1
|
Zhao Y, Du L, Han L, Liu F, Chen S, Li Z, Wang F. Exosomal hsa_circ_0093884 derived from endothelial progenitor cells promotes therapeutic neovascularization via miR-145/SIRT1 pathway. Biomed Pharmacother 2024; 173:116343. [PMID: 38428311 DOI: 10.1016/j.biopha.2024.116343] [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: 11/15/2023] [Revised: 02/16/2024] [Accepted: 02/23/2024] [Indexed: 03/03/2024] Open
Abstract
Therapeutic neovascularization is a strategy to promote blood vessel growth and improve blood flow, which is critical to tissue repair and regeneration in ischemic diseases. Here, we investigated the role of endothelial progenitor cell - derived exosomes (EPC-Exos) in therapeutic neovascularization and clarified the mechanism of hsa_circ_0093884 in EPC-Exos mediated neovascularization. Injection of EPC-Exos improved mouse ischemic hindlimb perfusion, promoted angiogenesis in Matrigel plugs and mouse skin wound healing. In vitro coculture with EPC-Exos improved HUVEC proliferation, angiogenic and migration ability, while alleviated hypoxia-induced apoptosis. hsa_circ_0093884 was identified from eleven types of circRNA derived from SIRT1 and proved to be enriched in EPC-Exos. Overexpression of hsa_circ_0093884 in EPC-Exos further enhanced the angiogenic capacity, while knockdown of hsa_circ_0093884 abolished the benefits. Mechanistically, EPC-Exos mediated shuttling of hsa_circ_0093884 induced cytoplasmic sponge of miR-145, thereby releasing repression of SIRT1. In vitro co-transfection indicated silence of miR-145 further strengthened the angiogenic effect of hsa_circ_0093884, while overexpression of miR-145 inhibited hsa_circ_0093884 mediated angiogenesis and abolished the beneficial effect of EPC-Exos. Furthermore, in vivo experiments using endothelial specific SIRT1 conditional knockout mice indicated hsa_circ_0093884 overexpressing EPC-Exos failed to promote therapeutic neovascularization in SIRT1cKO mice. Collectively, our results demonstrated that EPC-Exos promoted therapeutic neovascularization through hsa_circ_0093884/miR-145/SIRT1 axis.
Collapse
Affiliation(s)
- Yuhao Zhao
- Department of Geriatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Du
- Department of Geriatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Han
- Department of Geriatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang Liu
- Department of Geriatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuyan Chen
- Department of Geriatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhen Li
- Department of Geriatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Fei Wang
- Department of Geriatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
2
|
Wu Q, Qi B, Duan X, Ming X, Yan F, He Y, Bu X, Sun S, Zhu H. MicroRNA-126 enhances the biological function of endothelial progenitor cells under oxidative stress via PI3K/Akt/GSK3β and ERK1/2 signaling pathways. Bosn J Basic Med Sci 2021; 21:71-80. [PMID: 31999938 PMCID: PMC7861621 DOI: 10.17305/bjbms.2019.4493] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 12/26/2019] [Indexed: 12/15/2022] Open
Abstract
Endothelial progenitor cell (EPC) transplantation is a safe and effective method to treat acute myocardial infarction (AMI). However, oxidative stress leads to the death of a large number of EPCs in the early stage of transplantation, severely weakening the therapeutic effect. Previous studies demonstrated that microRNAs regulate the biological function of EPCs. The aim of the current study was to investigate the effect of microRNA on the biological function of EPCs under oxidative stress. Quantitative reverse transcription PCR was performed to detect the expression of miR-126, miR-508-5p, miR-150, and miR-16 in EPCs from rats, among which miR-126 showed a relatively higher expression. Treatment with H2O2 decreased miR-126 expression in EPCs in a dose-dependent manner. EPCs were further transfected with miR-126 mimics or inhibitors, followed by H2O2 treatment. Overexpression of miR-126 enhanced the proliferation, migration, and tube formation of H2O2-treated EPCs. MiR-126 overexpression also inhibited reactive oxygen species and malondialdehyde levels and enhanced superoxide dismutase levels, as well as increased angiopoietin (Ang)1 expression and decreased Ang2 expression in H2O2-treated EPCs. Moreover, miR-126 participated in the regulation of phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/glycogen synthase kinase 3β (GSK3β) and extracellular signal-regulated kinase 1/2 (ERK1/2) signaling in EPCs, where both pathways were activated after miR-126 overexpression in H2O2-treated EPCs. Overall, we showed that miR-126 promoted the biological function of EPCs under H2O2-induced oxidative stress by activating the PI3K/Akt/GSK3β and ERK1/2 signaling pathway, which may serve as a new therapeutic approach to treat AMI.
Collapse
Affiliation(s)
- Qinqin Wu
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Benling Qi
- Department of Geriatrics, Institute of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyu Duan
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyan Ming
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengqin Yan
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingxia He
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaofen Bu
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan Sun
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Zhu
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Endothelial Progenitor Cells Induce Angiogenesis: a Potential Mechanism Underlying Neovascularization in Encephaloduroarteriosynangiosis. Transl Stroke Res 2020; 12:357-365. [PMID: 32632776 DOI: 10.1007/s12975-020-00834-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/10/2020] [Accepted: 07/01/2020] [Indexed: 12/12/2022]
Abstract
Encephaloduroarteriosynangiosis (EDAS) is one of the most commonly used indirect vascular reconstruction methods. EDAS aids in the formation of collateral vessels from the extracranial to the intracranial circulation in patients with moyamoya disease (MMD). However, the underlying mechanism of collateral vessel formation is not well understood. Endothelial progenitor cells (EPCs) differentiate to form the vascular endothelial cells and play a very important role in angiogenesis. We designed this prospective clinical trial to investigate the presence of EPCs in patients with MMD and to explore the neovascularization mechanism mediated by the EPCs in EDAS. The patients who were diagnosed with MMD were recruited between February 5, 2017, and January 7, 2018. The blood samples were obtained from an antecubital vein and were analyzed using flow cytometry. EPCs were defined as CD34brCD133+CD45dimKDR+. All the patients enrolled in the study underwent EDAS. Cerebral arteriography was performed 6 months post-EDAS to assess the efficacy of synangiosis. The correlation between EPC count and good collateral circulation was evaluated. Among the 116 patients with MMD enrolled in this study, 73 were women and 43 were men. The average age of the patients was 33.8 ± 15.2 years. The EPC count of the patients with MMD was 0.071% ± 0.050% (expressed as percentage of the peripheral blood mononuclear cells). The EPC count in the good postoperative collateral circulation group was significantly higher (0.085% ± 0.054%) than that in the poor collateral circulation group (0.048% ± 0.034%) (P = 0.000). The age, modified Suzuki-Mugikura grade, and EPC count were significantly correlated with the good collateral circulation post-EDAS in the multivariate analysis (P = 0.018, P = 0.007, and P = 0.003, respectively). The formation of collateral vessels by EDAS is primarily driven by angiogenesis. The EPC count may be the most critical factor for collateral circulation. The therapeutic effect of EDAS is more likely to benefit younger or severe ischemic patients with MMD.
Collapse
|
4
|
Duan Y, Prasad R, Feng D, Beli E, Li Calzi S, Longhini ALF, Lamendella R, Floyd JL, Dupont M, Noothi SK, Sreejit G, Athmanathan B, Wright J, Jensen AR, Oudit GY, Markel TA, Nagareddy PR, Obukhov AG, Grant MB. Bone Marrow-Derived Cells Restore Functional Integrity of the Gut Epithelial and Vascular Barriers in a Model of Diabetes and ACE2 Deficiency. Circ Res 2019; 125:969-988. [PMID: 31610731 DOI: 10.1161/circresaha.119.315743] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
RATIONALE There is incomplete knowledge of the impact of bone marrow cells on the gut microbiome and gut barrier function. OBJECTIVE We postulated that diabetes mellitus and systemic ACE2 (angiotensin-converting enzyme 2) deficiency would synergize to adversely impact both the microbiome and gut barrier function. METHODS AND RESULTS Bacterial 16S rRNA sequencing and metatranscriptomic analysis were performed on fecal samples from wild-type, ACE2-/y, Akita (type 1 diabetes mellitus), and ACE2-/y-Akita mice. Gut barrier integrity was assessed by immunofluorescence, and bone marrow cell extravasation into the small intestine was evaluated by flow cytometry. In the ACE2-/y-Akita or Akita mice, the disrupted barrier was associated with reduced levels of myeloid angiogenic cells, but no increase in inflammatory monocytes was observed within the gut parenchyma. Genomic and metatranscriptomic analysis of the microbiome of ACE2-/y-Akita mice demonstrated a marked increase in peptidoglycan-producing bacteria. When compared with control cohorts treated with saline, intraperitoneal administration of myeloid angiogenic cells significantly decreased the microbiome gene expression associated with peptidoglycan biosynthesis and restored epithelial and endothelial gut barrier integrity. Also indicative of diabetic gut barrier dysfunction, increased levels of peptidoglycan and FABP-2 (intestinal fatty acid-binding protein 2) were observed in plasma of human subjects with type 1 diabetes mellitus (n=21) and type 2 diabetes mellitus (n=23) compared with nondiabetic controls (n=23). Using human retinal endothelial cells, we determined that peptidoglycan activates a noncanonical TLR-2 (Toll-like receptor 2) associated MyD88 (myeloid differentiation primary response protein 88)-ARNO (ADP-ribosylation factor nucleotide-binding site opener)-ARF6 (ADP-ribosylation factor 6) signaling cascade, resulting in destabilization of p120-catenin and internalization of VE-cadherin as a mechanism of deleterious impact of peptidoglycan on the endothelium. CONCLUSIONS We demonstrate for the first time that the defect in gut barrier function and dysbiosis in ACE2-/y-Akita mice can be favorably impacted by exogenous administration of myeloid angiogenic cells.
