatory and necrotic related processes, although the CB group presented the slowest ischemic progression along the assay. Moreover, many proteins involved in the repairing mechanisms promoted by cell treatments were identified. CONCLUSIONS The combination of AT-MSCs with AT-ECFCs or with CB-ECFCs promoted similar revascularization in CLTI mice, by restoring blood flow levels, together with the modulation of the inflammatory and necrotic processes, and reduction of muscle damage. The protein changes identified are representative of the molecular mechanisms involved in ECFCs and MSCs-induced revascularization (immune response, vascular repair, muscle regeneration, etc.).
Collapse
Affiliation(s)
- Marta Rojas-Torres
- Biomedicine, Biotechnology and Public Health Department, University of Cadiz, Cadiz, 11002, Spain
- Biomedical Research and Innovation Institute of Cadiz (INiBICA), Cadiz, 11002, Spain
| | - Lucía Beltrán-Camacho
- Cell Biology, Physiology and Immunology Department, University of Cordoba, Cordoba, 14004, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, 14004, Spain
| | - Ana Martínez-Val
- National Center of Cardiovascular Research Carlos III (CNIC), Madrid, 28029, Spain
| | - Ismael Sánchez-Gomar
- Biomedicine, Biotechnology and Public Health Department, University of Cadiz, Cadiz, 11002, Spain
- Biomedical Research and Innovation Institute of Cadiz (INiBICA), Cadiz, 11002, Spain
| | - Sara Eslava-Alcón
- Biomedicine, Biotechnology and Public Health Department, University of Cadiz, Cadiz, 11002, Spain
- Biomedical Research and Innovation Institute of Cadiz (INiBICA), Cadiz, 11002, Spain
| | - Antonio Rosal-Vela
- Biomedicine, Biotechnology and Public Health Department, University of Cadiz, Cadiz, 11002, Spain
- Biomedical Research and Innovation Institute of Cadiz (INiBICA), Cadiz, 11002, Spain
| | - Margarita Jiménez-Palomares
- Biomedicine, Biotechnology and Public Health Department, University of Cadiz, Cadiz, 11002, Spain
- Biomedical Research and Innovation Institute of Cadiz (INiBICA), Cadiz, 11002, Spain
| | - Esther Doiz-Artázcoz
- Angiology & Vascular Surgery Unit, Hospital Universitario Puerta del Mar, Cadiz, Spain
| | - Mario Martínez-Torija
- Pathophysiology and Regenerative Medicine Group, Hospital Nacional de Parapléjicos (SESCAM), Toledo, 45071, Spain
- Nursing department, Hospital Universitario de Toledo (SESCAM), Toledo, 45071, Spain
| | - Rafael Moreno-Luna
- Pathophysiology and Regenerative Medicine Group, Hospital Nacional de Parapléjicos (SESCAM), Toledo, 45071, Spain.
- Cooperative Research Network Orientated to Health Results, Vascular Brain Diseases, RICORS-ICTUS, SESCAM, Toledo, Spain.
| | - Jesper V Olsen
- Novo Nordisk Foundation Center for Protein Research, Copenhagen, Denmark
| | - Ma Carmen Duran-Ruiz
- Biomedicine, Biotechnology and Public Health Department, University of Cadiz, Cadiz, 11002, Spain.
- Biomedical Research and Innovation Institute of Cadiz (INiBICA), Cadiz, 11002, Spain.
- Biomedicine, Biotechnology and Public Health Department, Science Faculty, Cádiz University. Torre Sur. Avda. República Saharaui S/N, Polígono Río San Pedro, Puerto Real, Cádiz, 11519, Spain.
| |
Collapse
|
5
|
Liu Y, Lyons CJ, Ayu C, O’Brien T. Enhancing endothelial colony-forming cells for treating diabetic vascular complications: challenges and clinical prospects. Front Endocrinol (Lausanne) 2024; 15:1396794. [PMID: 39076517 PMCID: PMC11284052 DOI: 10.3389/fendo.2024.1396794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 06/14/2024] [Indexed: 07/31/2024] Open
Abstract
Diabetes mellitus (DM) is a metabolic disease characterized by hyperglycemia, leading to various vascular complications. Accumulating evidence indicates that endothelial colony-forming cells (ECFCs) have attractive prospects for repairing and restoring blood vessels. Thus, ECFCs may be a novel therapeutic option for diabetic patients with vascular complications who require revascularization therapy. However, it has been reported that the function of ECFCs is impaired in DM, which poses challenges for the autologous transplantation of ECFCs. In this review, we summarize the molecular mechanisms that may be responsible for ECFC dysfunction and discuss potential strategies for improving the therapeutic efficacy of ECFCs derived from patients with DM. Finally, we discuss barriers to the use of ECFCs in human studies in light of the fact that there are no published reports using these cells in humans.
Collapse
Affiliation(s)
| | | | | | - Timothy O’Brien
- Regenerative Medicine Institute (REMEDI), Biomedical Sciences Building, University of Galway, Galway, Ireland
| |
Collapse
|
6
|
Salem S, Leach L. Umbilical cord mesenchymal stem cells from gestational diabetes show impaired ability to up-regulate paracellular permeability from sub-endothelial niche. Clin Sci (Lond) 2024; 138:87-102. [PMID: 38168704 PMCID: PMC10794701 DOI: 10.1042/cs20230657] [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: 06/14/2023] [Revised: 12/14/2023] [Accepted: 01/02/2024] [Indexed: 01/05/2024]
Abstract
In vitro studies have shown that Wharton's jelly mesenchymal stem cells (WJ-MSCs) can cross umbilical and uterine endothelial barriers and up-regulate endothelial junctional integrity from sub-endothelial niches. This pericytic behaviour may be lost in pregnancies complicated by gestational diabetes (GDM), where increased vascular permeability and junctional disruption are reported. The aim of the present study was to investigate whether WJ-MSCs isolated from GDM pregnancies displayed any changes in morphology, proliferation, VEGF-A secretion, and their ability to influence paracellular junctional composition and permeability. WJ-MSCs were isolated from human umbilical cords from normal pregnancies (nWJ-MSCs, n=13) and those complicated by GDM (gWJ-MSCs), either diet-controlled (d-GDM, n=13) or metformin-treated (m-GDM, n=9). We recorded that 4-fold more WJ-MSCs migrated from m-GDM, and 2.5-fold from d-GDM cord samples compared with the normal pregnancy. gWJ-MSCs showed a less predominance of spindle-shaped morphology and secreted 3.8-fold more VEGF-A compared with nWJ-MSCs. The number of cells expressing CD105 (Endoglin) was higher in gWJ-MSCs compared with nWJ-MSCs (17%) at P-2. The tracer leakage after 24 h across the HUVEC + gWJ-MSCs bilayer was 22.13% and 11.2% higher in the m-GDM and d-GDM, respectively, HUVEC + nWJ-MSCs. Transfection studies with siRNAs that target Endoglin were performed in n-WJ-MSCs; transfected cells were co-cultured with HUVEC followed by permeability studies and VE-cadherin analyses. Loss of Endoglin also led to increased VEGF-A secretion, increased permeability and affected endothelial stabilization. These results reinforce the pericytic role of nWJ-MSCs to promote vascular repair and the deficient ability of gWJ-MSCs to maintain endothelial barrier integrity.
Collapse
Affiliation(s)
- Samar Salem
- School of Life Sciences, Division of Physiology, Pharmacology and Neuroscience, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, U.K
| | - Lopa Leach
- School of Life Sciences, Division of Physiology, Pharmacology and Neuroscience, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, U.K
| |
Collapse
|
7
|
Smadja DM, Rossi E, Haviari S, Bieche I, Cras A, Gaussem P. Thrombin receptor PAR1 silencing in endothelial colony-forming cells modifies stemness and vasculogenic properties. J Thromb Haemost 2023; 21:3640-3648. [PMID: 37678550 DOI: 10.1016/j.jtha.2023.08.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/09/2023] [Accepted: 08/28/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND The involvement of thrombin receptor PAR1 in blood vessel development has been largely demonstrated in knockout mice; however, its implication in adult mouse angiogenesis seems very moderate. OBJECTIVES We aimed to explore the potential relationships between PAR1, stemness, and angiogenic properties of human endothelial colony-forming cells (ECFCs). METHODS AND RESULTS PAR1 activation on ECFCs using the selective PAR1-activating peptide induced a significant decrease in CD133 expression (RTQ-PCR analysis). In line, silencing of PAR1 gene expression with siRNA increased CD133 mRNA as well as intracellular CD133 protein expression. To confirm the link between CD133 and PAR1, we explored the association between PAR1 and CD133 levels in fast and slow fibroblasts prone to reprogramming. An imbalance between PAR1 and CD133 levels was evidenced, with a decreased expression of PAR1 in fast reprogramming fibroblasts expressing a high CD133 level. Regarding in vitro ECFC angiogenic properties, PAR1 silencing with specific siRNA induced cell proliferation evidenced by the overexpression of Ki67. However, it did not impact migration properties nor ECFC adhesion on smooth muscle cells or human arterial endothelial cells. In a mouse model of hind-limb ischemia, PAR1 silencing in ECFCs significantly increased postischemic revascularization compared to siCtrl-ECFCs along with a significant increase in cutaneous blood flows (P < .0001), microvessel density (P = .02), myofiber regeneration (P < .0001), and human endothelial cell incorporation in muscle (P < .0001). CONCLUSION In conclusion, our work describes for the first time a link between PAR1, stemness, and vasculogenesis in human ECFCs.
