51
|
Feitz WJC, van de Kar NCAJ, Cheong I, van der Velden TJAM, Ortiz-Sandoval CG, Orth-Höller D, van den Heuvel LPJW, Licht C. Primary Human Derived Blood Outgrowth Endothelial Cells: An Appropriate In Vitro Model to Study Shiga Toxin Mediated Damage of Endothelial Cells. Toxins (Basel) 2020; 12:toxins12080483. [PMID: 32751286 PMCID: PMC7472281 DOI: 10.3390/toxins12080483] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/19/2020] [Accepted: 07/21/2020] [Indexed: 11/30/2022] Open
Abstract
Hemolytic uremic syndrome (HUS) is a rare disease primarily characterized by hemolytic anemia, thrombocytopenia, and acute renal failure. Endothelial damage is the hallmark of the pathogenesis of HUS with an infection with the Shiga toxin (Stx) producing Escherichia coli (STEC-HUS) as the main underlying cause in childhood. In this study, blood outgrowth endothelial cells (BOECs) were isolated from healthy donors serving as controls and patients recovered from STEC-HUS. We hypothesized that Stx is more cytotoxic for STEC-HUS BOECs compared to healthy donor control BOECs explained via a higher amount of Stx bound to the cell surface. Binding of Shiga toxin-2a (Stx2a) was investigated and the effect on cytotoxicity, protein synthesis, wound healing, and cell proliferation was studied in static conditions. Results show that BOECs are highly susceptible for Stx2a. Stx2a is able to bind to the cell surface of BOECs with cytotoxicity in a dose-dependent manner as a result. Pre-treatment with tumor necrosis factor alpha (TNF-α) results in enhanced Stx binding with 20–30% increased lactate dehydrogenase (LDH) release. Endothelial wound healing is delayed in a Stx2a-rich environment; however, this is not caused by an effect on the proliferation rate of BOECs. No significant differences were found between control BOECs and BOECs from recovered STEC-HUS patients in terms of Stx2a binding and inhibition of protein synthesis.
Collapse
Affiliation(s)
- Wouter J. C. Feitz
- Department of Pediatric Nephrology, Amalia Children’s Hospital, Radboud Institute for Molecular Life Sciences, Radboudumc, 6525 GA Nijmegen, The Netherlands; (W.J.C.F.); (N.C.A.J.v.d.K.); (T.J.A.M.v.d.V.); (L.P.J.W.v.d.H.)
- Cell Biology Program, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; (I.C.); (C.G.O.-S.)
| | - Nicole C. A. J. van de Kar
- Department of Pediatric Nephrology, Amalia Children’s Hospital, Radboud Institute for Molecular Life Sciences, Radboudumc, 6525 GA Nijmegen, The Netherlands; (W.J.C.F.); (N.C.A.J.v.d.K.); (T.J.A.M.v.d.V.); (L.P.J.W.v.d.H.)
| | - Ian Cheong
- Cell Biology Program, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; (I.C.); (C.G.O.-S.)
| | - Thea J. A. M. van der Velden
- Department of Pediatric Nephrology, Amalia Children’s Hospital, Radboud Institute for Molecular Life Sciences, Radboudumc, 6525 GA Nijmegen, The Netherlands; (W.J.C.F.); (N.C.A.J.v.d.K.); (T.J.A.M.v.d.V.); (L.P.J.W.v.d.H.)
| | - Carolina G. Ortiz-Sandoval
- Cell Biology Program, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; (I.C.); (C.G.O.-S.)
| | - Dorothea Orth-Höller
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - Lambert P. J. W. van den Heuvel
- Department of Pediatric Nephrology, Amalia Children’s Hospital, Radboud Institute for Molecular Life Sciences, Radboudumc, 6525 GA Nijmegen, The Netherlands; (W.J.C.F.); (N.C.A.J.v.d.K.); (T.J.A.M.v.d.V.); (L.P.J.W.v.d.H.)
- Department of Development and Regeneration, Department of Pediatric Nephrology, KU, 3000 Leuven, Belgium
| | - Christoph Licht
- Cell Biology Program, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; (I.C.); (C.G.O.-S.)
- Division of Nephrology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
- Department of Pediatrics, University of Toronto, Toronto, ON M5G 1X8, Canada
- Correspondence: ; Tel.: +1-416-813-7654 (ext. 309343)
| |
Collapse
|
52
|
Tripathi DM, Hassan M, Siddiqui H, Kaur I, Rawal P, Bihari C, Kaur S, Sarin SK. Cirrhotic Endothelial Progenitor Cells Enhance Liver Angiogenesis and Fibrosis and Aggravate Portal Hypertension in Bile Duct-Ligated Cirrhotic Rats. Front Physiol 2020; 11:617. [PMID: 32595520 PMCID: PMC7300214 DOI: 10.3389/fphys.2020.00617] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 05/15/2020] [Indexed: 12/21/2022] Open
Abstract
Background Circulating cirrhotic endothelial progenitor cells (EPC) interact with both liver sinusoidal endothelial cells (LSEC) and hepatic stellate cells (HSC) and promote angiogenesis in vitro. This study evaluated the effect of cirrhotic and control EPCs on hepatic angiogenesis, microcirculation, and fibrosis in vivo in rat models of cirrhosis. Methodology Animal models of cirrhosis were prepared by bile duct ligation (BDL). Circulating EPCs isolated from healthy human and cirrhotic blood were characterized by flow cytometry, cultured and administered through the tail vein in BDL rats after 2 weeks of ligation. The cells were given thrice a week for 2 weeks. The untreated group of BDL rats received only saline. Fibrosis was evaluated by Masson’s trichrome staining. Dedifferentiated LSECs were identified by the expression of CD31, and activated HSCs were marked as alpha-SMA-positive cells and were studied by immunohistochemistry and western blotting in saline-, healthy EPC-, and cirrhotic EPC-treated rats. In vivo, hepatic and systemic hemodynamic parameters were evaluated. Liver functions were evaluated. Results In comparison to controls, BDL rats revealed an increase of fibrosis and angiogenesis. Among the treated rats, cirrhotic EPC-treated rats had increased fibrosis grade as compared to healthy EPC-treated and saline-treated rats. There was an increase of both fibrosis and angiogenesis markers, alpha-SMA and CD31 in cirrhotic EPC-treated rats as compared to healthy EPC-treated and saline-treated rats in immunohistochemistry and western blot studies. Cirrhotic EPC-treated BDL rats had high portal pressure and portal blood flow with significantly elevated hepatic vascular resistance in comparison with healthy EPC- and saline-treated BDL animals, without significant differences in mean arterial pressure. Cirrhotic EPC-treated BDL rats also showed a substantial increase in the hepatic expression of angiogenic receptors, VEGFR2 and CXCR4 in comparison with saline-treated rats. Conclusion The study suggests that transplantation of cirrhotic EPCs enhances LSEC differentiation and angiogenesis, activates HSCs and worsens fibrosis, thus resulting in hepatic hemodynamic derangements in BDL-induced cirrhosis.
Collapse
Affiliation(s)
- Dinesh Mani Tripathi
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Mohsin Hassan
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Hamda Siddiqui
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Impreet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Preety Rawal
- School of Biotechnology, Gautam Buddha University, Greater Noida, India
| | - Chaggan Bihari
- Department of Pathology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Savneet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Shiv K Sarin
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India.,Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| |
Collapse
|
53
|
Abstract
Peripheral artery disease is a common disorder and a major cause of morbidity and mortality worldwide. Therapy is directed at reducing the risk of major adverse cardiovascular events and at ameliorating symptoms. Medical therapy is effective at reducing the incidence of myocardial infarction and stroke to which these patients are prone but is inadequate in relieving limb-related symptoms, such as intermittent claudication, rest pain, and ischemic ulceration. Limb-related morbidity is best addressed with surgical and endovascular interventions that restore perfusion. Current medical therapies have only modest effects on limb blood flow. Accordingly, there is an opportunity to develop medical approaches to restore limb perfusion. Vascular regeneration to enhance limb blood flow includes methods to enhance angiogenesis, arteriogenesis, and vasculogenesis using angiogenic cytokines and cell therapies. We review the molecular mechanisms of these processes; briefly discuss what we have learned from the clinical trials of angiogenic and cell therapies; and conclude with an overview of a potential new approach based upon transdifferentiation to enhance vascular regeneration in peripheral artery disease.
Collapse
Affiliation(s)
- John P Cooke
- From the Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute, TX
| | - Shu Meng
- From the Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute, TX
| |
Collapse
|
54
|
Wing TT, Erikson DW, Burghardt RC, Bazer FW, Bayless KJ, Johnson GA. OPN binds alpha V integrin to promote endothelial progenitor cell incorporation into vasculature. Reproduction 2020; 159:465-478. [PMID: 31990676 PMCID: PMC10792589 DOI: 10.1530/rep-19-0358] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 01/28/2020] [Indexed: 11/08/2022]
Abstract
Angiogenesis is fundamental to the expansion of the placental vasculature during pregnancy. Integrins are associated with vascular formation; and osteopontin is a candidate ligand for integrins to promote angiogenesis. Endothelial progenitor cells (EPCs) are released from bone marrow into the blood and incorporate into newly vascularized tissue where they differentiate into mature endothelium. Results of studies in women suggest that EPCs may play an important role in maintaining placental vascular integrity during pregnancy, although little is known about how EPCs are recruited to these tissues. Our goal was to determine the αv integrin mediated effects of osteopontin on EPC adhesion and incorporation into angiogenic vascular networks. EPCs were isolated from 6 h old piglets. RT-PCR revealed that EPCs initially had a monocyte-like phenotype in culture that became more endothelial-like with cell passage. Immunofluorescence microscopy confirmed that the EPCs express platelet endothelial cell adhesion molecule, vascular endothelial cadherin, and von Willebrand factor. When EPCs were cultured on OPN-coated slides, the αv integrin subunit was observed in focal adhesions at the basal surface of EPCs. Silencing of αv integrin reduced EPC binding to OPN and focal adhesion assembly. In vitro siRNA knockdown in EPCs,demonstrated that OPN stimulates EPC incorporation into human umbilical vein endothelial cell (HUVEC) networks via αv-containing integrins. Finally, in situ hybridization and immunohistochemistry localized osteopontin near placental blood vessels. In summary, OPN binds the αv integrin subunit on EPCs to support EPC adhesion and increase EPC incorporation into angiogenic vascular networks.
Collapse
Affiliation(s)
- Theodore T. Wing
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843
| | - David W. Erikson
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843
| | - Robert C. Burghardt
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843
| | - Fuller W. Bazer
- Department of Animal Science, Texas A&M University, College Station, TX 77843
| | - Kayla J. Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843
| | - Greg A. Johnson
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843
| |
Collapse
|
55
|
Deutsch MA, Brunner S, Grabmaier U, David R, Ott I, Huber BC. Cardioprotective Potential of Human Endothelial-Colony Forming Cells from Diabetic and Nondiabetic Donors. Cells 2020; 9:cells9030588. [PMID: 32131432 PMCID: PMC7140510 DOI: 10.3390/cells9030588] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/24/2020] [Accepted: 02/27/2020] [Indexed: 12/20/2022] Open
Abstract
Objective: The potential therapeutic role of endothelial progenitor cells (EPCs) in ischemic heart disease for myocardial repair and regeneration is subject to intense investigation. The aim of the study was to investigate the proregenerative potential of human endothelial colony-forming cells (huECFCs), a very homogenous and highly proliferative endothelial progenitor cell subpopulation, in a myocardial infarction (MI) model of severe combined immunodeficiency (SCID) mice. Methods: CD34+ peripheral blood mononuclear cells were isolated from patient blood samples using immunomagnetic beads. For generating ECFCs, CD34+ cells were plated on fibronectin-coated dishes and were expanded by culture in endothelial-specific cell medium. Either huECFCs (5 × 105) or control medium were injected into the peri-infarct region after surgical MI induction in SCID/beige mice. Hemodynamic function was assessed invasively by conductance micromanometry 30 days post-MI. Hearts of sacrificed animals were analyzed by immunohistochemistry to assess cell fate, infarct size, and neovascularization (huECFCs n = 15 vs. control n = 10). Flow-cytometric analysis of enzymatically digested whole heart tissue was used to analyze different subsets of migrated CD34+/CD45+ peripheral mononuclear cells as well as CD34−/CD45− cardiac-resident stem cells two days post-MI (huECFCs n = 10 vs. control n = 6). Results: Transplantation of human ECFCs after MI improved left ventricular (LV) function at day 30 post-MI (LVEF: 30.43 ± 1.20% vs. 22.61 ± 1.73%, p < 0.001; ΔP/ΔTmax 5202.28 ± 316.68 mmHg/s vs. 3896.24 ± 534.95 mmHg/s, p < 0.05) when compared to controls. In addition, a significantly reduced infarct size (50.3 ± 4.5% vs. 66.1 ± 4.3%, p < 0.05) was seen in huECFC treated animals compared to controls. Immunohistochemistry failed to show integration and survival of transplanted cells. However, anti-CD31 immunohistochemistry demonstrated an increased vascular density within the infarct border zone (8.6 ± 0.4 CD31+ capillaries per HPF vs. 6.2 ± 0.5 CD31+ capillaries per HPF, p < 0.001). Flow cytometry at day two post-MI showed a trend towards increased myocardial homing of CD45+/CD34+ mononuclear cells (1.1 ± 0.3% vs. 0.7 ± 0.1%, p = 0.2). Interestingly, we detected a significant increase in the population of CD34−/CD45−/Sca1+ cardiac resident stem cells (11.7 ± 1.7% vs. 4.7 ± 1.7%, p < 0.01). In a subgroup analysis no significant differences were seen in the cardioprotective effects of huECFCs derived from diabetic or nondiabetic patients. Conclusions: In a murine model of myocardial infarction in SCID mice, transplantation of huECFCs ameliorated myocardial function by attenuation of adverse post-MI remodeling, presumably through paracrine effects. Cardiac repair is enhanced by increasing myocardial neovascularization and the pool of Sca1+ cardiac resident stem cells. The use of huECFCs for treating ischemic heart disease warrants further investigation.