Collapse
Affiliation(s)
- Yaqian Duan
- From the Department of Anatomy, Cell Biology and Physiology (Y.D., A.G.O.), Indiana University School of Medicine, Indianapolis.,Department of Endocrinology, The Second Affiliated Hospital of Chongqing Medical University, China (Y.D.)
| | - Ram Prasad
- Department of Ophthalmology and Visual Sciences (R.P., S.L.C., A.L.F.L., J.L.F., M.D., S.K.N., M.B.G.), University of Alabama at Birmingham
| | - Dongni Feng
- Department of Ophthalmology, The Eugene and Marilyn Glick Eye Institute (D.F., E.B.), Indiana University School of Medicine, Indianapolis
| | - Eleni Beli
- Department of Ophthalmology, The Eugene and Marilyn Glick Eye Institute (D.F., E.B.), Indiana University School of Medicine, Indianapolis
| | - Sergio Li Calzi
- Department of Ophthalmology and Visual Sciences (R.P., S.L.C., A.L.F.L., J.L.F., M.D., S.K.N., M.B.G.), University of Alabama at Birmingham
| | - Ana Leda F Longhini
- Department of Ophthalmology and Visual Sciences (R.P., S.L.C., A.L.F.L., J.L.F., M.D., S.K.N., M.B.G.), University of Alabama at Birmingham
| | - Regina Lamendella
- Ohio State University, Wright Labs, LLC, Huntingdon, PA (R.L., J.W.)
| | - Jason L Floyd
- Department of Ophthalmology and Visual Sciences (R.P., S.L.C., A.L.F.L., J.L.F., M.D., S.K.N., M.B.G.), University of Alabama at Birmingham
| | - Mariana Dupont
- Department of Ophthalmology and Visual Sciences (R.P., S.L.C., A.L.F.L., J.L.F., M.D., S.K.N., M.B.G.), University of Alabama at Birmingham
| | - Sunil K Noothi
- Department of Ophthalmology and Visual Sciences (R.P., S.L.C., A.L.F.L., J.L.F., M.D., S.K.N., M.B.G.), University of Alabama at Birmingham
| | | | | | - Justin Wright
- Ohio State University, Wright Labs, LLC, Huntingdon, PA (R.L., J.W.)
| | - Amanda R Jensen
- Riley Hospital for Children, Pediatric Surgery (A.R.J., T.A.M.), Indiana University School of Medicine, Indianapolis
| | - Gavin Y Oudit
- Ohio State University, Wright Labs, LLC, Huntingdon, PA (R.L., J.W.)
| | - Troy A Markel
- Riley Hospital for Children, Pediatric Surgery (A.R.J., T.A.M.), Indiana University School of Medicine, Indianapolis
| | | | - Alexander G Obukhov
- From the Department of Anatomy, Cell Biology and Physiology (Y.D., A.G.O.), Indiana University School of Medicine, Indianapolis
| | - Maria B Grant
- Department of Ophthalmology and Visual Sciences (R.P., S.L.C., A.L.F.L., J.L.F., M.D., S.K.N., M.B.G.), University of Alabama at Birmingham
| |
Collapse
|
5
|
Mathur T, Singh KA, R Pandian NK, Tsai SH, Hein TW, Gaharwar AK, Flanagan JM, Jain A. Organ-on-chips made of blood: endothelial progenitor cells from blood reconstitute vascular thromboinflammation in vessel-chips. LAB ON A CHIP 2019; 19:2500-2511. [PMID: 31246211 PMCID: PMC6650325 DOI: 10.1039/c9lc00469f] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Development of therapeutic approaches to treat vascular dysfunction and thrombosis at disease- and patient-specific levels is an exciting proposed direction in biomedical research. However, this cannot be achieved with animal preclinical models alone, and new in vitro techniques, like human organ-on-chips, currently lack inclusion of easily obtainable and phenotypically-similar human cell sources. Therefore, there is an unmet need to identify sources of patient primary cells and apply them in organ-on-chips to increase personalized mechanistic understanding of diseases and to assess drugs. In this study, we provide a proof-of-feasibility of utilizing blood outgrowth endothelial cells (BOECs) as a disease-specific primary cell source to analyze vascular inflammation and thrombosis in vascular organ-chips or "vessel-chips". These blood-derived BOECs express several factors that confirm their endothelial identity. The vessel-chips are cultured with BOECs from healthy or diabetic patients and form an intact 3D endothelial lumen. Inflammation of the BOEC endothelium with exogenous cytokines reveals vascular dysfunction and thrombosis in vitro similar to in vivo observations. Interestingly, our study with vessel-chips also reveals that unstimulated BOECs of type 1 diabetic pigs show phenotypic behavior of the disease - high vascular dysfunction and thrombogenicity - when compared to control BOECs or normal primary endothelial cells. These results demonstrate the potential of organ-on-chips made from autologous endothelial cells obtained from blood in modeling vascular pathologies and therapeutic outcomes at a disease and patient-specific level.
Collapse
Affiliation(s)
- Tanmay Mathur
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell St, College Station, TX 77843, USA.
| | - Kanwar Abhay Singh
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell St, College Station, TX 77843, USA.
| | - Navaneeth K R Pandian
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell St, College Station, TX 77843, USA.
| | - Shu-Huai Tsai
- Department of Medical Physiology, Texas A&M University System Health Science Center, Temple, USA
| | - Travis W Hein
- Department of Medical Physiology, Texas A&M University System Health Science Center, Temple, USA
| | - Akhilesh K Gaharwar
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell St, College Station, TX 77843, USA. and Center for Remote Health Technologies and Systems, Texas A&M University, College Station, USA and Department of Materials Science and Engineering, Texas A&M University, College Station, USA
| | - Jonathan M Flanagan
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, USA
| | - Abhishek Jain
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell St, College Station, TX 77843, USA.
| |
Collapse
|
6
|
Qiu C, Zhang D, Chi Y, Chen Q, Xu L, Xie Q. Clinical significance of 5-(and 6)-carboxyfluorescein diacetate succinimidyl ester-labeled microspheres for detecting endothelial progenitor cells in human peripheral blood. Exp Ther Med 2017; 14:1659-1664. [PMID: 28810633 DOI: 10.3892/etm.2017.4657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 02/14/2017] [Indexed: 11/06/2022] Open
Abstract
The aims of the present study were to establish a single-platform flow cytometry method using 5-(and 6)-carboxyfluorescein diacetate succinimidyl ester (CFSE)-labeled microspheres as the reference for determining endothelial progenitor cell (EPC) number and to evaluate the efficacy of this detection method. Single-platform flow cytometry was used to count cell numbers using CFSE-stained fluorescent microspheres as the internal reference and the EPC numbers in specimens using this novel method were compared with an in vitro clonogenic counting assay. The results of the two counting methods were consistent and compared with the in vitro clonogenic counting assay, the time and cost of the novel method was markedly reduced, as were the corresponding technical requirements. The present findings indicated that single-platform flow cytometry, with CFSE-labeled microspheres as the reference, provides faster and improved detection of EPCs in human peripheral blood specimens, with reduced time and cost, making it more suitable for routine clinical application.
Collapse
Affiliation(s)
- Chaolin Qiu
- Clinical Laboratory Department, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, P.R. China
| | - Denghai Zhang
- Central Laboratory Department, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, P.R. China
| | - Yongbin Chi
- Clinical Laboratory Department, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, P.R. China
| | - Qing Chen
- Clinical Laboratory Department, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, P.R. China
| | - Limin Xu
- Clinical Laboratory Department, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, P.R. China
| | - Qiuhua Xie
- Clinical Laboratory Department, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, P.R. China
| |
Collapse
|
7
|
Stoltz JF, Bensoussan D, De Isla N, Zhang L, Han Z, Magdalou J, Huselstein C, Ye J, Leballe B, Decot V, Reppel L. Stem cells and vascular regenerative medicine: A mini review. Clin Hemorheol Microcirc 2017; 64:613-633. [DOI: 10.3233/ch-168036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- J.-F. Stoltz
- CNRS, UMR 7365, Biopole, Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CHRU de Nancy, Unité de Thérapie Cellulaire et Tissulaire (UTCT) (FR CNRS-INSERM-UHP-CHU), Vandoeuvre-Lès-Nancy, France
| | - D. Bensoussan
- CNRS, UMR 7365, Biopole, Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CHRU de Nancy, Unité de Thérapie Cellulaire et Tissulaire (UTCT) (FR CNRS-INSERM-UHP-CHU), Vandoeuvre-Lès-Nancy, France
| | - N. De Isla
- CNRS, UMR 7365, Biopole, Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
| | - L. Zhang
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- Centre de Recherche, Calmette Hospital, Kunming, China
| | - Z. Han
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- Centre de Recvherche sur les cellules souches, Beijing et Tianjin, China
| | - J. Magdalou
- CNRS, UMR 7365, Biopole, Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
| | - C. Huselstein
- CNRS, UMR 7365, Biopole, Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
| | - J.S. Ye
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- Centre de Recherche, Calmette Hospital, Kunming, China
| | | | - V. Decot
- CNRS, UMR 7365, Biopole, Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CHRU de Nancy, Unité de Thérapie Cellulaire et Tissulaire (UTCT) (FR CNRS-INSERM-UHP-CHU), Vandoeuvre-Lès-Nancy, France
| | - L. Reppel
- CNRS, UMR 7365, Biopole, Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CHRU de Nancy, Unité de Thérapie Cellulaire et Tissulaire (UTCT) (FR CNRS-INSERM-UHP-CHU), Vandoeuvre-Lès-Nancy, France
| |
Collapse
|
8
|
Ferensztajn-Rochowiak E, Kucharska-Mazur J, Samochowiec J, Ratajczak MZ, Michalak M, Rybakowski JK. The effect of long-term lithium treatment of bipolar disorder on stem cells circulating in peripheral blood. World J Biol Psychiatry 2017; 18:54-62. [PMID: 27071327 DOI: 10.3109/15622975.2016.1174301] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVES To investigate the effect of long-term lithium treatment on very small embryonic-like stem cells (VSELs), haematopoietic stem cells (HSCs), mesenchymal stem cells (MSCs) and endothelial progenitor cells (EPCs) circulating in peripheral blood (PB), in bipolar disorder (BD). METHODS The study included 15 BD patients (aged 55 ± 6 years) treated with lithium for 8-40 years (mean 16 years), 15 BD patients (aged 53 ± 7 years) with duration of illness >10 years, who had never received lithium, and 15 healthy controls (aged 50 ± 5 years). The VSELs, HSCs, MSCs and EPCs were measured by flow cytometric analysis. RESULTS In BD subjects not taking lithium the number of CD34+ VSELs was significantly higher, and MSCs and EPCs numerically higher, than in control subjects and the number of CD34+ VSELs correlated with the duration of illness. In lithium-treated patients these values were similar to controls and the number of CD34+ VSELs correlated negatively with the duration of lithium treatment and serum lithium concentration. CONCLUSIONS Long-term treatment with lithium may suppress the activation of regenerative processes by reducing the number of VSELs circulating in PB. These cells, in BD patients not treated with lithium, may provide a new potential biological marker of the illness and its clinical progress.