Collapse
Affiliation(s)
- David M Smadja
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Paris, France; Université Paris-Cité, INSERM UMR-S 1140, Innovative Therapies in Haemostasis, Paris, France.
| | - Elisa Rossi
- Université Paris-Cité, INSERM UMR-S 1140, Innovative Therapies in Haemostasis, Paris, France
| | - Skerdi Haviari
- Université Paris-Cité, INSERM UMR-S 1137 (IAME), Paris, France; Département Épidémiologie Biostatistiques et Recherche Clinique, AP-HP, Hôpital Bichat, Paris, France
| | | | - Audrey Cras
- Université Paris-Cité, INSERM UMR-S 1140, Innovative Therapies in Haemostasis, Paris, France; Cell Therapy Unit, AP-HP, Saint Louis Hospital, Paris, France
| | - Pascale Gaussem
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Paris, France; Université Paris-Cité, INSERM UMR-S 1140, Innovative Therapies in Haemostasis, Paris, France
| |
Collapse
|
8
|
Rossi E, Bernabeu C. Novel vascular roles of human endoglin in pathophysiology. J Thromb Haemost 2023; 21:2327-2338. [PMID: 37315795 DOI: 10.1016/j.jtha.2023.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/19/2023] [Accepted: 06/02/2023] [Indexed: 06/16/2023]
Abstract
Endoglin, alias CD105, is a human membrane glycoprotein highly expressed in vascular endothelial cells. It is involved in angiogenesis and angiogenesis-related diseases, including the rare vascular pathology known as hereditary hemorrhagic telangiectasia type 1. Although endoglin acts as an accessory receptor for members of the transforming growth factor-β family, in recent years, emerging evidence has shown a novel functional role for this protein beyond the transforming growth factor-β system. In fact, endoglin has been found to be an integrin counterreceptor involved in endothelial cell adhesion processes during pathological inflammatory conditions and primary hemostasis. Furthermore, a circulating form of endoglin, also named as soluble endoglin, whose levels are abnormally increased in different pathological conditions, such as preeclampsia, seems to act as an antagonist of membrane-bound endoglin and as a competitor of the fibrinogen-integrin interaction in platelet-dependent thrombus formation. These studies suggest that membrane-bound endoglin and circulating endoglin are important components involved in vascular homeostasis and hemostasis.
Collapse
Affiliation(s)
- Elisa Rossi
- Université Paris Cité, INSERM U1140, Innovative Therapies in Haemostasis, Paris, France.
| | - Carmelo Bernabeu
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
9
|
Tripathi H, Domingues A, Donahue R, Cras A, Guerin CL, Gao E, Levitan B, Ratajczak MZ, Smadja DM, Abdel-Latif A, Tarhuni WM. Combined Transplantation of Human MSCs and ECFCs Improves Cardiac Function and Decrease Cardiomyocyte Apoptosis After Acute Myocardial Infarction. Stem Cell Rev Rep 2023; 19:573-577. [PMID: 36271311 DOI: 10.1007/s12015-022-10468-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Ischemic heart disease, often caused by an acute myocardial infarction (AMI) is one of the leading causes of morbidity and mortality worldwide. Despite significant advances in medical and procedural therapies, millions of AMI patients progress to develop heart failure every year. METHODS Here, we examine the combination therapy of human mesenchymal stromal cells (MSCs) and endothelial colony-forming cells (ECFCs) to reduce the early ischemic damage (MSCs) and enhance angiogenesis (ECFCs) in a pre-clinical model of acute myocardial infarction. NOD/SCID mice were subjected to AMI followed by transplantation of MSCs and ECFCs either alone or in combination. Cardiomyocyte apoptosis and cardiac functional recovery were assessed in short- and long-term follow-up studies. RESULTS At 1 day after AMI, MSC- and ECFC-treated animals demonstrated significantly lower cardiomyocyte apoptosis compared to vehicle-treated animals. This phenomenon was associated with a significant reduction in infarct size, cardiac fibrosis, and improvement in functional cardiac recovery 4 weeks after AMI. CONCLUSIONS The use of ECFCs, MSCs, and the combination of both cell types reduce cardiomyocyte apoptosis, scar size, and adverse cardiac remodeling, compared to vehicle, in a pre-clinical model of AMI. These results support the use of this combined cell therapy approach in future human studies during the acute phase of ischemic cardiac injury.
Collapse
Affiliation(s)
- Himi Tripathi
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY, USA
| | - Alison Domingues
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA.,Université de Paris, INSERM, Innovative Therapies in Haemostasis, 75006, Paris, France
| | - Renee Donahue
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY, USA
| | - Audrey Cras
- Université de Paris, INSERM, Innovative Therapies in Haemostasis, 75006, Paris, France.,Cell Therapy Department, AP-HP, Hôpital Saint Louis, 75010, Paris, France
| | - Coralie L Guerin
- Université de Paris, INSERM, Innovative Therapies in Haemostasis, 75006, Paris, France.,Curie Institute, Paris, France
| | - Erhe Gao
- The Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Bryana Levitan
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY, USA
| | - Mariusz Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - David M Smadja
- Université de Paris, INSERM, Innovative Therapies in Haemostasis, 75006, Paris, France.,Hematology Department and Biosurgical Research Lab (Carpentier Foundation), AP-HP, Hôpital Européen Georges Pompidou, 75015, Paris, France
| | - Ahmed Abdel-Latif
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY, USA. .,Department of Internal Medicine, Division of Cardiology, University of Michigan, Ann Arbor, MI, USA. .,Division of Cardiovascular Medicine, University of Michigan and the Ann Arbor VA Medical Center, Ann Arbor, MI, USA.
| | - Wadea M Tarhuni
- Canadian Cardiac Research Center, Department of Internal Medicine, Division of Cardiology, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
10
|
Rojas-Torres M, Sánchez-Gomar I, Rosal-Vela A, Beltrán-Camacho L, Eslava-Alcón S, Alonso-Piñeiro JÁ, Martín-Ramírez J, Moreno-Luna R, Durán-Ruiz MC. Assessment of endothelial colony forming cells delivery routes in a murine model of critical limb threatening ischemia using an optimized cell tracking approach. Stem Cell Res Ther 2022; 13:266. [PMID: 35729651 PMCID: PMC9210810 DOI: 10.1186/s13287-022-02943-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/07/2022] [Indexed: 01/15/2023] Open
Abstract
Background Endothelial colony forming cells (ECFCs), alone or in combination with mesenchymal stem cells, have been selected as potential therapeutic candidates for critical limb-threatening ischemia (CLTI), mainly for those patients considered as “no-option,” due to their capability to enhance revascularization and perfusion recovery of ischemic tissues. Nevertheless, prior to translating cell therapy to the clinic, biodistribution assays are required by regulatory guidelines to ensure biosafety as well as to discard undesired systemic translocations. Different approaches, from imaging technologies to qPCR-based methods, are currently applied. Methods In the current study, we have optimized a cell-tracking assay based on DiR fluorescent cell labeling and near-infrared detection for in vivo and ex vivo assays. Briefly, an improved protocol for DiR staining was set up, by incubation of ECFCs with 6.67 µM DiR and intensive washing steps prior cell administration. The minimal signal detected for the residual DiR, remaining after these washes, was considered as a baseline signal to estimate cell amounts correlated to the DiR intensity values registered in vivo. Besides, several assays were also performed to determine any potential effect of DiR over ECFCs functionality. Furthermore, the optimized protocol was applied in combination with qPCR amplification of specific human Alu sequences to assess the final distribution of ECFCs after intramuscular or intravenous administration to a murine model of CLTI. Results The optimized DiR labeling protocol indicated that ECFCs administered intramuscularly remained mainly within the hind limb muscle while cells injected intravenously were found in the spleen, liver and lungs. Conclusion Overall, the combination of DiR labeling and qPCR analysis in biodistribution assays constitutes a highly sensitive approach to systemically track cells in vivo. Thereby, human ECFCs administered intramuscularly to CLTI mice remained locally within the ischemic tissues, while intravenously injected cells were found in several organs. Our data corroborate the need to perform biodistribution assays in order to define specific parameters such as the optimal delivery route for ECFCs before their application into the clinic. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02943-8.
