101
|
Hypoxia: Turning vessels into vassals of cancer immunotolerance. Cancer Lett 2020; 487:74-84. [PMID: 32470491 DOI: 10.1016/j.canlet.2020.05.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 04/27/2020] [Accepted: 05/12/2020] [Indexed: 12/12/2022]
Abstract
Hypoxia is a universal feature of solid cancers caused by a mismatch between cellular oxygen supply and consumption. To meet the increased demand for oxygen, hypoxic cancer cells (CCs) induce a multifaceted process known as angiogenesis, wherein new vessels are formed by the sprouting of pre-existing ones. In addition to providing oxygen for growth and an exit route for dissemination, angiogenic vessels and factors are co-opted by CCs to enable the generation of an immunotolerant, hypoxic tumor microenvironment, leading to therapeutic failure and mortality. In this review, we discuss how hypoxia-inducible factors (HIFs), the mechanistic target of rapamycin (mTOR), and the unfolded protein response (UPR) control angiogenic factors serving both vascular and immunomodulatory functions in the tumor microenvironment. Possible therapeutic strategies, wherein targeting oxygen sensing might enhance anti-angiogenic and immunologically-mediated anti-cancer responses, are suggested.
Collapse
|
102
|
Abstract
Peripheral artery disease is a common disorder and a major cause of morbidity and mortality worldwide. Therapy is directed at reducing the risk of major adverse cardiovascular events and at ameliorating symptoms. Medical therapy is effective at reducing the incidence of myocardial infarction and stroke to which these patients are prone but is inadequate in relieving limb-related symptoms, such as intermittent claudication, rest pain, and ischemic ulceration. Limb-related morbidity is best addressed with surgical and endovascular interventions that restore perfusion. Current medical therapies have only modest effects on limb blood flow. Accordingly, there is an opportunity to develop medical approaches to restore limb perfusion. Vascular regeneration to enhance limb blood flow includes methods to enhance angiogenesis, arteriogenesis, and vasculogenesis using angiogenic cytokines and cell therapies. We review the molecular mechanisms of these processes; briefly discuss what we have learned from the clinical trials of angiogenic and cell therapies; and conclude with an overview of a potential new approach based upon transdifferentiation to enhance vascular regeneration in peripheral artery disease.
Collapse
Affiliation(s)
- John P Cooke
- From the Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute, TX
| | - Shu Meng
- From the Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute, TX
| |
Collapse
|
103
|
He L, Lui KO, Zhou B. The Formation of Coronary Vessels in Cardiac Development and Disease. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a037168. [PMID: 31636078 DOI: 10.1101/cshperspect.a037168] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Understanding how coronary blood vessels form and regenerate during development and progression of cardiac diseases will shed light on the development of new treatment options targeting coronary artery diseases. Recent studies with the state-of-the-art technologies have identified novel origins of, as well as new, cellular and molecular mechanisms underlying the formation of coronary vessels in the postnatal heart, including collateral artery formation, endocardial-to-endothelial differentiation and mesenchymal-to-endothelial transition. These new mechanisms of coronary vessel formation and regeneration open up new possibilities targeting neovascularization for promoting cardiac repair and regeneration. Here, we highlight some recent studies on cellular mechanisms of coronary vessel formation, and discuss the potential impact and significance of the findings on basic research and clinical application for treating ischemic heart disease.
Collapse
Affiliation(s)
- Lingjuan He
- The State Key Laboratory of Cell Biology, CAS Center for Excellence on Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Kathy O Lui
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR 999077, China
| | - Bin Zhou
- The State Key Laboratory of Cell Biology, CAS Center for Excellence on Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
104
|
Endothelialization of arterial vascular grafts by circulating monocytes. Nat Commun 2020; 11:1622. [PMID: 32238801 PMCID: PMC7113268 DOI: 10.1038/s41467-020-15361-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 03/05/2020] [Indexed: 12/15/2022] Open
Abstract
Recently our group demonstrated that acellular tissue engineered vessels (A-TEVs) comprised of small intestinal submucosa (SIS) immobilized with heparin and vascular endothelial growth factor (VEGF) could be implanted into the arterial system of a pre-clinical ovine animal model, where they endothelialized within one month and remained patent. Here we report that immobilized VEGF captures blood circulating monocytes (MC) with high specificity under a range of shear stresses. Adherent MC differentiate into a mixed endothelial (EC) and macrophage (Mφ) phenotype and further develop into mature EC that align in the direction of flow and produce nitric oxide under high shear stress. In-vivo, newly recruited cells on the vascular lumen express MC markers and at later times they co-express MC and EC-specific proteins and maintain graft patency. This novel finding indicates that the highly prevalent circulating MC contribute directly to the endothelialization of acellular vascular grafts under the right chemical and biomechanical cues. Acellular tissue engineered vessels functionalised with VEGF are coated with a layer of endothelial cells after in vivo implantation, but the source of the cells are unknown. Here the authors provide evidence that monocytes expressing VEGF receptors can transdifferentiate into endothelial cells via a macrophage intermediate.
Collapse
|
105
|
Golab-Janowska M, Paczkowska E, Machalinski B, Kotlega D, Meller A, Safranow K, Wankowicz P, Nowacki P. Elevated Inflammatory Parameter Levels Negatively Impact Populations of Circulating Stem Cells (CD133+), Early Endothelial Progenitor Cells (CD133+/VEGFR2+), and Fibroblast Growth Factor in Stroke Patients. Curr Neurovasc Res 2020; 16:19-26. [PMID: 30706812 DOI: 10.2174/1567202616666190129164906] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/13/2019] [Accepted: 01/14/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Endothelial Progenitor Cells (EPCs) are important players in neovascularization, mobilized through signalling by Angiogenic Growth Factors (AGFs) such as Vascular Endothelial Growth Factor (VEGF) and fibroblast growth factor (FGF). In vitro, inflammatory parameters impair the function and influence of EPCs on AGFs. However, this connection is not clear in vivo. To understand the mechanisms of augmented arteriogenesis and angiogenesis in acute ischemic stroke (AIS) patients, we investigated whether circulating stem cells (CD133+), early endothelial progenitor cells (CD133+/VEGFR2+), and endothelial cells (ECs; CD34¯/CD133¯/VEGFR2+) were increasingly mobilized during AIS, and whether there were correlations between EPC levels, growth factor levels and inflammatory parameters. METHODS Data on demographics, classical vascular risk factors, neurological deficit information (assessed using the National Institutes of Health Stroke Scale), and treatment were collected from 43 consecutive AIS patients (group I). Risk factor control patients (group II) included 22 nonstroke subjects matched by age, gender, and traditional vascular risk factors. EPCs were measured by flow cytometry and the populations of circulating stem cells (CD133+), early EPCs (CD133+/VEGFR2+), and ECs (CD34¯/CD133¯/VEGFR2+) were analysed. Correlations between EPC levels and VEGF and FGF vascular growth factor levels as well as the influence of inflammatory parameters on EPCs and AGFs were assessed. RESULTS Patient ages ranged from 54 to 92 years (mean age 75.2 ± 11.3 years). The number of circulating CD34¯/CD133¯/VEGF-R2+ cells was significantly higher in AIS patients than in control patients (p < 0.05). VEGF plasma levels were also significantly higher in AIS patients compared to control patients on day 7 (p < 0.05). FGF plasma levels in patients with AIS were significantly higher than those in the control group on day 3 (p < 0.05). There were no correlations between increased VEGF and FGF levels and the number of CD133+, CD133+/VEGFR2+, or CD34¯/CD133¯/VEGFR2+ cells. Leukocyte levels, FGF plasma levels, and the number of early EPCs were negatively correlated on day 3. High sensitivity C-reactive protein levels and the number of CD133+ and CD133+/VEGFR2+ cells were negatively correlated on day 7. In addition, there was a negative correlation between fibrinogen levels and FGF plasma levels as well as the number of early EPCs (CD133+/VEGFR2+). CONCLUSION AIS patients exhibited increased numbers of early EPCs (CD133+/VEGFR2+) and AGF (VEGF and FGF) levels. A negative correlation between inflammatory parameters and AGFs and EPCs indicated the unfavourable influence of inflammatory factors on EPC differentiation and survival. Moreover, these correlations represented an important mechanism linking inflammation to vascular disease.
Collapse
Affiliation(s)
| | - Edyta Paczkowska
- Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Boguslaw Machalinski
- Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Dariusz Kotlega
- Department of Neurology, Pomeranian Medical University, Szczecin, Poland
| | - Agnieszka Meller
- Department of Neurology, Pomeranian Medical University, Szczecin, Poland
| | - Krzysztof Safranow
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Szczecin, Poland
| | - Pawel Wankowicz
- Department of Neurology, Pomeranian Medical University, Szczecin, Poland
| | - Przemyslaw Nowacki
- Department of Neurology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
106
|
Yanishi K, Shoji K, Fujioka A, Hori Y, Yukawa A, Matoba S. Impact of Therapeutic Angiogenesis Using Autologous Bone Marrow-derived Mononuclear Cell Implantation in Patients with No-option Critical Limb Ischemia. Ann Vasc Dis 2020; 13:13-22. [PMID: 32273917 PMCID: PMC7140169 DOI: 10.3400/avd.ra.20-00002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Recently, the limb salvage rate of patients with critical limb ischemia (CLI) has been improved due to the development of revascularization and wound care treatment. However, many patients with CLI are refractory to standard treatments, including revascularization such as endovascular treatment or surgical bypass. Establishment of a new cell therapy is required to improve the limb salvage rate and prognosis in patients with CLI. In 1997, endothelial progenitor cells were found to be derived from the bone marrow to circulate as CD34 surface antigen positive cells in peripheral blood and to affect therapeutic angiogenesis in ischemic tissues. Later, therapeutic angiogenesis using autologous bone marrow-derived mononuclear cell (BM-MNC) implantation was performed for patients with no-option CLI in clinical practice. Several reports showed the safety and efficacy of the BM-MNC implantation in patients with CLI caused by arteriosclerosis obliterans, thromboangiitis obliterans (TAO), and collagen diseases. In particular, in patients with CLI caused by TAO, limb salvage rate was significantly improved compared with standard treatments. The BM-MNC implantation may be feasible and safe in patients with no-option CLI. Here, we review the efficacy of BM-MNC implantation in no-option CLI, with a focus on therapeutic angiogenesis.