Collapse
Affiliation(s)
- Marcus-André Deutsch
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, Ruhr-University Bochum, Georgstr. 11, D-32545 Bad Oeynhausen, Germany;
| | - Stefan Brunner
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Marchioninistr. 15, D-81377 Munich, Germany; (S.B.); (U.G.)
| | - Ulrich Grabmaier
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Marchioninistr. 15, D-81377 Munich, Germany; (S.B.); (U.G.)
| | - Robert David
- Reference- and Translation Center for Cardiac Stem Cell Therapy (RTC), Rostock University Medical Center, Department of Cardiac Surgery, Department Life, Light & Matter (LL&M), 18057 Rostock, Germany;
| | - Ilka Ott
- Department of Internal Medicine, Division of Cardiology, Helios Klinikum Pforzheim, Kanzlerstraße 2-6, D-75175 Pforzheim, Germany;
| | - Bruno C. Huber
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Marchioninistr. 15, D-81377 Munich, Germany; (S.B.); (U.G.)
- Correspondence: ; Tel.: +49-89-44-000
| |
Collapse
|
56
|
Naito H, Iba T, Takakura N. Mechanisms of new blood-vessel formation and proliferative heterogeneity of endothelial cells. Int Immunol 2020; 32:295-305. [DOI: 10.1093/intimm/dxaa008] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 01/27/2020] [Indexed: 12/26/2022] Open
Abstract
Abstract
The vast blood-vessel network of the circulatory system is crucial for maintaining bodily homeostasis, delivering essential molecules and blood cells, and removing waste products. Blood-vessel dysfunction and dysregulation of new blood-vessel formation are related to the onset and progression of many diseases including cancer, ischemic disease, inflammation and immune disorders. Endothelial cells (ECs) are fundamental components of blood vessels and their proliferation is essential for new vessel formation, making them good therapeutic targets for regulating the latter. New blood-vessel formation occurs by vasculogenesis and angiogenesis during development. Induction of ECs termed tip, stalk and phalanx cells by interactions between vascular endothelial growth factor A (VEGF-A) and its receptors (VEGFR1–3) and between Notch and Delta-like Notch ligands (DLLs) is crucial for regulation of angiogenesis. Although the importance of angiogenesis is unequivocal in the adult, vasculogenesis effected by endothelial progenitor cells (EPCs) may also contribute to post-natal vessel formation. However, the definition of these cells is ambiguous and they include several distinct cell types under the simple classification of ‘EPC’. Furthermore, recent evidence indicates that ECs within the intima show clonal expansion in some situations and that they may harbor vascular-resident endothelial stem cells. In this article, we summarize recent knowledge on vascular development and new blood-vessel formation in the adult. We also introduce concepts of EC heterogeneity and EC clonal expansion, referring to our own recent findings.
Collapse
Affiliation(s)
- Hisamichi Naito
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Tomohiro Iba
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Nobuyuki Takakura
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Laboratory of Signal Transduction, World Premier Institute Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
57
|
Pei CZ, Liu B, Li YT, Fang L, Zhang Y, Li YG, Meng S. MicroRNA-126 protects against vascular injury by promoting homing and maintaining stemness of late outgrowth endothelial progenitor cells. Stem Cell Res Ther 2020; 11:28. [PMID: 31964421 PMCID: PMC6975061 DOI: 10.1186/s13287-020-1554-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/24/2019] [Accepted: 01/07/2020] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Endothelial progenitor cells (EPCs) contribute to reendothelialization and neovascularization and protect against vascular injury and ischemia of various organs. We have previously shown downregulation of microRNA (miR)-126 in EPCs from diabetic patients, which contributes to dysfunction of EPCs including impaired migratory ability. The aims of the present study were to examine (1) in vitro the effects of miR-126 on the homing and stemness of late outgrowth EPCs (LOCs), along with relevant signaling pathways, and (2) in vivo the effects of modulating LOCs by manipulating miR-126 expression on LOC homing and reendothelialization of injured arteries in GK rats (a non-obese diabetes model). METHODS Rat bone marrow-derived LOCs were transfected with miR-126 inhibitor or lentiviral vectors expressing miR-126. LOC migration was determined by transwell migration assay. CXCR4 expression was measured by real-time PCR, Western blotting, and confocal microscopy while related signaling pathway proteins were measured by Western Blotting. Stemness gene expression, and gene and protein expression and promoter activity of KLF-8 were also measured. LOCs transfected with lenti-miR-126 or miR-126 inhibitor were injected into GK rats with carotid artery injury, and then vascular reendothelialization and the extent of intimal hyperplasia were examined. RESULTS Lenti-miR-126 increased while miR-126 inhibitor decreased LOC migration and CXCR4 expression on LOCs. miR-126 positively regulated p-ERK, VEGF, p-Akt, and eNOS protein expression, and inhibitors of these proteins blocked miR-126-induced CXCR4 expression and also reduced LOC migration. Overexpression of miR-126 promoted while inhibition of miR-126 suppressed stemness gene expression in LOCs. miR-126 also inhibited gene and protein expression and promoter activity of KLF-8 while shRNA-mediated knockdown of KLF-8 increased stemness gene expression. Upregulation of stemness gene expression by miR-126 overexpression was completely abrogated by co-transfection of lenti-KLF-8 and lenti-miR-126 into LOCs. In GK rats, transplantation of LOCs overexpressing miR-126 enhanced LOC homing and reendothelialization and decreased intimal hyperplasia of injured arteries. CONCLUSION Our results indicate that miR-126 protects against vascular injury by promoting CXCR4 expression and LOC homing via ERK/VEGF and Akt/eNOS signaling pathways and maintaining stemness via targeting KLF-8.
Collapse
Affiliation(s)
- Chong Zhe Pei
- Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Bo Liu
- Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ye Ting Li
- Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Lu Fang
- Haematopoiesis and Leukocyte Biology Laboratory, Baker Heart and Diabetes Research Institute, Melbourne, VIC, Australia
| | - Yi Zhang
- Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yi Gang Li
- Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| | - Shu Meng
- Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| |
Collapse
|
58
|
Gupta SK, Liu Z, Sims EC, Repass MJ, Haneline LS, Yoder MC. Endothelial Colony-Forming Cell Function Is Reduced During HIV Infection. J Infect Dis 2020; 219:1076-1083. [PMID: 30239747 DOI: 10.1093/infdis/jiy550] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 09/12/2018] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Human immunodeficiency virus (HIV) may be related to cardiovascular disease through monocyte activation-associated endothelial dysfunction. METHODS Blood samples from 15 HIV-negative participants (the uninfected group), 8 HIV-positive participants who were not receiving antiretroviral therapy (ART) (the infected, untreated group), and 15 HIV-positive participants who were receiving ART (the infected, treated group) underwent flow cytometry of endothelial colony-forming cells (ECFCs) and monocyte proportions. IncuCyte live cell imaging of 8 capillary proliferative capacity parameters were obtained from cord blood ECFCs treated with participant plasma. RESULTS The ECFC percentage determined by flow cytometry was not different between the study groups; however, values of the majority of capillary proliferative capacity parameters (ie, cell area, network length, network branch points, number of networks, and average tube width uniformity) were significantly lower in infected, untreated participants as compared to values for uninfected participants or infected, treated participants (P < .00625 for all comparisons). CD14+CD16+ intermediate monocytes and soluble CD163 were significantly and negatively correlated with several plasma-treated, cord blood ECFC proliferative capacity parameters in the combined HIV-positive groups but not in the uninfected group. CONCLUSIONS Cord blood ECFC proliferative capacity was significantly impaired by plasma from infected, untreated patients, compared with plasma from uninfected participants and from infected, treated participants. Several ECFC functional parameters were adversely associated with monocyte activation in the HIV-positive groups, thereby suggesting a mechanism by which HIV-related inflammation may impair vascular reparative potential and consequently increase the risk of cardiovascular disease during HIV infection.
Collapse
Affiliation(s)
- Samir K Gupta
- Department of Medicine, Indiana University School of Medicine, Indianapolis
| | - Ziyue Liu
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis
| | - Emily C Sims
- Department of Medicine, Indiana University School of Medicine, Indianapolis
| | - Matthew J Repass
- Department of Medicine, Indiana University School of Medicine, Indianapolis
| | - Laura S Haneline
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis
| | - Mervin C Yoder
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis
| |
Collapse
|
59
|
Yang G, Mahadik B, Choi JY, Fisher JP. Vascularization in tissue engineering: fundamentals and state-of-art. ACTA ACUST UNITED AC 2020; 2. [PMID: 34308105 DOI: 10.1088/2516-1091/ab5637] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Vascularization is among the top challenges that impede the clinical application of engineered tissues. This challenge has spurred tremendous research endeavor, defined as vascular tissue engineering (VTE) in this article, to establish a pre-existing vascular network inside the tissue engineered graft prior to implantation. Ideally, the engineered vasculature can be integrated into the host vasculature via anastomosis to supply nutrient to all cells instantaneously after surgery. Moreover, sufficient vascularization is of great significance in regenerative medicine from many other perspectives. Due to the critical role of vascularization in successful tissue engineering, we aim to provide an up-to-date overview of the fundamentals and VTE strategies in this article, including angiogenic cells, biomaterial/bio-scaffold design and bio-fabrication approaches, along with the reported utility of vascularized tissue complex in regenerative medicine. We will also share our opinion on the future perspective of this field.
Collapse
Affiliation(s)
- Guang Yang
- Tissue Engineering and Biomaterials Laboratory, Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD, United States of America.,Center for Engineering Complex Tissues, University of Maryland, College Park, MD, United States of America
| | - Bhushan Mahadik
- Tissue Engineering and Biomaterials Laboratory, Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD, United States of America.,Center for Engineering Complex Tissues, University of Maryland, College Park, MD, United States of America
| | - Ji Young Choi
- Tissue Engineering and Biomaterials Laboratory, Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD, United States of America
| | - John P Fisher
- Tissue Engineering and Biomaterials Laboratory, Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD, United States of America.,Center for Engineering Complex Tissues, University of Maryland, College Park, MD, United States of America
| |
Collapse
|
60
|
Gouveia-Fernandes S. Monocytes and Macrophages in Cancer: Unsuspected Roles. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1219:161-185. [PMID: 32130699 DOI: 10.1007/978-3-030-34025-4_9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The behavior of cancer is undoubtedly affected by stroma. Macrophages belong to this microenvironment and their presence correlates with reduced survival in most cancers. After a tumor-induced "immunoediting", these monocytes/macrophages, originally the first line of defense against tumor cells, undergo a phenotypic switch and become tumor-supportive and immunosuppressive.The influence of these tumor-associated macrophages (TAMs) on cancer is present in all traits of carcinogenesis. These cells participate in tumor initiation and growth, migration, vascularization, invasion and metastasis. Although metastasis is extremely clinically relevant, this step is always reliant on the angiogenic ability of tumors. Therefore, the formation of new blood vessels in tumors assumes particular importance as a limiting step for disease progression.Herein, the once unsuspected roles of macrophages in cancer will be discussed and their importance as a promising strategy to treat this group of diseases will be reminded.
Collapse
Affiliation(s)
- Sofia Gouveia-Fernandes
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School | Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
61
|
Fenofibrate Reverses Dysfunction of EPCs Caused by Chronic Heart Failure. J Cardiovasc Transl Res 2019; 13:158-170. [PMID: 31701352 DOI: 10.1007/s12265-019-09889-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 04/12/2019] [Indexed: 12/18/2022]
Abstract
The enhanced activity of endothelial progenitor cells (EPCs) by AMP-activated protein kinase (AMPK) agonists might explain the reversal of chronic heart failure (CHF)-mediated endothelial dysfunction. We studied baseline circulating EPC numbers in patients with heart failure and clarified the effect of fenofibrate on both circulating angiogenic cell (CAC) and late EPC activity. The numbers of circulating EPCs in CHF patients were quantified by flow cytometry. Blood-derived mononuclear cells were cultured, and CAC and late EPC functions, including fibronectin adhesion, tube formation, and migration, were evaluated. We focused on the effect of fenofibrate, an AMPK agonist, on EPC function and Akt/eNOS cascade activation in vitro. The number of circulating EPCs (CD34+/KDR+) was significantly lower in CHF patients (ischemic cardiomyopathy (ICMP): 0.07%, dilated cardiomyopathy (DCMP): 0.068%; p < 0.05) than in healthy subjects (0.102% of the gating region). In CACs, fibronectin adhesion function was reversed by fenofibrate treatment (p < 0.05). Similar results were also found for tube formation and migration in late EPCs, which were significantly improved by fenofibrate in an AMPK-dependent manner (p < 0.05), suggesting that fenofibrate reversed CACs and late EPC dysfunction in CHF patients. The present findings reveal the potential application of the AMPK agonist fenofibrate to reverse endothelial dysfunction in CHF patients.
