1
|
Gurel M, Zomer H, McFetridge C, Murfee WL, McFetridge PS. Physiologically-Modeled Dynamic Stimulation and Growth Factors Induce Differentiation of Mesenchymal Stem Cells to a Vascular Endothelial Cell Phenotype. Microcirculation 2025; 32:e70007. [PMID: 40120632 PMCID: PMC12012511 DOI: 10.1111/micc.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 03/04/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
OBJECTIVE Mesenchymal stem cells (MSCs) represent an attractive option as an endothelial cell (EC) source for regenerative medicine therapies. However, the differentiation of MSCs toward an ECs phenotype can be regulated by a complex and dynamic microenvironment, including specific growth factors as well as local mechanical cues. The objective of this work was to evaluate whether Physiologically-modeled dynamic stimulation (PMDS) characterized by continuous variability in pulse frequencies mimicking the dynamic temporal range of cardiac function would enhance MSC differentiation toward ECs compared to a constant frequency stimulation. METHODS Mesenchymal stem cells were grown in a complex growth factor cocktail versus standard culture media to initiate the endothelial differentiation process, then subsequently exposed to PMDS that vary in duration and constant flow (CF) at a fixed 10 dynes/cm2 shear stress and 1.3 Hz frequency. RESULTS Both PMDS and media type strongly influence cell differentiation and function. Cells were shown to significantly upregulate eNOS activity and displayed lower TNF-a induced leukocyte adhesion compared to cells cultured under CF, consistent with a more quiescent ECs phenotype that regulates anti-inflammatory and anti-thrombotic states. CONCLUSION These findings suggest that the dynamic microenvironment created by perfusion, in contrast to constant frequency, combined with growth factors, enhances MSCs differentiation toward a vascular endothelial-like phenotype.
Collapse
Affiliation(s)
- Mediha Gurel
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
- Biotechnology Research and Application Center, Cukurova University, Adana, Turkey
- Electronic and Automation Department, Bitlis Eren University, Bitlis, Turkey
| | - Helena Zomer
- Department of Physiological Sciences, University of Florida, Gainesville, Florida, USA
| | - Calum McFetridge
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Walter L Murfee
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Peter S McFetridge
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
2
|
Wang Y, Dong H, Dong T, Zhao L, Fan W, Zhang Y, Yao W. Treatment of cytokine release syndrome-induced vascular endothelial injury using mesenchymal stem cells. Mol Cell Biochem 2024; 479:1149-1164. [PMID: 37392343 DOI: 10.1007/s11010-023-04785-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/04/2023] [Indexed: 07/03/2023]
Abstract
Cytokine release syndrome (CRS) is an acute systemic inflammatory reaction in which hyperactivated immune cells suddenly release a large amount of cytokines, leading to exaggerated inflammatory responses, multiple organ dysfunction, and even death. Although palliative treatment strategies have significantly reduced the overall mortality, novel targeted treatment regimens with superior therapy efficacy are urgently needed. Vascular endothelial cells (ECs) are important target cells of systemic inflammation, and their destruction is considered to be the initiating event underlying many serious complications of CRS. Mesenchymal stem/stromal cells (MSCs) are multipotent cells with self-renewing differentiation capacity and immunomodulatory properties. MSC transplantation can effectively suppress the activation of immune cells, reduce the bulk release of cytokines, and repair damaged tissues and organs. Here, we review the molecular mechanisms underlying CRS-induced vascular endothelial injury and discuss potential treatments using MSCs. Preclinical studies demonstrate that MSC therapy can effectively repair endothelium damage and thus reduce the incidence and severity of ensuing CRS-induced complications. This review highlights the therapeutic role of MSCs in fighting against CRS-induced EC damage, and summarizes the possible therapeutic formulations of MSCs for improved efficacy in future clinical trials.
Collapse
Affiliation(s)
- Yuyan Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Yangtze University, Jingzhou, China
- Health Science Center, Yangtze University, Jingzhou, China
| | - Haibo Dong
- Wuhan Optics Valley Vcanbiopharma Co., Ltd, Wuhan, China
- Key Industrial Base for Stem Cell Engineering Products, Tianjin, China
| | - Tengyun Dong
- Wuhan Optics Valley Vcanbiopharma Co., Ltd, Wuhan, China
- Key Industrial Base for Stem Cell Engineering Products, Tianjin, China
| | - Lulu Zhao
- Wuhan Optics Valley Vcanbiopharma Co., Ltd, Wuhan, China
- Key Industrial Base for Stem Cell Engineering Products, Tianjin, China
| | - Wen Fan
- Department of Laboratory Medicine, The First Affiliated Hospital of Yangtze University, Jingzhou, China.
| | - Yu Zhang
- Wuhan Optics Valley Vcanbiopharma Co., Ltd, Wuhan, China.
- Key Industrial Base for Stem Cell Engineering Products, Tianjin, China.
- Haihe Laboratory of Cell Ecosystem, Tianjin, China.
| | - Weiqi Yao
- Wuhan Optics Valley Vcanbiopharma Co., Ltd, Wuhan, China.
- Key Industrial Base for Stem Cell Engineering Products, Tianjin, China.
- Department of Biology and Medicine, Hubei University of Technology, Wuhan, China.
| |
Collapse
|
3
|
Davoodi P, Rezaei N, Hassan M, Hay DC, Vosough M. Bioengineering vascularized liver tissue for biomedical research and application. Scand J Gastroenterol 2024; 59:623-629. [PMID: 38319110 DOI: 10.1080/00365521.2024.2310172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/18/2024] [Accepted: 01/20/2024] [Indexed: 02/07/2024]
Abstract
The liver performs a wide range of biological functions that are essential to body homeostasis. Damage to liver tissue can result in reduced organ function, and if chronic in nature can lead to organ scarring and progressive disease. Currently, donor liver transplantation is the only longterm treatment for end-stage liver disease. However, orthotopic organ transplantation suffers from several drawbacks that include organ scarcity and lifelong immunosuppression. Therefore, new therapeutic strategies are required. One promising strategy is the engineering of implantable and vascularized liver tissue. This resource could also be used to build the next generation of liver tissue models to better understand human health, disease and aging in vitro. This article reviews recent progress in the field of liver tissue bioengineering, including microfluidic-based systems, bio-printed vascularized tissue, liver spheroids and organoid models, and the induction of angiogenesis in vivo.
Collapse
Affiliation(s)
- Parsa Davoodi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Niloofar Rezaei
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - David C Hay
- Centre for Regenerative Medicine, Institute for Repair and Regeneration, University of Edinburgh, Edinburgh, UK
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
4
|
Moradi-Gharibvand N, Hashemibeni B. The Effect of Stem Cells and Vascular Endothelial Growth Factor on Cancer Angiogenesis. Adv Biomed Res 2023; 12:124. [PMID: 37434939 PMCID: PMC10331557 DOI: 10.4103/abr.abr_378_21] [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: 12/02/2021] [Revised: 04/17/2022] [Accepted: 04/24/2022] [Indexed: 07/13/2023] Open
Abstract
The formation of new vessels from pre-existing vessels is known as angiogenesis. The process is controlled by stimuli and inhibitors. Angiogenesis starts as a result of the unbalance of these factors, where balance has a tendency toward the stimulus. One of the most important factors promoting angiogenesis is the vascular endothelial growth factor (VEGF). In addition to being involved in vascular regeneration in normal tissues, VEGF also takes part in tumor tissue angiogenesis. These factors affect endothelial cells (ECs) directly as well as differentiate tumor cells from endothelial cells and play an active role in tumor tissue angiogenesis. Angiogenesis partakes in the growth and proliferation of tumor tissue. Because anti-angiogenic treatment is favorable in existing cancer therapies, the potential benefits should be considered. One of these new therapies is cell therapy using mesenchymal stem cells (MSCs). Research on MSCs remains controversial because much of the earlier research on MSCs has shown their effectiveness, but more recent research has identified harmful effects of these cells. This article reviews the role of stem cells and their secretions in the angiogenesis of tumor tissues.
Collapse
Affiliation(s)
- Nahid Moradi-Gharibvand
- Abadan University of Medical Sciences, Abadan, Iran
- Department of Anatomical Sciences and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Batool Hashemibeni
- Department of Anatomical Sciences and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
5
|
Ahmed TA, Eldaly B, Eldosuky S, Elkhenany H, El-Derby AM, Elshazly MF, El-Badri N. The interplay of cells, polymers, and vascularization in three-dimensional lung models and their applications in COVID-19 research and therapy. Stem Cell Res Ther 2023; 14:114. [PMID: 37118810 PMCID: PMC10144893 DOI: 10.1186/s13287-023-03341-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 04/14/2023] [Indexed: 04/30/2023] Open
Abstract
Millions of people have been affected ever since the emergence of the corona virus disease of 2019 (COVID-19) outbreak, leading to an urgent need for antiviral drug and vaccine development. Current experimentation on traditional two-dimensional culture (2D) fails to accurately mimic the in vivo microenvironment for the disease, while in vivo animal model testing does not faithfully replicate human COVID-19 infection. Human-based three-dimensional (3D) cell culture models such as spheroids, organoids, and organ-on-a-chip present a promising solution to these challenges. In this report, we review the recent 3D in vitro lung models used in COVID-19 infection and drug screening studies and highlight the most common types of natural and synthetic polymers used to generate 3D lung models.
Collapse
Affiliation(s)
- Toka A Ahmed
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt
- Egypt Center for Research and Regenerative Medicine (ECRRM), Cairo, Egypt
| | - Bassant Eldaly
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt
| | - Shadwa Eldosuky
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt
| | - Hoda Elkhenany
- Department of Surgery, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 22785, Egypt
| | - Azza M El-Derby
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt
| | - Muhamed F Elshazly
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt.
| |
Collapse
|
6
|
Mahapatra C, Kumar P, Paul MK, Kumar A. Angiogenic stimulation strategies in bone tissue regeneration. Tissue Cell 2022; 79:101908. [DOI: 10.1016/j.tice.2022.101908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/24/2022] [Accepted: 08/22/2022] [Indexed: 11/28/2022]
|
7
|
Tracy EP, Stielberg V, Rowe G, Benson D, Nunes SS, Hoying JB, Murfee WL, LeBlanc AJ. State of the field: cellular and exosomal therapeutic approaches in vascular regeneration. Am J Physiol Heart Circ Physiol 2022; 322:H647-H680. [PMID: 35179976 PMCID: PMC8957327 DOI: 10.1152/ajpheart.00674.2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 01/19/2023]
Abstract
Pathologies of the vasculature including the microvasculature are often complex in nature, leading to loss of physiological homeostatic regulation of patency and adequate perfusion to match tissue metabolic demands. Microvascular dysfunction is a key underlying element in the majority of pathologies of failing organs and tissues. Contributing pathological factors to this dysfunction include oxidative stress, mitochondrial dysfunction, endoplasmic reticular (ER) stress, endothelial dysfunction, loss of angiogenic potential and vascular density, and greater senescence and apoptosis. In many clinical settings, current pharmacologic strategies use a single or narrow targeted approach to address symptoms of pathology rather than a comprehensive and multifaceted approach to address their root cause. To address this, efforts have been heavily focused on cellular therapies and cell-free therapies (e.g., exosomes) that can tackle the multifaceted etiology of vascular and microvascular dysfunction. In this review, we discuss 1) the state of the field in terms of common therapeutic cell population isolation techniques, their unique characteristics, and their advantages and disadvantages, 2) common molecular mechanisms of cell therapies to restore vascularization and/or vascular function, 3) arguments for and against allogeneic versus autologous applications of cell therapies, 4) emerging strategies to optimize and enhance cell therapies through priming and preconditioning, and, finally, 5) emerging strategies to bolster therapeutic effect. Relevant and recent clinical and animal studies using cellular therapies to restore vascular function or pathologic tissue health by way of improved vascularization are highlighted throughout these sections.