Collapse
Affiliation(s)
- Kenji Yanishi
- Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Kyoto, Japan
| | - Keisuke Shoji
- Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Kyoto, Japan
| | - Ayumu Fujioka
- Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Kyoto, Japan
| | - Yusuke Hori
- Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Kyoto, Japan
| | - Arito Yukawa
- Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Kyoto, Japan
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Kyoto, Japan
| |
Collapse
|
107
|
Beltran-Camacho L, Jimenez-Palomares M, Rojas-Torres M, Sanchez-Gomar I, Rosal-Vela A, Eslava-Alcon S, Perez-Segura MC, Serrano A, Antequera-González B, Alonso-Piñero JA, González-Rovira A, Extremera-García MJ, Rodriguez-Piñero M, Moreno-Luna R, Larsen MR, Durán-Ruiz MC. Identification of the initial molecular changes in response to circulating angiogenic cells-mediated therapy in critical limb ischemia. Stem Cell Res Ther 2020; 11:106. [PMID: 32143690 PMCID: PMC7060566 DOI: 10.1186/s13287-020-01591-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/10/2020] [Accepted: 02/06/2020] [Indexed: 12/18/2022] Open
Abstract
Background Critical limb ischemia (CLI) constitutes the most aggressive form of peripheral arterial occlusive disease, characterized by the blockade of arteries supplying blood to the lower extremities, significantly diminishing oxygen and nutrient supply. CLI patients usually undergo amputation of fingers, feet, or extremities, with a high risk of mortality due to associated comorbidities. Circulating angiogenic cells (CACs), also known as early endothelial progenitor cells, constitute promising candidates for cell therapy in CLI due to their assigned vascular regenerative properties. Preclinical and clinical assays with CACs have shown promising results. A better understanding of how these cells participate in vascular regeneration would significantly help to potentiate their role in revascularization. Herein, we analyzed the initial molecular mechanisms triggered by human CACs after being administered to a murine model of CLI, in order to understand how these cells promote angiogenesis within the ischemic tissues. Methods Balb-c nude mice (n:24) were distributed in four different groups: healthy controls (C, n:4), shams (SH, n:4), and ischemic mice (after femoral ligation) that received either 50 μl physiological serum (SC, n:8) or 5 × 105 human CACs (SE, n:8). Ischemic mice were sacrificed on days 2 and 4 (n:4/group/day), and immunohistochemistry assays and qPCR amplification of Alu-human-specific sequences were carried out for cell detection and vascular density measurements. Additionally, a label-free MS-based quantitative approach was performed to identify protein changes related. Results Administration of CACs induced in the ischemic tissues an increase in the number of blood vessels as well as the diameter size compared to ischemic, non-treated mice, although the number of CACs decreased within time. The initial protein changes taking place in response to ischemia and more importantly, right after administration of CACs to CLI mice, are shown. Conclusions Our results indicate that CACs migrate to the injured area; moreover, they trigger protein changes correlated with cell migration, cell death, angiogenesis, and arteriogenesis in the host. These changes indicate that CACs promote from the beginning an increase in the number of vessels as well as the development of an appropriate vascular network. Graphical abstract ![]()
Collapse
Affiliation(s)
- Lucia Beltran-Camacho
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | - Margarita Jimenez-Palomares
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | - Marta Rojas-Torres
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | - Ismael Sanchez-Gomar
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | - Antonio Rosal-Vela
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | - Sara Eslava-Alcon
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | | | - Ana Serrano
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain
| | - Borja Antequera-González
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | - Jose Angel Alonso-Piñero
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | - Almudena González-Rovira
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | - Mª Jesús Extremera-García
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | | | - Rafael Moreno-Luna
- Laboratory of Neuroinflammation, Hospital Nacional de Paraplejicos, SESCAM, Toledo, Spain
| | - Martin Røssel Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Mª Carmen Durán-Ruiz
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain. .,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain.
| |
Collapse
|
108
|
Jia J, Ma B, Wang S, Feng L. Therapeutic Potential of Endothelial Colony Forming Cells Derived from Human Umbilical Cord Blood. Curr Stem Cell Res Ther 2020; 14:460-465. [PMID: 30767752 DOI: 10.2174/1574888x14666190214162453] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 01/06/2019] [Accepted: 01/24/2019] [Indexed: 02/08/2023]
Abstract
Endothelial progenitor cells (EPCs) are implicated in multiple biologic processes such as vascular homeostasis, neovascularization and tissue regeneration, and tumor angiogenesis. A subtype of EPCs is referred to as endothelial colony-forming cells (ECFCs), which display robust clonal proliferative potential and can form durable and functional blood vessels in animal models. In this review, we provide a brief overview of EPCs' characteristics, classification and origins, a summary of the progress in preclinical studies with regard to the therapeutic potential of human umbilical cord blood derived ECFCs (CB-ECFCs) for ischemia repair, tissue engineering and tumor, and highlight the necessity to select high proliferative CB-ECFCs and to optimize their recovery and expansion conditions.
Collapse
Affiliation(s)
- Jing Jia
- Department of Obstetrics and Gynaecology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R., China
| | - Baitao Ma
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R., China
| | - Shaoshuai Wang
- Department of Obstetrics and Gynaecology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R., China
| | - Ling Feng
- Department of Obstetrics and Gynaecology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R., China
| |
Collapse
|
109
|
Lee JB, Kim DH, Yoon JK, Park DB, Kim HS, Shin YM, Baek W, Kang ML, Kim HJ, Sung HJ. Microchannel network hydrogel induced ischemic blood perfusion connection. Nat Commun 2020; 11:615. [PMID: 32001693 PMCID: PMC6992688 DOI: 10.1038/s41467-020-14480-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 01/12/2020] [Indexed: 12/13/2022] Open
Abstract
Angiogenesis induction into damaged sites has long been an unresolved issue. Local treatment with pro-angiogenic molecules has been the most common approach. However, this approach has critical side effects including inflammatory coupling, tumorous vascular activation, and off-target circulation. Here, the concept that a structure can guide desirable biological function is applied to physically engineer three-dimensional channel networks in implant sites, without any therapeutic treatment. Microchannel networks are generated in a gelatin hydrogel to overcome the diffusion limit of nutrients and oxygen three-dimensionally. Hydrogel implantation in mouse and porcine models of hindlimb ischemia rescues severely damaged tissues by the ingrowth of neighboring host vessels with microchannel perfusion. This effect is guided by microchannel size-specific regenerative macrophage polarization with the consequent functional recovery of endothelial cells. Multiple-site implantation reveals hypoxia and neighboring vessels as major causative factors of the beneficial function. This technique may contribute to the development of therapeutics for hypoxia/inflammatory-related diseases.
Collapse
Affiliation(s)
- Jung Bok Lee
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Dae-Hyun Kim
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jeong-Kee Yoon
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Dan Bi Park
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hye-Seon Kim
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Young Min Shin
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Wooyeol Baek
- Department of Plastic & Reconstructive Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Mi-Lan Kang
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- TMD LAB Co. Ltd., 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hyun Jung Kim
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Hak-Joon Sung
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
110
|
Yamamoto K, Nishimura R, Kato F, Naito A, Suda R, Sekine A, Jujo T, Shigeta A, Sakao S, Tanabe N, Tatsumi K. Protective role of endothelial progenitor cells stimulated by riociguat in chronic thromboembolic pulmonary hypertension. Int J Cardiol 2020; 299:263-270. [DOI: 10.1016/j.ijcard.2019.07.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 06/06/2019] [Accepted: 07/04/2019] [Indexed: 12/18/2022]
|
111
|
Gouveia-Fernandes S. Monocytes and Macrophages in Cancer: Unsuspected Roles. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1219:161-185. [PMID: 32130699 DOI: 10.1007/978-3-030-34025-4_9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The behavior of cancer is undoubtedly affected by stroma. Macrophages belong to this microenvironment and their presence correlates with reduced survival in most cancers. After a tumor-induced "immunoediting", these monocytes/macrophages, originally the first line of defense against tumor cells, undergo a phenotypic switch and become tumor-supportive and immunosuppressive.The influence of these tumor-associated macrophages (TAMs) on cancer is present in all traits of carcinogenesis. These cells participate in tumor initiation and growth, migration, vascularization, invasion and metastasis. Although metastasis is extremely clinically relevant, this step is always reliant on the angiogenic ability of tumors. Therefore, the formation of new blood vessels in tumors assumes particular importance as a limiting step for disease progression.Herein, the once unsuspected roles of macrophages in cancer will be discussed and their importance as a promising strategy to treat this group of diseases will be reminded.
Collapse
Affiliation(s)
- Sofia Gouveia-Fernandes
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School | Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
112
|
Amirsadeghi A, Jafari A, Eggermont LJ, Hashemi SS, Bencherif SA, Khorram M. Vascularization strategies for skin tissue engineering. Biomater Sci 2020; 8:4073-4094. [DOI: 10.1039/d0bm00266f] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Lack of proper vascularization after skin trauma causes delayed wound healing. This has sparked the development of various tissue engineering strategies to improve vascularization.
Collapse
Affiliation(s)
- Armin Amirsadeghi
- Department of Chemical Engineering
- School of Chemical and Petroleum Engineering
- Shiraz University
- Shiraz 71348-51154
- Iran
| | - Arman Jafari
- Department of Chemical Engineering
- School of Chemical and Petroleum Engineering
- Shiraz University
- Shiraz 71348-51154
- Iran
| | | | - Seyedeh-Sara Hashemi
- Burn & Wound Healing Research Center
- Shiraz University of Medical Science
- Shiraz 71345-1978
- Iran
| | - Sidi A. Bencherif
- Department of Chemical Engineering
- Northeastern University
- Boston
- USA
- Department of Bioengineering
| | - Mohammad Khorram
- Department of Chemical Engineering
- School of Chemical and Petroleum Engineering
- Shiraz University
- Shiraz 71348-51154
- Iran
| |
Collapse
|
113
|
Qu C, Brohlin M, Kingham PJ, Kelk P. Evaluation of growth, stemness, and angiogenic properties of dental pulp stem cells cultured in cGMP xeno-/serum-free medium. Cell Tissue Res 2019; 380:93-105. [PMID: 31889209 DOI: 10.1007/s00441-019-03160-1] [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] [Received: 01/22/2019] [Accepted: 12/12/2019] [Indexed: 12/15/2022]
Abstract
This study was aimed to investigate the effects of cGMP xeno-/serum-free medium (XSF, Irvine Scientific) on the properties of human dental pulp stem cells (DPSCs). DPSCs, from passage 2, were cultured in XSF or fetal bovine serum (FBS)-supplemented medium, and sub-cultured up to passage 8. Cumulative population doublings (PDs) and the number of colony-forming-units (CFUs) were determined. qRT-PCR, ELISA, and in vitro assays were used to assess angiogenic capacity. Flow cytometry was used to measure CD73, CD90, and CD105 expression. Differentiation into osteo-, adipo-, and chondrogenic cell lineages was performed. DPSCs showed more elongated morphology, a reduced rate of proliferation at later passages, and lower CFU counts in XSF compared with FBS. Expression of angiogenic factors at the gene and protein levels varied in the two media and with passage number, but cells grown in XSF had more in vitro angiogenic activity. The majority of early and late passage DPSCs cultured in XSF expressed CD73 and CD90. In contrast, the percentage of CD105 positive DPSCs in XSF medium was significantly lower with increased passage whereas the majority of cells cultured in FBS were CD105 positive. Switching XSF-cultured DPSCs to medium supplemented with human serum restored the expression of CD105. The tri-lineage differentiation of DPSCs cultured under XSF and FBS conditions was similar. We showed that despite reduced CD105 expression levels, DPSCs expanded in XSF medium maintained a functional MSC phenotype. Furthermore, restoration of CD105 expression is likely to occur upon in vivo transplantation, when cells are exposed to human serum.