Collapse
|
62
|
Song W, Chiu A, Wang LH, Schwartz RE, Li B, Bouklas N, Bowers DT, An D, Cheong SH, Flanders JA, Pardo Y, Liu Q, Wang X, Lee VK, Dai G, Ma M. Engineering transferrable microvascular meshes for subcutaneous islet transplantation. Nat Commun 2019; 10:4602. [PMID: 31601796 PMCID: PMC6787187 DOI: 10.1038/s41467-019-12373-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 09/03/2019] [Indexed: 12/21/2022] Open
Abstract
The success of engineered cell or tissue implants is dependent on vascular regeneration to meet adequate metabolic requirements. However, development of a broadly applicable strategy for stable and functional vascularization has remained challenging. We report here highly organized and resilient microvascular meshes fabricated through a controllable anchored self-assembly method. The microvascular meshes are scalable to centimeters, almost free of defects and transferrable to diverse substrates, ready for transplantation. They promote formation of functional blood vessels, with a density as high as ~220 vessels mm-2, in the poorly vascularized subcutaneous space of SCID-Beige mice. We further demonstrate the feasibility of fabricating microvascular meshes from human induced pluripotent stem cell-derived endothelial cells, opening a way to engineer patient-specific microvasculature. As a proof-of-concept for type 1 diabetes treatment, we combine microvascular meshes and subcutaneously transplanted rat islets and achieve correction of chemically induced diabetes in SCID-Beige mice for 3 months.
Collapse
Affiliation(s)
- Wei Song
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Alan Chiu
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Long-Hai Wang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Robert E Schwartz
- Division of Gastroenterology & Hepatology, Department of Medicine, Weill Cornell Medical College, New York, NY, 10021, USA
| | - Bin Li
- Department of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Nikolaos Bouklas
- Department of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Daniel T Bowers
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Duo An
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Soon Hon Cheong
- Department of Clinical Sciences, Cornell University, Ithaca, NY, 14853, USA
| | - James A Flanders
- Department of Clinical Sciences, Cornell University, Ithaca, NY, 14853, USA
| | - Yehudah Pardo
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Qingsheng Liu
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Xi Wang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Vivian K Lee
- Department of Bioengineering, Northeastern University, Boston, MA, 02120, USA
| | - Guohao Dai
- Department of Bioengineering, Northeastern University, Boston, MA, 02120, USA
| | - Minglin Ma
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
63
|
de Jong A, Weijers E, Dirven R, de Boer S, Streur J, Eikenboom J. Variability of von Willebrand factor-related parameters in endothelial colony forming cells. J Thromb Haemost 2019; 17:1544-1554. [PMID: 31265169 PMCID: PMC6852380 DOI: 10.1111/jth.14558] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 06/03/2019] [Accepted: 06/25/2019] [Indexed: 01/01/2023]
Abstract
Essentials Endothelial colony forming cells (ECFCs) are a powerful tool to study vascular diseases ex vivo. Separate ECFC lines show variations in morphology and von Willebrand factor-related parameters. Maximum cell density is correlated with von Willebrand factor expression in ECFCs. Variations in ECFC lines are dependent on the age and mesenchymal state of the cells. ABSTRACT: Background Endothelial colony forming cells (ECFCs) are cultured endothelial cells derived from peripheral blood. ECFCs are a powerful tool to study pathophysiological mechanisms underlying vascular diseases, including von Willebrand disease. In prior research, however, large variations between ECFC lines were observed in, among others, von Willebrand factor (VWF) expression. Objective Understand the relation between phenotypic characteristics and VWF-related parameters of healthy control ECFCs. Methods ECFC lines (n = 16) derived from six donors were studied at maximum cell density. Secreted and intracellular VWF antigen were measured by ELISA. The angiogenic capacity of ECFCs was investigated by the Matrigel tube formation assay. Differences in expression of genes involved in angiogenesis, aging, and endothelial to mesenchymal transition (EndoMT) were measured by quantitative PCR. Results Different ECFC lines show variable morphologies and cell density at maximum confluency and cell lines with a low maximum cell density show a mixed and more mesenchymal phenotype. We identified a significant positive correlation between maximum cell density and VWF production, both at protein and mRNA level. Also, significant correlations were observed between maximum cell density and several angiogenic, aging and EndoMT parameters. Conclusions We observed variations in morphology, maximum cell density, VWF production, and angiogenic potential between healthy control ECFCs. These variations seem to be attributable to differences in aging and EndoMT. Because variations correlate with cell density, we believe that ECFCs maintain a powerful tool to study vascular diseases. It is however important to compare cell lines with the same characteristics and perform experiments at maximum cell density.
Collapse
Affiliation(s)
- Annika de Jong
- Department of Internal medicinedivision of Thrombosis and HemostasisEinthoven Laboratory for Vascular and Regenerative MedicineLeiden University Medical CenterLeidenthe Netherlands
| | - Ester Weijers
- Department of Internal medicinedivision of Thrombosis and HemostasisEinthoven Laboratory for Vascular and Regenerative MedicineLeiden University Medical CenterLeidenthe Netherlands
| | - Richard Dirven
- Department of Internal medicinedivision of Thrombosis and HemostasisEinthoven Laboratory for Vascular and Regenerative MedicineLeiden University Medical CenterLeidenthe Netherlands
| | - Suzan de Boer
- Department of Internal medicinedivision of Thrombosis and HemostasisEinthoven Laboratory for Vascular and Regenerative MedicineLeiden University Medical CenterLeidenthe Netherlands
| | - Jasmin Streur
- Department of Internal medicinedivision of Thrombosis and HemostasisEinthoven Laboratory for Vascular and Regenerative MedicineLeiden University Medical CenterLeidenthe Netherlands
| | - Jeroen Eikenboom
- Department of Internal medicinedivision of Thrombosis and HemostasisEinthoven Laboratory for Vascular and Regenerative MedicineLeiden University Medical CenterLeidenthe Netherlands
| |
Collapse
|
64
|
Wei Z, Volkova E, Blatchley MR, Gerecht S. Hydrogel vehicles for sequential delivery of protein drugs to promote vascular regeneration. Adv Drug Deliv Rev 2019; 149-150:95-106. [PMID: 31421149 PMCID: PMC6889011 DOI: 10.1016/j.addr.2019.08.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 07/04/2019] [Accepted: 08/12/2019] [Indexed: 12/12/2022]
Abstract
In recent years, as the mechanisms of vasculogenesis and angiogenesis have been uncovered, the functions of various pro-angiogenic growth factors (GFs) and cytokines have been identified. Therefore, therapeutic angiogenesis, by delivery of GFs, has been sought as a treatment for many vascular diseases. However, direct injection of these protein drugs has proven to have limited clinical success due to their short half-lives and systemic off-target effects. To overcome this, hydrogel carriers have been developed to conjugate single or multiple GFs with controllable, sustained, and localized delivery. However, these attempts have failed to account for the temporal complexity of natural angiogenic pathways, resulting in limited therapeutic effects. Recently, the emerging ideas of optimal sequential delivery of multiple GFs have been suggested to better mimic the biological processes and to enhance therapeutic angiogenesis. Incorporating sequential release into drug delivery platforms will likely promote the formation of neovasculature and generate vast therapeutic potential.
Collapse
Affiliation(s)
- Zhao Wei
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology Physical-Sciences Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Eugenia Volkova
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology Physical-Sciences Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Michael R Blatchley
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology Physical-Sciences Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology Physical-Sciences Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
65
|
Somani A, Nair SL, Milbauer LC, Zhu G, Sajja S, Solovey A, Chen Y, Hebbel RP. Blood outgrowth endothelial cells overexpressing eNOS mitigate pulmonary hypertension in rats: a unique carrier cell enabling autologous cell-based gene therapy. Transl Res 2019; 210:1-7. [PMID: 31082372 PMCID: PMC6741773 DOI: 10.1016/j.trsl.2019.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 04/10/2019] [Accepted: 04/11/2019] [Indexed: 12/22/2022]
Abstract
We have investigated a unique cell type, blood outgrowth endothelial cells (BOEC), as a cell-based gene therapy approach to pulmonary hypertension. BOEC are bona fide endothelial cells, obtained from peripheral blood, that can be expanded to vast numbers, and are amenable to both cryopreservation and genetic modification. We established primary cultures of rat BOEC and genetically altered them to over-express human eNOS plus green fluorescent protein (rBOEC/eNOS) or to express GFP only (rBOEC/GFP). We gave monocrotaline to rats on day 0, and they developed severe pulmonary hypertension. As a Prevention model, we infused saline or rBOEC/GFP or rBOEC/eNOS on day 3, and then examined endpoints on day 24. The rBOEC/eNOS recipients developed elevated NOx (serum and lung) and less severe: elevation of right ventricular systolic pressure (RVSP), right ventricular hypertrophy, and pulmonary arteriolar muscularization and loss of alveolar density. As an Intervention model, we waited until day 21 to give the test infusions, and we examined endpoints on day 35. The rBOEC/eNOS recipients again developed elevated NOx and manifested the same improvements. Indeed, rBOEC/eNOS infusion not only prevented worsening of RVSP but also partially reversed established arteriolar muscularization. These data suggest that BOEC may be useful as a carrier cell for genetic strategies targeting pulmonary hypertension. Their properties render BOEC amenable to preclinical and scale-up studies, available for autologous therapies, and tolerant of modification and storage for potential future use in patients at risk for PAH, eg, as defined by genetics or medical condition.
Collapse
Affiliation(s)
- Arif Somani
- Pediatric Critical Care Medicine, University of Minnesota Medical School, Minneapolis, Minnesota.
| | - Sethu L Nair
- Pediatric Critical Care Medicine, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Liming C Milbauer
- Division of Hematology-Oncology-Transplantation, Department of Medicine; and Vascular Biology Center, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Guangshuo Zhu
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Suchitra Sajja
- Pediatric Critical Care Medicine, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Anna Solovey
- Division of Hematology-Oncology-Transplantation, Department of Medicine; and Vascular Biology Center, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Yingjie Chen
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Robert P Hebbel
- Division of Hematology-Oncology-Transplantation, Department of Medicine; and Vascular Biology Center, University of Minnesota Medical School, Minneapolis, Minnesota
| |
Collapse
|
66
|
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: 58] [Impact Index Per Article: 9.7] [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
|
67
|
Elkhodiry MA, Boulanger MD, Bashth O, Tanguay JF, Laroche G, Hoesli CA. Isolating and expanding endothelial progenitor cells from peripheral blood on peptide-functionalized polystyrene surfaces. Biotechnol Bioeng 2019; 116:2598-2609. [PMID: 31286475 DOI: 10.1002/bit.27107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/19/2019] [Accepted: 07/01/2019] [Indexed: 11/11/2022]
Abstract
The expansion of human peripheral blood endothelial progenitor cells to obtain therapeutically relevant endothelial colony-forming cells (ECFCs) has been commonly performed on xeno-derived extracellular matrix proteins. For cellular therapy applications, xeno-free culture conditions are desirable to improve product safety and reduce process variability. We have previously described a novel fluorophore-tagged RGD peptide (RGD-TAMRA) that enhanced the adhesion of mature endothelial cells in vitro. To investigate whether this peptide can replace animal-derived extracellular matrix proteins in the isolation and expansion of ECFCs, peripheral blood mononuclear cells from 22 healthy adult donors were seeded on RGD-TAMRA-modified polystyrene culture surfaces. Endothelial colony formation was significantly enhanced on RGD-TAMRA-modified surfaces compared to the unmodified control. No phenotypic differences were detected between ECFCs obtained on RGD-TAMRA compared to ECFCs obtained on rat-tail collagen-coated surfaces. Compared with collagen-coated surfaces and unmodified surfaces, RGD-TAMRA surfaces promoted ECFC adhesion, cell spreading, and clonal expansion. This study presents a platform that allows for a comprehensive in vitro evaluation of peptide-based biofunctionalization as a promising avenue for ex vivo ECFC expansion.
Collapse
Affiliation(s)
- Mohamed A Elkhodiry
- Department of Chemical Engineering, McGill University, Montréal, Quebec, Canada
| | - Mariève D Boulanger
- Department of Chemical Engineering, McGill University, Montréal, Quebec, Canada
| | - Omar Bashth
- Department of Chemical Engineering, McGill University, Montréal, Quebec, Canada
| | - Jean-François Tanguay
- Coronary Care Unit, Montréal Heart Institute, Montréal, Quebec, Canada.,Department of Medicine, Université de Montréal, Montréal, Quebec, Canada
| | - Gaétan Laroche
- Département de Génie des Mines, des Matériaux et de la Métallurgie, Centre de Recherche du CHU de Québec, Université Laval, Quebec City, Quebec, Canada
| | - Corinne A Hoesli
- Department of Chemical Engineering, McGill University, Montréal, Quebec, Canada
| |
Collapse
|
68
|
Munisso MC, Yamaoka T. Circulating endothelial progenitor cells in small-diameter artificial blood vessel. J Artif Organs 2019; 23:6-13. [DOI: 10.1007/s10047-019-01114-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 06/06/2019] [Indexed: 01/19/2023]
|
69
|
Patschan S, Vogt M, Bakhtiari D, Bramlage CP, Henze E, Muller GA, Krause A, Patschan D. Humoral and Cellular Patterns of Early Endothelial Progenitor Cells in Relation to the Cardiovascular Risk in Axial Spondylarthritis. J Clin Med Res 2019; 11:391-400. [PMID: 31143305 PMCID: PMC6522236 DOI: 10.14740/jocmr3441w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 04/30/2018] [Indexed: 11/21/2022] Open
Abstract
Background Spondylarthritis (SpA) significantly affects sacroiliac, intervertebral and peripheral joints. Patients with SpA suffer from increased cardiovascular risk (CVR). The endothelial progenitor cell (EPC) system critically perpetuates vascular repair. The aim of the study was to evaluate circulating EPCs in axial (ax)SpA with special attention on parameters of disease activity and CVR. Methods Disease activity and functional impairment were quantified in 50 axSpA patients by using standardized parameters (Bath ankylosing spondylitis disease activity index (BASDAI), C-reactive protein (CRP), finger-floor distance (FFD) and Ott’ sign). Circulating EPCs and EPC regeneration were analyzed (fluorescence-activated cell sorting (FACS) and colony-forming unit (CFU) assay). Serum vasomodulatory mediators were quantified by enzyme-linked immunosorbent assay (ELISA). Results EPC colony numbers were lower in axSpA as compared to controls. Females displayed more colonies than males. In addition, fewer colonies were observed in smokers, in patients with a BASDAI of below 4 and in hypertension. Circulating CD133+/KDR+ cells did not differ between the groups. Follow-up analysis (33 months later) did not show any differences in gender, colony formation, CD133+/KDR+ cells or serum levels of vasomodulatory mediators if related to the categories of BASDAI, Ott’ sign or FFD. Conclusions EPC colony formation is significantly affected in axSpA with particularly low levels in males. EPC-related parameters do not allow predicting disease activity-related or functional parameters nor are they useful for CVR assessment in SpA.