Collapse
Affiliation(s)
- Evan Paul Tracy
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Virginia Stielberg
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Gabrielle Rowe
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Daniel Benson
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
- Department of Bioengineering, University of Louisville, Louisville, Kentucky
| | - Sara S Nunes
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Heart & Stroke/Richard Lewar Centre of Excellence, University of Toronto, Toronto, Ontario, Canada
| | - James B Hoying
- Advanced Solutions Life Sciences, Manchester, New Hampshire
| | - Walter Lee Murfee
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida
| | - Amanda Jo LeBlanc
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| |
Collapse
|
8
|
Huang Y, Qian JY, Cheng H, Li XM. Effects of shear stress on differentiation of stem cells into endothelial cells. World J Stem Cells 2021; 13:894-913. [PMID: 34367483 PMCID: PMC8316872 DOI: 10.4252/wjsc.v13.i7.894] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/20/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023] Open
Abstract
Stem cell transplantation is an appealing potential therapy for vascular diseases and an indispensable key step in vascular tissue engineering. Substantial effort has been made to differentiate stem cells toward vascular cell phenotypes, including endothelial cells (ECs) and smooth muscle cells. The microenvironment of vascular cells not only contains biochemical factors that influence differentiation but also exerts hemodynamic forces, such as shear stress and cyclic strain. More recently, studies have shown that shear stress can influence the differentiation of stem cells toward ECs. A deep understanding of the responses and underlying mechanisms involved in this process is essential for clinical translation. This review highlights current data supporting the role of shear stress in stem cell differentiation into ECs. Potential mechanisms and signaling cascades for transducing shear stress into a biological signal are proposed. Further study of stem cell responses to shear stress will be necessary to apply stem cells for pharmacological applications and cardiovascular implants in the realm of regenerative medicine.
Collapse
Affiliation(s)
- Yan Huang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Jia-Yi Qian
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Hong Cheng
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Xiao-Ming Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
| |
Collapse
|
9
|
Xie J, Li X, Zhang Y, Tang T, Chen G, Mao H, Gu Z, Yang J. VE-cadherin-based matrix promoting the self-reconstruction of pro-vascularization microenvironments and endothelial differentiation of human mesenchymal stem cells. J Mater Chem B 2021; 9:3357-3370. [PMID: 33881442 DOI: 10.1039/d1tb00017a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Regulating the secretion and endothelial differentiation of human mesenchymal stem cells (hMSCs) plays an important role in the vascularization in tissue engineering and regenerative medicine. In this study, a recombinant cadherin fusion protein consisting of a human vascular endothelial-cadherin extracellular domain and immunoglobulin IgG Fc region (hVE-cad-Fc) was developed as a bioartificial matrix for modulating hMSCs. The hVE-cad-Fc matrix significantly enhanced the secretion of angiogenic factors, activated the VE-cadherin-VEGFR2/FAK-AKT/PI3K signaling pathway in hMSCs, and promoted the endothelial differentiation of hMSCs even without extra VEGF. Furthermore, the hVE-cad-Fc matrix was applied for the surface modification of a poly (lactic-co-glycolic acid) (PLGA) porous scaffold, which significantly improved the hemocompatibility and vascularization of the PLGA scaffold in vivo. These results revealed that the hVE-cad-Fc matrix should be a superior bioartificial ECM for remodeling the pro-vascularization extracellular microenvironment by regulating the secretion of hMSCs, and showed great potential for the vascularization in tissue engineering.
Collapse
Affiliation(s)
- Jinghui Xie
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, 300071, China.
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Browne S, Gill EL, Schultheiss P, Goswami I, Healy KE. Stem cell-based vascularization of microphysiological systems. Stem Cell Reports 2021; 16:2058-2075. [PMID: 33836144 PMCID: PMC8452487 DOI: 10.1016/j.stemcr.2021.03.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/11/2021] [Accepted: 03/15/2021] [Indexed: 12/27/2022] Open
Abstract
Microphysiological systems (MPSs) (i.e., tissue or organ chips) exploit microfluidics and 3D cell culture to mimic tissue and organ-level physiology. The advent of human induced pluripotent stem cell (hiPSC) technology has accelerated the use of MPSs to study human disease in a range of organ systems. However, in the reduction of system complexity, the intricacies of vasculature are an often-overlooked aspect of MPS design. The growing library of pluripotent stem cell-derived endothelial cell and perivascular cell protocols have great potential to improve the physiological relevance of vasculature within MPS, specifically for in vitro disease modeling. Three strategic categories of vascular MPS are outlined: self-assembled, interface focused, and 3D biofabricated. This review discusses key features and development of the native vasculature, linking that to how hiPSC-derived vascular cells have been generated, the state of the art in vascular MPSs, and opportunities arising from interdisciplinary thinking.
Collapse
Affiliation(s)
- Shane Browne
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA 94720, USA
| | - Elisabeth L Gill
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA 94720, USA
| | - Paula Schultheiss
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA 94720, USA
| | - Ishan Goswami
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA 94720, USA; Department of Materials Science and Engineering, University of California, Berkeley, CA 94720, USA
| | - Kevin E Healy
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA 94720, USA; Department of Materials Science and Engineering, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
11
|
Kurdi BA, Ababneh NA, Abuharfeil N, Al Demour S, Awidi AS. Use of conditioned media (CM) and xeno-free serum substitute on human adipose-derived stem cells (ADSCs) differentiation into urothelial-like cells. PeerJ 2021; 9:e10890. [PMID: 33850639 PMCID: PMC8019311 DOI: 10.7717/peerj.10890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 01/12/2021] [Indexed: 01/01/2023] Open
Abstract
Background Congenital abnormalities, cancers as well as injuries can cause irreversible damage to the urinary tract, which eventually requires tissue reconstruction. Smooth muscle cells, endothelial cells, and urothelial cells are the major cell types required for the reconstruction of lower urinary tract. Adult stem cells represent an accessible source of unlimited repertoire of untransformed cells. Aim Fetal bovine serum (FBS) is the most vital supplement in the culture media used for cellular proliferation and differentiation. However, due to the increasing interest in manufacturing xeno-free stem cell-based cellular products, optimizing the composition of the culture media and the serum-type used is of paramount importance. In this study, the effects of FBS and pooled human platelet (pHPL) lysate were assessed on the capacity of human adipose-derived stem cells (ADSCs) to differentiate into urothelial-like cells. Also, we aimed to compare the ability of both conditioned media (CM) and unconditioned urothelial cell media (UCM) to induce urothelial differentiation of ADCS in vitro. Methods ADSCs were isolated from human lipoaspirates and characterized by flow cytometry for their ability to express the most common mesenchymal stem cell (MSCs) markers. The differentiation potential was also assessed by differentiating them into osteogenic and adipogenic cell lineages. To evaluate the capacity of ADSCs to differentiate towards the urothelial-like lineage, cells were cultured with either CM or UCM, supplemented with either 5% pHPL, 2.5% pHPL or 10% FBS. After 14 days of induction, cells were utilized for gene expression and immunofluorescence analysis. Results ADSCs cultured in CM and supplemented with FBS exhibited the highest upregulation levels of the urothelial cell markers; cytokeratin-18 (CK-18), cytokeratin-19 (CK-19), and Uroplakin-2 (UPK-2), with a 6.7, 4.2- and a 2-folds increase in gene expression, respectively. Meanwhile, the use of CM supplemented with either 5% pHPL or 2.5% pHPL, and UCM supplemented with either 5% pHPL or 2.5% pHPL showed low expression levels of CK-18 and CK-19 and no upregulation of UPK-2 level was observed. In contrast, the use of UCM with FBS has increased the levels of CK-18 and CK-19, however to a lesser extent compared to CM. At the cellular level, CK-18 and UPK-2 were only detected in CM/FBS supplemented group. Growth factor analysis revealed an increase in the expression levels of EGF, VEGF and PDGF in all of the differentiated groups. Conclusion Efficient ADSCs urothelial differentiation is dependent on the use of conditioned media. The presence of high concentrations of proliferation-inducing growth factors present in the pHPL reduces the efficiency of ADSCs differentiation towards the urothelial lineage. Additionally, the increase in EGF, VEGF and PDGF during the differentiation implicates them in the mechanism of urothelial cell differentiation.
Collapse
Affiliation(s)
- Ban Al- Kurdi
- Cell Therapy Center, University of Jordan, Amman, Jordan.,Department of Hematology and Oncology, Jordan University Hospital, Amman, Jordan
| | | | - Nizar Abuharfeil
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, Jordan
| | - Saddam Al Demour
- Department of Urology, School of medicine, University of Jordan, Amman, Jordan, University of Jordan, Amman, Jordan
| | - Abdalla S Awidi
- Cell Therapy Center, University of Jordan, Amman, Jordan.,Department of Hematology and Oncology, Jordan University Hospital, Amman, Jordan
| |
Collapse
|
12
|
Rafique M, Wei T, Sun Q, Midgley AC, Huang Z, Wang T, Shafiq M, Zhi D, Si J, Yan H, Kong D, Wang K. The effect of hypoxia-mimicking responses on improving the regeneration of artificial vascular grafts. Biomaterials 2021; 271:120746. [PMID: 33725586 DOI: 10.1016/j.biomaterials.2021.120746] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 02/16/2021] [Accepted: 02/28/2021] [Indexed: 12/12/2022]
Abstract
Cellular transition to hypoxia following tissue injury, has been shown to improve angiogenesis and regeneration in multiple tissues. To take advantage of this, many hypoxia-mimicking scaffolds have been prepared, yet the oxygen access state of implanted artificial small-diameter vascular grafts (SDVGs) has not been investigated. Therefore, the oxygen access state of electrospun PCL grafts implanted into rat abdominal arteries was assessed. The regions proximal to the lumen and abluminal surfaces of the graft walls were normoxic and only the interior of the graft walls was hypoxic. In light of this differential oxygen access state of the implanted grafts and the critical role of vascular regeneration on SDVG implantation success, we investigated whether modification of SDVGs with HIF-1α stabilizer dimethyloxalylglycine (DMOG) could achieve hypoxia-mimicking responses resulting in improving vascular regeneration throughout the entirety of the graft wall. Therefore, DMOG-loaded PCL grafts were fabricated by electrospinning, to support the sustained release of DMOG over two weeks. In vitro experiments indicated that DMOG-loaded PCL mats had significant biological advantages, including: promotion of human umbilical vein endothelial cells (HUVECs) proliferation, migration and production of pro-angiogenic factors; and the stimulation of M2 macrophage polarization, which in-turn promoted macrophage regulation of HUVECs migration and smooth muscle cells (SMCs) contractile phenotype. These beneficial effects were downstream of HIF-1α stabilization in HUVECs and macrophages in normoxic conditions. Our results indicated that DMOG-loaded PCL grafts improved endothelialization, contractile SMCs regeneration, vascularization and modulated the inflammatory reaction of grafts in abdominal artery replacement models, thus promoting vascular regeneration.
Collapse
Affiliation(s)
- Muhammad Rafique
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Tingting Wei
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Qiqi Sun
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Adam C Midgley
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Ziqi Huang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Ting Wang
- Tianjin Key Laboratory of Urban Transport Emission Research, College of Environmental Science and Engineering, Nankai University, Tianjin, 300071, China
| | - Muhammad Shafiq
- Department of Biotechnology, Faculty of Life Sciences, University of Central Punjab, Lahore, 54000, Pakistan
| | - Dengke Zhi
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jianghua Si
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Hongyu Yan
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Deling Kong
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Kai Wang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
13
|
Nwabo Kamdje AH, Seke Etet PF, Simo Tagne R, Vecchio L, Lukong KE, Krampera M. Tumor Microenvironment Uses a Reversible Reprogramming of Mesenchymal Stromal Cells to Mediate Pro-tumorigenic Effects. Front Cell Dev Biol 2020; 8:545126. [PMID: 33330442 PMCID: PMC7710932 DOI: 10.3389/fcell.2020.545126] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 10/28/2020] [Indexed: 12/13/2022] Open
Abstract
The role of mesenchymal stromal cells (MSCs) in the tumor microenvironment is well described. Available data support that MSCs display anticancer activities, and that their reprogramming by cancer cells in the tumor microenvironment induces their switch toward pro-tumorigenic activities. Here we discuss the recent evidence of pro-tumorigenic effects of stromal cells, in particular (i) MSC support to cancer cells through the metabolic reprogramming necessary to maintain their malignant behavior and stemness, and (ii) MSC role in cancer cell immunosenescence and in the establishment and maintenance of immunosuppression in the tumor microenvironment. We also discuss the mechanisms of tumor microenvironment mediated reprogramming of MSCs, including the effects of hypoxia, tumor stiffness, cancer-promoting cells, and tumor extracellular matrix. Finally, we summarize the emerging strategies for reprogramming tumor MSCs to reactivate anticancer functions of these stromal cells.