Collapse
Affiliation(s)
- Marta Rojas-Torres
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cádiz, Spain.,Institute of Research and Innovation in Biomedical Sciences of Cádiz (INiBICA), Cádiz, Spain
| | - Ismael Sánchez-Gomar
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cádiz, Spain.,Institute of Research and Innovation in Biomedical Sciences of Cádiz (INiBICA), Cádiz, Spain
| | - Antonio Rosal-Vela
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cádiz, Spain.,Institute of Research and Innovation in Biomedical Sciences of Cádiz (INiBICA), Cádiz, Spain
| | - Lucía Beltrán-Camacho
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cádiz, Spain.,Institute of Research and Innovation in Biomedical Sciences of Cádiz (INiBICA), Cádiz, Spain
| | - Sara Eslava-Alcón
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cádiz, Spain.,Institute of Research and Innovation in Biomedical Sciences of Cádiz (INiBICA), Cádiz, Spain
| | - José Ángel Alonso-Piñeiro
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cádiz, Spain.,Institute of Research and Innovation in Biomedical Sciences of Cádiz (INiBICA), Cádiz, Spain
| | | | - Rafael Moreno-Luna
- Laboratory of Neuroinflammation, Hospital Nacional de Paraplejicos, SESCAM, Toledo, Spain
| | - Mª Carmen Durán-Ruiz
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cádiz, Spain. .,Institute of Research and Innovation in Biomedical Sciences of Cádiz (INiBICA), Cádiz, Spain.
| |
Collapse
|
11
|
Shi H, Zhao Z, Jiang W, Zhu P, Zhou N, Huang X. A Review Into the Insights of the Role of Endothelial Progenitor Cells on Bone Biology. Front Cell Dev Biol 2022; 10:878697. [PMID: 35686054 PMCID: PMC9173585 DOI: 10.3389/fcell.2022.878697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/11/2022] [Indexed: 11/23/2022] Open
Abstract
In addition to its important transport functions, the skeletal system is involved in complex biological activities for the regulation of blood vessels. Endothelial progenitor cells (EPCs), as stem cells of endothelial cells (ECs), possess an effective proliferative capacity and a powerful angiogenic capacity prior to their differentiation. They demonstrate synergistic effects to promote bone regeneration and vascularization more effectively by co-culturing with multiple cells. EPCs demonstrate a significant therapeutic potential for the treatment of various bone diseases by secreting a combination of growth factors, regulating cellular functions, and promoting bone regeneration. In this review, we retrospect the definition and properties of EPCs, their interaction with mesenchymal stem cells, ECs, smooth muscle cells, and immune cells in bone regeneration, vascularization, and immunity, summarizing their mechanism of action and contribution to bone biology. Additionally, we generalized their role and potential mechanisms in the treatment of various bone diseases, possibly indicating their clinical application.
Collapse
Affiliation(s)
- Henglei Shi
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Disease Treatment, Guangxi Clinical Research Center for Craniofacia Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surg Deformity, Nanning, China
| | - Zhenchen Zhao
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Disease Treatment, Guangxi Clinical Research Center for Craniofacia Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surg Deformity, Nanning, China
| | - Weidong Jiang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Disease Treatment, Guangxi Clinical Research Center for Craniofacia Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surg Deformity, Nanning, China
| | - Peiqi Zhu
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Disease Treatment, Guangxi Clinical Research Center for Craniofacia Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surg Deformity, Nanning, China
| | - Nuo Zhou
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Disease Treatment, Guangxi Clinical Research Center for Craniofacia Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surg Deformity, Nanning, China
| | - Xuanping Huang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Disease Treatment, Guangxi Clinical Research Center for Craniofacia Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surg Deformity, Nanning, China
| |
Collapse
|
12
|
Luo Y, Liang F, Wan X, Liu S, Fu L, Mo J, Meng X, Mo Z. Hyaluronic Acid Facilitates Angiogenesis of Endothelial Colony Forming Cell Combining With Mesenchymal Stem Cell via CD44/ MicroRNA-139-5p Pathway. Front Bioeng Biotechnol 2022; 10:794037. [PMID: 35350177 PMCID: PMC8957954 DOI: 10.3389/fbioe.2022.794037] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/19/2022] [Indexed: 12/13/2022] Open
Abstract
Stem cells and progenitor cells have been identified as potential new therapeutic options for severe limb ischemia to induce angiogenesis, and hyaluronic acid (HA) is commonly applied as a biomaterial in tissue engineering. However, the efficiency of HA combined with human umbilical cord blood-derived endothelial colony forming cells (ECFCs) and human umbilical-derived mesenchymal stem cells (MSCs) on angiogenesis is unclear. In the present study, we showed that HA promoted angiogenesis induced by MSCs-ECFCs in Matrigel plugs and promoted blood perfusion of murine ischemic muscles. Laser confocal microscopy revealed that human-derived cells grew into the host vasculature and formed connections, as shown by mouse-specific CD31+/human-specific CD31+ double staining. In vitro assays revealed that HA supported cell proliferation and migration, enhanced CD44 expression and reduced microRNA (miR)-139-5p expression. Further analysis revealed that miR-139-5p expression was negatively regulated by CD44 in ECFCs. Flow cytometry assays showed that HA increased CD31 positive cells proportion in MSC-ECFC and could be reversed by miR-139-5p mimics transfection. Moreover, the improvement of MSC-ECFC proliferation and migration induced by HA could be blocked by upregulation of miR-139-5p expression. In conclusion, HA facilitates angiogenesis of MSCs-ECFCs, and this positive effect be associated with activation of the CD44/miR-139-5p pathway, providing a promising strategy for improving severe limb ischemia.
Collapse
Affiliation(s)
- Yufang Luo
- Department of Endocrinology, Third Xiangya Hospital of Central South University and Diabetic Foot Research Center of Central South University, Changsha, China
| | - Fang Liang
- Department of Endocrinology, Third Xiangya Hospital of Central South University and Diabetic Foot Research Center of Central South University, Changsha, China
| | - Xinxing Wan
- Department of Endocrinology, Third Xiangya Hospital of Central South University and Diabetic Foot Research Center of Central South University, Changsha, China
| | - Shengping Liu
- Department of Endocrinology, Third Xiangya Hospital of Central South University and Diabetic Foot Research Center of Central South University, Changsha, China
| | - Lanfang Fu
- Department of Endocrinology, Haikou People’s Hospital and Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Jiake Mo
- School of Medicine, Hunan Normal University, Changsha, China
| | - Xubiao Meng
- Department of Endocrinology, Haikou People’s Hospital and Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
- *Correspondence: Xubiao Meng, ; Zhaohui Mo,
| | - Zhaohui Mo
- Department of Endocrinology, Third Xiangya Hospital of Central South University and Diabetic Foot Research Center of Central South University, Changsha, China
- *Correspondence: Xubiao Meng, ; Zhaohui Mo,
| |
Collapse
|
13
|
Tian T, Li F, Chen R, Wang Z, Su X, Yang C. Therapeutic Potential of Exosomes Derived From circRNA_0002113 Lacking Mesenchymal Stem Cells in Myocardial Infarction. Front Cell Dev Biol 2022; 9:779524. [PMID: 35127703 PMCID: PMC8807507 DOI: 10.3389/fcell.2021.779524] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 12/23/2021] [Indexed: 12/27/2022] Open
Abstract
Exosomes are participated in the pathogenesis of cardiovascular diseases and can be secreted by mesenchymal stem cells (MSCs). However, the effects of circRNA, delivered by exosomes derived from MSCs, on myocardial injury remain unclear. Hence, this study aims to explore the therapeutic potential of exosomes derived from circRNA_0002113 lacking MSCs in the treatment of myocardial injury in vitro and in vivo. Our results reveal that exosomes derived from circRNA_0002113 lacking MSCs decreased cell apoptosis in anoxia-reoxygenation (A/R) model cells, and reduced myocardial injury by inhibiting nuclear translocation of RUNX1 in vitro and in vivo. Moreover, miR-188-3p, which targets RUNX1 in cardiomyocytes was also found to interact with circRNA_0002113. In conclusion, exosomes derived from circRNA_0002113 lacking MSCs could suppress myocardial infarction by sponging miR-188-3p to regulate RUNX1 nuclear translocation. The circRNA_0002113/miR-188-3p/RUNX1 axis mediated alleviation of apoptosis serves as a novel strategy to treat myocardial I/R injury.