Collapse
Affiliation(s)
| | | | - Jerzy Samochowiec
- b Department of Psychiatry , Pomeranian University of Medicine , Szczecin , Poland
| | - Mariusz Z Ratajczak
- c Department of Physiology , Pomeranian University of Medicine , Szczecin , Poland.,d Stem Cell Biology Program at the James Graham Brown Cancer Center , University of Louisville , Louisville , KY , 40202 , USA
| | - Michal Michalak
- e Department of Computer Science and Statistics , Poznan University of Medical Sciences , Poznan , Poland
| | - Janusz K Rybakowski
- a Department of Adult Psychiatry , Poznan University of Medical Sciences , Poznan , Poland
| |
Collapse
|
9
|
Möbius MA, Thébaud B. Cell Therapy for Bronchopulmonary Dysplasia: Promises and Perils. Paediatr Respir Rev 2016; 20:33-41. [PMID: 27425012 DOI: 10.1016/j.prrv.2016.06.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 06/07/2016] [Indexed: 12/13/2022]
Abstract
Despite great achievements in neonatal and perinatal medicine over the past decades, the immature lung remains the most critical organ to care for after premature birth. As a consequence, bronchopulmonary dysplasia (BPD) remains the most common complication of extreme prematurity. BPD impairs normal development and may cause lifelong morbidities. At present, there is no effective treatment for BPD - including preventing premature birth. Recent insights into the biology of stem and progenitor cells have ignited the hope of protecting the immature lung, and even regenerating an already damaged lung by using exogenous stem- or progenitor cells as therapeutics. These therapies are still experimental, and knowledge on the exact mechanisms behind the beneficial effects seen in various animal models of BPD is limited. Nevertheless, early phase clinical trials have started, and encouraging steps towards the therapeutic use of these cells are being made. This review aims to (I) provide an overview of the role of stem/progenitor cells in development and therapy of BPD for the practicing clinician, (II) discuss the potential clinical applications of cell products as therapeutic agents to prevent neonatal lung injury and (III) examine potential obstacles towards the manufacturing of clinical grade cell products for use in the care for premature infants.
Collapse
Affiliation(s)
- Marius Alexander Möbius
- Department of Neonatology and Pediatric Critical Care Medicine, Medical Faculty, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; DFG Research Center and Cluster of Excellence for Regenerative Therapies (CRTD), Technische Universität Dresden, Dresden, Germany; Sinclair Centre for Regenerative Medicine, Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada.
| | - Bernard Thébaud
- Sinclair Centre for Regenerative Medicine, Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada; Division of Neonatology, Department of Pediatrics, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
10
|
Bruyneel AAN, Sehgal A, Malandraki-Miller S, Carr C. Stem Cell Therapy for the Heart: Blind Alley or Magic Bullet? J Cardiovasc Transl Res 2016; 9:405-418. [PMID: 27542008 PMCID: PMC5153828 DOI: 10.1007/s12265-016-9708-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 08/05/2016] [Indexed: 12/15/2022]
Abstract
When stressed by ageing or disease, the adult human heart is unable to regenerate, leading to scarring and hypertrophy and eventually heart failure. As a result, stem cell therapy has been proposed as an ultimate therapeutic strategy, as stem cells could limit adverse remodelling and give rise to new cardiomyocytes and vasculature. Unfortunately, the results from clinical trials to date have been largely disappointing. In this review, we discuss the current status of the field and describe various limitations and how future work may attempt to resolve these to make way to successful clinical translation.
Collapse
Affiliation(s)
- Arne A N Bruyneel
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, UK
| | | | | | - Carolyn Carr
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
11
|
Xie C, Gao X, Luo Y, Pang Y, Li M. Electroacupuncture modulates stromal cell-derived factor-1α expression and mobilization of bone marrow endothelial progenitor cells in focal cerebral ischemia/reperfusion model rats. Brain Res 2016; 1648:119-126. [PMID: 27453543 DOI: 10.1016/j.brainres.2016.07.038] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 05/27/2016] [Accepted: 07/20/2016] [Indexed: 01/07/2023]
Abstract
Stromal cell-derived factor-1α(SDF-1α) plays a crucial role in regulating the mobilization, migration and homing of endothelial progenitor cells(EPCs). Electroacupuncture(EA), a modern version of Traditional Chinese Medicine, can improve neurological recovery and angiogenesis in cerebral ischemic area. This study aimed to investigate the effects of electroacupuncture(EA) on the mobilization and migration of bone marrow EPCs and neurological functional recovery in rats model after focal cerebral ischemia/reperfusion and the potentially involved mechanisms. Sprague-Dawley rats received filament occlusion of the right middle cerebral artery for 2h followed by reperfusion for 12h, 1d, 2d, 3d, 7d respectively. Rats were randomly divided into sham group, model group and EA group. After 2h of the reperfusion, EA was given at the "Baihui" (GV 20)/Siguan ("Hegu" (LI 4)/"Taichong" (LR 3)) acupoints in the EA group. Modified neurological severity score (mNSS) was used to assess the neurological functional recovery. EPCs number and SDF-1α level in bone marrow(BM) and peripheral blood(PB) were detected by using fluorescence-activated cell sorting (FACS) analysis and quantitative real time polymerase chain reaction (qRT-PCR) respectively. An mNSS test showed that EA treatment significantly improved the neurological functional outcome. EPCs number in PB and BM were obviously increased in the EA group. After cerebral ischemia, the SDF-1α level was decreased in BM while it was increased in PB, which implied a gradient of SDF-1α among BM and PB after ischemia. It suggested that the forming of SDF-1α concentration gradient can induce the mobilization and homing of EPCs. Eletroacupuncture as a treatment can accelerate and increase the forming of SDF-1α concentration gradient to further induce the mobilization of EPCs and angiogenesis in ischemic brain and improve the neurological function recovery.
Collapse
Affiliation(s)
- Chenchen Xie
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China; Department of Neurology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610018, China.
| | - Xiang Gao
- Department of Nephrology, The Eleventh People's Hospital of Chengdu, Chengdu, Sichuan 610018, China.
| | - Yong Luo
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China.
| | - Yueshan Pang
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China.
| | - Man Li
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China.
| |
Collapse
|
12
|
Altabas V, Altabas K, Kirigin L. Endothelial progenitor cells (EPCs) in ageing and age-related diseases: How currently available treatment modalities affect EPC biology, atherosclerosis, and cardiovascular outcomes. Mech Ageing Dev 2016; 159:49-62. [PMID: 26919825 DOI: 10.1016/j.mad.2016.02.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 01/25/2016] [Accepted: 02/22/2016] [Indexed: 12/15/2022]
Abstract
Endothelial progenitor cells (EPCs) are mononuclear cells that circulate in the blood and are derived from different tissues, expressing cell surface markers that are similar to mature endothelial cells. The discovery of EPCs has lead to new insights in vascular repair and atherosclerosis and also a new theory for ageing. EPCs from the bone marrow and some other organs aid in vascular repair by migrating to distant vessels where they differentiate into mature endothelial cells and replace old and injured endothelial cells. The ability of EPCs to repair vascular damage depends on their number and functionality. Currently marketed drugs used in a variety of diseases can modulate these characteristics. In this review, the effect of currently available treatment options for cardiovascular and metabolic disorders on EPC biology will be discussed. The various EPC-based therapies that will be discussed include lipid-lowering agents, antihypertensive agents, antidiabetic drugs, phosphodiesteraze inhibitors, hormones, as well as EPC capturing stents.
Collapse
Affiliation(s)
- Velimir Altabas
- Department of Internal Medicine, University Clinical Hospital "Sestre milosrdnice", Zagreb, Croatia.
| | - Karmela Altabas
- Department of Internal Medicine, University Clinical Hospital "Sestre milosrdnice", Zagreb, Croatia.
| | - Lora Kirigin
- Department of Internal Medicine, University Clinical Hospital "Sestre milosrdnice", Zagreb, Croatia.
| |
Collapse
|
13
|
Gao X, Wang YS, Li XQ, Hou HY, Su JB, Yao LB, Zhang J. Macrophages promote vasculogenesis of retinal neovascularization in an oxygen-induced retinopathy model in mice. Cell Tissue Res 2016; 364:599-610. [DOI: 10.1007/s00441-015-2353-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 12/16/2015] [Indexed: 01/06/2023]
|
14
|
Conditioned Medium from Early-Outgrowth Bone Marrow Cells Is Retinal Protective in Experimental Model of Diabetes. PLoS One 2016; 11:e0147978. [PMID: 26836609 PMCID: PMC4737492 DOI: 10.1371/journal.pone.0147978] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 01/11/2016] [Indexed: 11/19/2022] Open
Abstract
Bone marrow-derived cells were demonstrated to improve organ function, but the lack of cell retention within injured organs suggests that the protective effects are due to factors released by the cells. Herein, we tested cell therapy using early outgrowth cells (EOCs) or their conditioned media (CM) to protect the retina of diabetic animal models (type 1 and type 2) and assessed the mechanisms by in vitro study. Control and diabetic (db/db) mice (8 weeks of age) were randomized to receive a unique intravenous injection of 5×105EOCs or 0.25 ml thrice weekly tail-vein injections of 10x concentrated CM and Wystar Kyoto rats rendered diabetic were randomized to receive 0.50 ml thrice weekly tail-vein injections of 10x concentrated CM. Four weeks later, the animals were euthanized and the eyes were enucleated. Rat retinal Müller cells (rMCs) were exposed for 24 h to high glucose (HG), combined or not with EOC-conditioned medium (EOC-CM) from db/m EOC cultures. Diabetic animals showed increase in diabetic retinopathy (DR) and oxidative damage markers; the treatment with EOCs or CM infusions significantly reduced this damage and re-established the retinal function. In rMCs exposed to diabetic milieu conditions (HG), the presence of EOC-CM reduced reactive oxygen species production by modulating the NADPH-oxidase 4 system, thus upregulating SIRT1 activity and deacetylating Lys-310-p65-NFκB, decreasing GFAP and VEGF expressions. The antioxidant capacity of EOC-CM led to the prevention of carbonylation and nitrosylation posttranslational modifications on the SIRT1 molecule, preserving its activity. The pivotal role of SIRT1 on the mode of action of EOCs or their CM was also demonstrated on diabetic retina. These findings suggest that EOCs are effective as a form of systemic delivery for preventing the early molecular markers of DR and its conditioned medium is equally protective revealing a novel possibility for cell-free therapy for the treatment of DR.
Collapse
|
15
|
Wilson HK, Canfield SG, Shusta EV, Palecek SP. Concise review: tissue-specific microvascular endothelial cells derived from human pluripotent stem cells. Stem Cells 2015; 32:3037-45. [PMID: 25070152 DOI: 10.1002/stem.1797] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 06/22/2014] [Indexed: 01/06/2023]
Abstract
Accumulating evidence suggests that endothelial cells (ECs) display significant heterogeneity across tissue types, playing an important role in tissue regeneration and homeostasis. Recent work demonstrating the derivation of tissue-specific microvascular endothelial cells (TS-MVECs) from human pluripotent stem cells (hPSCs) has ignited the potential to generate tissue-specific models which may be applied to regenerative medicine and in vitro modeling applications. Here, we review techniques by which hPSC-derived TS-MVECs have been made to date and discuss how current hPSC-EC differentiation protocols may be directed toward tissue-specific fates. We begin by discussing the nature of EC tissue specificity in vivo and review general hPSC-EC differentiation protocols generated over the last decade. Finally, we describe how specificity can be integrated into hPSC-EC protocols to generate hPSC-derived TS-MVECs in vitro, including EC and parenchymal cell coculture, directed differentiation, and direct reprogramming strategies.