Collapse
Affiliation(s)
- Susann Patschan
- Clinic of Nephrology and Rheumatology, University Hospital of Gottingen, Gottingen, Germany.,Department of Cardiology, Pulmonology, Angiology and Nephrology, Brandenburg Medical School, University Hospital Brandenburg, Brandenburg, Germany
| | - Maria Vogt
- Clinic of Nephrology and Rheumatology, University Hospital of Gottingen, Gottingen, Germany
| | - Donia Bakhtiari
- Clinic of Nephrology and Rheumatology, University Hospital of Gottingen, Gottingen, Germany
| | - Carsten Peter Bramlage
- Clinic of Nephrology and Rheumatology, University Hospital of Gottingen, Gottingen, Germany
| | - Elvira Henze
- Clinic of Nephrology and Rheumatology, University Hospital of Gottingen, Gottingen, Germany
| | - Gerhard Anton Muller
- Clinic of Nephrology and Rheumatology, University Hospital of Gottingen, Gottingen, Germany
| | - Andreas Krause
- Rheumatology and Clinical Immunology, Immanuel-Krankenhaus Berlin, Berlin, Germany
| | - Daniel Patschan
- Department of Cardiology, Pulmonology, Angiology and Nephrology, Brandenburg Medical School, University Hospital Brandenburg, Brandenburg, Germany
| |
Collapse
|
70
|
Spiegler S, Rath M, Much CD, Sendtner BS, Felbor U. Precise CCM1 gene correction and inactivation in patient-derived endothelial cells: Modeling Knudson's two-hit hypothesis in vitro. Mol Genet Genomic Med 2019; 7:e00755. [PMID: 31124307 PMCID: PMC6625102 DOI: 10.1002/mgg3.755] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 03/26/2019] [Accepted: 04/27/2019] [Indexed: 12/20/2022] Open
Abstract
Background The CRISPR/Cas9 system has opened new perspectives to study the molecular basis of cerebral cavernous malformations (CCMs) in personalized disease models. However, precise genome editing in endothelial and other hard‐to‐transfect cells remains challenging. Methods In a proof‐of‐principle study, we first isolated blood outgrowth endothelial cells (BOECs) from a CCM1 mutation carrier with multiple CCMs. In a CRISPR/Cas9 gene correction approach, a high‐fidelity Cas9 variant was then transfected into patient‐derived BOECs using a ribonucleoprotein complex and a single‐strand DNA oligonucleotide. In addition, patient‐specific CCM1 knockout clones were expanded after CRISPR/Cas9 gene inactivation. Results Deep sequencing demonstrated correction of the mutant allele in nearly 33% of all cells whereas no CRISPR/Cas9‐induced mutations in predicted off‐target loci were identified. Corrected BOECs could be cultured in cell mixtures but demonstrated impaired clonal survival. In contrast, CCM1‐deficient BOECs displayed increased resistance to stress‐induced apoptotic cell death and could be clonally expanded to high passages. When cultured together, CCM1‐deficient BOECs largely replaced corrected as well as heterozygous BOECs. Conclusion We here demonstrate that a non‐viral CRISPR/Cas9 approach can not only be used for gene knockout but also for precise gene correction in hard‐to‐transfect endothelial cells (ECs). Comparing patient‐derived isogenic CCM1+/+, CCM1+/−, and CCM1−/− ECs, we show that the inactivation of the second allele results in clonal evolution of ECs lacking CCM1 which likely reflects the initiation phase of CCM genesis.
Collapse
Affiliation(s)
- Stefanie Spiegler
- Department of Human Genetics, University Medicine Greifswald and Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Matthias Rath
- Department of Human Genetics, University Medicine Greifswald and Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Christiane D Much
- Department of Human Genetics, University Medicine Greifswald and Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Barbara S Sendtner
- Department of Human Genetics, University Medicine Greifswald and Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Ute Felbor
- Department of Human Genetics, University Medicine Greifswald and Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| |
Collapse
|
71
|
|
72
|
Hydrogen sulfide improves vascular repair by promoting endothelial nitric oxide synthase-dependent mobilization of endothelial progenitor cells. J Hypertens 2019; 37:972-984. [DOI: 10.1097/hjh.0000000000001983] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
73
|
Lee SH, Ra JC, Oh HJ, Kim MJ, Setyawan EMN, Choi YB, Yang JW, Kang SK, Han SH, Kim GA, Lee BC. Clinical Assessment of Intravenous Endothelial Progenitor Cell Transplantation in Dogs. Cell Transplant 2019; 28:943-954. [PMID: 31018670 PMCID: PMC6719494 DOI: 10.1177/0963689718821686] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Endothelial progenitor cells (EPCs) have been applied for cell therapy because of their roles in angiogenesis and neovascularization in ischemic tissue. However, adverse responses caused by EPC therapy have not been fully investigated. In this study, a human peripheral blood sample was collected from a healthy donor and peripheral blood mononuclear cells were separated using Ficoll-Hypaque. There were four experimental groups: 10 ml saline infusion group (injection rate; 3 ml/min), 10 ml saline bolus group (injection rate; 60 ml/min), 10 ml EPCs infusion group (2 x 105 cells/ml, injection rate; 3 ml/min), 10 ml EPCs bolus group (2 × 105 cells/ml, injection rate; 60 ml/min). Clinical assessment included physical examination and laboratory examination for intravenous human EPC transplantation in dogs. The results revealed no remarkable findings in vital signs among the dogs used. In blood analysis, platelet counts in saline infusion groups were significantly higher than in the EPC groups within normal ranges, and no significant differences were observed except K+, Cl- and blood urea nitrogen/urea. In ELISA assay, no significant difference was observed in serum tumor necrosis factor alpha. The serum concentration of vascular endothelial growth factor was significantly higher in EPC groups than in saline groups, and interleukin 10 was significantly up-regulated in the EPC infusion group compared with other groups. In conclusion, we demonstrated that no clinical abnormalities were detected after intravenous transplantation of human EPCs in dogs. The transplanted xenogenic EPCs might be involved in anti-inflammatory and angiogenic functions in dogs.
Collapse
Affiliation(s)
- Seok Hee Lee
- 1 Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Republic of Korea
| | - Jeong Chan Ra
- 2 Biostar Stem Cell Research Institute, R Bio Co., Seoul, Republic of Korea
| | - Hyun Ju Oh
- 1 Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Republic of Korea
| | - Min Jung Kim
- 1 Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Republic of Korea
| | - Erif Maha Nugraha Setyawan
- 1 Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Republic of Korea
| | - Yoo Bin Choi
- 1 Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Republic of Korea
| | - Jung Won Yang
- 2 Biostar Stem Cell Research Institute, R Bio Co., Seoul, Republic of Korea
| | - Sung Keun Kang
- 2 Biostar Stem Cell Research Institute, R Bio Co., Seoul, Republic of Korea
| | - Seung Hyup Han
- 2 Biostar Stem Cell Research Institute, R Bio Co., Seoul, Republic of Korea
| | - Geon A Kim
- 2 Biostar Stem Cell Research Institute, R Bio Co., Seoul, Republic of Korea
| | - Byeong Chun Lee
- 1 Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Republic of Korea
| |
Collapse
|
74
|
Alexandru N, Safciuc F, Constantin A, Nemecz M, Tanko G, Filippi A, Dragan E, Bãdilã E, Georgescu A. Platelets of Healthy Origins Promote Functional Improvement of Atherosclerotic Endothelial Progenitor Cells. Front Pharmacol 2019; 10:424. [PMID: 31068820 PMCID: PMC6491786 DOI: 10.3389/fphar.2019.00424] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 04/03/2019] [Indexed: 11/13/2022] Open
Abstract
The purpose was to evaluate the effect of platelets on functional properties of late endothelial progenitor cells (EPCs), in the direct co-culture conditions, and to investigate the involved mediators, in experimental induced atherosclerosis. The late EPCs obtained from two animal groups, hypertensive-hyperlipidemic (HH) and control (C) hamsters, named late EPCs-HH and late EPCs-C, were co-incubated with or without platelets isolated from both groups. Our results have showed that exposure to platelets from control animals: (i) promoted the late EPCs-C capacity to form colonies and capillary-like structures, and also to proliferate and migrate; (ii) improved the functional properties of late EPCs-HH; (iii) strengthened the direct binding EPCs-platelets; (iv) increased SDF-1α,VEGF, PDGF, and reduced CD40L, IL-1β,-6,-8 levels; and (v) enhanced miR-223 and IGF-1R expressions. Platelets from HH group diminished functional abilities for both EPC types and had opposite effects on these pro-angiogenic and pro-inflammatory molecules. Furthermore, testing the direct effect of miR-223 and IGF-1R on late EPCs disclosed that these molecular factors improve late EPC functional properties in atherosclerosis in terms of stimulation of the proliferation and migration abilities. In conclusion, in vitro exposure to platelets of healthy origins had a positive effect on functional properties of atherosclerotic late EPCs. The most likely candidates mediating EPC-platelet interaction can be SDF-1α, VEGF, CD40L, PDGF, IL-1β,-6,-8, miR-223, and IGF-1R. The current study brings evidences that the presence of healthy origin platelets is of utmost importance on functional improvement of EPCs in atherosclerosis.
Collapse
Affiliation(s)
- Nicoleta Alexandru
- Institute of Cellular Biology and Pathology 'Nicolae Simionescu' of the Romanian Academy, Bucharest, Romania
| | - Florentina Safciuc
- Institute of Cellular Biology and Pathology 'Nicolae Simionescu' of the Romanian Academy, Bucharest, Romania
| | - Alina Constantin
- Institute of Cellular Biology and Pathology 'Nicolae Simionescu' of the Romanian Academy, Bucharest, Romania
| | - Miruna Nemecz
- Institute of Cellular Biology and Pathology 'Nicolae Simionescu' of the Romanian Academy, Bucharest, Romania
| | - Gabriela Tanko
- Institute of Cellular Biology and Pathology 'Nicolae Simionescu' of the Romanian Academy, Bucharest, Romania
| | - Alexandru Filippi
- Institute of Cellular Biology and Pathology 'Nicolae Simionescu' of the Romanian Academy, Bucharest, Romania
| | - Emanuel Dragan
- Institute of Cellular Biology and Pathology 'Nicolae Simionescu' of the Romanian Academy, Bucharest, Romania
| | - Elisabeta Bãdilã
- Internal Medicine Clinic, Emergency Clinical Hospital, Bucharest, Romania.,'Carol Davila' University of Medicine and Pharmacy, Bucharest, Romania
| | - Adriana Georgescu
- Institute of Cellular Biology and Pathology 'Nicolae Simionescu' of the Romanian Academy, Bucharest, Romania
| |
Collapse
|
75
|
Huuskes BM, DeBuque RJ, Kerr PG, Samuel CS, Ricardo SD. The Use of Live Cell Imaging and Automated Image Analysis to Assist With Determining Optimal Parameters for Angiogenic Assay in vitro. Front Cell Dev Biol 2019; 7:45. [PMID: 31024908 PMCID: PMC6468051 DOI: 10.3389/fcell.2019.00045] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 03/15/2019] [Indexed: 11/16/2022] Open
Abstract
Testing angiogenic potential and function of cells in culture is important for the understanding of the mechanisms that can modulate angiogenesis, especially when discovering novel anti- or pro-angiogenic therapeutics. Commonly used angiogenic assays include tube formation, proliferation, migration, and wound healing, and although well-characterized, it is important that methodology is standardized and reproducible. Human endothelial progenitor cells (EPCs) are critical for post-natal vascular homeostasis and can be isolated from human peripheral blood. Endothelial colony forming cells (ECFCs) are a subset of EPCs and are of interest as a possible therapeutic target for hypoxic diseases such as kidney disease, as they have a high angiogenic potential. However, once ECFCs are identified in culture, the exact timing of passaging has not been well-described and the optimal conditions to perform angiogenic assays such as seeding density, growth media (GM) concentrations and end-points of these assays is widely varied in the literature. Here, we describe the process of isolating, culturing and passaging ECFCs from patients with end-stage renal disease (ESRD), aided by image analysis. We further describe optimal conditions, for human bladder endothelial cells (hBECs), challenged in angiogenic assays and confirm that cell density is a limiting factor in accurately detecting angiogenic parameters. Furthermore, we show that GM along is enough to alter the angiogenic potential of cells, seeded at the same density. Lastly, we report on the success of human ECFCs in angiogenic assays and describe the benefits of live-cell imaging combined with time-lapse microscopy for this type of investigation.