Collapse
Affiliation(s)
- Tiantian Tian
- Hainan Yiling Medical Industry Development Company Ltd., Qionghai, China
- Center for Biological Science and Technology, Beijing Normal University, Zhuhai, China
| | - Feng Li
- Hainan Yiling Medical Industry Development Company Ltd., Qionghai, China
| | - Ruihua Chen
- Hainan Yiling Medical Industry Development Company Ltd., Qionghai, China
| | - Zhiwei Wang
- Hainan Yiling Medical Industry Development Company Ltd., Qionghai, China
| | - Xueming Su
- Hainan Yiling Medical Industry Development Company Ltd., Qionghai, China
- *Correspondence: Chao Yang, ; Xueming Su,
| | - Chao Yang
- Hainan Yiling Medical Industry Development Company Ltd., Qionghai, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, China
- *Correspondence: Chao Yang, ; Xueming Su,
| |
Collapse
|
14
|
Wang X, Jiang H, Guo L, Wang S, Cheng W, Wan L, Zhang Z, Xing L, Zhou Q, Yang X, Han H, Chen X, Wu X. SDF-1 secreted by mesenchymal stem cells promotes the migration of endothelial progenitor cells via CXCR4/PI3K/AKT pathway. J Mol Histol 2021; 52:1155-1164. [PMID: 34642827 DOI: 10.1007/s10735-021-10008-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 08/07/2021] [Indexed: 01/07/2023]
Abstract
Cell-based therapeutics bring great hope in areas of unmet medical needs. Mesenchymal stem cells (MSCs) have been suggested to facilitate neovascularization mainly by paracrine action. Endothelial progenitor cells (EPCs) can migrate to ischemic sites and participate in angiogenesis. The combination cell therapy that includes MSCs and EPCs has a favorable effect on ischemic limbs. However, the mechanism of combination cell therapy remains unclear. Herein, we investigate whether stromal cell-derived factor (SDF)-1 secreted by MSCs contributes to EPC migration to ischemic sites via CXCR4/Phosphoinositide 3-Kinases (PI3K)/protein kinase B (termed as AKT) signaling pathway. First, by a "dual-administration" approach, intramuscular MSC injections were supplemented with intravenous Qdot® 525 labeled-EPC injections in the mouse model of hind limb ischemia. Then, the mechanism of MSC effect on EPC migration was detected by the transwell system, tube-like structure formation assays, western blot assays in vitro. Results showed that the combination delivery of MSCs and EPCs enhanced the incorporation of EPCs into the vasculature and increased the capillary density in mouse ischemic hind limb. The numbers of CXCR4-positive EPCs increased after incubation with MSC-conditioned medium (CM). MSCs contributed to EPC migration and tube-like structure formation, both of which were suppressed by AMD3100 and wortmannin. Phospho-AKT induced by MSC-CM was attenuated when EPCs were pretreated with AMD3100 and wortmannin. In conclusion, we confirmed that MSCs contributes to EPC migration, which is mediated via CXCR4/PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Xiaoyi Wang
- Department of Pediatric Hematology and Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, Henan, China.,Laboratory of Translational Medicine, Medical School of Shihezi University, No. 59 North 2 Road, Shihezi, 832002, Xinjiang, China
| | - Huijiao Jiang
- Laboratory of Translational Medicine, Medical School of Shihezi University, No. 59 North 2 Road, Shihezi, 832002, Xinjiang, China
| | - Lijiao Guo
- Laboratory of Translational Medicine, Medical School of Shihezi University, No. 59 North 2 Road, Shihezi, 832002, Xinjiang, China
| | - Sibo Wang
- Laboratory of Translational Medicine, Medical School of Shihezi University, No. 59 North 2 Road, Shihezi, 832002, Xinjiang, China
| | - Wenzhe Cheng
- Laboratory of Translational Medicine, Medical School of Shihezi University, No. 59 North 2 Road, Shihezi, 832002, Xinjiang, China
| | - Longfei Wan
- Laboratory of Translational Medicine, Medical School of Shihezi University, No. 59 North 2 Road, Shihezi, 832002, Xinjiang, China
| | - Zhongzhou Zhang
- Laboratory of Translational Medicine, Medical School of Shihezi University, No. 59 North 2 Road, Shihezi, 832002, Xinjiang, China
| | - Lihang Xing
- Laboratory of Translational Medicine, Medical School of Shihezi University, No. 59 North 2 Road, Shihezi, 832002, Xinjiang, China
| | - Qing Zhou
- Laboratory of Translational Medicine, Medical School of Shihezi University, No. 59 North 2 Road, Shihezi, 832002, Xinjiang, China
| | - Xiongfeng Yang
- Laboratory of Translational Medicine, Medical School of Shihezi University, No. 59 North 2 Road, Shihezi, 832002, Xinjiang, China
| | - Huanhuan Han
- Laboratory of Translational Medicine, Medical School of Shihezi University, No. 59 North 2 Road, Shihezi, 832002, Xinjiang, China
| | - Xueling Chen
- Department of Immunology, Medical School of Shihezi University, No. 59 North 2 Road, Shihezi, 832002, Xinjiang, China.
| | - Xiangwei Wu
- Laboratory of Translational Medicine, Medical School of Shihezi University, No. 59 North 2 Road, Shihezi, 832002, Xinjiang, China. .,Department of General Surgery, The First Affiliated Hospital of Shihezi University, No. 107 North 2 Road, Shihezi, 832008, Xinjiang, China.
| |
Collapse
|
15
|
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
|
16
|
Paris GC, Azevedo AA, Ferreira AL, Azevedo YMA, Rainho MA, Oliveira GP, Silva KR, Cortez EAC, Stumbo AC, Carvalho SN, de Carvalho L, Thole AA. Therapeutic potential of mesenchymal stem cells in multiple organs affected by COVID-19. Life Sci 2021; 278:119510. [PMID: 33865879 PMCID: PMC8049196 DOI: 10.1016/j.lfs.2021.119510] [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: 03/03/2021] [Revised: 04/05/2021] [Accepted: 04/06/2021] [Indexed: 01/08/2023]
Abstract
Currently, the world has been devastated by an unprecedented pandemic in this century. The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), the agent of coronavirus disease 2019 (COVID-19), has been causing disorders, dysfunction and morphophysiological alterations in multiple organs as the disease evolves. There is a great scientific community effort to obtain a therapy capable of reaching the multiple affected organs in order to contribute for tissue repair and regeneration. In this regard, mesenchymal stem cells (MSCs) have emerged as potential candidates concerning the promotion of beneficial actions at different stages of COVID-19. MSCs are promising due to the observed therapeutic effects in respiratory preclinical models, as well as in cardiac, vascular, renal and nervous system models. Their immunomodulatory properties and secretion of paracrine mediators, such as cytokines, chemokines, growth factors and extracellular vesicles allow for long range tissue modulation and, particularly, blood-brain barrier crossing. This review focuses on SARS-CoV-2 impact to lungs, kidneys, heart, vasculature and central nervous system while discussing promising MSC's therapeutic mechanisms in each tissue. In addition, MSC's therapeutic effects in high-risk groups for COVID-19, such as obese, diabetic and hypertensive patients are also explored.
Collapse
Affiliation(s)
- Gustavo C Paris
- LPCT - Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Aline A Azevedo
- LPCT - Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Adriana L Ferreira
- LPCT - Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Yanca M A Azevedo
- LPCT - Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Mateus A Rainho
- LPCT - Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Genilza P Oliveira
- LPCT - Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Karina R Silva
- LPCT - Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Erika A C Cortez
- LPCT - Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Ana C Stumbo
- LPCT - Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Simone N Carvalho
- LPCT - Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Lais de Carvalho
- LPCT - Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Alessandra A Thole
- LPCT - Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
17
|
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
|
18
|
Age-Dependent Dysregulation of Muscle Vasculature and Blood Flow Recovery after Hindlimb Ischemia in the mdx Model of Duchenne Muscular Dystrophy. Biomedicines 2021; 9:biomedicines9050481. [PMID: 33925757 PMCID: PMC8145677 DOI: 10.3390/biomedicines9050481] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/23/2021] [Indexed: 02/04/2023] Open
Abstract
Duchenne muscular dystrophy (DMD), caused by a lack of functional dystrophin, is characterized by progressive muscle degeneration. Interestingly, dystrophin is also expressed in endothelial cells (ECs), and insufficient angiogenesis has already been hypothesized to contribute to DMD pathology, however, its status in mdx mice, a model of DMD, is still not fully clear. Our study aimed to reveal angiogenesis-related alterations in skeletal muscles of mdx mice compared to wild-type (WT) counterparts. By investigating 6- and 12-week-old mice, we sought to verify if those changes are age-dependent. We utilized a broad spectrum of methods ranging from gene expression analysis, flow cytometry, and immunofluorescence imaging to determine the level of angiogenic markers and to assess muscle blood vessel abundance. Finally, we implemented the hindlimb ischemia (HLI) model, more biologically relevant in the context of functional studies evaluating angiogenesis/arteriogenesis processes. We demonstrated that both 6- and 12-week-old dystrophic mice exhibited dysregulation of several angiogenic factors, including decreased vascular endothelial growth factor A (VEGF) in different muscle types. Nonetheless, in younger, 6-week-old mdx animals, neither the abundance of CD31+α-SMA+ double-positive blood vessels nor basal blood flow and its restoration after HLI was affected. In 12-week-old mdx mice, although a higher number of CD31+α-SMA+ double-positive blood vessels and an increased percentage of skeletal muscle ECs were found, the abundance of pericytes was diminished, and blood flow was reduced. Moreover, impeded perfusion recovery after HLI associated with a blunted inflammatory and regenerative response was evident in 12-week-old dystrophic mice. Hence, our results reinforce the hypothesis of age-dependent angiogenic dysfunction in dystrophic mice. In conclusion, we suggest that older mdx mice constitute an appropriate model for preclinical studies evaluating the effectiveness of vascular-based therapies aimed at the restoration of functional angiogenesis to mitigate DMD severity.
Collapse
|
19
|
Abstract
SARS-CoV-2 viruses are positive single-stranded RNA viruses, whose infection can be asymptomatic or lead to the coronavirus disease 2019 (Covid-19). Covid-19 is a respiratory infection with a significant impact on the hematopoietic system and hemostasis leading to several cardiovascular complications. Hematologic consequences of this new infection allowed medical community to start new treatment approaches concerning infection going from targeted anti-inflammatory drugs to anticoagulation or stem cell therapies. A better understanding of Covid-19 pathophysiology, in particular hematological disorders, will help to choose appropriate treatment strategies.
Collapse
|
20
|
Current Status of Angiogenic Cell Therapy and Related Strategies Applied in Critical Limb Ischemia. Int J Mol Sci 2021; 22:ijms22052335. [PMID: 33652743 PMCID: PMC7956816 DOI: 10.3390/ijms22052335] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/20/2021] [Accepted: 02/23/2021] [Indexed: 02/06/2023] Open
Abstract
Critical limb ischemia (CLI) constitutes the most severe form of peripheral arterial disease (PAD), it is characterized by progressive blockade of arterial vessels, commonly correlated to atherosclerosis. Currently, revascularization strategies (bypass grafting, angioplasty) remain the first option for CLI patients, although less than 45% of them are eligible for surgical intervention mainly due to associated comorbidities. Moreover, patients usually require amputation in the short-term. Angiogenic cell therapy has arisen as a promising alternative for these "no-option" patients, with many studies demonstrating the potential of stem cells to enhance revascularization by promoting vessel formation and blood flow recovery in ischemic tissues. Herein, we provide an overview of studies focused on the use of angiogenic cell therapies in CLI in the last years, from approaches testing different cell types in animal/pre-clinical models of CLI, to the clinical trials currently under evaluation. Furthermore, recent alternatives related to stem cell therapies such as the use of secretomes, exosomes, or even microRNA, will be also described.