Collapse
Affiliation(s)
- Chengjuan Qu
- Department of Integrative Medical Biology, Umeå University, 901 87, Umeå, Sweden
| | - Maria Brohlin
- Department of Clinical Microbiology, Infection and Immunology, Umeå University, 901 87, Umeå, Sweden.,Division of Clinical Immunology and Transfusion Medicine, Tissue Establishment, Cell Therapy Unit, Department of Laboratory Medicine, Umeå University Hospital, Daniel Naezéns väg, 907 37, Umeå, Sweden
| | - Paul J Kingham
- Department of Integrative Medical Biology, Umeå University, 901 87, Umeå, Sweden
| | - Peyman Kelk
- Department of Integrative Medical Biology, Umeå University, 901 87, Umeå, Sweden.
| |
Collapse
|
114
|
Haider N, Boscá L, Zandbergen HR, Kovacic JC, Narula N, González-Ramos S, Fernandez-Velasco M, Agrawal S, Paz-García M, Gupta S, DeLeon-Pennell K, Fuster V, Ibañez B, Narula J. Transition of Macrophages to Fibroblast-Like Cells in Healing Myocardial Infarction. J Am Coll Cardiol 2019; 74:3124-3135. [PMID: 31856969 PMCID: PMC7425814 DOI: 10.1016/j.jacc.2019.10.036] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 10/06/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Macrophages and fibroblasts are 2 major cell types involved in healing after myocardial infarction (MI), contributing to myocardial remodeling and fibrosis. Post-MI fibrosis progression is characterized by a decrease in cardiac macrophage content. OBJECTIVES This study explores the potential of macrophages to express fibroblast genes and the direct role of these cells in post-MI cardiac fibrosis. METHODS Prolonged in vitro culture of human macrophages was used to evaluate the capacity to express fibroblast markers. Infiltrating cardiac macrophages was tracked in vivo after experimental MI of LysM(Cre/+);ROSA26(EYFP/+) transgenic mice. The expression of Yellow Fluorescent Protein (YFP) in these animals is restricted to myeloid lineage allowing the identification of macrophage-derived fibroblasts. The expression in YFP-positive cells of fibroblast markers was determined in myocardial tissue sections of hearts from these mice after MI. RESULTS Expression of the fibroblast markers type I collagen, prolyl-4-hydroxylase, fibroblast specific protein-1, and fibroblast activation protein was evidenced in YFP-positive cells in the heart after MI. The presence of fibroblasts after MI was evaluated in the hearts of animals after depletion of macrophages with clodronate liposomes. This macrophage depletion significantly reduced the number of Mac3+Col1A1+ cells in the heart after MI. CONCLUSIONS The data provide both in vitro and in vivo evidence for the ability of macrophages to transition and adopt a fibroblast-like phenotype. Therapeutic manipulation of this macrophage-fibroblast transition may hold promise for favorably modulating the fibrotic response after MI and after other cardiovascular pathological processes.
Collapse
Affiliation(s)
- Nezam Haider
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Division of Vascular Surgery, University of Arizona, Tucson, Arizona
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Melchor Fernández Almagro, Madrid, Spain.
| | - H Reinier Zandbergen
- Department of Cardiothoracic Surgery, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Jason C Kovacic
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Navneet Narula
- Department of Pathology, New York University Langone Medical Center, New York, New York
| | - Silvia González-Ramos
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Melchor Fernández Almagro, Madrid, Spain
| | - María Fernandez-Velasco
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Melchor Fernández Almagro, Madrid, Spain; Instituto de Investigación Biomédica LaPaz, Paseo de la Castellana, Madrid, Spain
| | - Sudhanshu Agrawal
- Division of Basic and Clinical Immunology, University of California, Irvine, California
| | - Marta Paz-García
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, Madrid, Spain
| | - Sudhir Gupta
- Division of Basic and Clinical Immunology, University of California, Irvine, California
| | - Kristine DeLeon-Pennell
- Division of Cardiology, Medical University of South Carolina, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Valentin Fuster
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Borja Ibañez
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Melchor Fernández Almagro, Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Madrid, Spain. https://twitter.com/Borjaibanez1
| | - Jagat Narula
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
115
|
Ota Y, Kuwana M. Endothelial cells and endothelial progenitor cells in the pathogenesis of systemic sclerosis. Eur J Rheumatol 2019; 7:S139-S146. [PMID: 31922471 DOI: 10.5152/eurjrheum.2019.19158] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 11/25/2019] [Indexed: 12/27/2022] Open
Abstract
Systemic sclerosis (SSc) is a connective tissue disease characterized by excessive fibrosis, microvasculopathy, and autoimmunity. Endothelial cell (EC) injury and subsequent endothelial cell dysfunction is believed to be an initial event that eventually leads to a vicious pathogenic cycle. This process is further enhanced by defective angiogenesis and vasculogenesis, as the vascular repair machinery does not work properly. Endothelial progenitor cells (EPCs) are functionally and quantitatively insufficient to recover the endothelium in SSc patients. The dysfunctional ECs and EPCs not only trigger the formation of typical vascular lesions, such as progressive intimal fibrosis in small arteries and the loss of capillaries, but also promote a series of inflammatory and profibrotic processes, such as endothelial-mesenchymal transition and recruitment and accumulation of monocytic EPCs with profibrotic properties. These processes together contribute to the accumulation of extracellular matrix in the affected tissue. This review features current insights into the roles of ECs and EPCs in the pathogenesis of SSc.
Collapse
Affiliation(s)
- Yuko Ota
- Department of Allergy and Rheumatology, Nippon Medical School Graduate School of Medicine, Tokyo, Japan
| | - Masataka Kuwana
- Department of Allergy and Rheumatology, Nippon Medical School Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
116
|
Coalson E, Bishop E, Liu W, Feng Y, Spezia M, Liu B, Shen Y, Wu D, Du S, Li AJ, Ye Z, Zhao L, Cao D, Li A, Hagag O, Deng A, Liu W, Li M, Haydon RC, Shi L, Athiviraham A, Lee MJ, Wolf JM, Ameer GA, He TC, Reid RR. Stem cell therapy for chronic skin wounds in the era of personalized medicine: From bench to bedside. Genes Dis 2019; 6:342-358. [PMID: 31832514 PMCID: PMC6888708 DOI: 10.1016/j.gendis.2019.09.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 09/07/2019] [Accepted: 09/09/2019] [Indexed: 02/06/2023] Open
Abstract
With the significant financial burden of chronic cutaneous wounds on the healthcare system, not to the personal burden mention on those individuals afflicted, it has become increasingly essential to improve our clinical treatments. This requires the translation of the most recent benchtop approaches to clinical wound repair as our current treatment modalities have proven insufficient. The most promising potential treatment options rely on stem cell-based therapies. Stem cell proliferation and signaling play crucial roles in every phase of the wound healing process and chronic wounds are often associated with impaired stem cell function. Clinical approaches involving stem cells could thus be utilized in some cases to improve a body's inhibited healing capacity. We aim to present the laboratory research behind the mechanisms and effects of this technology as well as current clinical trials which showcase their therapeutic potential. Given the current problems and complications presented by chronic wounds, we hope to show that developing the clinical applications of stem cell therapies is the rational next step in improving wound care.
Collapse
Affiliation(s)
- Elam Coalson
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Elliot Bishop
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Surgery, Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Wei Liu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Key Laboratory of Diagnostic Medicine (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Yixiao Feng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Key Laboratory of Diagnostic Medicine (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Mia Spezia
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Bo Liu
- Department of Surgery, Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Key Laboratory of Diagnostic Medicine (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Yi Shen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery, Xiangya Second Hospital of Central South University, Changsha 410011, China
| | - Di Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Scott Du
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Student Inquiry Research Program, Illinois Mathematics and Science Academy (IMSA), Aurora, IL 60506, USA
| | - Alexander J. Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Zhenyu Ye
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Ling Zhao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Key Laboratory of Diagnostic Medicine (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Daigui Cao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery, Chongqing General Hospital, Chongqing 400013, China
| | - Alissa Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Ofir Hagag
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Alison Deng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Student Inquiry Research Program, Illinois Mathematics and Science Academy (IMSA), Aurora, IL 60506, USA
| | - Winny Liu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Student Inquiry Research Program, Illinois Mathematics and Science Academy (IMSA), Aurora, IL 60506, USA
| | - Mingyang Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Student Inquiry Research Program, Illinois Mathematics and Science Academy (IMSA), Aurora, IL 60506, USA
| | - Rex C. Haydon
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Lewis Shi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Aravind Athiviraham
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Michael J. Lee
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jennifer Moriatis Wolf
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Guillermo A. Ameer
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60616, USA
- Center for Advanced Regenerative Engineering (CARE), Evanston, IL 60208, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Center for Advanced Regenerative Engineering (CARE), Evanston, IL 60208, USA
| | - Russell R. Reid
- Department of Surgery, Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Center for Advanced Regenerative Engineering (CARE), Evanston, IL 60208, USA
| |
Collapse
|
117
|
Pinheiro D, Dias I, Ribeiro Silva K, Stumbo AC, Thole A, Cortez E, de Carvalho L, Weiskirchen R, Carvalho S. Mechanisms Underlying Cell Therapy in Liver Fibrosis: An Overview. Cells 2019; 8:cells8111339. [PMID: 31671842 PMCID: PMC6912561 DOI: 10.3390/cells8111339] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 12/19/2022] Open
Abstract
Fibrosis is a common feature in most pathogenetic processes in the liver, and usually results from a chronic insult that depletes the regenerative capacity of hepatocytes and activates multiple inflammatory pathways, recruiting resident and circulating immune cells, endothelial cells, non-parenchymal hepatic stellate cells, and fibroblasts, which become activated and lead to excessive extracellular matrix accumulation. The ongoing development of liver fibrosis results in a clinically silent and progressive loss of hepatocyte function, demanding the constant need for liver transplantation in clinical practice, and motivating the search for other treatments as the chances of obtaining compatible viable livers become scarcer. Although initially cell therapy has emerged as a plausible alternative to organ transplantation, many factors still challenge the establishment of this technique as a main or even additional therapeutic tool. Herein, the authors discuss the most recent advances and point out the corners and some controversies over several protocols and models that have shown promising results as potential candidates for cell therapy for liver fibrosis, presenting the respective mechanisms proposed for liver regeneration in each case.