Collapse
Affiliation(s)
- Hannah K Wilson
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | | | | |
Collapse
|
16
|
Ikutomi M, Sahara M, Nakajima T, Minami Y, Morita T, Hirata Y, Komuro I, Nakamura F, Sata M. Diverse contribution of bone marrow-derived late-outgrowth endothelial progenitor cells to vascular repair under pulmonary arterial hypertension and arterial neointimal formation. J Mol Cell Cardiol 2015; 86:121-35. [DOI: 10.1016/j.yjmcc.2015.07.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 07/10/2015] [Accepted: 07/24/2015] [Indexed: 01/29/2023]
|
17
|
Möbius MA, Thébaud B. Stem Cells and Their Mediators - Next Generation Therapy for Bronchopulmonary Dysplasia. Front Med (Lausanne) 2015; 2:50. [PMID: 26284246 PMCID: PMC4520239 DOI: 10.3389/fmed.2015.00050] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 07/15/2015] [Indexed: 01/13/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) remains a major complication of premature birth. Despite great achievements in perinatal medicine over the past decades, there is no treatment for BPD. Recent insights into the biology of stem/progenitor cells have ignited the hope of regenerating damaged organs. Animal experiments revealed promising lung protection/regeneration with stem/progenitor cells in experimental models of BPD and led to first clinical studies in infants. However, these therapies are still experimental and knowledge on the exact mechanisms of action of these cells is limited. Furthermore, heterogeneity of the therapeutic cell populations and missing potency assays currently limit our ability to predict a cell product’s efficacy. Here, we review the therapeutic potential of mesenchymal stromal, endothelial progenitor, and amniotic epithelial cells for BPD. Current knowledge on the mechanisms behind the beneficial effects of stem cells is briefly summarized. Finally, we discuss the obstacles constraining their transition from bench-to-bedside and present potential approaches to overcome them.
Collapse
Affiliation(s)
- Marius A Möbius
- Department of Neonatology and Pediatric Critical Care Medicine, Medical Faculty, University Hospital Carl Gustav Carus, Technische Universität Dresden , Dresden , Germany ; DFG Research Center and Cluster of Excellence for Regenerative Therapies (CRTD), Technische Universität Dresden , Dresden , Germany ; Regenerative Medicine Program, Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, University of Ottawa , Ottawa, ON , Canada
| | - Bernard Thébaud
- Regenerative Medicine Program, Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, University of Ottawa , Ottawa, ON , Canada ; Division of Neonatology, Department of Pediatrics, Children's Hospital of Eastern Ontario, University of Ottawa , Ottawa, ON , Canada
| |
Collapse
|
18
|
Atherogenic Cytokines Regulate VEGF-A-Induced Differentiation of Bone Marrow-Derived Mesenchymal Stem Cells into Endothelial Cells. Stem Cells Int 2015; 2015:498328. [PMID: 26106428 PMCID: PMC4464597 DOI: 10.1155/2015/498328] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 05/11/2015] [Indexed: 11/17/2022] Open
Abstract
Coronary artery stenting or angioplasty procedures frequently result in long-term endothelial dysfunction or loss and complications including arterial thrombosis and myocardial infarction. Stem cell-based therapies have been proposed to support endothelial regeneration. Mesenchymal stem cells (MSCs) differentiate into endothelial cells (ECs) in the presence of VEGF-A in vitro. Application of VEGF-A and MSC-derived ECs at the interventional site is a complex clinical challenge. In this study, we examined the effect of atherogenic cytokines (IL-6, TNFα, and Ang II) on EC differentiation and function. MSCs (CD44(+), CD73(+), CD90(+), CD14(-), and CD45(-)) were isolated from the bone marrow of Yucatan microswine. Naïve MSCs cultured in differentiation media containing VEGF-A (50 ng/mL) demonstrated increased expression of EC-specific markers (vWF, PECAM-1, and VE-cadherin), VEGFR-2 and Sox18, and enhanced endothelial tube formation. IL-6 or TNFα caused a dose-dependent attenuation of EC marker expression in VEGF-A-stimulated MSCs. In contrast, Ang II enhanced EC marker expression in VEGF-A-stimulated MSCs. Addition of Ang II to VEGF-A and IL-6 or TNFα was sufficient to rescue the EC phenotype. Thus, Ang II promotes but IL-6 and TNFα inhibit VEGF-A-induced differentiation of MSCs into ECs. These findings have important clinical implications for therapies intended to increase cardiac vascularity and reendothelialize coronary arteries following intervention.
Collapse
|
19
|
Poloni A, Maurizi G, Anastasi S, Mondini E, Mattiucci D, Discepoli G, Tiberi F, Mancini S, Partelli S, Maurizi A, Cinti S, Olivieri A, Leoni P. Plasticity of human dedifferentiated adipocytes toward endothelial cells. Exp Hematol 2015; 43:137-146. [PMID: 25448487 DOI: 10.1016/j.exphem.2014.10.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 10/01/2014] [Accepted: 10/11/2014] [Indexed: 12/28/2022]
Abstract
The process of cellular differentiation in terminally differentiated cells is thought to be irreversible, and these cells are thought to be incapable of differentiating into distinct cell lineages. Our previous study showed that mature adipocytes represent an alternative source of mesenchymal stem cells. Here, results showed the capacity of mature adipocytes to differentiate into endothelial-like cells, using the ability of these cells to revert into an immature phase without any relievable chromosomal alterations. Mature adipocytes were isolated from human omental and subcutaneous fat and were dedifferentiated in vitro. The resulting cells were subcultivated for endothelial differentiation and were analyzed for their expression of specific genes and proteins. Endothelial-like cells were harvested from the differentiation medium and were traditionally cultured to evaluate the endothelial markers and the karyotype. Cells cultured in specific medium formed tube-like structures and expressed several endothelial marker genes and proteins. The endothelial-like cells expressed significantly higher levels of vascular endothelium growth factor receptor 2, vascular endothelial cadherin, Von Willebrand factor, and CD133 than the untreated cells. These cells were positively stained for CD31 and vascular endothelial cadherin, markers of mature endothelial cells. Moreover, the low-density lipoprotein-uptake assay demonstrated a functionally endothelial differentiation of these cells. When these cells were harvested and reseeded in basal medium, they lost the endothelial markers and reacquired the typical mesenchymal stem cell markers and the ability to expand in a short time period. Moreover, karyotype analysis showed that these cells reverted into an immature phase without any karyotype alterations. In conclusion, the results showed that adipocytes exhibited a great plasticity toward the endothelial lineage, suggesting their possible use in cell therapy applications for vascular disease.
Collapse
Affiliation(s)
- Antonella Poloni
- Clinica di Ematologia, Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, Italy.
| | - Giulia Maurizi
- Clinica di Ematologia, Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, Italy
| | - Sara Anastasi
- Clinica di Ematologia, Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, Italy
| | - Eleonora Mondini
- Dipartimento di Medicina Sperimentale e Clinica, Università Politecnica delle Marche, Ancona, Italy
| | - Domenico Mattiucci
- Clinica di Ematologia, Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, Italy
| | - Giancarlo Discepoli
- Laboratorio di Citogenetica e Genetica Molecolare, Clinica di Pediatria, Università Politecnica delle Marche, Ancona, Italy
| | - Fabiola Tiberi
- Laboratorio di Citogenetica e Genetica Molecolare, Clinica di Pediatria, Università Politecnica delle Marche, Ancona, Italy
| | - Stefania Mancini
- Clinica di Ematologia, Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, Italy
| | - Stefano Partelli
- Clinica Chirurgia del Pancreas, Università Politecnica delle Marche, Ospedali Riuniti, Ancona, Italy
| | - Angela Maurizi
- Clinica Chirurgia del Pancreas, Università Politecnica delle Marche, Ospedali Riuniti, Ancona, Italy
| | - Saverio Cinti
- Dipartimento di Medicina Sperimentale e Clinica, Università Politecnica delle Marche, Ancona, Italy
| | - Attilio Olivieri
- Clinica di Ematologia, Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, Italy
| | - Pietro Leoni
- Clinica di Ematologia, Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
20
|
Piatkowski A, Grieb G, Simons D, Bernhagen J, van der Hulst RR. Endothelial progenitor cells--potential new avenues to improve neoangiogenesis and reendothelialization. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 306:43-81. [PMID: 24016523 DOI: 10.1016/b978-0-12-407694-5.00002-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The term endothelial progenitor cell (EPC) was established more than 10 years ago and is used to refer to a group of circulating cells that display endothelial lineage qualities and are able to home to areas of ischemia or vascular injury and to facilitate the repair of damaged blood vessels or develop new vessels as needed. This chapter reviews the current lineage relationships among all the cells called EPC and will clear the terminology used in EPC research. Furthermore, an overview of the clinical and in vitro research, as well as cytokine and drug interactions and potential EPC applications, is given.
Collapse
Affiliation(s)
- Andrzej Piatkowski
- Department of Plastic Surgery, academisch ziekenhuis Maastricht, MUMC+, Maastricht, The Netherlands.
| | | | | | | | | |
Collapse
|
21
|
McNiece I. Endothelial cells and regenerative medicine. Cytotherapy 2014; 16:1169-70. [PMID: 25108649 DOI: 10.1016/j.jcyt.2014.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Ian McNiece
- The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA.
| |
Collapse
|
22
|
Nakamura N, Naruse K, Kobayashi Y, Matsuki T, Hamada Y, Nakashima E, Kamiya H, Hata M, Nishikawa T, Enomoto A, Takahashi M, Murohara T, Matsubara T, Oiso Y, Nakamura J. High glucose impairs the proliferation and increases the apoptosis of endothelial progenitor cells by suppression of Akt. J Diabetes Investig 2014; 2:262-70. [PMID: 24843496 PMCID: PMC4014965 DOI: 10.1111/j.2040-1124.2010.00093.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
UNLABELLED Aims/Introduction: Endothelial progenitor cells (EPC) play a critical role in adult vasculogenesis and vascular repair. Previous studies have described the dysfunction of EPC in diabetic patients, but the precise mechanism is still unclear. To elucidate the dysfunction of EPC in diabetic patients, we investigated the functions and intracellular signaling of EPC under normal or high glucose conditions. We also examined the number of EPC in the peripheral blood of Japanese type 2 diabetic patients. MATERIALS AND METHODS EPC were cultured with normal or high glucose. Subsequently, the proliferation and the apoptosis of EPC were assessed in the presence or absence of vascular endothelial growth factor (VEGF). The phosphorylation of Akt was assessed by western blot analyses. We compared the number of CD34(+)CD45(low) progenitor cells, which is considered as a marker of EPC in non-diabetic and type 2 diabetic subjects, using flow cytometry. RESULTS High glucose decreased the proliferation of EPC and increased the number of apoptotic cells. VEGF significantly increased the proliferation and suppressed the apoptosis of EPC, both of which were abolished by PI 3-kinase inhibitor, LY294002. High glucose significantly suppressed the basal and VEGF-stimulated phosphorylation of Akt in EPC. Furthermore, the number of circulating EPC was decreased in type 2 diabetic patients, although there were no significant differences in the serum levels of VEGF between control subjects and diabetic patients. CONCLUSIONS These findings suggest that high glucose impairs the functions of EPC through the suppression of Akt phosphorylation stimulated by VEGF. (J Diabetes Invest, doi: 10.1111/j.2040-1124.2010.00093.x, 2011).