Collapse
|
21
|
Shi W, Xin Q, Yuan R, Yuan Y, Cong W, Chen K. Neovascularization: The Main Mechanism of MSCs in Ischemic Heart Disease Therapy. Front Cardiovasc Med 2021; 8:633300. [PMID: 33575274 PMCID: PMC7870695 DOI: 10.3389/fcvm.2021.633300] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 01/05/2021] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cell (MSC) transplantation after myocardial infarction (MI) has been shown to effectively limit the infarct area in numerous clinical and preclinical studies. However, the primary mechanism associated with this activity in MSC transplantation therapy remains unclear. Blood supply is fundamental for the survival of myocardial tissue, and the formation of an efficient vascular network is a prerequisite for blood flow. The paracrine function of MSCs, which is throughout the neovascularization process, including MSC mobilization, migration, homing, adhesion and retention, regulates angiogenesis and vasculogenesis through existing endothelial cells (ECs) and endothelial progenitor cells (EPCs). Additionally, MSCs have the ability to differentiate into multiple cell lineages and can be mobilized and migrate to ischemic tissue to differentiate into ECs, pericytes and smooth muscle cells in some degree, which are necessary components of blood vessels. These characteristics of MSCs support the view that these cells improve ischemic myocardium through angiogenesis and vasculogenesis. In this review, the results of recent clinical and preclinical studies are discussed to illustrate the processes and mechanisms of neovascularization in ischemic heart disease.
Collapse
Affiliation(s)
- Weili Shi
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Qiqi Xin
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Rong Yuan
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Yahui Yuan
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Weihong Cong
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Keji Chen
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| |
Collapse
|
22
|
Gendron N, Rosa M, Blandinieres A, Sottejeau Y, Rossi E, Van Belle E, Idelcadi S, Lecourt S, Vincentelli A, Cras A, Jashari R, Chocron R, Baudouin Y, Pamart T, Bièche I, Nevo N, Cholley B, Rancic J, Staels B, Gaussem P, Dupont A, Carpentier A, Susen S, Smadja DM. Human Aortic Valve Interstitial Cells Display Proangiogenic Properties During Calcific Aortic Valve Disease. Arterioscler Thromb Vasc Biol 2021; 41:415-429. [PMID: 33147990 DOI: 10.1161/atvbaha.120.314287] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVE The study's aim was to analyze the capacity of human valve interstitial cells (VICs) to participate in aortic valve angiogenesis. Approach and Results: VICs were isolated from human aortic valves obtained after surgery for calcific aortic valve disease and from normal aortic valves unsuitable for grafting (control VICs). We examined VIC in vitro and in vivo potential to differentiate in endothelial and perivascular lineages. VIC paracrine effect was also examined on human endothelial colony-forming cells. A pathological VIC (VICp) mesenchymal-like phenotype was confirmed by CD90+/CD73+/CD44+ expression and multipotent-like differentiation ability. When VICp were cocultured with endothelial colony-forming cells, they formed microvessels by differentiating into perivascular cells both in vivo and in vitro. VICp and control VIC conditioned media were compared using serial ELISA regarding quantification of endothelial and angiogenic factors. Higher expression of VEGF (vascular endothelial growth factor)-A was observed at the protein level in VICp-conditioned media and confirmed at the mRNA level in VICp compared with control VIC. Conditioned media from VICp induced in vitro a significant increase in endothelial colony-forming cell proliferation, migration, and sprouting compared with conditioned media from control VIC. These effects were inhibited by blocking VEGF-A with blocking antibody or siRNA approach, confirming VICp involvement in angiogenesis by a VEGF-A dependent mechanism. CONCLUSIONS We provide here the first proof of an angiogenic potential of human VICs isolated from patients with calcific aortic valve disease. These results point to a novel function of VICp in valve vascularization during calcific aortic valve disease, with a perivascular differentiation ability and a VEGF-A paracrine effect. Targeting perivascular differentiation and VEGF-A to slow calcific aortic valve disease progression warrants further investigation.
Collapse
Affiliation(s)
- Nicolas Gendron
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, France (N.G., A.B., E.R., S.I., S.L., A. Cras, N.N., J.R., P.G., D.M.S.)
- Hematology Department and Biosurgical Research Lab (Carpentier Foundation) (N.G., A.B., E.R., S.L., N.N., J.R., P.G., D.M.S.), AH-HP, Georges Pompidou European Hospital, France
| | - Mickael Rosa
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, France (M.R., Y.S., E.V.B., A.V., T.P., B.S., A.D., S.S.)
| | - Adeline Blandinieres
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, France (N.G., A.B., E.R., S.I., S.L., A. Cras, N.N., J.R., P.G., D.M.S.)
- Hematology Department and Biosurgical Research Lab (Carpentier Foundation) (N.G., A.B., E.R., S.L., N.N., J.R., P.G., D.M.S.), AH-HP, Georges Pompidou European Hospital, France
| | - Yoann Sottejeau
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, France (M.R., Y.S., E.V.B., A.V., T.P., B.S., A.D., S.S.)
| | - Elisa Rossi
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, France (N.G., A.B., E.R., S.I., S.L., A. Cras, N.N., J.R., P.G., D.M.S.)
- Hematology Department and Biosurgical Research Lab (Carpentier Foundation) (N.G., A.B., E.R., S.L., N.N., J.R., P.G., D.M.S.), AH-HP, Georges Pompidou European Hospital, France
| | - Eric Van Belle
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, France (M.R., Y.S., E.V.B., A.V., T.P., B.S., A.D., S.S.)
| | - Salim Idelcadi
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, France (N.G., A.B., E.R., S.I., S.L., A. Cras, N.N., J.R., P.G., D.M.S.)
- Department of Anesthesia and Intensive Care and Biosurgical Research Lab (Carpentier Foundation) (S.I., B.C.), AH-HP, Georges Pompidou European Hospital, France
| | - Séverine Lecourt
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, France (N.G., A.B., E.R., S.I., S.L., A. Cras, N.N., J.R., P.G., D.M.S.)
- Hematology Department and Biosurgical Research Lab (Carpentier Foundation) (N.G., A.B., E.R., S.L., N.N., J.R., P.G., D.M.S.), AH-HP, Georges Pompidou European Hospital, France
| | - André Vincentelli
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, France (M.R., Y.S., E.V.B., A.V., T.P., B.S., A.D., S.S.)
| | - Audrey Cras
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, France (N.G., A.B., E.R., S.I., S.L., A. Cras, N.N., J.R., P.G., D.M.S.)
- Cell therapy Department, AH-HP, Saint Louis Hospital, Paris, France (A. Cras)
| | - Ramadan Jashari
- European Homograft Bank, Clinic Saint Jean, Brussels, Belgium (R.J.)
| | - Richard Chocron
- Emergency Medicine Department (R.C.), AH-HP, Georges Pompidou European Hospital, France
- Université de Paris, PARCC, INSERM, France (R.C.)
| | - Yaël Baudouin
- Hematology Department, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France (Y.B.)
| | - Thibault Pamart
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, France (M.R., Y.S., E.V.B., A.V., T.P., B.S., A.D., S.S.)
| | - Ivan Bièche
- Department of Genetics, Pharmacogenomics Unit, Institut Curie, Paris, France (I.B.)
| | - Nathalie Nevo
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, France (N.G., A.B., E.R., S.I., S.L., A. Cras, N.N., J.R., P.G., D.M.S.)
- Hematology Department and Biosurgical Research Lab (Carpentier Foundation) (N.G., A.B., E.R., S.L., N.N., J.R., P.G., D.M.S.), AH-HP, Georges Pompidou European Hospital, France
| | - Bernard Cholley
- Department of Anesthesia and Intensive Care and Biosurgical Research Lab (Carpentier Foundation) (S.I., B.C.), AH-HP, Georges Pompidou European Hospital, France
| | - Jeanne Rancic
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, France (N.G., A.B., E.R., S.I., S.L., A. Cras, N.N., J.R., P.G., D.M.S.)
- Hematology Department and Biosurgical Research Lab (Carpentier Foundation) (N.G., A.B., E.R., S.L., N.N., J.R., P.G., D.M.S.), AH-HP, Georges Pompidou European Hospital, France
| | - Bart Staels
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, France (M.R., Y.S., E.V.B., A.V., T.P., B.S., A.D., S.S.)
| | - Pascale Gaussem
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, France (N.G., A.B., E.R., S.I., S.L., A. Cras, N.N., J.R., P.G., D.M.S.)
- Hematology Department and Biosurgical Research Lab (Carpentier Foundation) (N.G., A.B., E.R., S.L., N.N., J.R., P.G., D.M.S.), AH-HP, Georges Pompidou European Hospital, France
| | - Annabelle Dupont
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, France (M.R., Y.S., E.V.B., A.V., T.P., B.S., A.D., S.S.)
| | - Alain Carpentier
- Université de Paris, Biosurgical Research Lab (Carpentier Foundation) (A. Carpentier), AH-HP, Georges Pompidou European Hospital, France
| | - Sophie Susen
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, France (M.R., Y.S., E.V.B., A.V., T.P., B.S., A.D., S.S.)
| | - David M Smadja
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, France (N.G., A.B., E.R., S.I., S.L., A. Cras, N.N., J.R., P.G., D.M.S.)