Collapse
Affiliation(s)
- Daphne Pinheiro
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro 20550-170, Brazil.
| | - Isabelle Dias
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro 20550-170, Brazil.
| | - Karina Ribeiro Silva
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro 20550-170, Brazil.
| | - Ana Carolina Stumbo
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro 20550-170, Brazil.
| | - Alessandra Thole
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro 20550-170, Brazil.
| | - Erika Cortez
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro 20550-170, Brazil.
| | - Lais de Carvalho
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro 20550-170, Brazil.
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, D-52074 Aachen, Germany.
| | - Simone Carvalho
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro 20550-170, Brazil.
| |
Collapse
|
118
|
Kaushik K, Das A. Endothelial progenitor cell therapy for chronic wound tissue regeneration. Cytotherapy 2019; 21:1137-1150. [PMID: 31668487 DOI: 10.1016/j.jcyt.2019.09.002] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/20/2019] [Accepted: 09/24/2019] [Indexed: 02/07/2023]
Abstract
Despite advancements in wound care, healing of chronic diabetic wounds remains a great challenge for the clinical fraternity because of the intricacies of the healing process. Due to the limitations of existing treatment strategies for chronic wounds, stem/progenitor cell transplantation therapies have been explored as an alternative for tissue regeneration at the wound site. The non-healing phenotype of chronic wounds is directly associated with lack of vascularization. Therefore, endothelial progenitor cell (EPC) transplantation is proving to be a promising approach for the treatment of hypo-vascular chronic wounds. With the existing knowledge in EPC biology, significant efforts have been made to enrich EPCs at the chronic wound site, generating EPCs from somatic cells, induced pluripotent stem cells (iPSCs) using transcription factors, or from adult stem cells using chemicals/drugs for use in transplantation, as well as modulating the endogenous dysfunctional/compromised EPCs under diabetic conditions. This review mainly focuses on the pre-clinical and clinical approaches undertaken to date with EPC-based translational therapy for chronic diabetic as well as non-diabetic wounds to evaluate their vascularity-mediated regeneration potential.
Collapse
Affiliation(s)
- Komal Kaushik
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-IICT Campus, Hyderabad, India
| | - Amitava Das
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-IICT Campus, Hyderabad, India.
| |
Collapse
|
119
|
Jiang P, Tang X, Wang H, Dai C, Su J, Zhu H, Song M, Liu J, Nan Z, Ru T, Li Y, Wang J, Yang J, Chen B, Dai J, Hu Y. Collagen-binding basic fibroblast growth factor improves functional remodeling of scarred endometrium in uterine infertile women: a pilot study. SCIENCE CHINA-LIFE SCIENCES 2019; 62:1617-1629. [PMID: 31515729 DOI: 10.1007/s11427-018-9520-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 03/05/2019] [Indexed: 12/26/2022]
Abstract
Intrauterine adhesion (IUA) is a common cause of uterine infertility and one of the most severe clinical features is endometrial fibrosis namely endometrial scarring for which there are few cures currently. Blocked angiogenesis is the main pathological change in the scarred endometrium. The fibroblast growth factor 2 (bFGF), a member of FGF family, is usually applied to promote healing of refractory ulcer and contributes to angiogenesis of tissues. In this study, the sustained-release system of bFGF 100 µg was administrated around scarred endometrium guiding by ultrasound every 4 weeks in 18 patients (2-4 times). Results showed that after treatment, the menstrual blood volume, endometrial thickness and the scarred endometrial area were improved. Histological study showed blood vessel density increased obviously. Three patients (3/18) achieved pregnancy over 20 gestational weeks. Therefore, administrating the bFGF surrounding scarred endometrium may provide a new therapeutic approach for the patients with endometrial fibrosis.
Collapse
Affiliation(s)
- Peipei Jiang
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Xiaoqiu Tang
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Huiyan Wang
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Chenyan Dai
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Jing Su
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Hui Zhu
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Minmin Song
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Jingyu Liu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Ziqing Nan
- Nanjing Drum Tower Hospital, Chinese Academy of Medical Science & Peking Union Medical Collage, Nanjing, 210008, China
| | - Tong Ru
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Yaling Li
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Jingmei Wang
- Department of Pathology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Jun Yang
- Department of Pathology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Bing Chen
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100190, China
| | - Jianwu Dai
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100190, China.
| | - Yali Hu
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| |
Collapse
|
120
|
Lin Y, Gil CH, Yoder MC. Identification of Circulating Endothelial Colony-Forming Cells from Murine Embryonic Peripheral Blood. Methods Mol Biol 2019; 1940:97-107. [PMID: 30788820 DOI: 10.1007/978-1-4939-9086-3_7] [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: 03/08/2023]
Abstract
Human umbilical cord blood contains highly proliferative circulating endothelial colony-forming cells (ECFC). These cells have promising therapeutic potential for various cardiovascular diseases by possessing robust in vitro clonal expansion potential and the ability to form functional blood vessels in vivo upon transplantation into recipient immunodeficient mice. However whether similar cells also exist in murine blood remains unresolved, which impedes the study of circulating ECFC biology using murine models. Here we describe a method to identify and culture murine embryonic peripheral blood-derived circulating ECFC through co-culture with OP9 stromal cells. Using this method, embryonic circulating ECFC can be identified by the formation of sheet-like or network-like endothelial colonies upon OP9 stromal cell monolayers.
Collapse
Affiliation(s)
- Yang Lin
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Chang-Hyun Gil
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mervin C Yoder
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA. .,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
121
|
Preliminary Study: Purple Sweet Potato Extract Seems to Be Superior to Increase the Migration of Impaired Endothelial Progenitor Cells Compared to l-Ascorbic Acid. Sci Pharm 2019. [DOI: 10.3390/scipharm87030016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Impairment of the endothelial progenitor cells (EPCs) ability to proliferate and migrate in the patients with coronary heart disease (CHD) is partly caused by oxidative stress. This research evaluates the effect of treatment with Ipomoea batatas L./purple sweet potato (PSP) extract and l-ascorbic acid on the proliferation and migration of impaired EPCs. EPCs were isolated from CHD patient’s peripheral blood. EPCs culture were cultivated and divided into control (untreated), PSP extract treatment (dose 1 and 25 μg/mL), and l-ascorbic acid treatment (dose 10 and 250 μg/mL) groups for 48 h. EPCs proliferation was analyzed with the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) cell proliferation assay, and migration was evaluated with the cell migration assay kit. Statistical tests were evaluated using SPSS 25.0. This research showed that EPCs proliferation and migration was significantly higher in all PSP extract and l-ascorbic acid treatment compared to the control (p < 0.001). EPCs migration on treatment with a PSP extract dose of 25 μg/mL was significantly higher compared to the treatment with l-ascorbic acid dose of 250 μg/mL (303,000 ± 1000 compared to 215,000 ± 3000 cells, p< 0.001). In conclusion, both treatments with PSP extract and l-ascorbic acid can improve the proliferation and migration of impaired EPCs. At the dose of 25 μg/mL, PSP extract seems to be superior to the l-ascorbic acid dose of 250 μg/mL to improve EPCs migration.
Collapse
|
122
|
Ex vivo expansion of cord blood-derived endothelial cells using a novel xeno-free culture media. Future Sci OA 2019; 5:FSO376. [PMID: 31245040 PMCID: PMC6554691 DOI: 10.2144/fsoa-2018-0103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 01/09/2019] [Indexed: 01/06/2023] Open
Abstract
Aim Endothelial cells (ECs), isolated from peripheral blood (PB), bone marrow (BM) and cord blood (CB), are limited in numbers and expansion has had limited success. We used a novel serum-free medium (EndoGo) to evaluate effects on ex vivo expansion of CB-derived ECs. Materials & methods Flow cytometry and matrigel were used to determine expansion of ECs and for determination of the EC progenitor cell. Results EndoGo™-containing cultures demonstrated superior expansion and stimulated proliferation of two distinct subpopulations, CD34+CD31+ and CD34-CD31+, which exhibited different morphology, phenotype and function. EndoGo also expanded the CB endothelial progenitor cells from freshly isolated CB. Conclusion These findings demonstrate the potential of EndoGo to expand CB ECs, which could generate increased numbers of ECs for therapeutic applications.
Collapse
|
123
|
Abstract
The ability to generate new microvessels in desired numbers and at desired locations has been a long-sought goal in vascular medicine, engineering, and biology. Historically, the need to revascularize ischemic tissues nonsurgically (so-called therapeutic vascularization) served as the main driving force for the development of new methods of vascular growth. More recently, vascularization of engineered tissues and the generation of vascularized microphysiological systems have provided additional targets for these methods, and have required adaptation of therapeutic vascularization to biomaterial scaffolds and to microscale devices. Three complementary strategies have been investigated to engineer microvasculature: angiogenesis (the sprouting of existing vessels), vasculogenesis (the coalescence of adult or progenitor cells into vessels), and microfluidics (the vascularization of scaffolds that possess the open geometry of microvascular networks). Over the past several decades, vascularization techniques have grown tremendously in sophistication, from the crude implantation of arteries into myocardial tunnels by Vineberg in the 1940s, to the current use of micropatterning techniques to control the exact shape and placement of vessels within a scaffold. This review provides a broad historical view of methods to engineer the microvasculature, and offers a common framework for organizing and analyzing the numerous studies in this area of tissue engineering and regenerative medicine. © 2019 American Physiological Society. Compr Physiol 9:1155-1212, 2019.
Collapse
Affiliation(s)
- Joe Tien
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
- Division of Materials Science and Engineering, Boston University, Brookline, Massachusetts, USA
| |
Collapse
|
124
|
Zhao H, Mao J, Yuan Y, Feng J, Cheng H, Fan G, Zhang Y, Li T. Sodium Dichloroacetate Stimulates Angiogenesis by Improving Endothelial Precursor Cell Function in an AKT/GSK-3β/Nrf2 Dependent Pathway in Vascular Dementia Rats. Front Pharmacol 2019; 10:523. [PMID: 31156438 PMCID: PMC6533549 DOI: 10.3389/fphar.2019.00523] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 04/25/2019] [Indexed: 01/25/2023] Open
Abstract
Sodium dichloroacetate (DCA) is a mitochondrial pyruvate dehydrogenase kinase inhibitor, and has been shown to display vasoprotective effects in chronic ischemic stroke. The purpose of this study was to evaluate the therapeutic effect of DCA on vascular dementia (VD) and endothelial progenitor cell (EPC)-mediated angiogenesis. After cerebral ischemia-reperfusion in rats, DCA was administered continuously for 21 days; following which, histological analysis, and cognitive functional tests were conducted. Rat bone marrow-derived EPCs were isolated, their function and quantity were measured, and the effects of long-term administration of DCA on EPCs in a rat model of VD was studied. We found that long-term DCA administration improved cognitive function in VD rats, reduced brain infarct size and brain atrophy, increased VEGF and bFGF levels in vivo, promoted angiogenesis in damaged areas, and significantly improved EPC function in VD rats. Compared with the VD group, AKT, Nrf2, eNOS expression, and intracellular NO levels were elevated in EPCs of DCA-treated VD rats. In addition, GSK3β and intracellular ROS levels were decreased. Simultaneously, it was found that DCA directly acted on EPCs, and improved EPC functional behavior. Taken together, these findings suggested that long-term DCA administration improved cognitive function in a rat model of VD, and did so in part, by improving EPC function. Observations suggest that prolonged DCA administration might be beneficial in treating VD.