Collapse
Affiliation(s)
- Armel H. Nwabo Kamdje
- Department of Physiological Sciences and Biochemistry, Faculty of Medicine and Biomedical Sciences (FMBS), University of Ngaoundéré, Ngaoundéré, Cameroon
| | - Paul F. Seke Etet
- Department of Physiological Sciences and Biochemistry, Faculty of Medicine and Biomedical Sciences (FMBS), University of Ngaoundéré, Ngaoundéré, Cameroon
- Center for Sustainable Health and Development, Garoua, Cameroon
| | - Richard Simo Tagne
- Department of Physiological Sciences and Biochemistry, Faculty of Medicine and Biomedical Sciences (FMBS), University of Ngaoundéré, Ngaoundéré, Cameroon
| | - Lorella Vecchio
- Center for Sustainable Health and Development, Garoua, Cameroon
| | - Kiven Erique Lukong
- Department of Biochemistry, Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Mauro Krampera
- Section of Hematology, Stem Cell Research Laboratory, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
14
|
Nwabo Kamdje AH, Seke Etet PF, Simo RT, Vecchio L, Lukong KE, Krampera M. Emerging data supporting stromal cell therapeutic potential in cancer: reprogramming stromal cells of the tumor microenvironment for anti-cancer effects. Cancer Biol Med 2020; 17:828-841. [PMID: 33299638 PMCID: PMC7721102 DOI: 10.20892/j.issn.2095-3941.2020.0133] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 08/20/2020] [Indexed: 02/03/2023] Open
Abstract
After more than a decade of controversy on the role of stromal cells in the tumor microenvironment, the emerging data shed light on pro-tumorigenic and potential anti-cancer factors, as well as on the roots of the discrepancies. We discuss the pro-tumorigenic effects of stromal cells, considering the effects of tumor drivers like hypoxia and tumor stiffness on these cells, as well as stromal cell-mediated adiposity and immunosuppression in the tumor microenvironment, and cancer initiating cells' cellular senescence and adaptive metabolism. We summarize the emerging data supporting stromal cell therapeutic potential in cancer, discuss the possibility to reprogram stromal cells of the tumor microenvironment for anti-cancer effects, and explore some causes of discrepancies on the roles of stromal cells in cancer in the available literature.
Collapse
Affiliation(s)
- Armel Hervé Nwabo Kamdje
- Department of Biomedical Sciences, University of Ngaoundere, Faculty of Science, Ngaoundere 454, Cameroon
| | - Paul Faustin Seke Etet
- Department of Physiological Sciences and Biochemistry, University of Ngaoundéré, Garoua 454, Cameroon
- Center for Sustainable Health and Development, Garoua 454, Cameroon
| | - Richard Tagne Simo
- Department of Biomedical Sciences, University of Ngaoundere, Faculty of Science, Ngaoundere 454, Cameroon
| | - Lorella Vecchio
- Center for Sustainable Health and Development, Garoua 454, Cameroon
| | - Kiven Erique Lukong
- Department of Biochemistry, Microbiology & Immunology, University of Saskatchewan, College of Medicine, Saskatoon SK S7N 5E5, Canada
| | - Mauro Krampera
- Department of Medicine, University of Verona, Section of Hematology, Stem Cell Research Laboratory, Verona 37134, Italy
| |
Collapse
|
15
|
Sun X, Luo L, Li J. LncRNA MALAT1 facilitates BM-MSCs differentiation into endothelial cells via targeting miR-206/VEGFA axis. Cell Cycle 2020; 19:3018-3028. [PMID: 33121336 DOI: 10.1080/15384101.2020.1829799] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Bone marrow-derived mesenchymal stem cells (BM-MSCs) implantation shows a repair effect on erectile function in diabetes mellitus-induced erectile dysfunction (DMED) due to its differentiative capacity into endothelial cells (ECs) that contributes to endothelial repair. This study was designed to explore the functional role and mechanism of long noncoding RNA (lncRNA)-metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) in BM-MSCs-mediated DMED repairing. The DMED rat model was established and the erectile function was evaluated by calculating the intracavernous pressure (ICP)/mean arterial pressure (MAP) ratio in the DMED models with or without BM-MSCs implantation. The differentiation of BM-MSCs toward ECs was assessed by measuring the expression of EC-specific genes. RNA pull-down and luciferase reporter assay were performed to explore the interaction between miR-206 and MALAT1 or VEGFA. BM-MSCs implantation improved the erectile function of DMED rats and increased MALAT1 expression. MALAT1 was time-dependently upregulated during the VEGF-induced BM-MSCs differentiation into ECs. Mechanistically, MALAT1 acted as a sponge of miR-206 to upregulate VEGFA expression, thereby promoting the differentiation of BM-MSCs into ECs. Moreover, MALAT1 silencing in vivo impaired the repairing effect of BM-MSCs on erectile dysfunction. Collectively, MALAT1 facilitates BM-MSCs differentiation into ECs via regulating miR-206/VEGFA axis.
Collapse
Affiliation(s)
- Xiang Sun
- Department of Urology, The First Affiliated Hospital of Nanchang University , Nanchang, Jiangxi People's Republic of China
| | - Longhua Luo
- Department of Urology, The First Affiliated Hospital of Nanchang University , Nanchang, Jiangxi People's Republic of China
| | - Jiarui Li
- Department of Urology, The First Affiliated Hospital of Nanchang University , Nanchang, Jiangxi People's Republic of China
| |
Collapse
|
16
|
Forghani A, Koduru SV, Chen C, Leberfinger AN, Ravnic DJ, Hayes DJ. Differentiation of Adipose Tissue-Derived CD34+/CD31- Cells into Endothelial Cells In Vitro. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2020; 6:101-110. [PMID: 33344757 PMCID: PMC7747864 DOI: 10.1007/s40883-019-00093-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 02/07/2019] [Indexed: 12/18/2022]
Abstract
In this study, CD34+/CD31- progenitor cells were isolated from the stromal vascular fraction (SVF) of adipose tissue using magnetic activated cell sorting. The endothelial differentiation capability of these cells in vitro was evaluated by culturing them in vascular endothelial growth factor (VEGF) induced medium for 14 days. Viability, proliferation, differentiation and tube formation of these cells were evaluated. Cell viability study revealed that both undifferentiated and endothelial differentiated cells remained healthy for 14 days. However, the proliferation rate was higher in undifferentiated cells compared to endothelial differentiated ones. Upregulation of endothelial characteristic genes (Von Willebrand Factor (vWF) and VE Cadherin) was observed in 2D culture. However, PECAM (CD31) was only found to be upregulated after the cells had formed tube-like structures in 3D Matrigel culture. These results indicate that adipose derived CD34+/CD31- cells when cultured in VEGF induced medium, are capable differentiation into endothelial-like lineages. Tube formation of the cells started 3h after seeding the cells on Matrigel and formed more stable and connected network 24 h post seeding in presence of VEGF.
Collapse
Affiliation(s)
- Anoosha Forghani
- Department of Biomedical Engineering, Millennium Science Complex, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Srinivas V Koduru
- Department of Surgery, College of Medicine, Penn State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Cong Chen
- Department of Biomedical Engineering, Millennium Science Complex, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Ashley N Leberfinger
- Department of Surgery, College of Medicine, Penn State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Dino J Ravnic
- Department of Surgery, College of Medicine, Penn State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Daniel J Hayes
- Department of Biomedical Engineering, Millennium Science Complex, Pennsylvania State University, University Park, Pennsylvania, USA
- Materials Research Institute, Materials Characterization Lab, Millennium Science Complex, Pennsylvania State University, University Park, Pennsylvania, USA
- The Huck Institute of the Life Sciences, Millennium Science Complex, Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
17
|
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
|
18
|
Xu W, Cheng W, Cui X, Xu G. Therapeutic effect against retinal neovascularization in a mouse model of oxygen-induced retinopathy: bone marrow-derived mesenchymal stem cells versus Conbercept. BMC Ophthalmol 2020; 20:7. [PMID: 31906900 PMCID: PMC6945477 DOI: 10.1186/s12886-019-1292-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 12/27/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND To study the therapeutic effect of bone marrow-derived mesenchymal stem cells (BMSC) against retinal neovascularization and to compare with anti-vascular endothelial growth factor (VEGF) therapy. METHODS Neonatal C57BL/6 mice were exposed in hyperoxygen and returned to room air to develop oxygen-induced retinopathy (OIR). Red fluorescent protein-labeled BMSC and Conbercept were intravitreally injected into OIR mice, respectively. Inhibition of neovascularization and apoptosis in OIR mice were assessed through retinal angiography, histopathology and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay. RESULTS BMSC were able to migrate and integrate into the host retina, significantly inhibit retinal neovascular tufts and remodel the capillary network after injecton. Treatment with BMSC increased the retinal vascular density, decreased the number of acellular capillaries and inhibited retinal cell death. This effect was not inferior to current anti-VEGF therapy by using Conbercept. CONCLUSIONS Intravitreal injection of BMSC exerts a protective effect against retinal neovascularization and offers a therapeutic strategy for oxygen-induced retinopathy.
Collapse
Affiliation(s)
- Wei Xu
- Department of Ophthalmology, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou City, 350005 China
| | - Weijing Cheng
- Department of Ophthalmology, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou City, 350005 China
- Fujian Institute of Ophthalmology, Fuzhou, China
| | - Xiaoyuan Cui
- Department of Ophthalmology, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou City, 350005 China
| | - Guoxing Xu
- Department of Ophthalmology, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou City, 350005 China
- Fujian Institute of Ophthalmology, Fuzhou, China
| |
Collapse
|
19
|
Shear Stress Promotes Arterial Endothelium-Oriented Differentiation of Mouse-Induced Pluripotent Stem Cells. Stem Cells Int 2019; 2019:1847098. [PMID: 31827524 PMCID: PMC6881757 DOI: 10.1155/2019/1847098] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 09/05/2019] [Accepted: 10/17/2019] [Indexed: 12/29/2022] Open
Abstract
Establishment of a functional vascular network, which is required in tissue repair and regeneration, needs large-scale production of specific arterial or venous endothelial cells (ECs) from stem cells. Previous in vitro studies by us and others revealed that shear stress induces EC differentiation of bone marrow-derived mesenchymal stem cells and embryonic stem cells. In this study, we focused on the impact of different magnitudes of shear stress on the differentiation of mouse-induced pluripotent stem cells (iPSCs) towards arterial or venous ECs. When iPSCs were exposed to shear stress (5, 10, and 15 dyne/cm2) with 50 ng/mL vascular endothelial growth factor and 10 ng/mL fibroblast growth factor, the expression levels of the general EC markers and the arterial markers increased, and the stress amplitude of 10 dyne/cm2 could be regarded as a proper promoter, whereas the venous and lymphatic markers had little or no expression. Further, shear stress caused cells to align parallel to the direction of the flow, induced cells forming functional tubes, and increased the secretion of nitric oxide. In addition, Notch1 was significantly upregulated, and the Notch ligand Delta-like 4 was activated in response to shear stress, while inhibition of Notch signaling by DAPT remarkably abolished the shear stress-induced arterial epithelium differentiation. Taken together, our results indicate that exposure to appropriate shear stress facilitated the differentiation of mouse iPSCs towards arterial ECs via Notch signaling pathways, which have potential applications for both disease modeling and regenerative medicine.