Collapse
Affiliation(s)
- Brooke M Huuskes
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Ryan J DeBuque
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Peter G Kerr
- Department of Nephrology, Monash Medical Centre, Monash University, Melbourne, VIC, Australia
| | - Chrishan S Samuel
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Sharon D Ricardo
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
76
|
Chhabra M, Sharma S. Potential role of Peroxisome Proliferator Activated Receptor gamma analogues in regulation of endothelial progenitor cells in diabetes mellitus: An overview. Diabetes Metab Syndr 2019; 13:1123-1129. [PMID: 31336454 DOI: 10.1016/j.dsx.2019.01.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 01/18/2019] [Indexed: 12/27/2022]
Abstract
Endothelial progenitor cells are recognized as the potential targets for the revascularization and angiogenesis because of their ability to get themselves transformed into mature endothelial cells. Underlying pathophysiology in diabetes mellitus leads to decrease in circulatory endothelial progenitor cells, resulting in diabetic macro-vascular and micro-vascular complications. Peroxisome Proliferator Activated Receptor (PPAR) gamma analogues serves as an effective therapy for controlling blood sugar levels and preventing its complications. Reports of clinical trials and meta-analysis of clinical trial suggests the beneficial aspects of PPAR gamma therapy in increasing the number and function of circulating endothelial progenitor cells. This review highlights the pleotropic effect of PPAR gamma analogs, apart from their antidiabetic action via reduction of oxidative stress, increasing expression of eNOS, reducing level of miR 22, miR 222 levels and positive modulation of rapamycin/Protein kinase B/phosphoinoside3-kinase pathways, preventing the early apoptosis, enhanced mobility proliferation and transformation into mature endothelial cells. PPAR gamma therapy in diabetes regulates endothelial progenitor cells, reduces complications of diabetes like retinopathy, nephropathy, neuropathy, cardiomyopathy, deep vein thrombosis, and maintains the healthy vasculature.
Collapse
Affiliation(s)
- Manik Chhabra
- PharmD Intern, Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab, India.
| | - Saurabh Sharma
- Department of Pharmacology, School of Pharmaceutical and Allied Medical Sciences, CT University, Ludhiana, Punjab, India
| |
Collapse
|
77
|
Phenotypic differences in early outgrowth angiogenic cells based on in vitro cultivation. Cytotechnology 2019; 71:665-670. [PMID: 30756209 DOI: 10.1007/s10616-019-00305-6] [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: 06/05/2018] [Accepted: 02/07/2019] [Indexed: 10/27/2022] Open
Abstract
Bone marrow-derived early outgrowth cells play an important role in endothelial repair. In vitro isolation techniques have identified two distinct morphological early outgrowth cell populations, but it is still unknown whether they present some functional phenotypic differences. Accordingly, the aim of the present study was to determine whether there are phenotypic differences in cellular function between two putative early outgrowth cells in culture. Peripheral blood samples were collected from 18 healthy adults. Thereafter, mononuclear cells were isolated by Ficoll density-gradient centrifugation and plated on 6-well plates coated with human fibronectin. After 2 and 7 days, respectively, non-adherent cells (NAC) and adherent cells (AC) underwent functional assays in order to measure the migratory capacity (Boyden chamber), angiogenic growth factor release (ELISA) and apoptosis (TUNEL). Migration to both VEGF (517 ± 74 vs. 273 ± 74 AU) and SDF-1 (517 ± 68 vs. 232 ± 68 AU) were approximately twofold higher (P < 0.05) in the NAC when compared to AC. Release of angiogenic factors, granulocyte colony-stimulating and hepatocyte growth factor, were not different between cell types. Apoptotic response to staurosporine was significantly lower in NAC (20 ± 32 vs. 125 ± 32%). In summary, NAC and AC demonstrated functional phenotypic differences in migratory capacity and apoptotic susceptibility, which makes it difficult to compare these two early outgrowth cell populations in literature.
Collapse
|
78
|
Pennings I, van Dijk LA, van Huuksloot J, Fledderus JO, Schepers K, Braat AK, Hsiao EC, Barruet E, Morales BM, Verhaar MC, Rosenberg AJWP, Gawlitta D. Effect of donor variation on osteogenesis and vasculogenesis in hydrogel cocultures. J Tissue Eng Regen Med 2019; 13:433-445. [PMID: 30650247 PMCID: PMC6593839 DOI: 10.1002/term.2807] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 01/02/2019] [Accepted: 01/09/2019] [Indexed: 12/29/2022]
Abstract
To introduce a functional vascular network into tissue-engineered bone equivalents, human endothelial colony forming cells (ECFCs) and multipotent mesenchymal stromal cells (MSCs) can be cocultured. Here, we studied the impact of donor variation of human bone marrow-derived MSCs and cord blood-derived ECFCs on vasculogenesis and osteogenesis using a 3D in vitro coculture model. Further, to make the step towards cocultures consisting of cells derived from a single donor, we tested how induced pluripotent stem cell (iPSC)-derived human endothelial cells (iECs) performed in coculture models. Cocultures with varying combinations of human donors of MSCs, ECFCs, or iECs were prepared in Matrigel. The constructs were cultured in an osteogenic differentiation medium. Following a 10-day culture period, the length of the prevascular structures and osteogenic differentiation were evaluated for up to 21 days of culture. The particular combination of MSC and ECFC donors influenced the vasculogenic properties significantly and induced variation in osteogenic potential. In addition, the use of iECs in the cocultures resulted in prevascular structure formation in osteogenically differentiated constructs. Together, these results showed that close attention to the source of primary cells, such as ECFCs and MSCs, is critical to address variability in vasculogenic and osteogenic potential. The 3D coculture model appeared to successfully generate prevascularized constructs and were sufficient in exceeding the ~200 μm diffusion limit. In addition, iPSC-derived cell lineages may decrease variability by providing a larger and potentially more uniform source of cells for future preclinical and clinical applications.
Collapse
Affiliation(s)
- Iris Pennings
- Department of Oral and Maxillofacial Surgery and Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Lukas A van Dijk
- Department of Oral and Maxillofacial Surgery and Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Juliet van Huuksloot
- Department of Oral and Maxillofacial Surgery and Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Joost O Fledderus
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Koen Schepers
- Department of Cell Biology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - A Koen Braat
- Department of Cell Biology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Edward C Hsiao
- Department of Medicine and the Institute for Human Genetics and the Program for Craniofacial Biology, University of California San Francisco, San Francisco, CA
| | - Emilie Barruet
- Department of Medicine and the Institute for Human Genetics and the Program for Craniofacial Biology, University of California San Francisco, San Francisco, CA
| | - Blanca M Morales
- Department of Medicine and the Institute for Human Genetics and the Program for Craniofacial Biology, University of California San Francisco, San Francisco, CA
| | - Marianne C Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Antoine J W P Rosenberg
- Department of Oral and Maxillofacial Surgery and Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery and Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
79
|
Filonov D, Tice R, Luo R, Grotegut C, Van Kanegan MJ, Ludlow JW, Il'yasova D, Kinev A. Initial Assessment of Variability of Responses to Toxicants in Donor-Specific Endothelial Colony Forming Cells. Front Public Health 2018; 6:369. [PMID: 30622937 PMCID: PMC6308159 DOI: 10.3389/fpubh.2018.00369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 12/03/2018] [Indexed: 12/14/2022] Open
Abstract
There is increased interest in using high throughput in vitro assays to characterize human population variability in response to toxicants and drugs. Utilizing primary human endothelial colony-forming cells (ECFCs) isolated from blood would be highly useful for this purpose because these cells are involved in neonatal and adult vasculogenesis. We characterized the cytotoxicity of four known toxic chemicals (NaAsO2, CdCl2, tributyltin [TBT], and menadione) and their four relatively nontoxic counterparts (Na2HAsO4, ZnCl2, SnCl2, and phytonadione, respectively) in eight ECFC clones representing four neonatal donors (2 male and 2 female donors, 2 clones per donor). ECFCs were exposed to 9 concentrations of each chemical in duplicate; cell viability was evaluated 48 h later using the fluorescent vital dye fluorescent dye 5-Carboxyfluorescein Diacetate (CFDA), yielding concentration-effect curves from each experiment. Technical (day-to-day) variability of the assay, assessed from three independent experiments, was low: p-values for the differences of results were 0.74 and 0.64 for the comparison of day 2 vs. day 1 and day 3 vs. day 1, respectively. The statistical analysis used to compare the entire concentration-effect curves has revealed significant differences in levels of cytotoxicity induced by the toxic and relatively nontoxic chemical counterparts, demonstrating that donor-specific ECFCs can clearly differentiate between these two groups of chemicals. Partitioning of the total variance in the nested design assessed the contributions of between-clone and between-donor variability for different levels of cytotoxicity. Individual ECFC clones demonstrated highly reproducible responses to the chemicals. The most toxic chemical was TBT, followed by NaAsO2, CdCl2, and Menadione. Nontoxic counterparts exhibited low cytotoxicity at the higher end of concentration ranges tested. Low variability was observed between ECFC clones obtained from the same donor or different donors for CdCl2, NaAsO2, and TBT, but for menadione, the between-donor variability was much greater than the between-clone variability. The low between-clone variability indicates that an ECFC clone may represent an individual donor in cell-based assays, although this finding must be confirmed using a larger number of donors. Such confirmation would demonstrate that an in vitro ECFC-based testing platform can be used to characterize the inter-individual variability of neonatal ECFCs exposed to drugs and/or environmental toxicants.
Collapse
Affiliation(s)
| | - Raymond Tice
- Creative Scientist, Inc.Durham, NC, United States
| | - Ruiyan Luo
- School of Public Health, Georgia State University, Atlanta, GA, United States
| | - Chad Grotegut
- Duke University Medical Center, Durham, NC, United States
| | | | | | - Dora Il'yasova
- School of Public Health, Georgia State University, Atlanta, GA, United States
| | | |
Collapse
|
80
|
Tasev D, Dekker-Vroling L, van Wijhe M, Broxterman HJ, Koolwijk P, van Hinsbergh VWM. Hypoxia Impairs Initial Outgrowth of Endothelial Colony Forming Cells and Reduces Their Proliferative and Sprouting Potential. Front Med (Lausanne) 2018; 5:356. [PMID: 30619865 PMCID: PMC6306419 DOI: 10.3389/fmed.2018.00356] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 12/06/2018] [Indexed: 01/09/2023] Open
Abstract
Vascular homeostasis and regeneration in ischemic tissue relies on intrinsic competence of the tissue to rapidly recruit endothelial cells for vascularization. The mononuclear cell (MNC) fraction of blood contains circulating progenitors committed to endothelial lineage. These progenitors give rise to endothelial colony-forming cells (ECFCs) that actively participate in neovascularization of ischemic tissue. To evaluate if the initial clonal outgrowth of ECFCs from cord (CB) and peripheral blood (PB) was stimulated by hypoxic conditions, MNCs obtained from CB and PB were subjected to 20 and 1% O2 cell culture conditions. Clonal outgrowth was followed during a 30 day incubation period. Hypoxia impaired the initial outgrowth of ECFC colonies from CB and also reduced their number that were developing from PB MNCs. Three days of oxygenation (20% O2) prior to hypoxia could overcome the initial CB-ECFC outgrowth. Once proliferating and subcultured the CB-ECFCs growth was only modestly affected by hypoxia; proliferation of PB-ECFCs was reduced to a similar extent (18-30% reduction). Early passages of subcultured CB- and PB-ECFCs contained only viable cells and few if any senescent cells. Tube formation by subcultured PB-ECFCs was also markedly inhibited by continuous exposure to 1% O2. Gene expression profiles point to regulation of the cell cycle and metabolism as major altered gene clusters. Finally we discuss our counterintuitive observations in the context of the important role that hypoxia has in promoting neovascularization.
Collapse
Affiliation(s)
- Dimitar Tasev
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Laura Dekker-Vroling
- Department of Medical Oncology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Michiel van Wijhe
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Henk J Broxterman
- Department of Medical Oncology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Pieter Koolwijk
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Victor W M van Hinsbergh
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, Netherlands
| |
Collapse
|
81
|
Keighron C, Lyons CJ, Creane M, O'Brien T, Liew A. Recent Advances in Endothelial Progenitor Cells Toward Their Use in Clinical Translation. Front Med (Lausanne) 2018; 5:354. [PMID: 30619864 PMCID: PMC6305310 DOI: 10.3389/fmed.2018.00354] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 12/03/2018] [Indexed: 12/28/2022] Open
Abstract
Since the discovery of Endothelial Progenitor Cells (EPC) by Asahara and colleagues in 1997, an increasing number of preclinical studies have shown that EPC based therapy is feasible, safe, and efficacious in multiple disease states. Subsequently, this has led to several, mainly early phase, clinical trials demonstrating the feasibility and safety profile of EPC therapy, with the suggestion of efficacy in several conditions including ischemic heart disease, pulmonary arterial hypertension and decompensated liver cirrhosis. Despite the use of the common term “EPC,” the characteristics, manufacturing methods and subset of the cell type used in these studies often vary significantly, rendering clinical translation challenging. It has recently been acknowledged that the true EPC is the endothelial colony forming cells (ECFC). The objective of this review was to summarize and critically appraise the registered and published clinical studies using the term “EPC,” which encompasses a heterogeneous cell population, as a therapeutic agent. Furthermore, the preclinical data using ECFC from the PubMed and Web of Science databases were searched and analyzed. We noted that despite the promising effect of ECFC on vascular regeneration, no clinical study has stemmed from these preclinical studies. We showed that there is a lack of information registered on www.clinicaltrials.gov for EPC clinical trials, specifically on cell culture methods. We also highlighted the importance of a detailed definition of the cell type used in EPC clinical trials to facilitate comparisons between trials and better understanding of the potential clinical benefit of EPC based therapy. We concluded our review by discussing the potential and limitations of EPC based therapy in clinical settings.