Collapse
Affiliation(s)
- Hui Zhao
- Department of Pharmacy, Punan Hospital, Shanghai, China.,College of Pharmacology, Anhui University of Chinese Medicine, Hefei, China
| | - Junqin Mao
- Department of Pharmacy, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Yuan Yuan
- College of Pharmacology, Anhui University of Chinese Medicine, Hefei, China
| | - Jingjing Feng
- College of Pharmacology, Anhui University of Chinese Medicine, Hefei, China
| | - Hao Cheng
- College of Pharmacology, Anhui University of Chinese Medicine, Hefei, China
| | - Guorong Fan
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yuefan Zhang
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China.,Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai, China
| | - Tiejun Li
- Department of Pharmacy, Punan Hospital, Shanghai, China.,College of Pharmacology, Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
125
|
Identification of Macrophage Genotype and Key Biological Pathways in Circulating Angiogenic Cell Transcriptome. Stem Cells Int 2019; 2019:9545261. [PMID: 31191690 PMCID: PMC6525806 DOI: 10.1155/2019/9545261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 01/14/2019] [Accepted: 02/13/2019] [Indexed: 11/17/2022] Open
Abstract
Background Circulating angiogenic cells (CAC) have been identified as important regulators of vascular biology. However, there is still considerable debate about the genotype and function of CAC. Methods and Results Data from publicly available gene expression data sets were used to analyse the transcriptome of in vitro cultured CAC (CACiv). Genes and pathways of interest were further evaluated using qPCR comparing CACiv versus CD14+ monocytic cells. The CACiv transcriptome strongly related to tissue macrophages, and more specifically to regulatory M2c macrophages. The cytokine expression profile of CACiv was predominantly immune modulatory and resembled the cytokine expression of tumor-associated macrophages (TAM). Pathway analysis revealed previously unrecognized biological processes in CACiv, such as riboflavin metabolism and liver X receptor (LXR)/retinoid X receptor (RXR) and farnesoid X receptor (FXR)/retinoid X receptor (RXR) pathways. Analysis of endothelial-specific genes did not show evidence for endothelial transdifferentiation. Conclusions CACiv are genotypically similar to regulatory M2c macrophages and lack signs of endothelial differentiation.
Collapse
|
126
|
Asadian S, Alibabrdel M, Daei N, Cheraghi H, Maedeh Jafari S, Noshadirad E, Jabarpour M, Siavashi V, Nassiri SM. Improved angiogenic activity of endothelial progenitor cell in diabetic patients treated with insulin plus metformin. J Cell Biochem 2019; 120:7115-7124. [PMID: 30378162 DOI: 10.1002/jcb.27985] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/08/2018] [Indexed: 01/24/2023]
Abstract
Type 2 diabetes (T2DM) is associated with an increased vascular disease. Moreover, endothelial progenitor cell (EPC) function is impaired in diabetic patients. Decreased EPC number plays a critical role in reduced endothelial repair and development of the vascular disorder. To determine the effect of metformin and insulin plus metformin on functional activity of EPCs, 130 participants were divided into three groups (group 1: healthy control; group 2: metformin; group 3: insulin plus metformin). The concentration of EPCs in the circulation was first quantified. Thereafter, circulating EPCs (cEPCs) were harvested and the biological features of these cells including proliferative, clonogenicity, tubulogenic, and migratory properties were analyzed after expansion. The serum protein levels of some proangiogenic factors were also measured. Our results showed greater numbers of cEPCs in control and in diabetic patients treated with insulin plus metformin than in metformin-treated patients. Insulin plus metformin therapy was associated with augmented proliferative, clonogenicity, migratory, and tubulogenic activity of cEPCs in patients with T2DM. Increased serum concentrations of angiogenic factors were also observed in patients treated with insulin plus metformin. Western blot analysis showed increased protein levels of pTie-2/Tie2 and Pakt/AKT in cEPCs harvested from T2DM, treated with insulin metformin plus. This study showed that treatment with insulin plus metformin in diabetic patients is associated with increased mobilization of EPCs into the circulation, with potential beneficial effect in vascular protection in diabetic patients.
Collapse
Affiliation(s)
- Simin Asadian
- Department of Pediatrics, Imam Reza Hospital, Kermanshah University of Medical Science, Kermanshah, Iran
| | - Mahdi Alibabrdel
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Nazanin Daei
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Hadi Cheraghi
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Seyedeh Maedeh Jafari
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Elnaz Noshadirad
- Department of Biology, Science and Research Branch´, Islamic Azad University, Tehran, Iran
| | - Masoome Jabarpour
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Vahid Siavashi
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Seyed Mahdi Nassiri
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
127
|
Lee SH, Ra JC, Oh HJ, Kim MJ, Setyawan EMN, Choi YB, Yang JW, Kang SK, Han SH, Kim GA, Lee BC. Clinical Assessment of Intravenous Endothelial Progenitor Cell Transplantation in Dogs. Cell Transplant 2019; 28:943-954. [PMID: 31018670 PMCID: PMC6719494 DOI: 10.1177/0963689718821686] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Endothelial progenitor cells (EPCs) have been applied for cell therapy because of their roles in angiogenesis and neovascularization in ischemic tissue. However, adverse responses caused by EPC therapy have not been fully investigated. In this study, a human peripheral blood sample was collected from a healthy donor and peripheral blood mononuclear cells were separated using Ficoll-Hypaque. There were four experimental groups: 10 ml saline infusion group (injection rate; 3 ml/min), 10 ml saline bolus group (injection rate; 60 ml/min), 10 ml EPCs infusion group (2 x 105 cells/ml, injection rate; 3 ml/min), 10 ml EPCs bolus group (2 × 105 cells/ml, injection rate; 60 ml/min). Clinical assessment included physical examination and laboratory examination for intravenous human EPC transplantation in dogs. The results revealed no remarkable findings in vital signs among the dogs used. In blood analysis, platelet counts in saline infusion groups were significantly higher than in the EPC groups within normal ranges, and no significant differences were observed except K+, Cl- and blood urea nitrogen/urea. In ELISA assay, no significant difference was observed in serum tumor necrosis factor alpha. The serum concentration of vascular endothelial growth factor was significantly higher in EPC groups than in saline groups, and interleukin 10 was significantly up-regulated in the EPC infusion group compared with other groups. In conclusion, we demonstrated that no clinical abnormalities were detected after intravenous transplantation of human EPCs in dogs. The transplanted xenogenic EPCs might be involved in anti-inflammatory and angiogenic functions in dogs.
Collapse
Affiliation(s)
- Seok Hee Lee
- 1 Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Republic of Korea
| | - Jeong Chan Ra
- 2 Biostar Stem Cell Research Institute, R Bio Co., Seoul, Republic of Korea
| | - Hyun Ju Oh
- 1 Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Republic of Korea
| | - Min Jung Kim
- 1 Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Republic of Korea
| | - Erif Maha Nugraha Setyawan
- 1 Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Republic of Korea
| | - Yoo Bin Choi
- 1 Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Republic of Korea
| | - Jung Won Yang
- 2 Biostar Stem Cell Research Institute, R Bio Co., Seoul, Republic of Korea
| | - Sung Keun Kang
- 2 Biostar Stem Cell Research Institute, R Bio Co., Seoul, Republic of Korea
| | - Seung Hyup Han
- 2 Biostar Stem Cell Research Institute, R Bio Co., Seoul, Republic of Korea
| | - Geon A Kim
- 2 Biostar Stem Cell Research Institute, R Bio Co., Seoul, Republic of Korea
| | - Byeong Chun Lee
- 1 Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Republic of Korea
| |
Collapse
|
128
|
Ex vivoexpansion of cord blood-derived endothelial cells using a novel xeno-free culture media. Future Sci OA 2019. [DOI: 10.4155/fsoa-2018-0103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
129
|
Wernly B, Mirna M, Rezar R, Prodinger C, Jung C, Podesser BK, Kiss A, Hoppe UC, Lichtenauer M. Regenerative Cardiovascular Therapies: Stem Cells and Beyond. Int J Mol Sci 2019; 20:E1420. [PMID: 30901815 PMCID: PMC6470623 DOI: 10.3390/ijms20061420] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/17/2019] [Accepted: 03/19/2019] [Indexed: 12/20/2022] Open
Abstract
Although reperfusion therapy has improved outcomes, acute myocardial infarction (AMI) is still associated with both significant mortality and morbidity. Once irreversible myocardial cell death due to ischemia and reperfusion sets in, scarring leads to reduction in left ventricular function and subsequent heart failure. Regenerative cardiovascular medicine experienced a boost in the early 2000s when regenerative effects of bone marrow stem cells in a murine model of AMI were described. Translation from an animal model to stem cell application in a clinical setting was rapid and the first large trials in humans suffering from AMI were conducted. However, high initial hopes were early shattered by inconsistent results of randomized clinical trials in patients suffering from AMI treated with stem cells. Hence, we provide an overview of both basic science and clinical trials carried out in regenerative cardiovascular therapies. Possible pitfalls in specific cell processing techniques and trial design are discussed as these factors influence both basic science and clinical outcomes. We address possible solutions. Alternative mechanisms and explanations for effects seen in both basic science and some clinical trials are discussed here, with special emphasis on paracrine mechanisms via growth factors, exosomes, and microRNAs. Based on these findings, we propose an outlook in which stem cell therapy, or therapeutic effects associated with stem cell therapy, such as paracrine mechanisms, might play an important role in the future. Optimizing stem cell processing and a better understanding of paracrine signaling as well as its effect on cardioprotection and remodeling after AMI might improve not only AMI research, but also our patients' outcomes.