Collapse
Affiliation(s)
| | - Keiko Naruse
- Department of Internal Medicine, School of Dentistry, Aichi-Gakuin University ; Endocrinology and Diabetes
| | - Yasuko Kobayashi
- Department of Internal Medicine, School of Dentistry, Aichi-Gakuin University
| | | | | | | | - Hideki Kamiya
- CKD Initiatives, Nagoya University School of Medicine
| | - Masaki Hata
- Department of Internal Medicine, School of Dentistry, Aichi-Gakuin University
| | - Toru Nishikawa
- Department of Internal Medicine, School of Dentistry, Aichi-Gakuin University
| | | | | | | | - Tatsuaki Matsubara
- Department of Internal Medicine, School of Dentistry, Aichi-Gakuin University
| | | | | |
Collapse
|
23
|
Moschetta M, Mishima Y, Sahin I, Manier S, Glavey S, Vacca A, Roccaro AM, Ghobrial IM. Role of endothelial progenitor cells in cancer progression. Biochim Biophys Acta Rev Cancer 2014; 1846:26-39. [PMID: 24709008 DOI: 10.1016/j.bbcan.2014.03.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 02/19/2014] [Accepted: 03/27/2014] [Indexed: 12/12/2022]
Abstract
Tumor-associated neovasculature is a critical therapeutic target; however, despite significant progress made in the clinical efficacy of anti-vessel drugs, the effect of these agents remains transient: over time, most patients develop resistance, which inevitably leads to tumor progression. To develop more effective treatments, it is imperative that we better understand the mechanisms involved in tumor vessel formation, how they participate to the tumor progression and metastasis, and the best way to target them. Several mechanisms contribute to the formation of tumor-associated vasculature: i) neoangiogenesis; ii) vascular co-option; iii) mosaicism; iv) vasculogenic mimicry, and v) postnatal vasculogenesis. These mechanisms can also play a role in the development of resistance to anti-angiogenic drugs, and could serve as targets for designing new anti-vascular molecules to treat solid as well as hematological malignancies. Bone marrow-derived endothelial progenitor cell (EPC)-mediated vasculogenesis represents an important new target, especially at the early stage of tumor growth (when EPCs are critical for promoting the "angiogenic switch"), and during metastasis, when EPCs promote the transition from micro- to macro-metastases. In hematologic malignancies, the EPC population could be related to the neoplastic clone, and both may share a common ontogeny. Thus, characterization of tumor-associated EPCs in blood cancers may provide clues for more specific anti-vascular therapy that has both direct and indirect anti-tumor effects. Here, we review the role of vasculogenesis, mediated by bone marrow-derived EPCs, in the progression of cancer, with a particular focus on the role of these cells in promoting progression of hematological malignancies.
Collapse
Affiliation(s)
- Michele Moschetta
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; University of Bari Medical School, Department of Biomedical Sciences and Human Oncology (DIMO), Section of Internal Medicine and Clinical Oncology, Bari, Italy
| | - Yuji Mishima
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Ilyas Sahin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Salomon Manier
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Siobhan Glavey
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Angelo Vacca
- University of Bari Medical School, Department of Biomedical Sciences and Human Oncology (DIMO), Section of Internal Medicine and Clinical Oncology, Bari, Italy
| | - Aldo M Roccaro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Irene M Ghobrial
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
24
|
Patel J, Seppanen E, Chong MSK, Yeo JSL, Teo EYL, Chan JKY, Fisk NM, Khosrotehrani K. Prospective surface marker-based isolation and expansion of fetal endothelial colony-forming cells from human term placenta. Stem Cells Transl Med 2013; 2:839-47. [PMID: 24106336 DOI: 10.5966/sctm.2013-0092] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The term placenta is a highly vascularized tissue and is usually discarded upon birth. Our objective was to isolate clinically relevant quantities of fetal endothelial colony-forming cells (ECFCs) from human term placenta and to compare them to the well-established donor-matched umbilical cord blood (UCB)-derived ECFCs. A sorting strategy was devised to enrich for CD45-CD34+CD31Lo cells prior to primary plating to obtain pure placental ECFCs (PL-ECFCs) upon culture. UCB-ECFCs were derived using a well-described assay. PL-ECFCs were fetal in origin and expressed the same cell surface markers as UCB-ECFCs. Most importantly, a single term placenta could yield as many ECFCs as 27 UCB donors. PL-ECFCs and UCB-ECFCs had similar in vitro and in vivo vessel forming capacities and restored mouse hind limb ischemia in similar proportions. Gene expression profiles were only minimally divergent between PL-ECFCs and UCB-ECFCs, probably reflecting a vascular source versus a circulating source. Finally, PL-ECFCs and UCB-ECFCs displayed similar hierarchies between high and low proliferative colonies. We report a robust strategy to isolate ECFCs from human term placentas based on their cell surface expression. This yielded much larger quantities of ECFCs than UCB, but the cells were comparable in immunophenotype, gene expression, and in vivo functional ability. We conclude that PL-ECFCs have significant bio-banking and clinical translatability potential.
Collapse
Affiliation(s)
- Jatin Patel
- University of Queensland Centre for Clinical Research, University of Queensland, Herston, Queensland, Australia
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Reed DM, Foldes G, Harding SE, Mitchell JA. Stem cell-derived endothelial cells for cardiovascular disease: a therapeutic perspective. Br J Clin Pharmacol 2013; 75:897-906. [PMID: 22703602 DOI: 10.1111/j.1365-2125.2012.04361.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 05/30/2012] [Indexed: 01/18/2023] Open
Abstract
Stem cell therapy and organ regeneration are therapeutic approaches that will, we suggest, become mainstream for the treatment of human disease. Endothelial cells, which line the luminal surface of every vessel in the body, are essential components in any organ regeneration programme. There are a number of potentially therapeutic endothelial cell types, including embryonic, adult progenitor and induced pluripotent stem cell-derived endothelial cells, as well as host vascular cells. The features (benefits as well as disadvantages) of each cell type that make them potentially useful in therapy are important to consider. The field of stem cell biology is well developed in terms of protocols for generating endothelium. However, where there is a distinct and urgent unmet need for knowledge concerning how the endothelial cells from these different sources function as endothelium and how susceptible they may be to inflammation and atherosclerosis. Furthermore, where stem cells have been used in clinical trials there is little commonality in protocols for deriving the cells (and thereby the specific phenotype of cells used), administering the cells, dosing the cells and/or in assessing efficacy attributed to the cells themselves. This review discusses these and other issues relating to stem cell-derived endothelial cells in cell therapy for cardiovascular disease.
Collapse
Affiliation(s)
- Daniel M Reed
- Department of Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College London, London, UK.
| | | | | | | |
Collapse
|
26
|
Roura S, Gálvez-Montón C, Bayes-Genis A. The challenges for cardiac vascular precursor cell therapy: lessons from a very elusive precursor. J Vasc Res 2013; 50:304-23. [PMID: 23860201 DOI: 10.1159/000353294] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 05/01/2013] [Indexed: 11/19/2022] Open
Abstract
There is compelling evidence that cardiovascular disorders arise and/or progress due mainly to endothelial dysfunction. Novel therapeutic strategies aim to generate new myocardial tissue using cells with regenerative potential, either alone or in combination with biomaterials, cytokines and advanced monitoring devices. Among the human adult progenitor cells used in such methods, those historically termed 'endothelial progenitor cells' show promise for vascular growth and repair. Asahara et al. [Science 1997;275:964-967] initially described putative endothelial cell precursors in 1997. Subsequently, distinct cell populations termed endothelial colony-forming units-Hill, circulating angiogenic cells and endothelial colony-forming cells were identified that varied in terms of phenotype, vascular homeostasis contribution and purity. Notably, most of these cells are not genuine vascular precursor cells belonging to the endothelial lineage. This review provides a broad overview of the main properties of the endothelium, focusing on the basis governing its growth and repair. We discuss efforts to identify true vascular precursors, a matter of debate for the past 15 years, as well as recent methodological advances in identifying new hierarchies of more homogeneous, clonogenic and proliferative vascular endothelial-lineage precursors. Consideration of these issues provides insights that may help develop more effective therapies against human diseases that involve vascular deficits.
Collapse
Affiliation(s)
- Santiago Roura
- ICREC Research Program, Health Research Institute Germans Trias i Pujol-IGTP, University Hospital Germans Trias i Pujol, Badalona, Spain.
| | | | | |
Collapse
|
27
|
Ichim TE, Warbington T, Cristea O, Chin JL, Patel AN. Intracavernous administration of bone marrow mononuclear cells: a new method of treating erectile dysfunction? J Transl Med 2013; 11:139. [PMID: 23758954 PMCID: PMC3718667 DOI: 10.1186/1479-5876-11-139] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 04/23/2013] [Indexed: 02/06/2023] Open
Abstract
While PDE5 inhibitors have revolutionized treatment of ED, approximately 30% of patients are non-responsive. A significant cause of this is vascular and smooth muscle dysfunction, as well as nerve atrophy. Autologous administration of bone marrow mononuclear cells (BMMC) has been performed in over 2000 cardiac patients without adverse effects, for stimulation of angiogenesis/regeneration. Despite its ease of access, and dependence on effective vasculature for function, comparatively little has been perform in terms of BMMC therapy for ED. Here we outline the rationale for use of autologous BMMC in patients with ED, as well as provide early safety data on the first use of this procedure clinically.