Collapse
|
20
|
Abdelrazik H, Giordano E, Barbanti Brodano G, Griffoni C, De Falco E, Pelagalli A. Substantial Overview on Mesenchymal Stem Cell Biological and Physical Properties as an Opportunity in Translational Medicine. Int J Mol Sci 2019; 20:5386. [PMID: 31671788 PMCID: PMC6862078 DOI: 10.3390/ijms20215386] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 10/25/2019] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSC) have piqued worldwide interest for their extensive potential to treat a large array of clinical indications, their unique and controversial immunogenic and immune modulatory properties allowing ample discussions and debates for their possible applications. Emerging data demonstrating that the interaction of biomaterials and physical cues with MSC can guide their differentiation into specific cell lineages also provide new interesting insights for further MSC manipulation in different clinical applications. Moreover, recent discoveries of some regulatory molecules and signaling pathways in MSC niche that may regulate cell fate to distinct lineage herald breakthroughs in regenerative medicine. Although the advancement and success in the MSC field had led to an enormous increase in the amount of ongoing clinical trials, we still lack defined clinical therapeutic protocols. This review will explore the exciting opportunities offered by human and animal MSC, describing relevant biological properties of these cells in the light of the novel emerging evidence mentioned above while addressing the limitations and challenges MSC are still facing.
Collapse
Affiliation(s)
- Heba Abdelrazik
- Department of Clinical Pathology, Cairo University, Cairo 1137, Egypt.
- Department of Diagnosis, central laboratory department, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Policlinico San Martino, 16131 Genoa, Italy.
| | - Emanuele Giordano
- Department of Electrical, Electronic and Information Engineering "Guglielmo Marconi" (DEI), University of Bologna, 47522 Cesena, Italy.
| | - Giovanni Barbanti Brodano
- Department of Oncological and Degenerative Spine Surgery, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| | - Cristiana Griffoni
- Department of Oncological and Degenerative Spine Surgery, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| | - Elena De Falco
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy.
- Mediterranea Cardiocentro, 80122 Napoli, Italy.
| | - Alessandra Pelagalli
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", 80131 Naples, Italy.
- Institute of Biostructures and Bioimages (IBB), National Research Council (CNR), 80131 Naples, Italy.
| |
Collapse
|
21
|
Liu J, Wada Y, Katsura M, Tozawa H, Erwin N, Kapron CM, Bao G, Liu J. Rho-Associated Coiled-Coil Kinase (ROCK) in Molecular Regulation of Angiogenesis. Am J Cancer Res 2018; 8:6053-6069. [PMID: 30613282 PMCID: PMC6299434 DOI: 10.7150/thno.30305] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 10/16/2018] [Indexed: 02/06/2023] Open
Abstract
Identified as a major downstream effector of the small GTPase RhoA, Rho-associated coiled-coil kinase (ROCK) is a versatile regulator of multiple cellular processes. Angiogenesis, the process of generating new capillaries from the pre-existing ones, is required for the development of various diseases such as cancer, diabetes and rheumatoid arthritis. Recently, ROCK has attracted attention for its crucial role in angiogenesis, making it a promising target for new therapeutic approaches. In this review, we summarize recent advances in understanding the role of ROCK signaling in regulating the permeability, migration, proliferation and tubulogenesis of endothelial cells (ECs), as well as its functions in non-ECs which constitute the pro-angiogenic microenvironment. The therapeutic potential of ROCK inhibitors in angiogenesis-related diseases is also discussed.
Collapse
|
22
|
Yong KW, Choi JR, Mohammadi M, Mitha AP, Sanati-Nezhad A, Sen A. Mesenchymal Stem Cell Therapy for Ischemic Tissues. Stem Cells Int 2018; 2018:8179075. [PMID: 30402112 PMCID: PMC6196793 DOI: 10.1155/2018/8179075] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 08/01/2018] [Accepted: 08/29/2018] [Indexed: 12/13/2022] Open
Abstract
Ischemic diseases such as myocardial infarction, ischemic stroke, and critical limb ischemia are immense public health challenges. Current pharmacotherapy and surgical approaches are insufficient to completely heal ischemic diseases and are associated with a considerable risk of adverse effects. Alternatively, human mesenchymal stem cells (hMSCs) have been shown to exhibit immunomodulation, angiogenesis, and paracrine secretion of bioactive factors that can attenuate inflammation and promote tissue regeneration, making them a promising cell source for ischemic disease therapy. This review summarizes the pathogenesis of ischemic diseases, discusses the potential therapeutic effects and mechanisms of hMSCs for these diseases, and provides an overview of challenges of using hMSCs clinically for treating ischemic diseases.
Collapse
Affiliation(s)
- Kar Wey Yong
- Pharmaceutical Production Research Facility, Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Jane Ru Choi
- Department of Mechanical Engineering, University of British Columbia, 2054-6250 Applied Science Lane, Vancouver, BC, V6T 1Z4, Canada
- Centre for Blood Research, Life Sciences Centre, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Mehdi Mohammadi
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
- Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Alim P. Mitha
- Department of Clinical Neurosciences, Foothills Medical Centre, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| | - Amir Sanati-Nezhad
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
- Center of Bioengineering Research and Education, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Arindom Sen
- Pharmaceutical Production Research Facility, Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
- Center of Bioengineering Research and Education, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| |
Collapse
|
23
|
Sun X, Luo LH, Feng L, Li DS. Down-regulation of lncRNA MEG3 promotes endothelial differentiation of bone marrow derived mesenchymal stem cells in repairing erectile dysfunction. Life Sci 2018; 208:246-252. [PMID: 30012476 DOI: 10.1016/j.lfs.2018.07.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 07/05/2018] [Accepted: 07/12/2018] [Indexed: 01/05/2023]
Abstract
AIMS In the treatment of diabetes mellitus associated erectile dysfunction (DMED), the intracavernous and periprostatic implantations of bone marrow derived mesenchymal stem cells (BM-MSCs) represent the new therapeutic approaches with great applied prospect. However, the specific mechanisms of BM-MSCs protecting erectile function remain largely unknown. MATERIALS AND METHODS The DMED rats were induced and the erectile function was assessed in the models with or without BM-MSCs implantation using intracavernous pressure (ICP)/mean arterial pressure (MAP) ratio. The differentiation of BM-MSCs toward endothelial cells (ECs) was induced by exogenous vascular endothelial growth factor (VEGF) in vitro. RNA pull-down and RIP assays were performed to explore the interaction between MEG3 and FOXM1 protein. KEY FINDINGS Intracavernous implantation of BM-MSCs effectively improved the erectile function of DMED rats, which was accompanied by a significant decrease in the expression of MEG3 in the corpus cavernosum tissues. Also, our study revealed that MEG3 expression was significantly down-regulated during the endothelial differentiation of BM-MSCs in vitro. The down-regulation of MEG3 was further confirmed to be conducive to the differentiation of BM-MSCs toward ECs. More importantly, MEG3 promoted the degradation of FOXM1 protein via facilitating FOXM1 ubiquitination, thereby decreasing VEGF expression, which ultimately regulated the endothelial differentiation of BM-MSCs. SIGNIFICANCE Taken together, our findings presented the vital role of MEG3 in the repairing processes of BM-MSCs for erectile function and provided new mechanistic insights into the BM-MSCs-mediated DMED repairing.
Collapse
Affiliation(s)
- Xiang Sun
- Department of Urology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China.
| | - Long-Hua Luo
- Department of Urology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Liang Feng
- Department of Urology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Dong-Shui Li
- Department of Urology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| |
Collapse
|
24
|
Lin J, Cao S, Wang Y, Hu Y, Liu H, Li J, Chen J, Li P, Liu J, Wang Q, Zheng L. Long non-coding RNA UBE2CP3 enhances HCC cell secretion of VEGFA and promotes angiogenesis by activating ERK1/2/HIF-1α/VEGFA signalling in hepatocellular carcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:113. [PMID: 29866133 PMCID: PMC5987644 DOI: 10.1186/s13046-018-0727-1] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 03/06/2018] [Indexed: 12/19/2022]
Abstract
Background Angiogenesis is considered as an important process in the development of malignancies and is associated with cancer progression and metastasis. Hepatocellular carcinoma (HCC) is the most common primary tumor of the liver and is recognized as a typical angiogenic tumor. Thus, it is of great importance to study the underlying mechanism of angiogenesis in HCC. The long non-coding RNA (lncRNA) ubiquitin conjugating enzyme E2C pseudogene 3 (UBE2CP3) has been reported as an oncogene that promotes tumor metastasis in HCC. However, the role and underlying mechanisms of UBE2CP3 in HCC angiogenesis are still unclear. Methods We measured the expression levels of UBE2CP3 by in situ hybridization (ISH) and quantitative real-time polymerase chain reaction (qRT-PCR) in HCC patient samples. We also concomitantly used CD31/PAS double-staining to measure endothelial vessel (EV) density and used qRT-PCR to measure the CD31 mRNA level. HepG2 and SMMC-7721 cells were transfected with Lv-UBE2CP3 or Sh-UBE2CP3 virus to obtain stably over-expressing or knocking-down UBE2CP3 cell lines. The indirect effects of UBE2CP3 on ECs were studied by establishing a co-culture system using Transwell chambers with a 0.4-μm pore size. HCC cells and ECs in the co-culture system were separated, but the cytokines and growth factors were able to communicate with each other. Following exposed to HCC cells, ECs were collected for functional studies. Finally, we studied the function of UBE2CP3 in vivo by chick embryo chorioallantoic membrane (CAM) angiogenesis assays and nude mouse tumorigenicity assays. Results In this study, we found that UBE2CP3 expression was higher in HCC tissues than in para-tumor tissues and was up-regulated in tissues with high EV density. Functionally, we found that in the co-culture systems, HCC cells overexpressing UBE2CP3 promoted HUVEC proliferation, migration and tube formation via the activation of ERK/HIF-1α/p70S6K/VEGFA signalling, increasing the level of VEGFA in HCC cell supernatant. In addition, the opposite results appeared when the expression of UBE2CP3 in HCC cells was knocked down. Consistent with these results, CAM angiogenesis assays and nude mouse tumorigenicity assays showed that UBE2CP3 expression up-regulated EV density in vivo. Conclusion Our study suggests that UBE2CP3 can enhance the interaction between HCC tumor cells and HUVECs and promote HCC tumorigenicity by facilitating angiogenesis. Electronic supplementary material The online version of this article (10.1186/s13046-018-0727-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jinduan Lin
- Department of Laboratory Medicine Center, Nanfang Hospital, Southern Medical University/The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China.,Department of Clinical Laboratory, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| | - Shunwang Cao
- Department of Laboratory Medicine Center, Nanfang Hospital, Southern Medical University/The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yu Wang
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University/The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yanwei Hu
- Department of Laboratory Medicine Center, Nanfang Hospital, Southern Medical University/The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Hongwei Liu
- Department of Clinical Laboratory, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| | - Jiehua Li
- Department of Clinical Laboratory, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| | - Jing Chen
- Department of Laboratory Medicine Center, Nanfang Hospital, Southern Medical University/The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Pan Li
- Department of Laboratory Medicine Center, Nanfang Hospital, Southern Medical University/The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jumei Liu
- Department of Laboratory Medicine Center, Nanfang Hospital, Southern Medical University/The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Qian Wang
- Department of Laboratory Medicine Center, Nanfang Hospital, Southern Medical University/The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Lei Zheng
- Department of Laboratory Medicine Center, Nanfang Hospital, Southern Medical University/The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
25
|
La A, Tranquillo RT. Hemocompatible tissue-engineered vascular grafts using adult mesenchymal stem cells. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2018; 5:66-73. [DOI: 10.1016/j.cobme.2018.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
26
|
Gu W, Hong X, Potter C, Qu A, Xu Q. Mesenchymal stem cells and vascular regeneration. Microcirculation 2018; 24. [PMID: 27681821 DOI: 10.1111/micc.12324] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 09/20/2016] [Indexed: 12/22/2022]
Abstract
In recent years, MSCs have emerged as a promising therapeutic cell type in regenerative medicine. They hold great promise for treating cardiovascular diseases, such as myocardial infarction and limb ischemia. MSCs may be utilized in both cell-based therapy and vascular graft engineering to restore vascular function, thereby providing therapeutic benefits to patients. The efficacy of MSCs lies in their multipotent differentiation ability toward vascular smooth muscle cells, endothelial cells and other cell types, as well as their capacity to secrete various trophic factors, which are potent in promoting angiogenesis, inhibiting apoptosis and modulating immunoreaction. Increasing our understanding of the mechanisms of MSC involvement in vascular regeneration will be beneficial in boosting present therapeutic approaches and developing novel ones to treat cardiovascular diseases. In this review, we aim to summarize current progress in characterizing the in vivo identity of MSCs, to discuss mechanisms involved in cell-based therapy utilizing MSCs, and to explore current and future strategies for vascular regeneration.