Collapse
Affiliation(s)
- Bernhard Wernly
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria.
| | - Moritz Mirna
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria.
| | - Richard Rezar
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria.
| | - Christine Prodinger
- Department of Dermatology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria.
| | - Christian Jung
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, University of Düsseldorf, 40225 Düsseldorf, Germany.
| | - Bruno K Podesser
- Ludwig Boltzmann Cluster for Cardiovascular Research, Center for Biomedical Research, Medical University Vienna, 1090 Vienna, Austria.
| | - Attila Kiss
- Ludwig Boltzmann Cluster for Cardiovascular Research, Center for Biomedical Research, Medical University Vienna, 1090 Vienna, Austria.
| | - Uta C Hoppe
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria.
| | - Michael Lichtenauer
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria.
| |
Collapse
|
130
|
Landers-Ramos RQ, Sapp RM, Shill DD, Hagberg JM, Prior SJ. Exercise and Cardiovascular Progenitor Cells. Compr Physiol 2019; 9:767-797. [PMID: 30892694 DOI: 10.1002/cphy.c180030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Autologous stem/progenitor cell-based methods to restore blood flow and function to ischemic tissues are clinically appealing for the substantial proportion of the population with cardiovascular diseases. Early preclinical and case studies established the therapeutic potential of autologous cell therapies for neovascularization in ischemic tissues. However, trials over the past ∼15 years reveal the benefits of such therapies to be much smaller than originally estimated and a definitive clinical benefit is yet to be established. Recently, there has been an emphasis on improving the number and function of cells [herein generally referred to as circulating angiogenic cells (CACs)] used for autologous cell therapies. CACs include of several subsets of circulating cells, including endothelial progenitor cells, with proangiogenic potential that is largely exerted through paracrine functions. As exercise is known to improve CV outcomes such as angiogenesis and endothelial function, much attention is being given to exercise to improve the number and function of CACs. Accordingly, there is a growing body of evidence that acute, short-term, and chronic exercise have beneficial effects on the number and function of different subsets of CACs. In particular, recent studies show that aerobic exercise training can increase the number of CACs in circulation and enhance the function of isolated CACs as assessed in ex vivo assays. This review summarizes the roles of different subsets of CACs and the effects of acute and chronic exercise on CAC number and function, with a focus on the number and paracrine function of circulating CD34+ cells, CD31+ cells, and CD62E+ cells. © 2019 American Physiological Society. Compr Physiol 9:767-797, 2019.
Collapse
Affiliation(s)
- Rian Q Landers-Ramos
- University of Maryland School of Public Health, Department of Kinesiology, College Park, Maryland, USA.,Education and Clinical Center, Baltimore Veterans Affairs Geriatric Research, Baltimore, Maryland, USA.,University of Maryland School of Medicine, Department of Medicine, Baltimore, Maryland, USA
| | - Ryan M Sapp
- University of Maryland School of Public Health, Department of Kinesiology, College Park, Maryland, USA
| | - Daniel D Shill
- University of Maryland School of Public Health, Department of Kinesiology, College Park, Maryland, USA
| | - James M Hagberg
- University of Maryland School of Public Health, Department of Kinesiology, College Park, Maryland, USA
| | - Steven J Prior
- University of Maryland School of Public Health, Department of Kinesiology, College Park, Maryland, USA.,Education and Clinical Center, Baltimore Veterans Affairs Geriatric Research, Baltimore, Maryland, USA.,University of Maryland School of Medicine, Department of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
131
|
Noh M, Choi YH, An YH, Tahk D, Cho S, Yoon JW, Jeon NL, Park TH, Kim J, Hwang NS. Magnetic Nanoparticle-Embedded Hydrogel Sheet with a Groove Pattern for Wound Healing Application. ACS Biomater Sci Eng 2019; 5:3909-3921. [PMID: 33438430 DOI: 10.1021/acsbiomaterials.8b01307] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Endothelial progenitor cells (EPCs) can induce a pro-angiogenic response during tissue repair. Recently, EPC transplantations have been widely investigated in wound healing applications. To maximize the healing efficacy by EPCs, a unique scaffold design that allows cell retention and function would be desirable for in situ delivery. Herein, we fabricated an alginate/poly-l-ornithine/gelatin (alginate-PLO-gelatin) hydrogel sheet with a groove pattern for use as a cell delivery platform. In addition, we demonstrate the topographical modification of the hydrogel sheet surface with a groove pattern to modulate cell proliferation, alignment, and elongation. We report that the patterned substrate prompted morphological changes of endothelial cells, increased cell-cell interaction, and resulted in the active secretion of growth factors such as PDGF-BB. Additionally, we incorporated magnetic nanoparticles (MNPs) into the patterned hydrogel sheet for the magnetic field-induced transfer of cell-seeded hydrogel sheets. As a result, enhanced wound healing was observed via efficient transplantation of the EPCs with an MNP-embedded patterned hydrogel sheet (MPS). Finally, enhanced vascularization and dermal wound repair were observed with EPC seeded MPS.
Collapse
Affiliation(s)
- Miyeon Noh
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea.,School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Young Hwan Choi
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Young-Hyeon An
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Dongha Tahk
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul 08826, Republic of Korea.,Institute of Advanced Machinery and Design, Seoul National University, Seoul 08826, Republic of Korea
| | - Sungwoo Cho
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Jung Won Yoon
- Department of Physiology, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea
| | - Noo Li Jeon
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea.,School of Mechanical and Aerospace Engineering, Seoul National University, Seoul 08826, Republic of Korea.,Institute of Advanced Machinery and Design, Seoul National University, Seoul 08826, Republic of Korea
| | - Tai Hyun Park
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea.,School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaeho Kim
- Department of Physiology, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea
| | - Nathaniel S Hwang
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea.,School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
132
|
Whitsett JA, Kalin TV, Xu Y, Kalinichenko VV. Building and Regenerating the Lung Cell by Cell. Physiol Rev 2019; 99:513-554. [PMID: 30427276 DOI: 10.1152/physrev.00001.2018] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The unique architecture of the mammalian lung is required for adaptation to air breathing at birth and thereafter. Understanding the cellular and molecular mechanisms controlling its morphogenesis provides the framework for understanding the pathogenesis of acute and chronic lung diseases. Recent single-cell RNA sequencing data and high-resolution imaging identify the remarkable heterogeneity of pulmonary cell types and provides cell selective gene expression underlying lung development. We will address fundamental issues related to the diversity of pulmonary cells, to the formation and function of the mammalian lung, and will review recent advances regarding the cellular and molecular pathways involved in lung organogenesis. What cells form the lung in the early embryo? How are cell proliferation, migration, and differentiation regulated during lung morphogenesis? How do cells interact during lung formation and repair? How do signaling and transcriptional programs determine cell-cell interactions necessary for lung morphogenesis and function?
Collapse
Affiliation(s)
- Jeffrey A Whitsett
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| | - Tanya V Kalin
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| | - Yan Xu
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| | - Vladimir V Kalinichenko
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| |
Collapse
|
133
|
Kuschnerus K, Straessler ET, Müller MF, Lüscher TF, Landmesser U, Kränkel N. Increased Expression of miR-483-3p Impairs the Vascular Response to Injury in Type 2 Diabetes. Diabetes 2019; 68:349-360. [PMID: 30257976 DOI: 10.2337/db18-0084] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 09/13/2018] [Indexed: 11/13/2022]
Abstract
Aggravated endothelial injury and impaired endothelial repair capacity contribute to the high cardiovascular risk in patients with type 2 diabetes (T2D), but the underlying mechanisms are still incompletely understood. Here we describe the functional role of a mature form of miRNA (miR) 483-3p, which limits endothelial repair capacity in patients with T2D. Expression of human (hsa)-miR-483-3p was higher in endothelial-supportive M2-type macrophages (M2MΦs) and in the aortic wall of patients with T2D than in control subjects without diabetes. Likewise, the murine (mmu)-miR-483* was higher in T2D than in nondiabetic murine carotid samples. Overexpression of miR-483-3p increased endothelial and macrophage apoptosis and impaired reendothelialization in vitro. The inhibition of hsa-miR-483-3p in human T2D M2MΦs transplanted to athymic nude mice (NMRI-Foxn1ν/Foxn1ν ) or systemic inhibition of mmu-miR-483* in B6.BKS(D)-Leprdb /J diabetic mice rescued diabetes-associated impairment of reendothelialization in the murine carotid-injury model. We identified the endothelial transcription factor vascular endothelial zinc finger 1 (VEZF1) as a direct target of miR-483-3p. VEZF1 expression was reduced in aortae of diabetic mice and upregulated in diabetic murine aortae upon systemic inhibition of mmu-483*. The miRNA miR-483-3p is a critical regulator of endothelial integrity in patients with T2D and may represent a therapeutic target to rescue endothelial regeneration after injury in patients with T2D.
Collapse
Affiliation(s)
- Kira Kuschnerus
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Deutsches Herzzentrum Berlin, Berlin, Germany
| | - Elisabeth T Straessler
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Maja F Müller
- University Hospital Zurich, Department of Cardiology, Zürich, Switzerland
| | - Thomas F Lüscher
- University Hospital Zurich, Department of Cardiology, Zürich, Switzerland
- Center of Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Ulf Landmesser
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- University Hospital Zurich, Department of Cardiology, Zürich, Switzerland
- Center of Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Nicolle Kränkel
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Center of Molecular Cardiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
134
|
Gao W, Jiang T, Liu YH, Ding WG, Guo CC, Cui XG. Endothelial progenitor cells attenuate the lung ischemia/reperfusion injury following lung transplantation via the endothelial nitric oxide synthase pathway. J Thorac Cardiovasc Surg 2019; 157:803-814. [PMID: 30391008 DOI: 10.1016/j.jtcvs.2018.08.092] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 07/28/2018] [Accepted: 08/11/2018] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Endothelial progenitor cells (EPCs) can improve endothelial integrity. This study aimed to examine the effects and the mechanism of EPCs on lung ischemia-reperfusion injury (LIRI). METHODS Wistar rats were randomized into the sham or the left lung transplantation group. The recipients were randomized and treated with vehicle as the LIRI group, with EPC as the EPC group, or with N5-(1-iminoethyl)-l-ornithine-pretreated EPC as the EPC/L group (n = 8 per group). The ratios of arterial oxygen partial pressure to fractional inspiratory oxygen were measured. The lung wet-to-dry weight ratios, protein levels, and injury, as well as the levels of plasma cytokines, were examined. The levels of endothelin (ET)-1, endothelial nitric oxide synthase (eNOS), phosphorylated eNOS, inducible NOS, phosphorylated myosin light chain, nuclear factor-κBp65, Bax, Bcl-2, cleaved caspase-3, and myeloperoxidase in the graft lungs were detected. RESULTS Compared with the LIRI group, EPC treatment significantly increased the ratios of arterial oxygen partial pressure to fractional inspiratory oxygen and decreased the lung wet-to-dry weight ratios and protein levels in the grafts, accompanied by increasing eNOS expression and phosphorylation, but decreasing endothelin-1, inducible NOS, phosphorylated nuclear factor-kBp65, phosphorylated myosin light chain expression, and myeloperoxidase activity. EPCs reduced lung tissue damage and apoptosis associated with decreased levels of Bax and cleaved caspase-3 expression, but increased Bcl-2 expression. EPC treatment significantly reduced the levels of serum proinflammatory factors, but elevated levels of interleukin-10. In contrast, the protective effect of EPCs were mitigated and abrogated by N5-(1-iminoethyl)-l-ornithine pretreatment. CONCLUSIONS Data indicated that EPC ameliorated LIRI by increasing eNOS expression.