Collapse
Affiliation(s)
- Thomas E Ichim
- Institute for Molecular Medicine, Huntington Beach, CA, USA.
| | | | | | | | | |
Collapse
|
28
|
Campioni D, Zauli G, Gambetti S, Campo G, Cuneo A, Ferrari R, Secchiero P. In vitro characterization of circulating endothelial progenitor cells isolated from patients with acute coronary syndrome. PLoS One 2013; 8:e56377. [PMID: 23409178 PMCID: PMC3569417 DOI: 10.1371/journal.pone.0056377] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Accepted: 01/07/2013] [Indexed: 02/07/2023] Open
Abstract
Background The current understanding of the functional characteristics of circulating endothelial progenitor cells (EPC) is limited, especially in patients affected by cardiovascular diseases. In this study, we have analyzed the in vitro clonogenic capacity of circulating EPC, also known as endothelial colony-forming cells (ECFC), in patients with acute coronary syndrome (ACS), in comparison to the colony forming unit-endothelial-like cells (CFU-EC) of hematopoietic/monocytic origin. Methodology/Principal Findings By culturing peripheral blood mononuclear cells (PBMC) of patients with ACS (n = 70), CFU-EC were frequently isolated (from 77% of ACS patients), while EPC/ECFC were obtained only in a small subset (13%) of PBMC samples, all harvested between 7–14 days after the acute cardiovascular event. Notably, ex-vivo generation of EPC/ECFC was correlated to a higher in vitro release of PDGF-AA by the corresponding ACS patient PBMC. By using specific endothelial culture media, EPC/ECFC displayed in vitro expansion capacity, allowing the phenotypic and functional characterization of the cells. Indeed, after expansion, EPC/ECFC exhibited a normal diploid chromosomal setting by FISH analysis and an immunophenotype characterized by: i) uniform positivity for the expression of CD105, CD31, CD146 and Factor VIII, i) variable expression of the CD34, CD106 and CD184 markers, and iii) negativity for CD45, CD90, CD117 and CD133. Of interest, in single-cell replanting assays EPC/ECFC exhibited clonogenic expansion capacity, forming secondary colonies characterized by variable proliferation capacities. Conclusion/Significance Our data indicate that a careful characterization of true EPC is needed in order to design future studies in the clinical autologous setting of patients with ACS.
Collapse
Affiliation(s)
- Diana Campioni
- Department of Medical Sciences, Section of Hematology, Azienda Ospedaliero-Universitaria, Arcispedale Sant’Anna, University of Ferrara, Ferrara, Italy
| | - Giorgio Zauli
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Stefania Gambetti
- Department of Medical Sciences, Cardiovascular Section, Azienda Ospedaliero-Universitaria, Arcispedale Sant’Anna, University of Ferrara, Ferrara, Italy
| | - Gianluca Campo
- Department of Medical Sciences, Cardiovascular Section, Azienda Ospedaliero-Universitaria, Arcispedale Sant’Anna, University of Ferrara, Ferrara, Italy
| | - Antonio Cuneo
- Department of Medical Sciences, Section of Hematology, Azienda Ospedaliero-Universitaria, Arcispedale Sant’Anna, University of Ferrara, Ferrara, Italy
| | - Roberto Ferrari
- Department of Medical Sciences, Cardiovascular Section, Azienda Ospedaliero-Universitaria, Arcispedale Sant’Anna, University of Ferrara, Ferrara, Italy
| | - Paola Secchiero
- Department of Morphology and Embryology and LTTA Centre, University of Ferrara, Ferrara, Italy
- * E-mail:
| |
Collapse
|
29
|
Differential expression of Tie2 receptor and VEGFR2 by endothelial clones derived from isolated bovine mononuclear cells. PLoS One 2012; 7:e53385. [PMID: 23300924 PMCID: PMC3534049 DOI: 10.1371/journal.pone.0053385] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 11/27/2012] [Indexed: 12/12/2022] Open
Abstract
The purpose of these experiments was to evaluate the expression of endothelial markers, such as Tie2 and VEGFR2 in endothelial cells derived from blood mononuclear endothelial progenitor cells. Bovine mononuclear cells were isolated using separation by centrifugation and were grown in endothelial specific media supplemented with growth factors. Isolation of the whole cell population of mononuclear cells (MNC) from bovine peripheral blood gave rise to progenitor-like cells (CD45−) that, although morphologically similar, have different phenotypes revealed by expression of endothelial specific markers Tie2 and VEGFR2. Plating of MNCs on collagen and fibronectin gave rise to more colonies than non-coated dishes. Occasional colonies from MNC isolations had a mural cell phenotype, negative for Tie2 and VEGFR2 but positive for smooth muscle actin and PDGFRβ. Although cells expressing high levels of VEGFR2 and low levels of Tie2, and vice versa were both able to form cords on Matrigel, cells with higher expression of Tie2 migrate faster in a scratch assay than ones with lower expression of Tie2. When these different clones of cells were introduced in mice through tail vein injections, they retained an ability to home to angiogenesis occurring in a subcutaneous Matrigel plug, regardless of their Tie2/VEGFR2 receptor expression patterns, but cells with high VEGFR2/low Tie2 were more likely to be CD31 positive. Therefore, we suggest that active sites of angiogenesis (such as wounds, tumors, etc.) can attract a variety of endothelial cell precursors that may differentially express Tie2 and VEGFR2 receptors, and thus affect our interpretation of EPCs as biomarkers or therapies for vascular disease.
Collapse
|
30
|
An emerging cell-based strategy in orthopaedics: endothelial progenitor cells. Knee Surg Sports Traumatol Arthrosc 2012; 20:1366-77. [PMID: 22402606 DOI: 10.1007/s00167-012-1940-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 12/15/2011] [Indexed: 12/17/2022]
Abstract
PURPOSE The purpose of this article was to analyze the results of studies in the literature, which evaluated the use of endothelial progenitor cells (EPCs) as a cell-based tissue engineering strategy. METHODS EPCs have been successfully used in regenerative medicine to augment neovascularization in patients after myocardial infarction and limb ischemia. EPCs' important role as vasculogenic progenitors presents them as a potential source for cell-based therapies to promote bone healing. RESULTS EPCs have been shown to have prominent effects in promoting bone regeneration in several animal models. Evidence indicates that EPCs promote bone regeneration by stimulating both angiogenesis and osteogenesis through a differentiation process toward endothelial cell lineage and formation of osteoblasts. Moreover, EPCs increase vascularization and osteogenesis by increased secretion of growth factors and cytokines through paracrine mechanisms. CONCLUSION EPCs offer the potential to emerge as a new strategy among other cell-based therapies to promote bone regeneration. Further investigations and human trials are required to address current questions with regard to biology and mechanisms of action of EPCs in bone tissue engineering.
Collapse
|
31
|
Comparative angiogenic activities of induced pluripotent stem cells derived from young and old mice. PLoS One 2012; 7:e39562. [PMID: 22761825 PMCID: PMC3384644 DOI: 10.1371/journal.pone.0039562] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 05/21/2012] [Indexed: 11/28/2022] Open
Abstract
Advanced age is associated with decreased stem cell activity. However, the effect of aging on the differentiation capacity of induced pluripotent stem (iPS) cells into cardiovascular cells has not been fully clarified. We investigated whether iPS cells derived from young and old mice are equally capable of differentiating into vascular progenitor cells, and whether these cells regulate vascular responses in vivo. iPS cells from mouse embryonic fibroblasts (young) or 21 month-old mouse bone marrow (old) were used. Fetal liver kinase-1 positive (Flk-1+) cells, as a vascular progenitor marker, were induced after 3 to 4 days of culture from iPS cells derived from young and old mice. These Flk-1+ cells were sorted and shown to differentiate into VE-cadherin+ endothelial cells and α-SMA+ smooth muscle cells. Tube-like formation was also successfully induced in both young and old murine Flk-1+ cells. Next, hindlimb ischemia was surgically induced, and purified Flk-1+ cells were directly injected into ischemic hindlimbs of nude mice. Revascularization of the ischemic hindlimb was significantly accelerated in mice transplanted with Flk-1+ cells derived from iPS cells from either young or old mice, as compared to control mice as evaluated by laser Doppler blood flowmetry. The degree of revascularization was similar in the two groups of ischemic mice injected with iPS cell-derived Flk-1+ cells from young or old mice. Transplantation of Flk-1+ cells from both young and old murine iPS cells also increased the expression of VEGF, HGF and IGF mRNA in ischemic tissue as compared to controls. iPS cell-derived Flk-1+ cells differentiated into vascular progenitor cells, and regulated angiogenic vascular responses both in vitro and in vivo. These properties of iPS cells derived from old mice are essentially the same as those of iPS cells from young mice, suggesting the functionality of generated iPS cells themselves to be unaffected by aging.
Collapse
|
32
|
Baumer Y, Leder C, Ziegler M, Schönberger T, Ochmann C, Perk A, Degen H, Schmid-Horch B, Elvers M, Münch G, Ungerer M, Schlosshauer B, Gawaz M. The recombinant bifunctional protein αCD133-GPVI promotes repair of the infarcted myocardium in mice. J Thromb Haemost 2012; 10:1152-64. [PMID: 22448969 DOI: 10.1111/j.1538-7836.2012.04710.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Bone-marrow-derived progenitor cells are important in myocardial repair mechanisms following prolonged ischemia. Cell-based therapy of diseased myocardium is limited by a low level of tissue engraftment. OBJECTIVES The aim of this study was the development of the bifunctional protein αCD133-glycoprotein (GP)VI as an effective treatment for supporting vascular and myocardial repair mechanisms. RESULTS We have generated and characterized a bifunctional molecule (αCD133-GPVI) that binds both to the subendothelium of the injured microvasculature and to CD133(+) progenitor cells with high affinity. αCD133-GPVI enhances progenitor cell adhesion to extracellular matrix proteins and differentiation into mature endothelial cells. In vivo studies showed that αCD133-GPVI favors adhesion of circulating progenitor cells to the injured vessel wall (intravital microscopy). Also, treatment of mice undergoing experimental myocardial infarction with αCD133-GPVI-labeled progenitor cells reduces infarction size and preserves myocardial function. CONCLUSIONS The bifunctional trapping protein αCD133-GPVI represents a novel and promising therapeutic option for limiting heart failure of the ischemic myocardium.
Collapse
Affiliation(s)
- Y Baumer
- NMI, Natural and Medical Sciences Institute at the University of Tübingen, Regenerative Medicine I, Reutlingen University Clinic of Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Rodriguez JP, Murphy MP, Hong S, Madrigal M, March KL, Minev B, Harman RJ, Chen CS, Timmons RB, Marleau AM, Riordan NH. Autologous stromal vascular fraction therapy for rheumatoid arthritis: rationale and clinical safety. Int Arch Med 2012; 5:5. [PMID: 22313603 PMCID: PMC3296619 DOI: 10.1186/1755-7682-5-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Accepted: 02/08/2012] [Indexed: 02/08/2023] Open
Abstract
Advancements in rheumatoid arthritis (RA) treatment protocols and introduction of targeted biological therapies have markedly improved patient outcomes, despite this, up to 50% of patients still fail to achieve a significant clinical response. In veterinary medicine, stem cell therapy in the form of autologous stromal vascular fraction (SVF) is an accepted therapeutic modality for degenerative conditions with 80% improvement and no serious treatment associated adverse events reported. Clinical translation of SVF therapy relies on confirmation of veterinary findings in targeted patient populations. Here we describe the rationale and preclinical data supporting the use of autologous SVF in treatment of RA, as well as provide 1, 3, 6, and 13 month safety outcomes in 13 RA patients treated with this approach.