- Hematology Department and Biosurgical Research Lab (Carpentier Foundation) (N.G., A.B., E.R., S.L., N.N., J.R., P.G., D.M.S.), AH-HP, Georges Pompidou European Hospital, France
| |
Collapse
|
23
|
Reddy LVK, Murugan D, Mullick M, Begum Moghal ET, Sen D. Recent Approaches for Angiogenesis in Search of Successful Tissue Engineering and Regeneration. Curr Stem Cell Res Ther 2020; 15:111-134. [PMID: 31682212 DOI: 10.2174/1574888x14666191104151928] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/28/2019] [Accepted: 08/06/2019] [Indexed: 02/06/2023]
Abstract
Angiogenesis plays a central role in human physiology from reproduction and fetal development to wound healing and tissue repair/regeneration. Clinically relevant therapies are needed for promoting angiogenesis in order to supply oxygen and nutrients after transplantation, thus relieving the symptoms of ischemia. Increase in angiogenesis can lead to the restoration of damaged tissues, thereby leading the way for successful tissue regeneration. Tissue regeneration is a broad field that has shown the convergence of various interdisciplinary fields, wherein living cells in conjugation with biomaterials have been tried and tested on to the human body. Although there is a prevalence of various approaches that hypothesize enhanced tissue regeneration via angiogenesis, none of them have been successful in gaining clinical relevance. Hence, the current review summarizes the recent cell-based and cell free (exosomes, extracellular vesicles, micro-RNAs) therapies, gene and biomaterial-based approaches that have been used for angiogenesis-mediated tissue regeneration and have been applied in treating disease models like ischemic heart, brain stroke, bone defects and corneal defects. This review also puts forward a concise report of the pre-clinical and clinical studies that have been performed so far; thereby presenting the credible impact of the development of biomaterials and their 3D concepts in the field of tissue engineering and regeneration, which would lead to the probable ways for heralding the successful future of angiogenesis-mediated approaches in the greater perspective of tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Lekkala Vinod Kumar Reddy
- Cellular and Molecular Therapeutics Laboratory, Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Durai Murugan
- Cellular and Molecular Therapeutics Laboratory, Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Madhubanti Mullick
- Cellular and Molecular Therapeutics Laboratory, Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Erfath Thanjeem Begum Moghal
- Cellular and Molecular Therapeutics Laboratory, Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Dwaipayan Sen
- Cellular and Molecular Therapeutics Laboratory, Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India.,University of Georgia, Athens, GA, United States
| |
Collapse
|
24
|
Combined Transplantation of Mesenchymal Stem Cells and Endothelial Colony-Forming Cells Accelerates Refractory Diabetic Foot Ulcer Healing. Stem Cells Int 2020; 2020:8863649. [PMID: 33061991 PMCID: PMC7545465 DOI: 10.1155/2020/8863649] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/19/2020] [Accepted: 08/26/2020] [Indexed: 12/17/2022] Open
Abstract
Background This study is aimed at investigating the effect of combined transplantation of umbilical cord mesenchymal stem cells (UCMSCs) and umbilical cord blood-derived endothelial colony-forming cells (ECFCs) on diabetic foot ulcer healing and at providing a novel therapy for chronic diabetic foot ulcer. Methods We reported the treatment of refractory diabetic foot ulcers in twelve patients. Among them, five patients had two or more wounds; thus, one wound in the same patient was treated with cell injection, and other wounds were regarded as self-controls. The remaining seven patients had only one wound; therefore, the difference between the area of wound before and after treatment was estimated. The UCMSCs and ECFCs were injected into the wound along with topically applied hyaluronic acid (HA). Results In this report, we compared the healing rate of multiple separate wounds in the same foot of the same patient: one treated with cell injection combined with topically applied HA-based hydrogel and was later covered by the hydrocolloid dressings, while the self-control wounds were only treated with conventional therapy and covered by the hydrocolloid dressings. The wound underwent cell injection showed accelerated healing in comparison to control wound within the first week after treatment. In other diabetic patients with only one refractory wound, the healing rate after cell transplantation was significantly faster than that before injection. Two large wounds healed without needing skin grafts after combination therapy of cell injection and HA. After four weeks of combination treatment, wound closure was reached in six patients, and the wounds of the other six patients were significantly reduced in size. Conclusions Our study suggests that the combination of UCMSCs, ECFCs, and HA can safely synergize the accelerated healing of refractory diabetic foot ulcers.
Collapse
|
25
|
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: 30] [Impact Index Per Article: 6.0] [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
|
26
|
Abbasi N, Lee RSB, Ivanovski S, Love RM, Hamlet S. In vivo bone regeneration assessment of offset and gradient melt electrowritten (MEW) PCL scaffolds. Biomater Res 2020; 24:17. [PMID: 33014414 PMCID: PMC7529514 DOI: 10.1186/s40824-020-00196-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 09/21/2020] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Biomaterial-based bone tissue engineering represents a promising solution to overcome reduced residual bone volume. It has been previously demonstrated that gradient and offset architectures of three-dimensional melt electrowritten poly-caprolactone (PCL) scaffolds could successfully direct osteoblast cells differentiation toward an osteogenic lineage, resulting in mineralization. The aim of this study was therefore to evaluate the in vivo osteoconductive capacity of PCL scaffolds with these different architectures. METHODS Five different calcium phosphate (CaP) coated melt electrowritten PCL pore sized scaffolds: 250 μm and 500 μm, 500 μm with 50% fibre offset (offset.50.50), tri layer gradient 250-500-750 μm (grad.250top) and 750-500-250 μm (grad.750top) were implanted into rodent critical-sized calvarial defects. Empty defects were used as a control. After 4 and 8 weeks of healing, the new bone was assessed by micro-computed tomography and immunohistochemistry. RESULTS Significantly more newly formed bone was shown in the grad.250top scaffold 8 weeks post-implantation. Histological investigation also showed that soft tissue was replaced with newly formed bone and fully covered the grad.250top scaffold. While, the bone healing did not happen completely in the 250 μm, offset.50.50 scaffolds and blank calvaria defects following 8 weeks of implantation. Immunohistochemical analysis showed the expression of osteogenic markers was present in all scaffold groups at both time points. The mineralization marker Osteocalcin was detected with the highest intensity in the grad.250top and 500 μm scaffolds. Moreover, the expression of the endothelial markers showed that robust angiogenesis was involved in the repair process. CONCLUSIONS These results suggest that the gradient pore size structure provides superior conditions for bone regeneration.
Collapse
Affiliation(s)
- Naghmeh Abbasi
- School of Dentistry and Oral Health, Griffith University, Gold Coast Campus, Southport, Queensland 4215 Australia
- Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Southport, Queensland 4215 Australia
| | - Ryan S. B. Lee
- School of Dentistry and Oral Health, Griffith University, Gold Coast Campus, Southport, Queensland 4215 Australia
- School of Dentistry, University of Queensland, Herston Campus, Herston, Queensland 4006 Australia
| | - Saso Ivanovski
- School of Dentistry, University of Queensland, Herston Campus, Herston, Queensland 4006 Australia
| | - Robert M. Love
- School of Dentistry and Oral Health, Griffith University, Gold Coast Campus, Southport, Queensland 4215 Australia
| | - Stephen Hamlet
- School of Dentistry and Oral Health, Griffith University, Gold Coast Campus, Southport, Queensland 4215 Australia
- Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Southport, Queensland 4215 Australia
| |
Collapse
|
27
|
Blervaque L, Pomiès P, Rossi E, Catteau M, Blandinières A, Passerieux E, Blaquière M, Ayoub B, Molinari N, Mercier J, Perez-Martin A, Marchi N, Smadja DM, Hayot M, Gouzi F. COPD is deleterious for pericytes: implications during training-induced angiogenesis in skeletal muscle. Am J Physiol Heart Circ Physiol 2020; 319:H1142-H1151. [PMID: 32986960 DOI: 10.1152/ajpheart.00306.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Improvements in skeletal muscle endurance and oxygen uptake are blunted in patients with chronic obstructive pulmonary disease (COPD), possibly because of a limitation in the muscle capillary oxygen supply. Pericytes are critical for capillary blood flow adaptation during angiogenesis but may be impaired by COPD systemic effects, which are mediated by circulating factors. This study compared the pericyte coverage of muscle capillaries in response to 10 wk of exercise training in patients with COPD and sedentary healthy subjects (SHS). Fourteen patients with COPD were compared with seven matched SHS. SHS trained at moderate intensity corresponding to an individualized moderate-intensity patient with COPD trained at the same relative (%V̇o2: COPD-RI) or absolute (mL·min-1·kg-1: COPD-AI) intensity as SHS. Capillary-to-fiber ratio (C/F) and NG2+ pericyte coverage were assessed from vastus lateralis muscle biopsies, before and after 5 and 10 wk of training. We also tested in vitro the effect of COPD and SHS serum on pericyte morphology and mesenchymal stem cell (MSC) differentiation into pericytes. SHS showed greater improvement in aerobic capacity (V̇o2VT) than both patients with COPD-RI and patients with COPD-AI (Group × Time: P = 0.004). Despite a preserved increase in the C/F ratio, NG2+ pericyte coverage did not increase in patients with COPD in response to training, contrary to SHS (Group × Time: P = 0.011). Conversely to SHS serum, COPD serum altered pericyte morphology (P < 0.001) and drastically reduced MSC differentiation into pericytes (P < 0.001). Both functional capacities and pericyte coverage responses to exercise training are blunted in patients with COPD. We also provide direct evidence of the deleterious effect of COPD circulating factors on pericyte morphology and differentiation.NEW & NOTEWORTHY This work confirms the previously reported impairment in the functional response to exercise training of patients with COPD compared with SHS. Moreover, it shows for the first time that pericyte coverage of the skeletal capillaries is drastically reduced in patients with COPD compared with SHS during training-induced angiogenesis. Finally, it provides experimental evidence that circulating factors are involved in the impaired pericyte coverage of patients with COPD.