Collapse
Affiliation(s)
- Wenduo Gu
- Cardiovascular Division, King's College London BHF Centre, London, UK
| | - Xuechong Hong
- Cardiovascular Division, King's College London BHF Centre, London, UK
| | - Claire Potter
- Cardiovascular Division, King's College London BHF Centre, London, UK
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China
| | - Qingbo Xu
- Cardiovascular Division, King's College London BHF Centre, London, UK
| |
Collapse
|
27
|
Luo Y, Xu Y, Wang ZY, Li X, Xing WB, Zhang TC. The Synergy of Two Factors on Insulin Expression. Cell Reprogram 2018; 20:49-54. [PMID: 29303357 DOI: 10.1089/cell.2017.0026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
As a potential cure for diabetes, more and more attentions have been paid to organ transplants to replace insulin therapy. As a result, many researchers have explored out many programs to get insulin-producing cells (IPCs) to replace the defective β cells. Currently, more and more new induction methods are being proposed, and at the same time, more and more possible induction molecular mechanisms are being revealed. The purpose of this study was to explore whether and how the two factors pdx-1 and myocardin affected the differentiation of rat mesenchymal stem cells (rMSCs) into IPCs. In this study, we investigated the process of transfecting myocardin and/or pdx-1 in rMSCs in vitro. The results showed that rMSCs were able to secrete insulin after cotransfected with myocardin and pdx-1. At the same time, we explored the possible mechanism that myocardin and pdx-1 coinduced rMSCs into IPCs by forming a complex to promote the transcriptional activity of insulin. Our results may provide a theoretical basis to the study of islet transplantation in the future.
Collapse
Affiliation(s)
- Ying Luo
- 1 Institute of Biology and Medicine, Wuhan University of Science and Technology , Wuhan, China
| | - Yao Xu
- 1 Institute of Biology and Medicine, Wuhan University of Science and Technology , Wuhan, China
| | - Zhen-Yu Wang
- 1 Institute of Biology and Medicine, Wuhan University of Science and Technology , Wuhan, China
| | - Xi Li
- 1 Institute of Biology and Medicine, Wuhan University of Science and Technology , Wuhan, China
| | - Wei-Bing Xing
- 1 Institute of Biology and Medicine, Wuhan University of Science and Technology , Wuhan, China
| | - Tong-Cun Zhang
- 1 Institute of Biology and Medicine, Wuhan University of Science and Technology , Wuhan, China .,2 Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology , Tianjin, China
| |
Collapse
|
28
|
Cerino G, Gaudiello E, Muraro MG, Eckstein F, Martin I, Scherberich A, Marsano A. Engineering of an angiogenic niche by perfusion culture of adipose-derived stromal vascular fraction cells. Sci Rep 2017; 7:14252. [PMID: 29079730 PMCID: PMC5660248 DOI: 10.1038/s41598-017-13882-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 10/02/2017] [Indexed: 01/01/2023] Open
Abstract
In vitro recapitulation of an organotypic stromal environment, enabling efficient angiogenesis, is crucial to investigate and possibly improve vascularization in regenerative medicine. Our study aims at engineering the complexity of a vascular milieu including multiple cell-types, a stromal extracellular matrix (ECM), and molecular signals. For this purpose, the human adipose stromal vascular fraction (SVF), composed of a heterogeneous mix of pericytes, endothelial/stromal progenitor cells, was cultured under direct perfusion flow on three-dimensional (3D) collagen scaffolds. Perfusion culture of SVF-cells reproducibly promoted in vitro the early formation of a capillary-like network, embedded within an ECM backbone, and the release of numerous pro-angiogenic factors. Compared to static cultures, perfusion-based engineered constructs were more rapidly vascularized and supported a superior survival of delivered cells upon in vivo ectopic implantation. This was likely mediated by pericytes, whose number was significantly higher (4.5-fold) under perfusion and whose targeted depletion resulted in lower efficiency of vascularization, with an increased host foreign body reaction. 3D-perfusion culture of SVF-cells leads to the engineering of a specialized milieu, here defined as an angiogenic niche. This system could serve as a model to investigate multi-cellular interactions in angiogenesis, and as a module supporting increased grafted cell survival in regenerative medicine.
Collapse
Affiliation(s)
- Giulia Cerino
- Departments of Biomedicine and Surgery, University of Basel and University Hospital of Basel, 4031, Basel, Switzerland
| | - Emanuele Gaudiello
- Departments of Biomedicine and Surgery, University of Basel and University Hospital of Basel, 4031, Basel, Switzerland
| | - Manuele Giuseppe Muraro
- Departments of Biomedicine and Surgery, University of Basel and University Hospital of Basel, 4031, Basel, Switzerland
| | - Friedrich Eckstein
- Departments of Biomedicine and Surgery, University of Basel and University Hospital of Basel, 4031, Basel, Switzerland
| | - Ivan Martin
- Departments of Biomedicine and Surgery, University of Basel and University Hospital of Basel, 4031, Basel, Switzerland
| | - Arnaud Scherberich
- Departments of Biomedicine and Surgery, University of Basel and University Hospital of Basel, 4031, Basel, Switzerland
| | - Anna Marsano
- Departments of Biomedicine and Surgery, University of Basel and University Hospital of Basel, 4031, Basel, Switzerland.
| |
Collapse
|
29
|
Peters EB. Endothelial Progenitor Cells for the Vascularization of Engineered Tissues. TISSUE ENGINEERING PART B-REVIEWS 2017; 24:1-24. [PMID: 28548628 DOI: 10.1089/ten.teb.2017.0127] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Self-assembled microvasculature from cocultures of endothelial cells (ECs) and stromal cells has significantly advanced efforts to vascularize engineered tissues by enhancing perfusion rates in vivo and producing investigative platforms for microvascular morphogenesis in vitro. However, to clinically translate prevascularized constructs, the issue of EC source must be resolved. Endothelial progenitor cells (EPCs) can be noninvasively supplied from the recipient through adult peripheral and umbilical cord blood, as well as derived from induced pluripotent stem cells, alleviating antigenicity issues. EPCs can also differentiate into all tissue endothelium, and have demonstrated potential for therapeutic vascularization. Yet, EPCs are not the standard EC choice to vascularize tissue constructs in vitro. Possible reasons include unresolved issues with EPC identity and characterization, as well as uncertainty in the selection of coculture, scaffold, and culture media combinations that promote EPC microvessel formation. This review addresses these issues through a summary of EPC vascular biology and the effects of tissue engineering design parameters upon EPC microvessel formation. Also included are perspectives to integrate EPCs with emerging technologies to produce functional, organotypic vascularized tissues.
Collapse
Affiliation(s)
- Erica B Peters
- Department of Chemical and Biological Engineering, University of Colorado , Boulder, Colorado
| |
Collapse
|
30
|
Liu M, Zhou Z, Chai Y, Zhang S, Wu X, Huang S, Su J, Jiang J. Synthesis of cell composite alginate microfibers by microfluidics with the application potential of small diameter vascular grafts. Biofabrication 2017; 9:025030. [PMID: 28485303 DOI: 10.1088/1758-5090/aa71da] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Fabrication of small diameter vascular grafts (SDVGs) with appropriate responses for clinical application is still challenging. In the present work, the production and characterization of solid alginate based microfibers as potential SDVG candidates through the method of microfluidics were considered original. A simple glass microfluidic device with a 'L-shape' cylindrical-flow channel in the microfluidic platform was developed. The gelation of microfibers occurred when the alginate solution and a CaCl2 solution were introduced as a core flow and as a sheath flow, respectively. The diameters of the microfibers could be controlled by varying the flow rates and the glass capillary tubes diameters at their tips. The generated microfibers had somewhat rough and porous surfaces, their suture retention strengths were comparable to the strength of other tissue engineered grafts. The encapsulated mesenchymal stem cells proliferated well in the microfibers, and showed a stable endothelialization under the angiogenesis effects of vascular endothelial growth factor and fibroblastic growth factor. The in vivo implant into the mice abdomens indicated that cell composite microfibers caused a mild host reaction. These encouraging results suggest great promise of the application of microfluidics as a future alternative in SDVGs engineering.
Collapse
Affiliation(s)
- Mingying Liu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education; State and Local Joint Engineering Laboratory for Vascular Implants; Bioengineering College, Chongqing University, Chongqing, 400044, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Engineering biomimetic periosteum with β-TCP scaffolds to promote bone formation in calvarial defects of rats. Stem Cell Res Ther 2017; 8:134. [PMID: 28583167 PMCID: PMC5460346 DOI: 10.1186/s13287-017-0592-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 04/26/2017] [Accepted: 05/18/2017] [Indexed: 01/01/2023] Open
Abstract
Background There is a critical need for the management of large bone defects. The purpose of this study was to engineer a biomimetic periosteum and to combine this with a macroporous β-tricalcium phosphate (β-TCP) scaffold for bone tissue regeneration. Methods Rat bone marrow-derived mesenchymal stem cells (rBMSCs) were harvested and cultured in different culture media to form undifferentiated rBMSC sheets (undifferentiated medium (UM)) and osteogenic cell sheets (osteogenic medium (OM)). Simultaneously, rBMSCs were differentiated to induced endothelial-like cells (iECs), and the iECs were further cultured on a UM to form a vascularized cell sheet. At the same time, flow cytometry was used to detect the conversion rates of rBMSCs to iECs. The pre-vascularized cell sheet (iECs/UM) and the osteogenic cell sheet (OM) were stacked together to form a biomimetic periosteum with two distinct layers, which mimicked the fibrous layer and cambium layer of native periosteum. The biomimetic periostea were wrapped onto porous β-TCP scaffolds (BP/β-TCP) and implanted in the calvarial bone defects of rats. As controls, autologous periostea with β-TCP (AP/β-TCP) and β-TCP alone were implanted in the calvarial defects of rats, with a no implantation group as another control. At 2, 4, and 8 weeks post-surgery, implants were retrieved and X-ray, microcomputed tomography (micro-CT), histology, and immunohistochemistry staining analyses were performed. Results Flow cytometry results showed that rBMSCs were partially differentiated into iECs with a 35.1% conversion rate in terms of CD31. There were still 20.97% rBMSCs expressing CD90. Scanning electron microscopy (SEM) results indicated that cells from the wrapped cell sheet on the β-TCP scaffold apparently migrated into the pores of the β-TCP scaffold. The histology and immunohistochemistry staining results from in vivo implantation indicated that the BP/β-TCP and AP/β-TCP groups promoted the formation of blood vessels and new bone tissues in the bone defects more than the other two control groups. In addition, micro-CT showed that more new bone tissue formed in the BP/β-TCP and AP/β-TCP groups than the other groups. Conclusions Inducing rBMSCs to iECs could be a good strategy to obtain an endothelial cell source for prevascularization. Our findings indicate that the biomimetic periosteum with porous β-TCP scaffold has a similar ability to promote osteogenesis and angiogenesis in vivo compared to the autologous periosteum. This function could result from the double layers of biomimetic periosteum. The prevascularized cell sheet served a mimetic fibrous layer and the osteogenic cell sheet served a cambium layer of native periosteum. The biomimetic periosteum with a porous ceramic scaffold provides a new promising method for bone healing.