Collapse
|
34
|
Shelton EL, Poole SD, Reese J, Bader DM. Omental grafting: a cell-based therapy for blood vessel repair. J Tissue Eng Regen Med 2012; 7:421-33. [PMID: 22318999 DOI: 10.1002/term.528] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Revised: 07/18/2011] [Accepted: 09/26/2011] [Indexed: 01/15/2023]
Abstract
Clinicians regularly transplant omental pedicles to repair a wide variety of injured tissues, but the basic mechanism underlying this efficacious procedure is not understood. One possibility that has not been addressed is the ability of omentum to directly contribute regenerative cells to injured tissues. We hypothesized that if omental progenitor cells could be mobilized to incorporate into damaged tissue, the power of this therapy would be greatly expanded. Labelled omental grafts were transplanted into a murine carotid artery injury model. Selected grafts were treated with thymosin β4 (Tβ4) prior to transplantation to investigate the effects of chemical potentiation on healing. We found treatment of grafts with Tβ4-induced progenitor cells to fully integrate into the wall of injured vessels and differentiate into vascular smooth muscle. Myographic studies determined that arteries receiving Tβ4-stimulated grafts were functionally indistinguishable from uninjured controls. Concurrent in vitro analyses showed that Tβ4 promoted proliferation, migration and trans-differentiation of cells via AKT signalling. This study is the first to demonstrate that omentum can provide progenitor cells for repair, thus revealing a novel and naturally occurring source of vascular smooth muscle for use in cell-based therapies. Furthermore, our data show that this system can be optimized with inducing factors, highlighting a more powerful therapeutic potential than that of its current clinical application. This is a paradigm-setting concept that lays the foundation for the use of chemical genetics to enhance therapeutic outcomes in a myriad of fields.
Collapse
Affiliation(s)
- Elaine L Shelton
- Stahlman Cardiovascular Research Laboratories, Program for Developmental Biology and Department of Medicine, Vanderbilt University, Medical Center, Nashville, TN, USA
| | | | | | | |
Collapse
|
35
|
Barrett JM, Parham KA, Pippal JB, Cockshell MP, Moretti PAB, Brice SL, Pitson SM, Bonder CS. Over-expression of sphingosine kinase-1 enhances a progenitor phenotype in human endothelial cells. Microcirculation 2012; 18:583-97. [PMID: 21672077 DOI: 10.1111/j.1549-8719.2011.00119.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
OBJECTIVES The use of endothelial progenitor cells in vascular therapies has been limited due to their low numbers present in the bone marrow and peripheral blood. The aim of this study was to investigate the effect of sphingosine kinase on the de-differentiation of mature human endothelial cells toward a progenitor phenotype. METHODS The lipid enzyme sphingosine kinase-1 was lentivirally over-expressed in human umbilical vein endothelial cells and cells were analyzed for progenitor phenotype and function. RESULTS Sphingosine kinase-1 mRNA expression was induced approximately 150-fold with a resultant 20-fold increase in sphingosine kinase-1 enzymatic activity. The mRNA expression of the progenitor cell markers CD34, CD133, and CD117 and transcription factor NANOG increased, while the endothelial cell markers analyzed were largely unchanged. The protein level of mature endothelial cell surface markers CD31, CD144, and von Willebrand factor significantly decreased compared to controls. In addition, functional assays provided further evidence for a de-differentiated phenotype with increased viability, reduced uptake of acetylated low-density lipoprotein and decreased tube formation in Matrigel in the cells over-expressing sphingosine kinase-1. CONCLUSIONS These findings suggest that over-expression of sphingosine kinase-1 in human endothelial cells promotes, in part, their de-differentiation to a progenitor cell phenotype, and is thus a potential tool for the generation of a large population of vascular progenitor cells for therapeutic use.
Collapse
Affiliation(s)
- Jeffrey M Barrett
- Human Immunology, Centre for Cancer Biology, SA Pathology, Adelaide, SA, Australia
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Mund JA, Estes ML, Yoder MC, Ingram DA, Case J. Flow cytometric identification and functional characterization of immature and mature circulating endothelial cells. Arterioscler Thromb Vasc Biol 2012; 32:1045-53. [PMID: 22282356 DOI: 10.1161/atvbaha.111.244210] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE We sought to identify and characterize 2 distinct populations of bona fide circulating endothelial cells, including the endothelial colony-forming cell (ECFC), by polychromatic flow cytometry (PFC), colony assays, immunomagnetic selection, and electron microscopy. METHODS AND RESULTS Mononuclear cells from human umbilical cord blood and peripheral blood were analyzed using our recently published PFC protocol. A population of cells containing both ECFCs and mature circulating endothelial cells was determined by varying expressions of CD34, CD31, and CD146 but not AC133 and CD45. After immunomagnetic separation, these cells failed to form hematopoietic colonies, yet clonogenic endothelial colonies with proliferative potential were obtained, thus verifying their identity as ECFCs. The frequency of ECFCs were increased in cord blood and were extremely rare in the peripheral blood of healthy adults. We also detected another mature endothelial cell population in the circulation that was apoptotic. Finally, when comparing this new protocol with a prior method, we determined that the present protocol identifies circulating endothelial cells, whereas the earlier protocol identified extracellular vesicles. CONCLUSIONS Two populations of circulating endothelial cells, including the functionally characterized ECFC, are now identifiable in human cord blood and peripheral blood by PFC.
Collapse
Affiliation(s)
- Julie A Mund
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, 46202, USA
| | | | | | | | | |
Collapse
|
37
|
Smadja DM, Duong-van-Huyen JP, Dal Cortivo L, Blanchard A, Bruneval P, Emmerich J, Gaussem P. Early endothelial progenitor cells in bone marrow are a biomarker of cell therapy success in patients with critical limb ischemia. Cytotherapy 2011; 14:232-9. [PMID: 22040109 DOI: 10.3109/14653249.2011.627917] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND AIMS Endothelial progenitor cells (EPC) have been proposed for autologous angiogenic therapy. The objectives of this study were to quantify EPC in the peripheral blood and bone marrow mononuclear cells (BM-MNC) of patients with critical limb ischemia that had received BM-MNC as a cell therapy product, and to study the putative relationship between the presence of EPC and the process of neovascularization in toe or transmetatarsal amputation specimens. METHODS Early and late endothelial progenitor cells (CFU-EC and ECFC) were cultivated and quantified according to published methods in peripheral blood and BM-MNC from patients with critical limb ischemia (CLI; n = 11) enrolled in the OPTIPEC trial ( http://clinicaltrials.gov/ct2/show/NCT00377897 ) to receive BM-MNC as a cell therapy product. RESULTS Eight out of the 11 patients had undergone amputations. Three of the patients displayed a neoangiogenic process that was associated with a higher number of CFU-EC in BM-MNC, while CD3+ , CFU-GM and CD34+ in BM-MNC, and EPC in peripheral blood, did not correlate with the appearance of newly formed vessels. As expected, circulating CFU-EC and ECFC counts were significantly lower in CLI patients compared with age-matched controls. CONCLUSIONS In patients with critical limb ischemia, EPC in peripheral blood were decreased compared with healthy individuals. However, in BM-MNC we found that relative numbers of CFU-EC could be used as an indicator to discriminate patients with neoangiogenic processes. These results need to be confirmed in a randomized study.
Collapse
Affiliation(s)
- David M Smadja
- Université Paris Descartes, Paris, France Sorbonne Paris Cite, France.
| | | | | | | | | | | | | |
Collapse
|
38
|
Borlongan CV, Glover LE, Tajiri N, Kaneko Y, Freeman TB. The great migration of bone marrow-derived stem cells toward the ischemic brain: therapeutic implications for stroke and other neurological disorders. Prog Neurobiol 2011; 95:213-28. [PMID: 21903148 PMCID: PMC3185169 DOI: 10.1016/j.pneurobio.2011.08.005] [Citation(s) in RCA: 163] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 08/10/2011] [Accepted: 08/15/2011] [Indexed: 02/08/2023]
Abstract
Accumulating laboratory studies have implicated the mobilization of bone marrow (BM)-derived stem cells in brain plasticity and stroke therapy. This mobilization of bone cells to the brain is an essential concept in regenerative medicine. Over the past ten years, mounting data have shown the ability of bone marrow-derived stem cells to mobilize from BM to the peripheral blood (PB) and eventually enter the injured brain. This homing action is exemplified in BM stem cell mobilization following ischemic brain injury. Various BM-derived cells, such as hematopoietic stem cells (HSCs), mesenchymal stem cells (MSCs), endothelial progenitor cells (EPCs) and very small embryonic-like cells (VSELs) have been demonstrated to exert therapeutic benefits in stroke. Here, we discuss the current status of these BM-derived stem cells in stroke therapy, with emphasis on possible cellular and molecular mechanisms of action that mediate the cells' beneficial effects in the ischemic brain. When possible, we also discuss the relevance of this therapeutic regimen in other central nervous system (CNS) disorders.
Collapse
Affiliation(s)
- Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd, Tampa, FL 33612, USA.
| | | | | | | | | |
Collapse
|
39
|
Grapensparr L, Olerud J, Vasylovska S, Carlsson PO. The therapeutic role of endothelial progenitor cells in Type 1 diabetes mellitus. Regen Med 2011; 6:599-605. [DOI: 10.2217/rme.11.45] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Pancreatic β-cells sense and adjust the blood glucose level by secretion of insulin. In Type 1 diabetes mellitus, these insulin-producing cells are destroyed, leaving the patients incapable of regulating blood glucose homeostasis. At the time of diagnosis, most patients still have 20–30% of their original β-cell mass remaining. These residual β-cells are targets for intervention therapies aimed at preventing further autoimmune destruction, in addition to increasing the number of existing β-cells. Such a therapeutic option is highly desirable since it may lead to a full recovery of newly diagnosed patients, with no need for further treatment with immunosuppressant drugs or exogenous insulin administration. In this article, we propose that endothelial progenitor cells, a cell type known to promote and support neovascularization following endothelial injury, may be used as part of a combinational stem cell therapy aimed to improve the vascularization, survival and proliferation of β-cells.