Collapse
Affiliation(s)
- Léo Blervaque
- PhyMedExp, INSERM-CNRS-Montpellier University, Montpellier, France
| | - Pascal Pomiès
- PhyMedExp, INSERM-CNRS-Montpellier University, Montpellier, France
| | - Elisa Rossi
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, Paris, France
| | - Matthias Catteau
- PhyMedExp, INSERM-CNRS-Montpellier University, Montpellier, France
| | - Adeline Blandinières
- Service d'Hématologie et Laboratoire de Recherches Biochirugicales (Fondation Carpentier), AH-HP, Georges Pompidou European Hospital, Paris, France
| | | | - Marine Blaquière
- Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS-U1191 INSERM, University of Montpellier), Montpellier, France
| | - Bronia Ayoub
- PhyMedExp, INSERM-CNRS-Montpellier University, CHU Montpellier, Montpellier, France
| | - Nicolas Molinari
- IMAG, CNRS, Montpellier University, CHU Montpellier, Montpellier, France
| | - Jacques Mercier
- PhyMedExp, INSERM-CNRS-Montpellier University, CHU Montpellier, Montpellier, France
| | - Antonia Perez-Martin
- Vascular Medicine Department and Laboratory, CHU Nîmes and EA2992 Research Unit, Montpellier University, Nimes, France
| | - Nicola Marchi
- Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS-U1191 INSERM, University of Montpellier), Montpellier, France
| | - David M Smadja
- Service d'Hématologie et Laboratoire de Recherches Biochirugicales (Fondation Carpentier), AH-HP, Georges Pompidou European Hospital, Paris, France
| | - Maurice Hayot
- PhyMedExp, INSERM-CNRS-Montpellier University, CHU Montpellier, Montpellier, France
| | - Fares Gouzi
- PhyMedExp, INSERM-CNRS-Montpellier University, CHU Montpellier, Montpellier, France
| |
Collapse
|
28
|
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: 31] [Impact Index Per Article: 6.2] [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
|
29
|
Lyons CJ, O’Brien T. The Functionality of Endothelial-Colony-Forming Cells from Patients with Diabetes Mellitus. Cells 2020; 9:cells9071731. [PMID: 32698397 PMCID: PMC7408543 DOI: 10.3390/cells9071731] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 07/15/2020] [Accepted: 07/18/2020] [Indexed: 12/18/2022] Open
Abstract
Endothelial-colony-forming cells (ECFCs) are a population of progenitor cells which have demonstrated promising angiogenic potential both in vitro and in vivo. However, ECFCs from diabetic patients have been shown to be dysfunctional compared to ECFCs from healthy donors. Diabetes mellitus itself presents with many vascular co-morbidities and it has been hypothesized that ECFCs may be a potential cell therapy option to promote revascularisation in these disorders. While an allogeneic cell therapy approach would offer the potential of an ‘off the shelf’ therapeutic product, to date little research has been carried out on umbilical cord-ECFCs in diabetic models. Alternatively, autologous cell therapy using peripheral blood-ECFCs allows the development of a personalised therapeutic approach to medicine; however, autologous diabetic ECFCs are dysfunctional and need to be repaired so they can effectively treat diabetic co-morbidities. Many different groups have modified autologous diabetic ECFCs to improve their function using a variety of methods including pre-treatment with different factors or with genetic modification. While the in vitro and in vivo data from the literature is promising, no ECFC therapy has proceeded to clinical trials to date, indicating that more research is needed for a potential ECFC therapy in the future to treat diabetic complications.
Collapse
|
30
|
Bachelier K, Bergholz C, Friedrich EB. Differentiation potential and functional properties of a CD34‑CD133+ subpopulation of endothelial progenitor cells. Mol Med Rep 2019; 21:501-507. [PMID: 31746407 DOI: 10.3892/mmr.2019.10831] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 09/02/2019] [Indexed: 11/05/2022] Open
Abstract
Endothelial progenitor cells (EPCs) promote angiogenesis and play an important role in myocardial and vascular repair after ischemia and infarction. EPCs consist of different subpopulations including CD34‑CD133+ EPCs, which are precursors of more mature CD34+CD133+ EPCs and functionally more active in terms of homing and endothelial regeneration. In the present study we analyzed the functional and differentiation abilities of CD34‑CD133+ EPCs. Isolation of EPC populations (CD34+CD133+, CD34‑CD133+) were performed by specific multi‑step magnetic depletion. After specific stimulation a significant higher adhesive and migrative capacity of CD34‑CD133+ cells could be detected compared to CD34+CD133+ cells (P<0.001, respectively). Next to this finding, not only significantly higher rates of proliferation (P<0.005) were detected among CD34‑CD133+ cells, but also a higher potential of cell‑differentiation capacity into other cell types. Next to a significant increase of CD34‑CD133+ EPCs differentiating into a fibroblast cell‑type (P<0.001), an enhancement into a hepatocytic cell‑type (P=0.033) and a neural cell‑type (P=0.016) could be measured in contrast to CD34+CD133+ cells. On the other hand, there was no significant difference in differentiation into a cardiomyocyte cell‑type between these EPC subpopulations (P=0.053). These results demonstrate that EPC subpopulations vary in their functional abilities and, to different degrees, have the capacity to transdifferentiate into unrelated cell‑types such as fibroblasts, hepatocytes, and neurocytes. This indicates that CD34‑CD133+ cells are more pluripotent compared to the CD34+CD133+ EPC subset, which may have important consequences for the therapy of vascular diseases.
Collapse
Affiliation(s)
- Katrin Bachelier
- Clinic of Internal Medicine III, Cardiology, Angiology and Intensive Care, University of The Saarland, D‑66421 Homburg/Saar, Germany
| | - Carolin Bergholz
- Clinic of Internal Medicine III, Cardiology, Angiology and Intensive Care, University of The Saarland, D‑66421 Homburg/Saar, Germany
| | - Erik B Friedrich
- Clinic of Internal Medicine III, Cardiology, Angiology and Intensive Care, University of The Saarland, D‑66421 Homburg/Saar, Germany
| |
Collapse
|
31
|
Chitlac-coated Thermosets Enhance Osteogenesis and Angiogenesis in a Co-culture of Dental Pulp Stem Cells and Endothelial Cells. NANOMATERIALS 2019; 9:nano9070928. [PMID: 31252684 PMCID: PMC6669739 DOI: 10.3390/nano9070928] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/25/2019] [Accepted: 06/26/2019] [Indexed: 12/14/2022]
Abstract
Dental pulp stem cells (DPSCs) represent a population of stem cells which could be useful in oral and maxillofacial reconstruction. They are part of the periendothelial niche, where their crosstalk with endothelial cells is crucial in the cellular response to biomaterials used for dental restorations. DPSCs and the endothelial cell line EA.hy926 were co-cultured in the presence of Chitlac-coated thermosets in culture conditions inducing, in turn, osteogenic or angiogenic differentiation. Cell proliferation was evaluated by 3-[4,5-dimethyl-thiazol-2-yl-]-2,5-diphenyl tetrazolium bromide (MTT) assay. DPSC differentiation was assessed by measuring Alkaline Phosphtase (ALP) activity and Alizarin Red S staining, while the formation of new vessels was monitored by optical microscopy. The IL-6 and PGE2 production was evaluated as well. When cultured together, the proliferation is increased, as is the DPSC osteogenic differentiation and EA.hy926 vessel formation. The presence of thermosets appears either not to disturb the system balance or even to improve the osteogenic and angiogenic differentiation. Chitlac-coated thermosets confirm their biocompatibility in the present co-culture model, being capable of improving the differentiation of both cell types. Furthermore, the assessed co-culture appears to be a useful tool to investigate cell response toward newly synthesized or commercially available biomaterials, as well as to evaluate their engraftment potential in restorative dentistry.
Collapse
|
32
|
Rossi E, Bernabeu C, Smadja DM. Endoglin as an Adhesion Molecule in Mature and Progenitor Endothelial Cells: A Function Beyond TGF-β. Front Med (Lausanne) 2019; 6:10. [PMID: 30761306 PMCID: PMC6363663 DOI: 10.3389/fmed.2019.00010] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 01/14/2019] [Indexed: 12/13/2022] Open
Abstract
Endoglin (ENG) is a transmembrane glycoprotein expressed on endothelial cells that functions as a co-receptor for several ligands of the transforming growth factor beta (TGF-β) family. ENG is also a recognized marker of angiogenesis and mutations in the endoglin gene are responsible for Hereditary Hemorrhagic Telangiectasia (HHT) type 1, a vascular disease characterized by defective angiogenesis, arteriovenous malformations, telangiectasia, and epistaxis. In addition to its involvement in the TGF-β family signaling pathways, several lines of evidence suggest that the extracellular domain of ENG has a role in integrin-mediated cell adhesion via its RGD motif. Indeed, we have described a role for endothelial ENG in leukocyte trafficking and extravasation via its binding to leukocyte integrins. We have also found that ENG is involved in vasculogenic properties of endothelial progenitor cells known as endothelial colony forming cells (ECFCs). Moreover, the binding of endothelial ENG to platelet integrins regulate the resistance to shear during platelet-endothelium interactions under inflammatory conditions. Because of the need for more effective treatments in HHT and the involvement of ENG in angiogenesis, current studies are aimed at identifying novel biological functions of ENG which could serve as a therapeutic target. This review focuses on the interaction between ENG and integrins with the aim to better understand the role of this protein in blood vessel formation driven by progenitor and mature endothelial cells.