Collapse
|
32
|
Nwabo Kamdje AH, Kamga PT, Simo RT, Vecchio L, Seke Etet PF, Muller JM, Bassi G, Lukong E, Goel RK, Amvene JM, Krampera M. Mesenchymal stromal cells' role in tumor microenvironment: involvement of signaling pathways. Cancer Biol Med 2017; 14:129-141. [PMID: 28607804 PMCID: PMC5444925 DOI: 10.20892/j.issn.2095-3941.2016.0033] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are adult multipotent stem cells residing as pericytes in various tissues and organs where they can differentiate into specialized cells to replace dying cells and damaged tissues. These cells are commonly found at injury sites and in tumors that are known to behave like " wounds that do not heal." In this article, we discuss the mechanisms of MSCs in migrating, homing, and repairing injured tissues. We also review a number of reports showing that tumor microenvironment triggers plasticity mechanisms in MSCs to induce malignant neoplastic tissue formation, maintenance, and chemoresistance, as well as tumor growth. The antitumor properties and therapeutic potential of MSCs are also discussed.
Collapse
Affiliation(s)
| | - Paul Takam Kamga
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| | - Richard Tagne Simo
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| | - Lorella Vecchio
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| | | | - Jean Marc Muller
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| | - Giulio Bassi
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| | - Erique Lukong
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| | - Raghuveera Kumar Goel
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| | - Jeremie Mbo Amvene
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| | - Mauro Krampera
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| |
Collapse
|
33
|
The Roles of Insulin-Like Growth Factors in Mesenchymal Stem Cell Niche. Stem Cells Int 2017; 2017:9453108. [PMID: 28298931 PMCID: PMC5337393 DOI: 10.1155/2017/9453108] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 12/22/2016] [Accepted: 01/18/2017] [Indexed: 12/12/2022] Open
Abstract
Many tissues contain adult mesenchymal stem cells (MSCs), which may be used in tissue regeneration therapies. However, the MSC availability in most tissues is limited which demands expansion in vitro following isolation. Like many developing cells, the state of MSCs is affected by the surrounding microenvironment, and mimicking this natural microenvironment that supports multipotent or differentiated state in vivo is essential to understand for the successful use of MSC in regenerative therapies. Many researchers are, therefore, optimizing cell culture conditions in vitro by altering growth factors, extracellular matrices, chemicals, oxygen tension, and surrounding pH to enhance stem cells self-renewal or differentiation. Insulin-like growth factors (IGFs) system has been demonstrated to play an important role in stem cell biology to either promote proliferation and self-renewal or enhance differentiation onset and outcome, depending on the cell culture conditions. In this review, we will describe the importance of IGFs, IGF-1 and IGF-2, in development and in the MSC niche and how they affect the pluripotency or differentiation towards multiple lineages of the three germ layers.
Collapse
|
34
|
Growth Hormone-Releasing Hormone and Its Analogues: Significance for MSCs-Mediated Angiogenesis. Stem Cells Int 2016; 2016:8737589. [PMID: 27774107 PMCID: PMC5059609 DOI: 10.1155/2016/8737589] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 06/19/2016] [Accepted: 07/03/2016] [Indexed: 02/08/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are promising candidates for regenerative medicine because of their multipotency, immune-privilege, and paracrine properties including the potential to promote angiogenesis. Accumulating evidence suggests that the inherent properties of cytoprotection and tissue repair by native MSCs can be enhanced by various preconditioning stimuli implemented prior to cell transplantation. Growth hormone-releasing hormone (GHRH), a stimulator in extrahypothalamus systems including tumors, has attracted great attentions in recent years because GHRH and its agonists could promote angiogenesis in various tissues. GHRH and its agonists are proangiogenic in responsive tissues including tumors, and GHRH antagonists have been tested as antitumor agents through their ability to suppress angiogenesis and cell growth. GHRH-R is expressed by MSCs and evolving work from our laboratory indicates that treatment of MSCs with GHRH agonists prior to cell transplantation markedly enhanced the angiogenic potential and tissue reparative properties of MSCs through a STAT3 signaling pathway. In this review we summarized the possible effects of GHRH analogues on cell growth and development, as well as on the proangiogenic properties of MSCs. We also discussed the relationship between GHRH analogues and MSC-mediated angiogenesis. The analyses provide new insights into molecular pathways of MSCs-based therapies and their augmentation by GHRH analogues.
Collapse
|
35
|
Castaldi A, Chesini GP, Taylor AE, Sussman MA, Brown JH, Purcell NH. Sphingosine 1-phosphate elicits RhoA-dependent proliferation and MRTF-A mediated gene induction in CPCs. Cell Signal 2016; 28:871-9. [PMID: 27094722 PMCID: PMC5004781 DOI: 10.1016/j.cellsig.2016.04.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 04/01/2016] [Accepted: 04/10/2016] [Indexed: 12/16/2022]
Abstract
Although c-kit(+) cardiac progenitor cells (CPCs) are currently used in clinical trials there remain considerable gaps in our understanding of the molecular mechanisms underlying their proliferation and differentiation. G-protein coupled receptors (GPCRs) play an important role in regulating these processes in mammalian cell types thus we assessed GPCR mRNA expression in c-kit(+) cells isolated from adult mouse hearts. Our data provide the first comprehensive overview of the distribution of this fundamental class of cardiac receptors in CPCs and reveal notable distinctions from that of adult cardiomyocytes. We focused on GPCRs that couple to RhoA activation in particular those for sphingosine-1-phosphate (S1P). The S1P2 and S1P3 receptors are the most abundant S1P receptor subtypes in mouse and human CPCs while cardiomyocytes express predominantly S1P1 receptors. Treatment of CPCs with S1P, as with thrombin and serum, increased proliferation through a pathway requiring RhoA signaling, as evidenced by significant attenuation when Rho was inhibited by treatment with C3 toxin. Further analysis demonstrated that both S1P- and serum-induced proliferation are regulated through the S1P2 and S1P3 receptor subtypes which couple to Gα12/13 to elicit RhoA activation. The transcriptional co-activator MRTF-A was activated by S1P as assessed by its nuclear accumulation and induction of a RhoA/MRTF-A luciferase reporter. In addition S1P treatment increased expression of cardiac lineage markers Mef2C and GATA4 and the smooth muscle marker GATA6 through activation of MRTF-A. In conclusion, we delineate an S1P-regulated signaling pathway in CPCs that introduces the possibility of targeting S1P2/3 receptors, Gα12/13 or RhoA to influence the proliferation and commitment of c-kit(+) CPCs and improve the response of the myocardium following injury.
Collapse
Affiliation(s)
- Alessandra Castaldi
- Department of Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0636, USA
| | - Gino P Chesini
- Department of Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0636, USA
| | - Amy E Taylor
- Department of Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0636, USA
| | - Mark A Sussman
- San Diego State Heart Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Joan Heller Brown
- Department of Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0636, USA.
| | - Nicole H Purcell
- Department of Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0636, USA
| |
Collapse
|
36
|
Ke X, Liu C, Wang Y, Ma J, Mao X, Li Q. Netrin-1 promotes mesenchymal stem cell revascularization of limb ischaemia. Diab Vasc Dis Res 2016; 13:145-156. [PMID: 26818229 DOI: 10.1177/1479164115611594] [Citation(s) in RCA: 14] [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] [Indexed: 11/15/2022] Open
Abstract
This study examines the effect and mechanism of action of Netrin-1 on bone marrow mesenchymal stem cells in angiogenesis. Tube formation and migration of bone marrow mesenchymal stem cells were observed in cell culture. Bone marrow mesenchymal stem cells or Netrin-1-bone marrow mesenchymal stem cells were injected into the ischaemic area of the rat hind limb on the first day after surgery. Laser Doppler perfusion imaging was performed to analyse the levels of vascular endothelial growth factor in plasma and muscles, and immunohistochemistry and immunofluorescence were used to analyse angiogenesis. Bone marrow mesenchymal stem cells in medium containing Netrin-1 markedly increased the number of tubes formed and the migration of bone marrow mesenchymal stem cells compared with the untreated control group. The function of Netrin-1 in tube formation and migration is similar to vascular endothelial growth factor, and combined with vascular endothelial growth factor, Netrin-1 has more enhanced effect than in the other three groups. The Netrin-1-bone marrow mesenchymal stem cell group had better augmented blood-perfusion scores and vessel densities, as well as improved function of the ischaemic limb than that of the group injected with bone marrow mesenchymal stem cells (treated with bone marrow mesenchymal stem cells individually) or the control group (treated with medium). These results suggest that Netrin-1 has the ability to augment the angiogenesis of bone marrow mesenchymal stem cells and improve the function of the ischaemic hind limb by increasing the level of vascular endothelial growth factor.
Collapse
Affiliation(s)
- Xianjin Ke
- Department of Neurology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Chenxiao Liu
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Ying Wang
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jianhua Ma
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiaoming Mao
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qian Li
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
37
|
Hong S, Lee JY, Hwang C, Shin JH, Park Y. Inhibition of Rho-Associated Protein Kinase Increases the Angiogenic Potential of Mesenchymal Stem Cell Aggregates via Paracrine Effects. Tissue Eng Part A 2016; 22:233-43. [PMID: 26592750 DOI: 10.1089/ten.tea.2015.0289] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The aggregation of multiple cells, such as mesenchymal condensation, is an important biological process in skeletal muscle development, osteogenesis, and adipogenesis. Due to limited in vivo study model systems, a simple and effective in vitro three-dimensional (3D) aggregation system is required to study the mechanisms of multicellular aggregation and its applications. We first generated controlled mesenchymal stem cell (MSC) aggregates using a bioprinting technique to monitor their aggregation and sprouting. We induced the angiogenic potential of the MSCs through chemical inhibition of the Rho/Rho-associated protein kinase (ROCK) pathway, which led to hairy sprouting in the aggregates. The angiogenic potential of this 3D construct was then tested by subcutaneously implanting the Matrigel with 3D MSC aggregates in a rat. Treatment of 3D MSCs with the ROCK inhibitor, Y27632, increased their angiogenic activity in vivo. The gene expressions and histological staining indicated that angiogenesis and neovascularization were mainly regulated by the paracrine factors secreted from human 3D MSC constructs. Our results demonstrate the enhancement of the angiogenic potential of the MSC constructs through the secretion of vascular endothelial growth factor (VEGF) and epidermal growth factor (EGF) by the inhibition of the Rho/ROCK pathway.