Collapse
Affiliation(s)
- Cameron Keighron
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science and Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
| | - Caomhán J Lyons
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science and Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
| | - Michael Creane
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science and Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
| | - Timothy O'Brien
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science and Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
| | - Aaron Liew
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science and Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
| |
Collapse
|
82
|
Son Y, Kwon SM, Cho JY. CD276 (B7-H3) Maintains Proliferation and Regulates Differentiation in Angiogenic Function in Late Endothelial Progenitor Cells. Stem Cells 2018; 37:382-394. [PMID: 30379377 DOI: 10.1002/stem.2944] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 09/22/2018] [Accepted: 10/16/2018] [Indexed: 12/24/2022]
Abstract
Endothelial progenitor cells (EPCs) provide an important source of recovery from blood vessel dysfunction. Late EPCs (LEPCs) are circulating blood cells that are capable of promoting vascular repair. Using transcriptome analysis, we identified distinctive LEPC profiles and found that CD276 (B7-H3) mRNA is strongly expressed in LEPCs. CD276 protein is present abundantly on the cell surface of LEPC when analyzed by fluorescence-activated cell sorter and immunocytochemistry. CD276, a B7 family member, is a type I transmembrane glycoprotein. The role of CD276 in LEPCs remains unknown. CD276 knockdown by lentivirus transduction in LEPCs significantly decreased proliferation and increased apoptosis of LEPCs in vitro. After CD276 silencing, the cell cycle of LEPCs was prone to remain at the G0/G1 phase, and the cell migration rates as well as transwell and wound-healing migration were decreased. CD276 knockdown in LEPCs increased the G1 phase regulators cyclin D2/D3/E1-cyclin-dependent kinases (CDK2/4/6), but decreased the S-G2-M phase regulators cyclin A/B-CDK1. However, LEPCs with CD276 knockdown resulted in increased tube formation in vitro and angiogenesis in a Matrigel plug assay in vivo. FoxC1/C2, an upstream signal of Notch in arterial cell proliferation, and Hey1/2, which is known to promote arterial differentiation in the vasculature, were upregulated in CD276 knockdown LEPCs. In LEPCS, CD276 has a positive effect on proliferation and migration of endothelial cells, but negative effects on angiogenesis, particularly endothelial cell differentiation. Our data indicate, for therapeutic purpose, that CD276 can be used to acquire and maintain cell populations of LEPCs and blocking CD276 will promote angiogenetic differentiation. We found that CD276 (B7-H3) is enriched on the cell membrane of LEPCs. CD276 knockdown reduced proliferation and migration of LEPCs by increasing cell cycle inhibitors such as p21cip1 and pRb and decreasing pErk1/2 and pAkt but promoted angiogenesis and endothelial cell differentiation by elevating vascular endothelial growth factor-vascular endothelial growth factor receptor 1 and p-p38. Stem Cells 2019;37:382-394.
Collapse
Affiliation(s)
- YeonSung Son
- Department of Biochemistry, BK21 Plus and Research Institute for Veterinary Science, School of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Sang-Mo Kwon
- Laboratory for Vascular Medicine & Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Je-Yoel Cho
- Department of Biochemistry, BK21 Plus and Research Institute for Veterinary Science, School of Veterinary Medicine, Seoul National University, Seoul, Korea
| |
Collapse
|
83
|
Castro PR, Barbosa AS, Pereira JM, Ranfley H, Felipetto M, Gonçalves CAX, Paiva IR, Berg BB, Barcelos LS. Cellular and Molecular Heterogeneity Associated with Vessel Formation Processes. BIOMED RESEARCH INTERNATIONAL 2018; 2018:6740408. [PMID: 30406137 PMCID: PMC6199857 DOI: 10.1155/2018/6740408] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 09/06/2018] [Indexed: 12/11/2022]
Abstract
The microvasculature heterogeneity is a complex subject in vascular biology. The difficulty of building a dynamic and interactive view among the microenvironments, the cellular and molecular heterogeneities, and the basic aspects of the vessel formation processes make the available knowledge largely fragmented. The neovascularisation processes, termed vasculogenesis, angiogenesis, arteriogenesis, and lymphangiogenesis, are important to the formation and proper functioning of organs and tissues both in the embryo and the postnatal period. These processes are intrinsically related to microvascular cells, such as endothelial and mural cells. These cells are able to adjust their activities in response to the metabolic and physiological requirements of the tissues, by displaying a broad plasticity that results in a significant cellular and molecular heterogeneity. In this review, we intend to approach the microvasculature heterogeneity in an integrated view considering the diversity of neovascularisation processes and the cellular and molecular heterogeneity that contribute to microcirculatory homeostasis. For that, we will cover their interactions in the different blood-organ barriers and discuss how they cooperate in an integrated regulatory network that is controlled by specific molecular signatures.
Collapse
Affiliation(s)
- Pollyana Ribeiro Castro
- Department of Physiology and Biophysics, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Alan Sales Barbosa
- Department of Physiology and Biophysics, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Jousie Michel Pereira
- Department of Physiology and Biophysics, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Hedden Ranfley
- Department of Physiology and Biophysics, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Mariane Felipetto
- Department of Physiology and Biophysics, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Carlos Alberto Xavier Gonçalves
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Isabela Ribeiro Paiva
- Department of Pharmacology, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Bárbara Betônico Berg
- Department of Pharmacology, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Luciola Silva Barcelos
- Department of Physiology and Biophysics, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Brazil
| |
Collapse
|
84
|
Shao Y, Li X, Wood JW, Ma JX. Mitochondrial dysfunctions, endothelial progenitor cells and diabetic retinopathy. J Diabetes Complications 2018; 32:966-973. [PMID: 30068485 DOI: 10.1016/j.jdiacomp.2018.06.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/18/2018] [Accepted: 06/28/2018] [Indexed: 12/11/2022]
Abstract
AIM Diabetic retinopathy (DR) is the leading cause of vision loss in the working age population. Endothelial progenitor cells (EPC) play a vital role in vascular damage repair. This article will review recent progress regarding mitochondrial and EPC dysfunction associated with DR. RESULTS EPCs represent a limited population of adult stem cells possessing vasculogenic potential postnatally; their number and function are changed in DR. Among all the function changes, mitochondrial dysfunction plays an important role in the dysregulation of EPCs, as mitochondria regulate energy balance, and cell fate determination. CONCLUSIONS Although the mechanism for the role of mitochondria dysregulation in EPC function remains elusive, mitochondria of EPCs represent a promising target for the treatment of the vasculopathy presented within DR.
Collapse
Affiliation(s)
- Yan Shao
- Tianjin Medical University Eye Hospital, Eye Institute & School of Optometry and Ophthalmology, Tianjin, China; Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73014, USA
| | - Xiaorong Li
- Tianjin Medical University Eye Hospital, Eye Institute & School of Optometry and Ophthalmology, Tianjin, China
| | - John W Wood
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73014, USA
| | - Jian-Xing Ma
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73014, USA.
| |
Collapse
|
85
|
Plein A, Fantin A, Denti L, Pollard JW, Ruhrberg C. Erythro-myeloid progenitors contribute endothelial cells to blood vessels. Nature 2018; 562:223-228. [PMID: 30258231 PMCID: PMC6289247 DOI: 10.1038/s41586-018-0552-x] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 08/17/2018] [Indexed: 12/16/2022]
Abstract
The earliest blood vessels in mammalian embryos are formed when endothelial cells differentiate from angioblasts and coalesce into tubular networks. Thereafter, the endothelium is thought to expand solely by proliferation of pre-existing endothelial cells. Here we show that a complementary source of endothelial cells is recruited into pre-existing vasculature after differentiation from the earliest precursors of erythrocytes, megakaryocytes and macrophages, the erythro-myeloid progenitors (EMPs) that are born in the yolk sac. A first wave of EMPs contributes endothelial cells to the yolk sac endothelium, and a second wave of EMPs colonizes the embryo and contributes endothelial cells to intraembryonic endothelium in multiple organs, where they persist into adulthood. By demonstrating that EMPs constitute a hitherto unrecognized source of endothelial cells, we reveal that embryonic blood vascular endothelium expands in a dual mechanism that involves both the proliferation of pre-existing endothelial cells and the incorporation of endothelial cells derived from haematopoietic precursors.
Collapse
Affiliation(s)
- Alice Plein
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Alessandro Fantin
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Laura Denti
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Jeffrey W Pollard
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | | |
Collapse
|
86
|
Dai NT, Huang WS, Chang FW, Wei LG, Huang TC, Li JK, Fu KY, Dai LG, Hsieh PS, Huang NC, Wang YW, Chang HI, Parungao R, Wang Y. Development of a Novel Pre-Vascularized Three-Dimensional Skin Substitute Using Blood Plasma Gel. Cell Transplant 2018; 27:1535-1547. [PMID: 30203684 PMCID: PMC6180730 DOI: 10.1177/0963689718797570] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Skin substitutes with existing vascularization are in great demand for the repair of
full-thickness skin defects. In the present study, we hypothesized that a pre-vascularized
skin substitute can potentially promote wound healing. Novel three-dimensional (3D) skin
substitutes were prepared by seeding a mixture of human endothelial progenitor cells
(EPCs) and fibroblasts into a human plasma/calcium chloride formed gel scaffold, and
seeding keratinocytes onto the surface of the plasma gel. The capacity of the EPCs to
differentiate into a vascular-like tubular structure was evaluated using
immunohistochemistry analysis and WST-8 assay. Experimental studies in mouse
full-thickness skin wound models showed that the pre-vascularized gel scaffold
significantly accelerated wound healing 7 days after surgery, and resembled normal skin
structures after 14 days post-surgery. Histological analysis revealed that
pre-vascularized gel scaffolds were well integrated in the host skin, resulting in the
vascularization of both the epidermis and dermis in the wound area. Moreover, mechanical
strength analysis demonstrated that the healed wound following the implantation of the
pre-vascularized gel scaffolds exhibited good tensile strength. Taken together, this novel
pre-vascularized human plasma gel scaffold has great potential in skin tissue
engineering.
Collapse
Affiliation(s)
- Niann-Tzyy Dai
- 1 Division of Plastic and Reconstructive Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Wen-Shyan Huang
- 2 Plastic and Reconstructive Surgery, Zouying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan, R.O.C
| | - Fang-Wei Chang
- 3 Department of Obstetrics & Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Lin-Gwei Wei
- 4 Division of Plastic and Reconstructive Surgery, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan, R.O.C
| | - Tai-Chun Huang
- 1 Division of Plastic and Reconstructive Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Jhen-Kai Li
- 1 Division of Plastic and Reconstructive Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Keng-Yen Fu
- 1 Division of Plastic and Reconstructive Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Lien-Guo Dai
- 5 Department of Orthopedics, Shuang Ho Hospital, Taipei Medical University, New Taipei, Taiwan, R.O.C
| | - Pai-Shan Hsieh
- 1 Division of Plastic and Reconstructive Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Nien-Chi Huang
- 1 Division of Plastic and Reconstructive Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Yi-Wen Wang
- 6 Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Hsin-I Chang
- 7 Department of Biochemical Science and Technology, National Chiayi University, Chiayi, Taiwan, R.O.C
| | - Roxanne Parungao
- 8 Burns Research Group, ANZAC Research Institute, Concord Hospital, University of Sydney, New South Wales, Australia
| | - Yiwei Wang
- 8 Burns Research Group, ANZAC Research Institute, Concord Hospital, University of Sydney, New South Wales, Australia
| |
Collapse
|
87
|
Lo Gullo A, Aragona CO, Scuruchi M, Versace AG, Saitta A, Imbalzano E, Loddo S, Campo GM, Mandraffino G. Endothelial progenitor cells and rheumatic disease modifying therapy. Vascul Pharmacol 2018; 108:8-14. [PMID: 29842927 DOI: 10.1016/j.vph.2018.05.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 04/12/2018] [Accepted: 05/18/2018] [Indexed: 02/07/2023]
Abstract
Rheumatic diseases are associated with accelerated atherosclerosis and with increased risk of cardiovascular morbidity and mortality. The mechanisms underlying the higher prevalence of cardiovascular disease are not completely clarified, but it is likely that a pivotal role is played by vascular inflammation and consequently to altered vascular endothelium homeostasis. Also, high prevalence of traditional risk factors, proatherogenic activation and endothelial dysfunction further contribute to vascular damage. Circulating endothelial progenitor cells (EPCs) can restore dysfunctional endothelium and protect against atherosclerotic vascular disease. However, abnormalities in number and function of these cells in patients with rheumatic condition have been extensively reported. During the last years, growing interest in the mechanisms of endothelial renewal and its potential as a therapy for CVD has been shown; in addition, pioneering studies show that EPC dysfunction might be improved with pharmacological strategies. However, how to restore EPC function, and whether achieving this aim may be effective in preventing cardiovascular complications in rheumatic disease, remain to be established. In this review we report an overview on the current stand of knowledge on the effect of pharmaceutical and lifestyle intervention in improving EPCs number and function in rheumatic disease.