Collapse
Affiliation(s)
- Liza Grapensparr
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Johan Olerud
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Svitlana Vasylovska
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Per-Ola Carlsson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
40
|
Ergün S, Tilki D, Klein D. Vascular wall as a reservoir for different types of stem and progenitor cells. Antioxid Redox Signal 2011; 15:981-95. [PMID: 20712422 DOI: 10.1089/ars.2010.3507] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tissue regeneration and several diseases such as tumor and atherosclerosis depend on new vessel formation by both angiogenesis and vasculogenesis. Endothelial cells (ECs) are widely considered to be the active cellular component in these processes, followed by contractile cells such as pericytes and smooth muscle cells. The best known sources providing these cell types or their progenitors are ECs lining the vessel lumen and bone marrow. As easily evident, the vessel wall was recognized as being a passive player to a great extent except ECs of the vascular intima. Particularly, the vascular adventitia has been considered as a passive layer rather than an active part of the vessel wall. But results provided during the last few years have led to a revision of this classical view because of an apparent stem cell niche function of the vascular adventitia. This review aims to sum up findings identifying the vessel wall as an important stem cell reservoir and discusses its impact on health and disease.
Collapse
Affiliation(s)
- Süleyman Ergün
- Institute of Anatomy, University Hospital Essen, Essen, Germany.
| | | | | |
Collapse
|
41
|
Borlongan CV. Bone marrow stem cell mobilization in stroke: a 'bonehead' may be good after all! Leukemia 2011; 25:1674-86. [PMID: 21727900 DOI: 10.1038/leu.2011.167] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mobilizing bone cells to the head, astutely referred to as 'bonehead' therapeutic approach, represents a major discipline of regenerative medicine. The last decade has witnessed mounting evidence supporting the capacity of bone marrow (BM)-derived cells to mobilize from BM to peripheral blood (PB), eventually finding their way to the injured brain. This homing action is exemplified in BM stem cell mobilization following ischemic brain injury. Here, I review accumulating laboratory studies implicating the role of therapeutic mobilization of transplanted BM stem cells for brain plasticity and remodeling in stroke.
Collapse
Affiliation(s)
- C V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA.
| |
Collapse
|
42
|
Kaposi’s sarcoma-associated herpesvirus infection of endothelial progenitor cells impairs angiogenic activity in vitro. J Microbiol 2011; 49:299-304. [DOI: 10.1007/s12275-011-0408-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Accepted: 11/29/2010] [Indexed: 10/18/2022]
|
43
|
Tang WL, Guo H, Yang J, Chen B, Wang X. Suppression of tissue inhibitors of metalloproteinases may reverse severe pulmonary arterial hypertension. Cytotherapy 2011; 13:499-502. [DOI: 10.3109/14653249.2010.536215] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
44
|
|
45
|
Capobianco S, Chennamaneni V, Mittal M, Zhang N, Zhang C. Endothelial progenitor cells as factors in neovascularization and endothelial repair. World J Cardiol 2010; 2:411-20. [PMID: 21191542 PMCID: PMC3011136 DOI: 10.4330/wjc.v2.i12.411] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Revised: 09/20/2010] [Accepted: 09/27/2010] [Indexed: 02/06/2023] Open
Abstract
Endothelial progenitor cells (EPCs) are a heterogeneous population of cells that are provided by the bone marrow and other adult tissue in both animals and humans. They express both hematopoietic and endothelial surface markers, which challenge the classic dogma that the presumed differentiation of cells into angioblasts and subsequent endothelial and vascular differentiation occurred exclusively in embryonic development. This breakthrough stimulated research to understand the mechanism(s) underlying their physiologic function to allow development of new therapeutic options. One focus has been on their ability to form new vessels in injured tissues, and another has been on their ability to repair endothelial damage and restore both monolayer integrity and endothelial function in denuded vessels. Moreover, measures of their density have been shown to be a better predictor of cardiovascular events, both in healthy and coronary artery disease populations than the classical tools used in the clinic to evaluate the risk stratification. In the present paper we review the effects of EPCs on revascularization and endothelial repair in animal models and human studies, in an attempt to better understand their function, which may lead to potential advancement in clinical management.
Collapse
Affiliation(s)
- Stefano Capobianco
- Stefano Capobianco, Department of Cardiology, Gaetano Rummo Hospital, Via Dell'Angelo 1, 82100 Benevento, Italy
| | | | | | | | | |
Collapse
|
46
|
|
47
|
Abstract
Endothelial cells provide the dynamic lining of blood vessels throughout the body and provide many tissue-specific functions, in addition to providing a nonthrombogenic surface for blood cells and conduit for oxygen and nutrient delivery. As might be expected, some endothelial cells are injured or become senescent and are sloughed into the bloodstream, and most circulating endothelial cells display evidence of undergoing apoptosis or necrosis. However, there are rare viable circulating endothelial cells that display properties consistent with those of a progenitor cell for the endothelial lineage. This article reviews historical and current literature to present some evidence that the endothelial lining of blood vessels may serve as a source for rare endothelial colony-forming cells that display clonal proliferative potential, self-renewal, and in vivo vessel forming ability. The article also discusses the current gaps in our knowledge to prove whether the colony-forming cells are in fact derived from vascular endothelium.
Collapse
Affiliation(s)
- Mervin C. Yoder
- From Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Ind
| |
Collapse
|
48
|
Kumar AHS, Caplice NM. Clinical potential of adult vascular progenitor cells. Arterioscler Thromb Vasc Biol 2010; 30:1080-7. [PMID: 20453166 DOI: 10.1161/atvbaha.109.198895] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Cell therapy to treat vascular and cardiovascular diseases has evolved over the past decade with improved understanding of progenitor cell mobilization, recruitment, and differentiation. The beneficial effects seen in several preclinical studies have prompted translation of adult vascular progenitor therapy to clinical trials. To date, progenitor cells isolated from bone marrow and peripheral blood have been tested in the context of acute myocardial infarction and chronic ischemic cardiomyopathy, with moderate benefit. This therapeutic effect occurs despite a relatively small number of injected progenitor cells and short-term residence in the target zone. Thus, indirect benefits, such as paracrine factors released from these cells, have been suggested as significant contributors to therapeutic efficacy. Several additional vascular progenitors of endothelial, smooth muscle, mesenchymal, and cardiac origin have been identified that may contribute to vasculogenesis. Indeed, a unifying paradigm for the most effective cell therapy strategies to date appears to be robust support of angiogenesis. Here we discuss a number of progenitor cells that currently show potential as cardiovascular therapeutics, either singly or in combination. We look at emerging cell types and disease targets that may be exploited for therapeutic benefit and future strategies that may maximize clinical efficacy.
Collapse
Affiliation(s)
- Arun H S Kumar
- Centre for Research in Vascular Biology, Biosciences Institute, University College Cork, Cork, Ireland
| | | |
Collapse
|
49
|
Senegaglia AC, Barboza LA, Dallagiovanna B, Aita CAM, Hansen P, Rebelatto CLK, Aguiar AM, Miyague NI, Shigunov P, Barchiki F, Correa A, Olandoski M, Krieger MA, Brofman PRS. Are purified or expanded cord blood-derived CD133+ cells better at improving cardiac function? Exp Biol Med (Maywood) 2010; 235:119-29. [PMID: 20404026 DOI: 10.1258/ebm.2009.009194] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Endothelial progenitor cells (EPCs), which express the CD133 marker, can differentiate into mature endothelial cells (ECs) and create new blood vessels. Normal angiogenesis is unable to repair the injured tissues that result from myocardial infarction (MI). Patients who have high cardiovascular risks have fewer EPCs and their EPCs exhibit greater in vitro senescence. Human umbilical cord blood (HUCB)-derived EPCs could be an alternative to rescue impaired stem cell function in the sick and elderly. The aim of this study was to purify HUCB-derived CD133(+) cells, expand them in vitro and evaluate the efficacy of the purified and expanded cells in treating MI in rats. CD133(+) cells were selected for using CD133-coupled magnetic microbeads. Purified cells stained positive for EPC markers. The cells were expanded and differentiated in media supplemented with fetal calf serum and basic fibroblast growth factor, insulin-like growth factor-I and vascular endothelial growth factor (VEGF). Differentiation was confirmed by lack of staining for EPC markers. These expanded cells exhibited increased expression of mature EC markers and formed tubule-like structures in vitro. Only the expanded cells expressed VEGF mRNA. Cells were expanded up to 70-fold during 60 days of culture, and they retained their functional activity. Finally, we evaluated the therapeutic potential of purified and expanded CD133(+) cells in treating MI by intramyocardially injecting them into a rat model of MI. Rats were divided into three groups: A (purified CD133(+) cells-injected); B (expanded CD133(+) cells-injected) and C (saline buffer-injected). We observed a significant improvement in left ventricular ejection fraction for groups A and B. In summary, CD133(+) cells can be purified from HUCB, expanded in vitro without loosing their biological activity, and both purified and expanded cells show promising results for use in cellular cardiomyoplasty. However, further pre-clinical testing should be performed to determine whether expanded CD133(+) cells have any clinical advantages over purified CD133(+) cells.
Collapse
Affiliation(s)
- Alexandra C Senegaglia
- Pontifícia Universidade Católica do Paraná, Institute for Health and Biological Sciences, Rua Imaculada Conceição, 1155 Curitiba, Paraná, 80215901, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Atorvastatin increases the number of endothelial progenitor cells after cardiac surgery: a randomized control study. J Cardiovasc Pharmacol 2010; 55:30-8. [PMID: 19834333 DOI: 10.1097/fjc.0b013e3181c37d4d] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Endothelial progenitor cells (EPCs) are a subtype of hematopoietic stem cells, which contribute to the repair of injured endothelium. Treatment with atorvastatin has been shown to increase EPC count in patients with coronary artery disease. Therefore, we investigated whether atorvastatin augments the number of EPCs after cardiopulmonary bypass (CPB) surgery. We conducted a randomized double-blind, placebo-controlled, 2-way crossover trial in 50 patients undergoing elective coronary surgery. Patients received either 3-week treatment with atorvastatin or placebo. EPCs were quantitated by flow cytometric phenotyping on blood samples. Levels of interleukin, IL-6 and IL-8; tumor necrosis factor alpha; SDF-1alpha; granulocyte colony-stimulating factor; and vascular endothelial growth factor were determined at recruitment, preoperatively, post-CPB, and 6, 12, and 24 hours postoperatively. The atorvastatin group showed a significantly higher amount of EPCs both pre- and postoperatively compared with the placebo, with a >4-fold increase compared with the baseline values. CPB induced an increase in all cytokines, but the levels of proinflammatory cytokines were significantly lower in the atorvastatin group (P < 0.05). Statin did not affect levels of SDF-1alpha, granulocyte colony-stimulating factor, and vascular endothelial growth factor. However, no correlation was found between plasma levels of any cytokine and number of EPCs, with the exception of SDF-1alpha. Pretreatment with atorvastatin significantly increases the amount of EPCs after CPB, by a mechanism independent of plasma levels of cytokines and cholesterol.
Collapse
|