Collapse
Affiliation(s)
- Soyoung Hong
- 1 Department of Biomedical Engineering, College of Medicine, Korea University , Seoul, Korea.,2 Biomedical Engineering Research Center, Asan Medical Center , Seoul, Korea
| | - Jae Yeon Lee
- 1 Department of Biomedical Engineering, College of Medicine, Korea University , Seoul, Korea
| | - Changmo Hwang
- 2 Biomedical Engineering Research Center, Asan Medical Center , Seoul, Korea
| | - Jennifer H Shin
- 3 Department of Mechanical Engineering, Graduate School of Medical Science and Engineering , KAIST, Daejeon, Korea
| | - Yongdoo Park
- 1 Department of Biomedical Engineering, College of Medicine, Korea University , Seoul, Korea
| |
Collapse
|
38
|
Inglis S, Christensen D, Wilson DI, Kanczler JM, Oreffo ROC. Human endothelial and foetal femur-derived stem cell co-cultures modulate osteogenesis and angiogenesis. Stem Cell Res Ther 2016; 7:13. [PMID: 26781715 PMCID: PMC4717648 DOI: 10.1186/s13287-015-0270-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 05/22/2015] [Accepted: 12/21/2015] [Indexed: 02/05/2023] Open
Abstract
Background A dynamic vasculature is a prerequisite for bone formation where the interaction of bone cells and endothelial cells is essential for both the development and the healing process of bone. Enhanced understanding of the specific mediators involved in bone cell and endothelial cell interactions offers new avenues for skeletal regenerative applications. This study has investigated the osteogenic and angiogenic potential of co-cultures of human foetal diaphyseal or epiphyseal cells with human umbilical vein endothelial cells (HUVEC) in the presence and absence of vascular endothelial growth factor (VEGF) supplementation. Methods Early osteogenic activities of the co-cultures (±VEGF) were assessed by alkaline phosphatase (ALP) activity. Osteogenic and angiogenic gene expression was measured using quantitative polymerase chain reaction. An ex vivo organotypic embryonic chick (E11) femur culture model was used to determine the osteogenic effects of VEGF as determined using micro-computed tomography (μCT) and Alcian blue/Sirius red histochemistry and immunocytochemistry for expression of CD31. Results ALP activity and gene expression of ALP and Type-1 collagen was enhanced in foetal skeletal/HUVECs co-cultures. In foetal diaphyseal/HUVECs co-cultures, VEGF reduced the levels of ALP activity and displayed a negligible effect on von Willebrand factor (vWF) and VEGF gene expression. In contrast, VEGF supplementation was observed to significantly increase FLT-1 and KDR gene expression in co-cultures with modulation of expression enhanced, compared to VEGF skeletal monocultures. In the organotypic chick model, addition of VEGF significantly enhanced bone formation, which coincided with elevated levels of CD31-positive cells in the mid-diaphyseal region of the femurs. Conclusion These studies demonstrate a differential skeletal response of early foetal skeletal cells, when co-cultured with endothelial cells and the potential of co-culture models for bone repair. The differential effect of VEGF supplementation on markers of angiogenesis and osteogenesis in co-cultures and organ cultures, demonstrate the importance of the intricate temporal coordination of osteogenic and angiogenic processes during bone formation and implications therein for effective approaches to bone regenerative therapies. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0270-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stefanie Inglis
- Bone & Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Institute of Developmental Sciences, University of Southampton, Southampton, SO16 6YD, UK.
| | - David Christensen
- Human Development and Health, Centre for Human Development, Stem Cells and Regeneration, University of Southampton, Southampton, SO16 6YD, UK.
| | - David I Wilson
- Human Development and Health, Centre for Human Development, Stem Cells and Regeneration, University of Southampton, Southampton, SO16 6YD, UK.
| | - Janos M Kanczler
- Bone & Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Institute of Developmental Sciences, University of Southampton, Southampton, SO16 6YD, UK.
| | - Richard O C Oreffo
- Bone & Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Institute of Developmental Sciences, University of Southampton, Southampton, SO16 6YD, UK.
| |
Collapse
|
39
|
Tao H, Han Z, Han ZC, Li Z. Proangiogenic Features of Mesenchymal Stem Cells and Their Therapeutic Applications. Stem Cells Int 2016; 2016:1314709. [PMID: 26880933 PMCID: PMC4736816 DOI: 10.1155/2016/1314709] [Citation(s) in RCA: 186] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 11/04/2015] [Accepted: 11/29/2015] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have shown their therapeutic potency for treatment of cardiovascular diseases owing to their low immunogenicity, ease of isolation and expansion, and multipotency. As multipotent progenitors, MSCs have revealed their ability to differentiate into various cell types and could promote endogenous angiogenesis via microenvironmental modulation. Studies on cardiovascular diseases have demonstrated that transplanted MSCs could engraft at the injured sites and differentiate into cardiomyocytes and endothelial cells as well. Accordingly, several clinical trials using MSCs have been performed and revealed that MSCs may improve relevant clinical parameters in patients with vascular diseases. To fully comprehend the characteristics of MSCs, understanding their intrinsic property and associated modulations in tuning their behaviors as well as functions is indispensable for future clinical translation of MSC therapy. This review will focus on recent progresses on endothelial differentiation and potential clinical application of MSCs, with emphasis on therapeutic angiogenesis for treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Hongyan Tao
- Department of Pathophysiology, Nankai University School of Medicine, Tianjin 300071, China
- The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University College of Life Science, Tianjin 300071, China
| | - Zhibo Han
- State Key Lab of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Tianjin 300020, China
| | - Zhong Chao Han
- State Key Lab of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Tianjin 300020, China
| | - Zongjin Li
- Department of Pathophysiology, Nankai University School of Medicine, Tianjin 300071, China
- The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University College of Life Science, Tianjin 300071, China
| |
Collapse
|
40
|
Hwang S, Lee HJ, Kim G, Won KJ, Park YS, Jo I. CCN1 acutely increases nitric oxide production via integrin αvβ3-Akt-S6K-phosphorylation of endothelial nitric oxide synthase at the serine 1177 signaling axis. Free Radic Biol Med 2015; 89:229-40. [PMID: 26393424 DOI: 10.1016/j.freeradbiomed.2015.08.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 07/14/2015] [Accepted: 08/05/2015] [Indexed: 11/30/2022]
Abstract
Although CCN1 (also known as cysteine-rich, angiogenic inducer 61, CYR61) has been reported to promote angiogenesis and neovascularization in endothelial cells (ECs), its effects on endothelial nitric oxide (NO) production have never been studied. Using human umbilical vein ECs, we investigated whether and how CCN1 regulates NO production. CCN1 acutely increased NO production in a time- and dose-dependent manner, which was accompanied by increased phosphorylation of endothelial NO synthase (eNOS) at serine 1177 (eNOS-Ser(1177)), but not that of eNOS-Thr(495) or eNOS-Ser(114). The level of total eNOS expression was unaltered. Treatment with either LY294002, a selective inhibitor of phosphoinositide 3-kinase known as an upstream kinase of Akt, or H-89, an inhibitor of protein kinase A, mitogen- and stress-activated protein kinase 1, Rho-associated protein kinase 2, and ribosomal protein S6 kinase (S6K), inhibited CCN1-stimulated eNOS-Ser(1177) phosphorylation and subsequent NO production. Ectopic expression of small interfering RNA against Akt and S6K significantly inhibited the effects of CCN1. Consistently, CCN1 increased the phosphorylation of Akt-Ser(473) and S6K-Thr(389). However, CCN1 did not alter the expression or secretion of VEGF, a known downstream factor of CCN1 and a potential upstream factor of Akt-mediated eNOS-Ser(1177) phosphorylation. Furthermore, neutralization of integrin αvβ3 with corresponding antibody completely reversed all of the observed effects of CCN1. Moreover, CCN1 increased acetylcholine-induced relaxation in the rat aortas. Finally, we also found that CCN1-stimulated eNOS-Ser(1177) phosphorylation and NO production are true for other types of EC tested. In conclusion, CCN1 acutely increases NO production via activation of a signaling axis in integrin αvβ3-Akt-S6K-eNOS-Ser(1177) phosphorylation, suggesting an important role for CCN1 in vasodilation.
Collapse
Affiliation(s)
- Soojin Hwang
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea
| | - Hyeon-Ju Lee
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea
| | - Gyungah Kim
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea
| | - Kyung-Jong Won
- Department of Medical Science, School of Medicine, Konkuk University, Chungju 380-701, Republic of Korea
| | - Yoon Shin Park
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea
| | - Inho Jo
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea.
| |
Collapse
|
41
|
Rammal H, Harmouch C, Lataillade JJ, Laurent-Maquin D, Labrude P, Menu P, Kerdjoudj H. Stem cells: a promising source for vascular regenerative medicine. Stem Cells Dev 2015; 23:2931-49. [PMID: 25167472 DOI: 10.1089/scd.2014.0132] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The rising and diversity of many human vascular diseases pose urgent needs for the development of novel therapeutics. Stem cell therapy represents a challenge in the medicine of the twenty-first century, an area where tissue engineering and regenerative medicine gather to provide promising treatments for a wide variety of diseases. Indeed, with their extensive regeneration potential and functional multilineage differentiation capacity, stem cells are now highlighted as promising cell sources for regenerative medicine. Their multilineage differentiation involves environmental factors such as biochemical, extracellular matrix coating, oxygen tension, and mechanical forces. In this review, we will focus on human stem cell sources and their applications in vascular regeneration. We will also discuss the different strategies used for their differentiation into both mature and functional smooth muscle and endothelial cells.
Collapse
Affiliation(s)
- Hassan Rammal
- 1 UMR 7365, Biopôle, Faculté de Médecine, CNRS-Université de Lorraine , Vandœuvre-lès-Nancy, France
| | | | | | | | | | | | | |
Collapse
|
42
|
Improving vascularization of engineered bone through the generation of pro-angiogenic effects in co-culture systems. Adv Drug Deliv Rev 2015; 94:116-25. [PMID: 25817732 DOI: 10.1016/j.addr.2015.03.012] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 02/27/2015] [Accepted: 03/20/2015] [Indexed: 01/31/2023]
Abstract
One of the major problems with bone tissue engineering is the development of a rapid vascularization after implantation to supply the growing osteoblast cells with the nutrients to grow and survive as well as to remove waste products. It has been demonstrated that capillary-like structures produced in vitro will anastomose rapidly after implantation and become functioning blood vessels. For this reason, in recent years many studies have examined a variety of human osteoblast and endothelial cell co-culture systems in order to distribute osteoblasts on all parts of the bone scaffold and at the same time provide conditions for the endothelial cells to migrate to form a network of capillary-like structures throughout the osteoblast-colonized scaffold. The movement and proliferation of endothelial cells to form capillary-like structures is known as angiogenesis and is dependent on a variety of pro-angiogenic factors. This review summarizes human 2- and 3-D co-culture models to date, the types and origins of cells used in the co-cultures and the proangiogenic factors that have been identified in the co-culture models.
Collapse
|
43
|
Use of a Human Artificial Chromosome for Delivering Trophic Factors in a Rodent Model of Amyotrophic Lateral Sclerosis. MOLECULAR THERAPY. NUCLEIC ACIDS 2015; 4:e253. [PMID: 26440597 PMCID: PMC4881756 DOI: 10.1038/mtna.2015.28] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Accepted: 08/17/2015] [Indexed: 12/13/2022]
Abstract
A human artificial chromosome (HAC) is maintained as an episome within a cell and avoids random integration into the host genome. It can transfer multiple and/or large transgenes along with their regulatory elements thereby resembling native chromosomes. Using this HAC system, we established mesenchymal stem cells (MSCs) that simultaneously expressed hepatocyte growth factor, glial cell line-derived neurotrophic factor, and insulin-like growth factor 1, termed HAC-MSCs. This cell line provides an opportunity for stable transplantation and thorough analyses. We then introduced the cells for the treatment of a neurodegenerative disorder, amyotrophic lateral sclerosis. The HAC-MSCs were transplanted via the fourth cerebral ventricle (CV) or intravenous (i.v.) infusion at various ages of recipient mice. Littermate- and sex-matched mice underwent a sham procedure. Compared to the controls, there was an encouraging trend of increased life span via CV transplantation and delayed onset in i.v. infusion 60 days after transplantation. Further, we confirmed a statistically significant increase in life span via CV transplantation at 100 days. This effect was not seen in mice transplanted with MSCs lacking the HAC. We successfully enhanced the trophic potential of the MSCs using the HAC. This strategy could be a promising direction for the treatment of neurodegenerative disorders.