Collapse
Affiliation(s)
- Wei Gao
- Department of Anesthesiology, the Second Affiliated Hospital of the Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Tao Jiang
- Department of Anesthesiology, the Second Affiliated Hospital of the Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yan-Hong Liu
- Department of Anesthesiology, the Second Affiliated Hospital of the Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Wen-Gang Ding
- Department of Anesthesiology, the Second Affiliated Hospital of the Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Chang-Chun Guo
- Department of Anesthesiology, the Second Affiliated Hospital of the Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xiao-Guang Cui
- Department of Anesthesiology, the Second Affiliated Hospital of the Harbin Medical University, Harbin, Heilongjiang Province, China.
| |
Collapse
|
135
|
Smith RJ, Yi T, Nasiri B, Breuer CK, Andreadis ST. Implantation of VEGF-functionalized cell-free vascular grafts: regenerative and immunological response. FASEB J 2019; 33:5089-5100. [PMID: 30629890 DOI: 10.1096/fj.201801856r] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Recently, our group demonstrated that immobilized VEGF can capture flowing endothelial cells (ECs) from the blood in vitro and promote endothelialization and patency of acellular tissue-engineered vessels (A-TEVs) into the arterial system of an ovine animal model. Here, we demonstrate implantability of submillimeter diameter heparin and VEGF-decorated A-TEVs in a mouse model and discuss the cellular and immunologic response. At 1 mo postimplantation, the graft lumen was fully endothelialized, as shown by expression of EC markers such as CD144, eNOS, CD31, and VEGFR2. Interestingly, the same cells coexpressed leukocyte/macrophage (Mϕ) markers CD14, CD16, VEGFR1, CD38, and EGR2. Notably, there was a stark difference in the cellular makeup between grafts containing VEGF and those containing heparin alone. In VEGF-containing grafts, infiltrating monocytes (MCs) converted into anti-inflammatory M2-Mϕs, and the grafts developed well-demarcated luminal and medial layers resembling those of native arteries. In contrast, in grafts containing only heparin, MCs converted primarily into M1-Mϕs, and the endothelial and smooth muscle layers were not well defined. Our results indicate that VEGF may play an important role in regulating A-TEV patency and regeneration, possibly by regulating the inflammatory response to the implants.-Smith, R. J., Jr., Yi, T., Nasiri, B., Breuer, C. K., Andreadis, S. T. Implantation of VEGF-functionalized cell-free vascular grafts: regenerative and immunological response.
Collapse
Affiliation(s)
- Randall J Smith
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Amherst, New York, USA
| | - Tai Yi
- Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Bita Nasiri
- Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Amherst, New York, USA; and
| | | | - Stelios T Andreadis
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Amherst, New York, USA.,Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Amherst, New York, USA; and.,Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, State University of New York, Amherst, New York, USA
| |
Collapse
|
136
|
Schito L. Hypoxia-Dependent Angiogenesis and Lymphangiogenesis in Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1136:71-85. [PMID: 31201717 DOI: 10.1007/978-3-030-12734-3_5] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hypoxia (low O2) is a ubiquitous feature of solid cancers, arising as a mismatch between cellular O2 supply and consumption. Hypoxia is associated to metastatic disease and mortality owing to its ability to stimulate the formation of blood (angiogenesis) and lymphatic vessels (lymphangiogenesis), thereby allowing cancer cells to escape the unfavorable tumor microenvironment and disseminate into secondary sites. This review outlines molecular mechanisms by which intratumoral hypoxia regulates the expression of motogenic and mitogenic factors that induce angiogenesis and lymphangiogenesis, whilst discussing their implications for metastatic cancers.
Collapse
Affiliation(s)
- Luana Schito
- Biological Sciences Platform, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
137
|
Antonyshyn JA, McFadden MJ, Gramolini AO, Hofer SO, Santerre JP. Limited Endothelial Plasticity of Mesenchymal Stem Cells Revealed by Quantitative Phenotypic Comparisons to Representative Endothelial Cell Controls. Stem Cells Transl Med 2019; 8:35-45. [PMID: 30269434 PMCID: PMC6312449 DOI: 10.1002/sctm.18-0127] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/03/2018] [Indexed: 12/30/2022] Open
Abstract
Considerable effort has been directed toward deriving endothelial cells (ECs) from adipose-derived mesenchymal stem cells (ASCs) since 2004, when it was first suggested that ECs and adipocytes share a common progenitor. While the capacity of ASCs to express endothelial markers has been repeatedly demonstrated, none constitute conclusive evidence of an endothelial phenotype as all reported markers have been detected in other, non-endothelial cell types. In this study, quantitative phenotypic comparisons to representative EC controls were used to determine the extent of endothelial differentiation being achieved with ASCs. ASCs were harvested from human subcutaneous abdominal white adipose tissue, and their endothelial differentiation was induced using well-established biochemical stimuli. Reverse transcription quantitative real-time polymerase chain reaction and parallel reaction monitoring mass spectrometry were used to quantify their expression of endothelial genes and corresponding proteins, respectively. Flow cytometry was used to quantitatively assess their uptake of acetylated low-density lipoprotein (AcLDL). Human umbilical vein, coronary artery, and dermal microvascular ECs were used as positive controls to reflect the phenotypic heterogeneity between ECs derived from different vascular beds. Biochemically conditioned ASCs were found to upregulate their expression of endothelial genes and proteins, as well as AcLDL uptake, but their abundance remained orders of magnitude lower than that observed in the EC controls despite their global proteomic heterogeneity. The findings of this investigation demonstrate the strikingly limited extent of endothelial differentiation being achieved with ASCs using well-established biochemical stimuli, and underscore the importance of quantitative phenotypic comparisons to representative primary cell controls in studies of differentiation. Stem Cells Translational Medicine 2019;8:35-45.
Collapse
Affiliation(s)
- Jeremy A. Antonyshyn
- Institute of Biomaterials and Biomedical Engineering, University of TorontoTorontoOntarioCanada
- Translational Biology and Engineering ProgramTed Rogers Centre for Heart ResearchTorontoOntarioCanada
| | - Meghan J. McFadden
- Institute of Biomaterials and Biomedical Engineering, University of TorontoTorontoOntarioCanada
- Translational Biology and Engineering ProgramTed Rogers Centre for Heart ResearchTorontoOntarioCanada
| | - Anthony O. Gramolini
- Translational Biology and Engineering ProgramTed Rogers Centre for Heart ResearchTorontoOntarioCanada
- Department of PhysiologyUniversity of TorontoTorontoOntarioCanada
| | - Stefan O.P. Hofer
- Department of Surgery, Division of Plastic and Reconstructive SurgeryUniversity of TorontoTorontoOntarioCanada
- Departments of Surgery and Surgical OncologyUniversity Health NetworkTorontoOntarioCanada
| | - J. Paul Santerre
- Institute of Biomaterials and Biomedical Engineering, University of TorontoTorontoOntarioCanada
- Translational Biology and Engineering ProgramTed Rogers Centre for Heart ResearchTorontoOntarioCanada
- Faculty of DentistryUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
138
|
Rodrigues M, Kosaric N, Bonham CA, Gurtner GC. Wound Healing: A Cellular Perspective. Physiol Rev 2019; 99:665-706. [PMID: 30475656 PMCID: PMC6442927 DOI: 10.1152/physrev.00067.2017] [Citation(s) in RCA: 1301] [Impact Index Per Article: 260.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 06/25/2018] [Accepted: 06/28/2018] [Indexed: 02/08/2023] Open
Abstract
Wound healing is one of the most complex processes in the human body. It involves the spatial and temporal synchronization of a variety of cell types with distinct roles in the phases of hemostasis, inflammation, growth, re-epithelialization, and remodeling. With the evolution of single cell technologies, it has been possible to uncover phenotypic and functional heterogeneity within several of these cell types. There have also been discoveries of rare, stem cell subsets within the skin, which are unipotent in the uninjured state, but become multipotent following skin injury. Unraveling the roles of each of these cell types and their interactions with each other is important in understanding the mechanisms of normal wound closure. Changes in the microenvironment including alterations in mechanical forces, oxygen levels, chemokines, extracellular matrix and growth factor synthesis directly impact cellular recruitment and activation, leading to impaired states of wound healing. Single cell technologies can be used to decipher these cellular alterations in diseased states such as in chronic wounds and hypertrophic scarring so that effective therapeutic solutions for healing wounds can be developed.
Collapse
Affiliation(s)
- Melanie Rodrigues
- Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Nina Kosaric
- Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Clark A Bonham
- Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Geoffrey C Gurtner
- Department of Surgery, Stanford University School of Medicine , Stanford, California
| |
Collapse
|
139
|
Mashimo T, Sato Y, Akita D, Toriumi T, Namaki S, Matsuzaki Y, Yonehara Y, Honda M. Bone marrow-derived mesenchymal stem cells enhance bone marrow regeneration in dental extraction sockets. J Oral Sci 2019; 61:284-293. [DOI: 10.2334/josnusd.18-0143] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Affiliation(s)
- Takayuki Mashimo
- Department of Oral and Maxillofacial Surgery, Juntendo University Faculty of Medicine
- Department of Oral and Maxillofacial Surgery, Nihon University School of Dentistry
| | - Yukio Sato
- Department of Emergency and Critical Care Medicine, Keio University School of Medicine
| | - Daisuke Akita
- Department of Partial Denture Prosthodontics, Nihon University School of Dentistry
| | - Taku Toriumi
- Department of Oral Anatomy, Aichi-Gakuin University School of Dentistry
| | - Shunsuke Namaki
- Department of Clinical Medicine, Nihon University School of Dentistry
| | - Yumi Matsuzaki
- Department of Life Science, Shimane University Faculty of Medicine
| | | | - Masaki Honda
- Department of Oral Anatomy, Aichi-Gakuin University School of Dentistry
| |
Collapse
|
140
|
Morikawa S, Iribar H, Gutiérrez-Rivera A, Ezaki T, Izeta A. Pericytes in Cutaneous Wound Healing. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1147:1-63. [DOI: 10.1007/978-3-030-16908-4_1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
141
|
KIM GA, LEE Y, KIM HJ, OH HJ, KANG SK, RA JC, LEE BC. Intravenous human endothelial progenitor cell administration into aged mice enhances embryo development and oocyte quality by reducing inflammation, endoplasmic reticulum stress and apoptosis. J Vet Med Sci 2018; 80:1905-1913. [PMID: 30369585 PMCID: PMC6305508 DOI: 10.1292/jvms.18-0242] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 09/29/2018] [Indexed: 11/22/2022] Open
Abstract
Stem cell therapy has been proposed to restore the function and structure of injured tissues. In the present study, we investigated the ability of human endothelial progenitor cells (hEPCs) to attenuate ovarian aging and dysfunction. Female ICR mice aged 4 and 6 months were injected with cultured hEPCs. Cultured hEPCs were injected intravenously twice with 5 × 104 cells with a 4 day interval. After pregnant mare serum gonadotropin and human chorionic gonadotropin stimulation, oocytes and ovaries of aged mice were collected, cumulus-free oocytes were activated by SrCl2 and gene expression levels related to inflammation, apoptosis, follicle development and endoplasmic reticulum (ER) stress in ovaries were compared. Administration of hEPCs attenuated the level of inflammatory cytokines and adverse apoptotic factor, as well as reducing ER stress in the ovaries. Increased cleavage and blastocyst formation rates and cell numbers in blastocysts from hEPCs-treated aged mice vs. same aged control mice demonstrated a protective function of hEPCs against reproductive aging. Based on these data, we suggest that treatment with hEPCs attenuates reproductive aging and dysfunction potentially via regulation of inflammation, apoptosis and ER stress.