Collapse
Affiliation(s)
- Alberto Lo Gullo
- Department of Clinical and Experimental Medicine, University of Messina, Italy.
| | | | - Michele Scuruchi
- Department of Clinical and Experimental Medicine, University of Messina, Italy
| | | | - Antonino Saitta
- Department of Clinical and Experimental Medicine, University of Messina, Italy
| | - Egidio Imbalzano
- Department of Clinical and Experimental Medicine, University of Messina, Italy
| | - Saverio Loddo
- Department of Clinical and Experimental Medicine, University of Messina, Italy
| | | | | |
Collapse
|
88
|
Profiling cellular morphodynamics by spatiotemporal spectrum decomposition. PLoS Comput Biol 2018; 14:e1006321. [PMID: 30071020 PMCID: PMC6091976 DOI: 10.1371/journal.pcbi.1006321] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 08/14/2018] [Accepted: 06/22/2018] [Indexed: 12/12/2022] Open
Abstract
Cellular morphology and associated morphodynamics are widely used for qualitative and quantitative assessments of cell state. Here we implement a framework to profile cellular morphodynamics based on an adaptive decomposition of local cell boundary motion into instantaneous frequency spectra defined by the Hilbert-Huang transform (HHT). Our approach revealed that spontaneously migrating cells with approximately homogeneous molecular makeup show remarkably consistent instantaneous frequency distributions, though they have markedly heterogeneous mobility. Distinctions in cell edge motion between these cells are captured predominantly by differences in the magnitude of the frequencies. We found that acute photo-inhibition of Vav2 guanine exchange factor, an activator of the Rho family of signaling proteins coordinating cell motility, produces significant shifts in the frequency distribution, but does not affect frequency magnitude. We therefore concluded that the frequency spectrum encodes the wiring of the molecular circuitry that regulates cell boundary movements, whereas the magnitude captures the activation level of the circuitry. We also used HHT spectra as multi-scale spatiotemporal features in statistical region merging to identify subcellular regions of distinct motion behavior. In line with our conclusion that different HHT spectra relate to different signaling regimes, we found that subcellular regions with different morphodynamics indeed exhibit distinct Rac1 activities. This algorithm thus can serve as an accurate and sensitive classifier of cellular morphodynamics to pinpoint spatial and temporal boundaries between signaling regimes. Many studies in cell biology employ global shape descriptors to probe mechanisms of cell morphogenesis. Here, we implement a framework in this paper to profile cellular morphodynamics very locally. We employ the Hilbert-Huang transform (HHT) to extract along the entire cell edge spectra of instantaneous edge motion frequency and magnitude and use them to classify overall cell behavior as well as subcellular edge sectors of distinct dynamics. We find in fibroblast-like COS7 cells that the marked heterogeneity in mobility of an unstimulated population is fully captured by differences in the magnitude spectra, while the frequency spectra are conserved between cells. Using optogenetics to acutely inhibit morphogenetic signaling pathways we find that these molecular shifts are reflected by changes in the frequency spectra but not in the magnitude spectra. After clustering cell edge sectors with distinct morphodynamics we observe in cells expressing a Rac1 activity biosensor that the sectors with different frequency spectra associate with different signaling intensity and dynamics. Together, these observations let us conclude that the frequency spectrum encodes the wiring of the molecular circuitry that regulates edge movements, whereas the magnitude captures the activation level of the circuitry.
Collapse
|
89
|
Affiliation(s)
- Yao Xie
- From the Cardiovascular Division, King's College London BHF Centre, London, UK (Y.X., Q.X.); and Institute of Respiratory, Xinqiao Hospital, Third Military Medical University, Chongqing, China (Y.F.)
| | - Ye Fan
- From the Cardiovascular Division, King's College London BHF Centre, London, UK (Y.X., Q.X.); and Institute of Respiratory, Xinqiao Hospital, Third Military Medical University, Chongqing, China (Y.F.)
| | - Qingbo Xu
- From the Cardiovascular Division, King's College London BHF Centre, London, UK (Y.X., Q.X.); and Institute of Respiratory, Xinqiao Hospital, Third Military Medical University, Chongqing, China (Y.F.).
| |
Collapse
|
90
|
Siddiquee AAM, Adaikan PG, Lau LC, Said BB, Chong M, Chan J, Teoh SH. Endothelial colony forming cells from human umbilical cord blood improved severe erectile dysfunction in obese type II diabetic rats. Life Sci 2018; 207:272-283. [PMID: 29920249 DOI: 10.1016/j.lfs.2018.06.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 06/07/2018] [Accepted: 06/15/2018] [Indexed: 01/03/2023]
Abstract
AIM To investigate the effect of intracavernous injection of human umbilical cord blood derived endothelial colony forming cells (HUCB ECFCs) on erectile dysfunction (ED) in Zucker Diabetic Fatty (ZDF) rat model. METHODS Erectile function was assessed by cavernous nerve electrostimulation in ZDF rats aged 20-28 weeks. Following confirmation of severe ED at the age of 28 weeks, 21 ZDF rats were randomly assigned to three experimental groups: 1 million ECFCs, 2 million ECFCs, and phosphate buffered saline (PBS). Four weeks after intracavernous injection, the efficacy of ECFCs was quantified by intracavernous pressure (ICP) measurement, Masson's trichrome staining, immunohistologic and immunoblot analyses and TUNEL assay. KEY FINDINGS Intracavernous ECFC administration improved ICP in a dose-dependent manner in comparison to the age-matched PBS group. Functional improvement in ICP was accompanied by a significant restoration of the cavernosal endothelial and smooth muscle cell content and cavernosal nerve function. The percentage eNOS and nNOS positive cavernosal cells, and their respective protein expression levels and nNOS positive cells in the dorsal penile nerve in 2 million ECFCs treated groups were significantly higher than the PBS group. TUNEL stain quantification showed a significant decrease in cavernosal apoptosis following ECFC treatment. SIGNIFICANCE The results are expected to provide a scientific basis to further study the clinical application of HUCB ECFCs in ameliorating ED in human. CONCLUSIONS HUCB ECFCs significantly improved severe ED in ZDF rats through improvement of the nerve and endothelium function and restoration of smooth muscle in the cavernosum by overcoming the cavernosal apoptosis.
Collapse
Affiliation(s)
- Abrar Al-Mahmood Siddiquee
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228
| | - P Ganesan Adaikan
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228.
| | - Lang Chu Lau
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228
| | - Baharudin Bin Said
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228
| | - Mark Chong
- School of Chemical & Biomedical Engineering, Nanyang Technological University, Singapore
| | - Jerry Chan
- Kandang Kerbau Women's & Children's Hospital, Singapore
| | - Swee Hin Teoh
- School of Chemical & Biomedical Engineering, Nanyang Technological University, Singapore
| |
Collapse
|
91
|
Del Papa N, Pignataro F. The Role of Endothelial Progenitors in the Repair of Vascular Damage in Systemic Sclerosis. Front Immunol 2018; 9:1383. [PMID: 29967618 PMCID: PMC6015881 DOI: 10.3389/fimmu.2018.01383] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 06/04/2018] [Indexed: 01/17/2023] Open
Abstract
Systemic sclerosis (SSc) is a connective tissue disease characterized by a complex pathological process where the main scenario is represented by progressive loss of microvascular bed, with the consequent progressive fibrotic changes in involved organ and tissues. Although most aspects of vascular injury in scleroderma are poorly understood, recent data suggest that the scleroderma impairment of neovascularization could be related to both angiogenesis and vasculogenesis failure. Particularly, compensatory angiogenesis does not occur normally in spite of an important increase in many angiogenic factors either in SSc skin or serum. Besides insufficient angiogenesis, the contribution of defective vasculogenesis to SSc vasculopathy has been extensively studied. Over the last decades, our understanding of the processes responsible for the formation of new vessels after tissue ischemia has increased. In the past, adult neovascularization was thought to depend mainly on angiogenesis (a process by which new vessels are formed by the proliferation and migration of mature endothelial cells). More recently, increased evidence suggests that stem cells mobilize from the bone marrow into the peripheral blood (PB), differentiate in circulating endothelial progenitors (EPCs), and home to site of ischemia to contribute to de novo vessel formation. Significant advances have been made in understanding the biology of EPCs, and molecular mechanisms regulating EPC function. Autologous EPCs now are becoming a novel treatment option for therapeutic vascularization and vascular repair, mainly in ischemic diseases. However, different diseases, such as cardiovascular diseases, diabetes, and peripheral artery ischemia are related to EPC dysfunction. Several studies have shown that EPCs can be detected in the PB of patients with SSc and are impaired in their function. Based on an online literature search (PubMed, EMBASE, and Web of Science, last updated December 2017) using keywords related to “endothelial progenitor cells” and “Systemic Sclerosis,” “scleroderma vasculopathy,” “angiogenesis,” “vasculogenesis,” this review gives an overview on the large body of data of current research in this issue, including controversies over the identity and functions of EPCs, their meaning as biomarker of SSc microangiopathy and their clinical potency.
Collapse
|
92
|
Besnier M, Gasparino S, Vono R, Sangalli E, Facoetti A, Bollati V, Cantone L, Zaccagnini G, Maimone B, Fuschi P, Da Silva D, Schiavulli M, Aday S, Caputo M, Madeddu P, Emanueli C, Martelli F, Spinetti G. miR-210 Enhances the Therapeutic Potential of Bone-Marrow-Derived Circulating Proangiogenic Cells in the Setting of Limb Ischemia. Mol Ther 2018; 26:1694-1705. [PMID: 29908843 DOI: 10.1016/j.ymthe.2018.06.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 06/05/2018] [Accepted: 06/05/2018] [Indexed: 12/28/2022] Open
Abstract
Therapies based on circulating proangiogenic cells (PACs) have shown promise in ischemic disease models but require further optimization to reach the bedside. Ischemia-associated hypoxia robustly increases microRNA-210 (miR-210) expression in several cell types, including endothelial cells (ECs). In ECs, miR-210 represses EphrinA3 (EFNA3), inducing proangiogenic responses. This study provides new mechanistic evidences for a role of miR-210 in PACs. PACs were obtained from either adult peripheral blood or cord blood. miR-210 expression was modulated with either an inhibitory complementary oligonucleotide (anti-miR-210) or a miRNA mimic (pre-miR-210). Scramble and absence of transfection served as controls. As expected, hypoxia increased miR-210 in PACs. In vivo, migration toward and adhesion to the ischemic endothelium facilitate the proangiogenic actions of transplanted PACs. In vitro, PAC migration toward SDF-1α/CXCL12 was impaired by anti-miR-210 and enhanced by pre-miR-210. Moreover, pre-miR-210 increased PAC adhesion to ECs and supported angiogenic responses in co-cultured ECs. These responses were not associated with changes in extracellular miR-210 and were abrogated by lentivirus-mediated EFNA3 overexpression. Finally, ex-vivo pre-miR-210 transfection predisposed PACs to induce post-ischemic therapeutic neovascularization and blood flow recovery in an immunodeficient mouse limb ischemia model. In conclusion, miR-210 modulates PAC functions and improves their therapeutic potential in limb ischemia.
Collapse
Affiliation(s)
- Marie Besnier
- Bristol Heart Institute, School of Clinical Science, University of Bristol, Bristol, UK
| | - Stefano Gasparino
- Laboratory of Cardiovascular Research, IRCCS MultiMedica, Milan, Italy
| | - Rosa Vono
- Laboratory of Cardiovascular Research, IRCCS MultiMedica, Milan, Italy
| | - Elena Sangalli
- Laboratory of Cardiovascular Research, IRCCS MultiMedica, Milan, Italy
| | - Amanda Facoetti
- Laboratory of Cardiovascular Research, IRCCS MultiMedica, Milan, Italy
| | - Valentina Bollati
- EPIGET Lab, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Laura Cantone
- EPIGET Lab, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Germana Zaccagnini
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato, Italy
| | - Biagina Maimone
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato, Italy
| | - Paola Fuschi
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato, Italy
| | - Daniel Da Silva
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato, Italy
| | - Michele Schiavulli
- AORN Santobono Pausilipon, Transfusion Medicine and Bone Marrow Transplantation Unit-Regional Reference Center for Coagulation Disorders, Napoli, Italy
| | - Sezin Aday
- Bristol Heart Institute, School of Clinical Science, University of Bristol, Bristol, UK
| | - Massimo Caputo
- Bristol Heart Institute, School of Clinical Science, University of Bristol, Bristol, UK
| | - Paolo Madeddu
- Bristol Heart Institute, School of Clinical Science, University of Bristol, Bristol, UK
| | - Costanza Emanueli
- Bristol Heart Institute, School of Clinical Science, University of Bristol, Bristol, UK; National Heart and Lung Institute, Imperial College London, London, UK
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato, Italy.
| | - Gaia Spinetti
- Laboratory of Cardiovascular Research, IRCCS MultiMedica, Milan, Italy.
| |
Collapse
|
93
|
Isolation and characterization of endothelial colony-forming cells from mononuclear cells of rat bone marrow. Exp Cell Res 2018; 370:116-126. [PMID: 29908162 DOI: 10.1016/j.yexcr.2018.06.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 06/12/2018] [Accepted: 06/13/2018] [Indexed: 12/25/2022]
Abstract
Transplantation of bone marrow-derived endothelial progenitor cells (BM-EPCs) has been used as a therapeutic strategy for vascular repair. However, it remains controversial whether BM-EPCs exhibit clonal endothelial colony-forming cell (ECFC) capacity, a characteristic of true EPCs. The aim of this study was to isolate and explore the cellular properties of BM-ECFCs. We isolated BM-ECFCs from rat bone marrow with high purity via an optimized method. This approach involved the removal of selective colonies based on the conventional differential adhesive culture method used to isolate ECFCs from peripheral and umbilical cord blood. Our results indicate that primary colony BM-ECFCs display a panel of surface antigen markers consistent with endothelial cells. These BM-ECFCs coexpress CD34, CD133, and VEGFR2 at high levels, and these levels decrease with passaging. These cells have high potential for proliferation, migration, and formation of capillary-like structures on Matrigel, and these abilities are retained during ex vivo expansion. Furthermore, BM-ECFCs cultured with 10% or 20% fetal bovine serum demonstrated two different patterns of spontaneous capillary-like structure formation. These results provide a foundation for isolation of ECFCs from human bone marrow for autologous cell transplantation and tissue engineering applications in the future.