Collapse
|
44
|
Production of Human Endothelial Cells Free from Soluble Xenogeneic Antigens for Bioartificial Small Diameter Vascular Graft Endothelization. BIOMED RESEARCH INTERNATIONAL 2015; 2015:652474. [PMID: 26146626 PMCID: PMC4471259 DOI: 10.1155/2015/652474] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 01/29/2015] [Indexed: 11/17/2022]
Abstract
Arterial bypass graft implantation remains the primary therapy for patients with advanced cardiovascular disease, but most lack adequate saphenous vein or other conduits for bypass procedures and would benefit from a bioartificial conduit. This study aimed to produce human endothelial cells (hECs) in large scale, free from xenogeneic antigens, to develop a small diameter, compatible vessel for potential use as a vascular graft. Human adipose-derived stromal cells (hASCs) were isolated, cultured, and differentiated in the presence of human serum and used for the reendothelization of a decellularized rat aorta. hASC derived ECs (hASC-ECs) expressed VEGFR2, vWf and CD31 endothelial cell markers, the latter in higher levels than hASCs and HUVECs, and were shown to be functional. Decellularization protocol yielded aortas devoid of cell nuclei, with preserved structure, including a preserved basement membrane. When seeded with hASC-ECs, the decellularized aorta was completely reendothelized, and the hASC-ECs maintained their phenotype in this new condition. hASCs can be differentiated into functional hECs without the use of animal supplements and are capable of reendothelizing a decellularized rat aorta while maintaining their phenotype. The preservation of the basement membrane following decellularization supported the complete reendothelization of the scaffold with no cell migration towards other layers. This approach is potentially useful for rapid obtention of compatible, xenogeneic-free conduit.
Collapse
|
45
|
A novel inhibitory mechanism of MRTF-A/B on the ICAM-1 gene expression in vascular endothelial cells. Sci Rep 2015; 5:10627. [PMID: 26024305 PMCID: PMC4448521 DOI: 10.1038/srep10627] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 04/22/2015] [Indexed: 01/09/2023] Open
Abstract
The roles of myocardin-related transcription factor A (MRTF-A) and MRTF-B in vascular endothelial cells are not completely understood. Here, we found a novel regulatory mechanism for MRTF-A/B function. MRTF-A/B tend to accumulate in the nucleus in arterial endothelial cells in vivo and human aortic endothelial cells (HAoECs) in vitro. In HAoECs, nuclear localization of MRTF-A/B was not significantly affected by Y27632 or latrunculin B, primarily due to the reduced binding of MRTF-A/B to G-actin and in part, to the low level of MRTF-A phosphorylation by ERK. MRTF-A/B downregulation by serum depletion or transfection of siRNA against MRTF-A and/or MRTF-B induced ICAM-1 expression in HAoECs. It is known that nuclear import of nuclear factor−κB (NF−κB) plays a key role in ICAM-1 gene transcription. However, nuclear accumulation of NF−κB p65 was not observed in MRTF-A/B-depleted HAoECs. Our present findings suggest that MRTF-A/B inhibit ICAM-1 mRNA expression by forming a complex with NF−κB p65 in the nucleus. Conversely, downregulation of MRTF-A/B alleviates this negative regulation without further translocation of NF−κB p65 into the nucleus. These results reveal the novel roles of MRTF-A/B in the homeostasis of vascular endothelium.
Collapse
|
46
|
Jian RL, Mao LB, Xu Y, Li XF, Wang FP, Luo XG, Zhou H, He HP, Wang N, Zhang TC. Generation of insulin-producing cells from C3H10T1/2 mesenchymal progenitor cells. Gene 2015; 562:107-116. [PMID: 25724395 DOI: 10.1016/j.gene.2015.02.061] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 02/10/2015] [Accepted: 02/18/2015] [Indexed: 12/29/2022]
Abstract
Mesenchymal stem cells (MSCs) have been reported to be an attractive source for the generation of transplantable surrogate β cells. A murine embryonic mesenchymal progenitor cell line C3H10T1/2 has been recognized as a model for MSCs, because of its multi-lineage differentiation potential. The purpose of this study was to explore whether C3H/10T1/2 cells have the potential to differentiate into insulin-producing cells (IPCs). Here, we investigated and compared the in vitro differentiation of rat MSCs and C3H10T1/2 cells into IPCs. After the cells underwent IPC differentiation, the expression of differentiation markers were detected by immunocytochemistry, reverse transcription-polymerase chain reaction (RT-PCR), quantitative real-time RT-PCR (qRT-PCR) and Western blotting. The insulin secretion was evaluated by enzyme-linked immunosorbent assay (ELISA). Furthermore, these differentiated cells were transplanted into streptozotocin-induced diabetic mice and their biological functions were tested in vivo. This study reports a 2-stage method to generate IPCs from C3H10T1/2 cells. Under specific induction conditions for 7-8 days, C3H10T1/2 cells formed three-dimensional spheroid bodies (SBs) and secreted insulin, while generation of IPCs derived from rat MSCs required a long time (more than 2 weeks). Furthermore, these IPCs derived from C3H10T1/2 cells were injected into diabetic mice and improves basal glucose, body weight and exhibited normal glucose tolerance test. The present study provided a simple and faithful in vitro model for further investigating the mechanism underlying IPC differentiation of MSCs and cell replacement therapy for diabetes.
Collapse
Affiliation(s)
- Ruo-Lei Jian
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Li-Bin Mao
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yao Xu
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Xiao-Fan Li
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Feng-Po Wang
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Xue-Gang Luo
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Hao Zhou
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Hong-Peng He
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Nan Wang
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China.
| | - Tong-Cun Zhang
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; Department of Biochemistry, Medical College, Wuhan University of Science and Technology, Wuhan 430081, China.
| |
Collapse
|
47
|
Zhang R, Wang N, Zhang M, Zhang LN, Guo ZX, Luo XG, Zhou H, He HP, Zhang TC. Rho/MRTF-A-Induced Integrin Expression Regulates Angiogenesis in Differentiated Multipotent Mesenchymal Stem Cells. Stem Cells Int 2015; 2015:534758. [PMID: 25949242 PMCID: PMC4408638 DOI: 10.1155/2015/534758] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/13/2015] [Accepted: 03/18/2015] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are known to undergo endothelial differentiation in response to treatment with vascular endothelial growth factor (VEGF), but their angiogenic ability is poorly characterized. In the present study, we aimed to further investigate the role of Rho/MRTF-A in angiogenesis by MSCs and the effect of the Rho/MRTF-A pathway on the expression of integrins α1β1 and α5β1, which are known to mediate physiological and pathological angiogenesis. Our results showed that increased expression of α1, α5, and β1 was observed during angiogenesis of differentiated MSCs, and the Rho/MRTF-A signaling pathway was demonstrated to be involved in regulating the expression of integrins α1, α5, and β1. Luciferase reporter assay and ChIP assay determined that MRTF-A could bind to and transactivate the integrin α1 and α5 promoters. Treatment with the Rho inhibitor C3 transferase, the Rho-associated protein kinase (ROCK) inhibitor Y27632 or with shMRTF-A inhibited both the upregulation of α1, α5, and β1 as well as angiogenesis. Furthermore, in human umbilical vein endothelial cells (HUVECs), MRTF-A deletion led to marked reductions in cell migration and vessel network formation compared with the control. These data demonstrate that Rho/MRTF-A signaling is an important mediator that controls integrin gene expression during MSC-mediated angiogenic processes.
Collapse
Affiliation(s)
- Rui Zhang
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Nan Wang
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Man Zhang
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Li-Nan Zhang
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Zhi-Xia Guo
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Xue-Gang Luo
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Hao Zhou
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Hong-Peng He
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Tong-Cun Zhang
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
- Department of Biochemistry, Medical College, Wuhan University of Science and Technology, Wuhan 430081, China
| |
Collapse
|
48
|
Shu XZ, Zhang LN, Zhang R, Zhang CJ, He HP, Zhou H, Wang N, Zhang TC. Histone acetyltransferase p300 promotes MRTF-A-mediates transactivation of VE-cadherin gene in human umbilical vein endothelial cells. Gene 2015; 563:17-23. [PMID: 25746323 DOI: 10.1016/j.gene.2015.02.076] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 02/16/2015] [Accepted: 02/27/2015] [Indexed: 11/15/2022]
Abstract
Vascular endothelial cadherin (VE-cadherin) is the major determinant of endothelial cell contact integrity and is required in vascular development and angiogenesis. Serum response factor (SRF) plays essential roles in postnatal retinal angiogenesis and adult neovascularization. It is unclear whether transcription of VE-cadherin is mediated by a SRF co-activator, myocardin-related transcription factor-A (MRTF-A). Here we have demonstrated that MRTF-A is a key regulatory factor to activate the transcription of VE-cadherin in human umbilical vein endothelial cells (HUVECs). siRNA-mediated knockdown of MRTF-A decreased the level of VE-cadherin in HUVECs. Vascular endothelial growth factor (VEGF) induced MRTF-A binding to the SRF-binding site (CArG box) within VE-cadherin promoter. Histone acetyltransferase p300 and MRTF-A could synergistically augment the expression of VE-cadherin by enhancing acetylation of histone3K9 (H3K9Ac), histone3K14 (H3K14Ac) and histone4 at the SRF-binding site within VE-cadherin promoter. Taken together, these data identified a detailed regulatory mechanism of VE-cadherin gene expression.
Collapse
Affiliation(s)
- Xiang-Zhu Shu
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, 300457, PR China
| | - Li-Nan Zhang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, 300457, PR China
| | - Rui Zhang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, 300457, PR China
| | - Cai-Jiao Zhang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, 300457, PR China
| | - Hong-Peng He
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, 300457, PR China
| | - Hao Zhou
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, 300457, PR China
| | - Nan Wang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, 300457, PR China.
| | - Tong-Cun Zhang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, 300457, PR China; Institute of Biology and Medicine, Wuhan University of Science and Technology, 430000, PR China.
| |
Collapse
|
49
|
Jiang Y. The expression of MRTF-A and AQP1 play important roles in the pathological vascular remodeling. Asian Pac J Cancer Prev 2015; 16:1375-1383. [PMID: 25743802 DOI: 10.7314/apjcp.2015.16.4.1375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Objective Myocardin-related transcription factor (MRTF)-A is a Rho signaling-responsive co-activator of serum response factor (SRF). The purpose of this study is to investigate the role of MRTF-A and AQP1 (aquaporin 1) in pathological vascular remodeling. MATERIALS AND METHODS MRTF-A, AQP1 and neointima expression was detected both in the wire injured femoral arteries of wild-type mice and the atherosclerotic aortic tissues of ApoE-/- mice. Expression of ICAM-1, matrix metallopeptidase 9 (MMP-9) and integrin β1 were also assayed. The intercourse relationship between the molecules were investigated by interfering RNA and inhibitor assay. RESULTS MRTF-A and AQP1 expression were significantly higher in the wire injured femoral arteries of wild-type mice and in the atherosclerotic aortic tissues of ApoE-/- mice than in healthy control tissues. Both in wire-injured femoral arteries in MRTF-A knockout (Mkl1-/-) mice and atherosclerotic lesions in Mkl1-/-; ApoE-/- mice, neointima formation were significantly attenuated and the expression of AQP1 were significantly decreased. Expression of ICAM-1, matrix metallopeptidase 9 (MMP-9) and integrin β1, three SRF targets and key regulators of cell migration, and AQP1 in injured arteries was significantly weaker in Mkl1-/- mice than in wild-type mice. In cultured vascular smooth muscle cells (VSMCs), knocking down MRTF-A reduced expression of these genes and significantly impaired cell migration. Underlying the increased MRTF-A expression in dedifferentiated VSMCs were the down-regulation of microRNA-300. Moreover, the MRTF-A inhibitor CCG1423 significantly reduced neointima formation following wire injury in mice. CONCLUSIONS MRTF-A could be a novel therapeutic target for the treatment of vascular diseases.
Collapse
Affiliation(s)
- Yong Jiang
- Laboratory Medical College, Jilin Medical College, Jilin, P.R.China E-mail :
| |
Collapse
|
50
|
Nassiri SM, Rahbarghazi R. Interactions of Mesenchymal Stem Cells with Endothelial Cells. Stem Cells Dev 2014; 23:319-32. [DOI: 10.1089/scd.2013.0419] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Seyed Mahdi Nassiri
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Reza Rahbarghazi
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|