Collapse
Affiliation(s)
- Elisa Rossi
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Inserm UMR-S1140, Paris, France
| | - Carmelo Bernabeu
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain
| | - David M Smadja
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Inserm UMR-S1140, Paris, France.,Department of Hematology, AP-HP, Hôpital Européen Georges Pompidou, Paris, France.,Laboratory of Biosurgical Research, Carpentier Foundation, Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
33
|
Blandinières A, Gille T, Sadoine J, Bièche I, Slimani L, Dizier B, Gaussem P, Chaussain C, Planes C, Dorfmüller P, Israël-Biet D, Smadja DM. Endothelial Colony-Forming Cells Do Not Participate to Fibrogenesis in a Bleomycin-Induced Pulmonary Fibrosis Model in Nude Mice. Stem Cell Rev Rep 2019; 14:812-822. [PMID: 30267203 DOI: 10.1007/s12015-018-9846-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating lung disease characterized by fibroblast proliferation, extracellular matrix deposition, destruction of pulmonary alveolar architecture and vascular remodeling. Apart pirfenidone or nintendanib that only slow down the fibrotic process, there is no curative treatment other than lung transplantation. Because cell therapy approaches have been proposed in IPF, we hypothesized that injection of endothelial colony-forming cells (ECFCs), the vasculogenic subtype of endothelial progenitor cells, could modulate fibrosis in a Nude mouse model of bleomycin induced-pulmonary fibrosis. Mice were injected with ECFCs isolated from cord blood and from peripheral blood of adult IPF patients at two time-points: during the development of the fibrosis or once the fibrosis was constituted. We assessed morbidity, weight variation, collagen deposition, lung imaging by microCT, Fulton score and microvascular density. Neither ECFCs isolated from cord blood nor from IPF patients were able to modulate fibrosis or vascular density during fibrogenesis or when fibrosis was constituted. These findings indicate that human ECFCs do not promote an adaptive regenerative response in the lung upon fibrosis or angiogenic process in the setting of bleomycin-induced pulmonary fibrosis in Nude mice.
Collapse
Affiliation(s)
- Adeline Blandinières
- AP-HP, European Georges Pompidou Hospital, Hematology Department , Paris, France
- Paris Descartes University, Sorbonne Paris Cité, Paris, France
- Inserm UMR-S1140, Paris, France
| | - Thomas Gille
- AP-HP, Avicenne Hospital, Physiology Department , Paris, France
| | - Jérémy Sadoine
- Laboratory EA 2496 Orofacial Pathologies, Imaging and Biotherapies, Montrouge, France
| | - Ivan Bièche
- Paris Descartes University, Sorbonne Paris Cité, Paris, France
- Pharmacogenomics Unit, Department of Genetics, Institut Curie, Paris, France
| | - Lofti Slimani
- Laboratory EA 2496 Orofacial Pathologies, Imaging and Biotherapies, Montrouge, France
| | - Blandine Dizier
- Paris Descartes University, Sorbonne Paris Cité, Paris, France
- Inserm UMR-S1140, Paris, France
| | - Pascale Gaussem
- AP-HP, European Georges Pompidou Hospital, Hematology Department , Paris, France
- Paris Descartes University, Sorbonne Paris Cité, Paris, France
- Inserm UMR-S1140, Paris, France
| | - Catherine Chaussain
- Laboratory EA 2496 Orofacial Pathologies, Imaging and Biotherapies, Montrouge, France
| | - Carole Planes
- AP-HP, Avicenne Hospital, Physiology Department , Paris, France
| | - Peter Dorfmüller
- Centre Chirurgical Marie Lannelongue, INSERM U999, Le Plessis Robinson, France
| | - Dominique Israël-Biet
- Paris Descartes University, Sorbonne Paris Cité, Paris, France
- Inserm UMR-S1140, Paris, France
- AP-HP, European Georges Pompidou Hospital, Respiratory Medicine Department , Paris, France
| | - David M Smadja
- AP-HP, European Georges Pompidou Hospital, Hematology Department , Paris, France.
- Paris Descartes University, Sorbonne Paris Cité, Paris, France.
- Inserm UMR-S1140, Paris, France.
| |
Collapse
|
34
|
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: 21] [Impact Index Per Article: 3.5] [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
|
35
|
Keighron C, Lyons CJ, Creane M, O'Brien T, Liew A. Recent Advances in Endothelial Progenitor Cells Toward Their Use in Clinical Translation. Front Med (Lausanne) 2018; 5:354. [PMID: 30619864 PMCID: PMC6305310 DOI: 10.3389/fmed.2018.00354] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 12/03/2018] [Indexed: 12/28/2022] Open
Abstract
Since the discovery of Endothelial Progenitor Cells (EPC) by Asahara and colleagues in 1997, an increasing number of preclinical studies have shown that EPC based therapy is feasible, safe, and efficacious in multiple disease states. Subsequently, this has led to several, mainly early phase, clinical trials demonstrating the feasibility and safety profile of EPC therapy, with the suggestion of efficacy in several conditions including ischemic heart disease, pulmonary arterial hypertension and decompensated liver cirrhosis. Despite the use of the common term “EPC,” the characteristics, manufacturing methods and subset of the cell type used in these studies often vary significantly, rendering clinical translation challenging. It has recently been acknowledged that the true EPC is the endothelial colony forming cells (ECFC). The objective of this review was to summarize and critically appraise the registered and published clinical studies using the term “EPC,” which encompasses a heterogeneous cell population, as a therapeutic agent. Furthermore, the preclinical data using ECFC from the PubMed and Web of Science databases were searched and analyzed. We noted that despite the promising effect of ECFC on vascular regeneration, no clinical study has stemmed from these preclinical studies. We showed that there is a lack of information registered on www.clinicaltrials.gov for EPC clinical trials, specifically on cell culture methods. We also highlighted the importance of a detailed definition of the cell type used in EPC clinical trials to facilitate comparisons between trials and better understanding of the potential clinical benefit of EPC based therapy. We concluded our review by discussing the potential and limitations of EPC based therapy in clinical settings.
Collapse
Affiliation(s)
- Cameron Keighron
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science and Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
| | - Caomhán J Lyons
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science and Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
| | - Michael Creane
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science and Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
| | - Timothy O'Brien
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science and Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
| | - Aaron Liew
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science and Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
| |
Collapse
|
36
|
O'Neill CL, McLoughlin KJ, Chambers SEJ, Guduric-Fuchs J, Stitt AW, Medina RJ. The Vasoreparative Potential of Endothelial Colony Forming Cells: A Journey Through Pre-clinical Studies. Front Med (Lausanne) 2018; 5:273. [PMID: 30460233 PMCID: PMC6232760 DOI: 10.3389/fmed.2018.00273] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/05/2018] [Indexed: 12/24/2022] Open
Abstract
For over a decade various cell populations have been investigated for their vasoreparative potential. Cells with the capacity to promote blood vessel regeneration are commonly known as endothelial progenitor cells (EPCs); although such a definition is currently considered too simple for the complexity of cell populations involved in the reparative angiogenic process. A subset of EPCs called endothelial colony forming cells (ECFCs) have emerged as a suitable candidate for cytotherapy, primarily due to their clonogenic progenitor characteristics, unequivocal endothelial phenotype, and inherent ability to promote vasculogenesis. ECFCs can be readily isolated from human peripheral and cord blood, expanded ex vivo and used to revascularize ischemic tissues. These cells have demonstrated efficacy in several in vivo preclinical models such as the ischemic heart, retina, brain, limb, lung and kidney. This review will summarize the current pre-clinical evidence for ECFC cytotherapy and discuss their potential for clinical application.
Collapse
Affiliation(s)
- Christina L O'Neill
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Kiran J McLoughlin
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Sarah E J Chambers
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Jasenka Guduric-Fuchs
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Alan W Stitt
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Reinhold J Medina
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
37
|
Sha Y, Yang L, Lv Y. MGF E peptide improves anterior cruciate ligament repair by inhibiting hypoxia‐induced cell apoptosis and accelerating angiogenesis. J Cell Physiol 2018; 234:8846-8861. [DOI: 10.1002/jcp.27546] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Accepted: 09/13/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Yongqiang Sha
- Key Laboratory of Biorheological Science and Technology Ministry of Education, Bioengineering College, Chongqing University Chongqing China
- Mechanobiology and Regenerative Medicine Laboratory Bioengineering College, Chongqing University Chongqing China
| | - Li Yang
- Key Laboratory of Biorheological Science and Technology Ministry of Education, Bioengineering College, Chongqing University Chongqing China
- Mechanobiology and Regenerative Medicine Laboratory Bioengineering College, Chongqing University Chongqing China
| | - Yonggang Lv
- Key Laboratory of Biorheological Science and Technology Ministry of Education, Bioengineering College, Chongqing University Chongqing China
- Mechanobiology and Regenerative Medicine Laboratory Bioengineering College, Chongqing University Chongqing China
| |
Collapse
|