Collapse
|
94
|
Yang JX, Pan YY, Wang XX, Qiu YG, Mao W. Endothelial progenitor cells in age-related vascular remodeling. Cell Transplant 2018; 27:786-795. [PMID: 29882417 PMCID: PMC6047273 DOI: 10.1177/0963689718779345] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Accumulating evidence has demonstrated that endothelial progenitor cells (EPCs) could facilitate the reendothelialization of injured arteries by replacing the dysfunctional endothelial cells, thereby suppressing the formation of neointima. Meanwhile, other findings suggest that EPCs may be involved in the pathogenesis of age-related vascular remodeling. This review is presented to summarize the characteristics of EPCs and age-related vascular remodeling. In addition, the role of EPCs in age-related vascular remodeling and possible solutions for improving the therapeutic effects of EPCs in the treatment of age-related diseases are discussed.
Collapse
Affiliation(s)
- Jin-Xiu Yang
- 1 Department of Cardiology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, P.R. China.,2 Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, P.R. China
| | - Yan-Yun Pan
- 1 Department of Cardiology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, P.R. China
| | - Xing-Xiang Wang
- 2 Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, P.R. China
| | - Yuan-Gang Qiu
- 1 Department of Cardiology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, P.R. China
| | - Wei Mao
- 1 Department of Cardiology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, P.R. China
| |
Collapse
|
95
|
Rasheed A, Tsai R, Cummins CL. Loss of the Liver X Receptors Disrupts the Balance of Hematopoietic Populations, With Detrimental Effects on Endothelial Progenitor Cells. J Am Heart Assoc 2018; 7:JAHA.117.007787. [PMID: 29739800 PMCID: PMC6015321 DOI: 10.1161/jaha.117.007787] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background The liver X receptors (LXRs; α/β) are nuclear receptors known to regulate cholesterol homeostasis and the production of select hematopoietic populations. The objective of this study was to determine the importance of LXRs and a high‐fat high‐cholesterol diet on global hematopoiesis, with special emphasis on endothelial progenitor cells (EPCs), a vasoreparative cell type that is derived from bone marrow hematopoietic stem cells. Methods and Results Wild‐type and LXR double‐knockout (Lxrαβ−/−) mice were fed a Western diet (WD) to increase plasma cholesterol levels. In WD‐fed Lxrαβ−/− mice, flow cytometry and complete blood cell counts revealed that hematopoietic stem cells, a myeloid progenitor, and mature circulating myeloid cells were increased; EPC numbers were significantly decreased. Hematopoietic stem cells from WD‐fed Lxrαβ−/− mice showed increased cholesterol content, along with increased myeloid colony formation compared with chow‐fed mice. In contrast, EPCs from WD‐fed Lxrαβ−/− mice also demonstrated increased cellular cholesterol content that was associated with greater expression of the endothelial lineage markers Cd144 and Vegfr2, suggesting accelerated differentiation of the EPCs. Treatment of human umbilical vein endothelial cells with conditioned medium collected from these EPCs increased THP‐1 monocyte adhesion. Increased monocyte adhesion to conditioned medium–treated endothelial cells was recapitulated with conditioned medium from Lxrαβ−/−EPCs treated with cholesterol ex vivo, suggesting cholesterol is the main component of the WD inducing EPC dysfunction. Conclusions LXRs are crucial for maintaining the balance of hematopoietic cells in a hypercholesterolemic environment and for mitigating the negative effects of cholesterol on EPC differentiation/secretome changes that promote monocyte‐endothelial adhesion.
Collapse
Affiliation(s)
- Adil Rasheed
- Department of Pharmaceutical Sciences, University of Toronto, Ontario, Canada
| | - Ricky Tsai
- Department of Pharmaceutical Sciences, University of Toronto, Ontario, Canada
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences, University of Toronto, Ontario, Canada .,Banting and Best Diabetes Centre, Toronto, Ontario, Canada.,The Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada
| |
Collapse
|
96
|
Shear stress: An essential driver of endothelial progenitor cells. J Mol Cell Cardiol 2018; 118:46-69. [PMID: 29549046 DOI: 10.1016/j.yjmcc.2018.03.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/08/2018] [Accepted: 03/09/2018] [Indexed: 02/06/2023]
Abstract
The blood flow through vessels produces a tangential, or shear, stress sensed by their innermost layer (i.e., endothelium) and representing a major hemodynamic force. In humans, endothelial repair and blood vessel formation are mainly performed by circulating endothelial progenitor cells (EPCs) characterized by a considerable expression of vascular endothelial growth factor receptor 2 (VEGFR2), CD34, and CD133, pronounced tube formation activity in vitro, and strong reendothelialization or neovascularization capacity in vivo. EPCs have been proposed as a promising agent to induce reendothelialization of injured arteries, neovascularization of ischemic tissues, and endothelialization or vascularization of bioartificial constructs. A number of preconditioning approaches have been suggested to improve the regenerative potential of EPCs, including the use of biophysical stimuli such as shear stress. However, in spite of well-defined influence of shear stress on mature endothelial cells (ECs), articles summarizing how it affects EPCs are lacking. Here we discuss the impact of shear stress on homing, paracrine effects, and differentiation of EPCs. Unidirectional laminar shear stress significantly promotes homing of circulating EPCs to endothelial injury sites, induces anti-thrombotic and anti-atherosclerotic phenotype of EPCs, increases their capability to form capillary-like tubes in vitro, and enhances differentiation of EPCs into mature ECs in a dose-dependent manner. These effects are mediated by VEGFR2, Tie2, Notch, and β1/3 integrin signaling and can be abrogated by means of complementary siRNA/shRNA or selective pharmacological inhibitors of the respective proteins. Although the testing of sheared EPCs for vascular tissue engineering or regenerative medicine applications is still an unaccomplished task, favorable effects of unidirectional laminar shear stress on EPCs suggest its usefulness for their preconditioning.
Collapse
|
97
|
Basile DP, Collett JA, Yoder MC. Endothelial colony-forming cells and pro-angiogenic cells: clarifying definitions and their potential role in mitigating acute kidney injury. Acta Physiol (Oxf) 2018; 222:10.1111/apha.12914. [PMID: 28656611 PMCID: PMC5745310 DOI: 10.1111/apha.12914] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 05/10/2017] [Accepted: 06/21/2017] [Indexed: 12/12/2022]
Abstract
Acute kidney injury (AKI) represents a significant clinical concern that is associated with high mortality rates and also represents a significant risk factor for the development of chronic kidney disease (CKD). This article will consider alterations in renal endothelial function in the setting of AKI that may underlie impairment in renal perfusion and how inefficient vascular repair may manifest post-AKI and contribute to the potential transition to CKD. We provide updated terminology for cells previously classified as 'endothelial progenitor' that may mediate vascular repair such as pro-angiogenic cells and endothelial colony-forming cells. We consider how endothelial repair may be mediated by these different cell types following vascular injury, particularly in models of AKI. We further summarize the potential ability of these different cells to mitigate the severity of AKI, improve perfusion and maintain vascular structure in pre-clinical studies.
Collapse
Affiliation(s)
- David P. Basile
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine
| | - Jason A. Collett
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine
| | - Mervin C. Yoder
- Department of Pediatrics, Indiana University School of Medicine
| |
Collapse
|
98
|
Abstract
BACKGROUND Burn blister fluid contains several angiogenic factors to promote wound neovascularization. In our previous study, we found that deep partial-thickness burn (DPTB) wounds showed higher expression levels of angiogenin to enhance vascularization compared with superficial partial-thickness burn wounds. Neovascularization is a complex process that involves an interaction between circulating angiogenic cells and mediators. We hypothesized that in addition to angiogenic factors burn blisters may contain specific cell types. The aim of the present study was to characterize the specific cells present in burn blisters. METHODS Twenty-four burn blister fluid samples were obtained with informed consent from patients with superficial partial-thickness burn (n = 16) or DPTB (n = 8) wounds. Blister cells were isolated from individual intact blisters and characterized with flow cytometry analysis using CD14, CD34, vascular endothelial growth factor receptor 2, and CD133 markers. CD14 and CD34 blister cells were also isolated using a magnetic-activated cell sorting system to examine their potential for endothelial differentiation. Angiogenin levels in the burn blister fluids were evaluated with enzyme-linked immunosorbent assay. RESULTS CD14 cells were the most highly represented cell type in the burn fluids of both groups, although a significantly greater percentage of CD14 cells were observed in DPTB fluids. CD14 blister cells had a higher potency to differentiate into functional endothelial cells as compared with CD34 cells. The proportion of CD14 cells gradually increased after burn injury. In contrast to CD14 cells, angiogenin showed the highest expression levels at day 1 postburn. With regard to burn wound neovascularization, angiogenin expression was partially correlated with CD14 blister cells in the burn fluids. CONCLUSIONS We provide the first report on the characterization of blister cells in burn fluids. Our data suggest that CD14 blister cells may play a role in burn wound neovascularization. Measurement of CD14 blister cells serves as a possible tool for assessing burn wound status.
Collapse
|
99
|
Huuskes BM, DeBuque RJ, Polkinghorne KR, Samuel CS, Kerr PG, Ricardo SD. Endothelial Progenitor Cells and Vascular Health in Dialysis Patients. Kidney Int Rep 2018; 3:205-211. [PMID: 29340332 PMCID: PMC5762957 DOI: 10.1016/j.ekir.2017.09.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 09/10/2017] [Accepted: 09/11/2017] [Indexed: 02/06/2023] Open
Affiliation(s)
- Brooke M Huuskes
- Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
| | - Ryan J DeBuque
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
| | - Kevan R Polkinghorne
- Department of Nephrology, Monash Medical Centre and Monash University, Melbourne, Victoria, Australia.,School of Public Health and Preventative Medicine, Monash University, Prahan, Melbourne, Australia
| | - Chrishan S Samuel
- Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Melbourne, Victoria, Australia
| | - Peter G Kerr
- Department of Nephrology, Monash Medical Centre and Monash University, Melbourne, Victoria, Australia
| | - Sharon D Ricardo
- Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
100
|
Laurenzana A, Margheri F, Chillà A, Biagioni A, Margheri G, Calorini L, Fibbi G, Del Rosso M. Endothelial Progenitor Cells as Shuttle of Anticancer Agents. Hum Gene Ther 2018; 27:784-791. [PMID: 27502560 DOI: 10.1089/hum.2016.066] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Cell therapies are treatments in which stem or progenitor cells are stimulated to differentiate into specialized cells able to home to and repair damaged tissues. After their discovery, endothelial progenitor cells (EPCs) stimulated worldwide interest as possible vehicles to perform autologous cell therapy of tumors. Taking into account the tumor-homing properties of EPCs, two different approaches to control cancer progression have been pursued by combining cell-based therapy with gene therapy or with nanomedicine. The first approach is based on the possibility of engineering EPCs to express different transgenes, and the second is based on the capacity of EPCs to take up nanomaterials. Here we review the most important progress covering the following issues: the characterization of bona fide endothelial progenitor cells, their role in tumor vascularization and metastasis, and preclinical data about their use in cell-based tumor therapy, considering antiangiogenic, suicide, immune-stimulating, and oncolytic virus gene therapy. The mixed approach of EPC cell therapy and nanomedicine is discussed in terms of plasmonic-dependent thermoablation and molecular imaging.
Collapse
Affiliation(s)
- Anna Laurenzana
- 1 Department of Clinical and Experimental Biomedical Sciences, University of Florence , Florence, Italy
| | - Francesca Margheri
- 1 Department of Clinical and Experimental Biomedical Sciences, University of Florence , Florence, Italy
| | - Anastasia Chillà
- 1 Department of Clinical and Experimental Biomedical Sciences, University of Florence , Florence, Italy
| | - Alessio Biagioni
- 1 Department of Clinical and Experimental Biomedical Sciences, University of Florence , Florence, Italy
| | - Giancarlo Margheri
- 2 Institute for Complex Systems , National Research Council, Sesto Fiorentino, Italy
| | - Lido Calorini
- 1 Department of Clinical and Experimental Biomedical Sciences, University of Florence , Florence, Italy.,3 Center of Excellence for the Study at Molecular and Clinical Levels of Chronic, Degenerative, and Neoplastic Diseases to Develop Novel Therapies (DENOTHE) , Florence, Italy
| | - Gabriella Fibbi
- 1 Department of Clinical and Experimental Biomedical Sciences, University of Florence , Florence, Italy
| | - Mario Del Rosso
- 1 Department of Clinical and Experimental Biomedical Sciences, University of Florence , Florence, Italy.,3 Center of Excellence for the Study at Molecular and Clinical Levels of Chronic, Degenerative, and Neoplastic Diseases to Develop Novel Therapies (DENOTHE) , Florence, Italy
| |
Collapse
|