Collapse
Affiliation(s)
- Geon A KIM
- Department of Theriogenology and Biotechnology, College of
Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yeonjae LEE
- Hankuk Academy of Foreign Studies, Yongin-si, Gyeonggi-do
17035, Republic of Korea
| | - Hyun Jin KIM
- Seoul National University, Seoul 08826, Republic of
Korea
| | - Hyun Ju OH
- Department of Theriogenology and Biotechnology, College of
Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sung Keun KANG
- Biostar Stem Cell Research Institute, R Bio Co., Ltd., Seoul
08506, Republic of Korea
| | - Jeong Chan RA
- Biostar Stem Cell Research Institute, R Bio Co., Ltd., Seoul
08506, Republic of Korea
| | - Byeong Chun LEE
- Department of Theriogenology and Biotechnology, College of
Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
142
|
Shen WC, Chou YH, Huang HP, Sheen JF, Hung SC, Chen HF. Induced pluripotent stem cell-derived endothelial progenitor cells attenuate ischemic acute kidney injury and cardiac dysfunction. Stem Cell Res Ther 2018; 9:344. [PMID: 30526689 PMCID: PMC6288873 DOI: 10.1186/s13287-018-1092-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 10/31/2018] [Accepted: 11/26/2018] [Indexed: 12/18/2022] Open
Abstract
Background Renal ischemia–reperfusion (I/R) injury is a major cause of acute kidney injury (AKI), which is associated with high morbidity and mortality. AKI is a serious and costly medical condition. Effective therapy for AKI is an unmet clinical need, and molecular mechanisms underlying the interactions between an injured kidney and distant organs remain unclear. Therefore, novel therapeutic strategies should be developed. Methods We directed the differentiation of human induced pluripotent stem (iPS) cells into endothelial progenitor cells (iEPCs), which were then applied for treating mouse AKI. The mouse model of AKI was induced by I/R injury. Results We discovered that intravenously infused iEPCs were recruited to the injured kidney, expressed the mature endothelial cell marker CD31, and replaced injured endothelial cells. Moreover, infused iEPCs produced abundant proangiogenic proteins, which entered into circulation. In AKI mice, blood urea nitrogen and plasma creatinine levels increased 2 days after I/R injury and reduced after the infusion of iEPCs. Tubular injury, cell apoptosis, and peritubular capillary rarefaction in injured kidneys were attenuated accordingly. In the AKI mice, iEPC therapy also ameliorated apoptosis of cardiomyocytes and cardiac dysfunction, as indicated by echocardiography. The therapy also ameliorated an increase in serum brain natriuretic peptide. Regarding the relevant mechanisms, indoxyl sulfate and interleukin-1β synergistically induced apoptosis of cardiomyocytes. Systemic iEPC therapy downregulated the proapoptotic protein caspase-3 and upregulated the anti-apoptotic protein Bcl-2 in the hearts of the AKI mice, possibly through the reduction of indoxyl sulfate and interleukin-1β. Conclusions Therapy using human iPS cell-derived iEPCs provided a protective effect against ischemic AKI and remote cardiac dysfunction through the repair of endothelial cells and the attenuation of cardiomyocyte apoptosis. Electronic supplementary material The online version of this article (10.1186/s13287-018-1092-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wen-Ching Shen
- Drug Development Center, Institute of New Drug Development, Institute of Biomedical Sciences, China Medical University, Taichung, 404, Taiwan.,Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yu-Hsiang Chou
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan.,Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Renal Division, Department of Internal Medicine, National Taiwan University Hospital Jin-Shan Branch, New Taipei City, Taiwan
| | - Hsiang-Po Huang
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Jenn-Feng Sheen
- Department of Biotechnology, National Formosa University, Yun-Lin, Taiwan
| | - Shih-Chieh Hung
- Drug Development Center, Institute of New Drug Development, Institute of Biomedical Sciences, China Medical University, Taichung, 404, Taiwan.,Integrative Stem Cell Center, Department of Orthopaedics, China Medical University Hospital, Taichung, 404, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, 105, Taiwan
| | - Hsin-Fu Chen
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei, Taiwan. .,Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
143
|
Harnessing Macrophages for Vascularization in Tissue Engineering. Ann Biomed Eng 2018; 47:354-365. [PMID: 30535815 DOI: 10.1007/s10439-018-02170-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 11/16/2018] [Indexed: 12/13/2022]
Abstract
In this review, we explore the roles of macrophages both in vessel development and in vascularization of tissue engineered constructs. Upon the implantation of tissue engineered constructs into the body, macrophages respond, invade and orchestrate the host's immune response. By altering their phenotype, macrophages can adopt a variety of roles. They can promote inflammation at the site of the implanted construct; they can also promote tissue repair. Macrophages support tissue repair by promoting angiogenesis through the secretion of pro-angiogenic cytokines and by behaving as support cells for nascent vasculature. Thus, the ability to manipulate the macrophage phenotype may yield macrophages capable of supporting vessel development. Moreover, macrophages are an easily isolated autologous cell source. For the generation of vascularized constructs outside of the body, these isolated macrophages can also be skewed to adopt a pro-angiogenic phenotype and enhance blood vessel development in the presence of endothelial cells. To assess the influence of macrophages on vessel development, both in vivo and in vitro models have been developed. Additionally, several groups have demonstrated the pro-angiogenic roles of macrophages in vascularization of tissue engineered constructs through the manipulation of macrophage phenotypes. This review comments on the roles of macrophages in promoting vascularization within these contexts.
Collapse
|
144
|
Baumann M, Gumpold C, Mueller-Felber W, Schoser B, Haberler C, Loescher WN, Rostásy K, Fischer MB, Wanschitz JV. Pattern of myogenesis and vascular repair in early and advanced lesions of juvenile dermatomyositis. Neuromuscul Disord 2018; 28:973-985. [DOI: 10.1016/j.nmd.2018.09.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 08/15/2018] [Accepted: 09/06/2018] [Indexed: 12/12/2022]
|
145
|
Zhang J, Zhang H, Chen Y, Fu J, Lei Y, Sun J, Tang B. Platelet‑derived growth factor D promotes the angiogenic capacity of endothelial progenitor cells. Mol Med Rep 2018; 19:125-132. [PMID: 30483778 PMCID: PMC6297765 DOI: 10.3892/mmr.2018.9692] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 10/09/2018] [Indexed: 01/04/2023] Open
Abstract
Neovascularization and re-endothelialization rely on endothelial progenitor cells (EPCs). However, the recruitment and angiogenic roles of EPCs are subject to regulation through the vascular microenvironment, which remains largely unknown. Platelet-derived growth factor D (PDGF-D) is a new member of the PDGF family that binds the PDGFR-β homodimer. However, it remains unknown whether and how it affects the angiogenic capacity of EPCs and participates in tube-like formation. EPCs were derived from rat bone marrow cells, and the gain-of-function approach was used to study the effects of PDGF-D on the biological activities of EPCs. EPCs that stably express PDGF-D were generated by lentiviral-mediated transduction, and the expression levels were evaluated by western blotting and reverse transcription, followed by real-time quantitative polymerase chain reaction (RT-qPCR). The biological activities of EPCs evaluated in the present study included proliferation, adhesion, migration, tube formation and senescence. Furthermore, the downstream signaling of PDGF-D was explored by western blot analysis. The results revealed that the lentiviral-mediated expression of PDGF-D in the microenvironment promoted the migration, proliferation, adhesion and tube formation of EPCs. In addition, PDGF-D suppressed the senescence of EPCs. Mechanistically, PDGF-D induced the phosphorylation of several signaling molecules, including STAT3, AKT, ERK1/2, mTOR and GSK-3β, suggesting that PDGF-D enhanced the angiogenic function of EPCs through PDGF receptor-dependent and -independent signaling pathways. In conclusion, PDGF-D promotes the angiogenic capacity of EPCs, including proliferation, migration, adhesion and tube formation, and thereby contributes to angiogenesis.
Collapse
Affiliation(s)
- Jianbo Zhang
- Department of Vascular Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Haolong Zhang
- Department of Vascular Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Yikuan Chen
- Department of Vascular Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Jian Fu
- Department of Vascular Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Yu Lei
- Department of Vascular Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Jianming Sun
- Department of Vascular Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Bo Tang
- Department of Vascular Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| |
Collapse
|
146
|
Son Y, Kwon SM, Cho JY. CD276 (B7-H3) Maintains Proliferation and Regulates Differentiation in Angiogenic Function in Late Endothelial Progenitor Cells. Stem Cells 2018; 37:382-394. [PMID: 30379377 DOI: 10.1002/stem.2944] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 09/22/2018] [Accepted: 10/16/2018] [Indexed: 12/24/2022]
Abstract
Endothelial progenitor cells (EPCs) provide an important source of recovery from blood vessel dysfunction. Late EPCs (LEPCs) are circulating blood cells that are capable of promoting vascular repair. Using transcriptome analysis, we identified distinctive LEPC profiles and found that CD276 (B7-H3) mRNA is strongly expressed in LEPCs. CD276 protein is present abundantly on the cell surface of LEPC when analyzed by fluorescence-activated cell sorter and immunocytochemistry. CD276, a B7 family member, is a type I transmembrane glycoprotein. The role of CD276 in LEPCs remains unknown. CD276 knockdown by lentivirus transduction in LEPCs significantly decreased proliferation and increased apoptosis of LEPCs in vitro. After CD276 silencing, the cell cycle of LEPCs was prone to remain at the G0/G1 phase, and the cell migration rates as well as transwell and wound-healing migration were decreased. CD276 knockdown in LEPCs increased the G1 phase regulators cyclin D2/D3/E1-cyclin-dependent kinases (CDK2/4/6), but decreased the S-G2-M phase regulators cyclin A/B-CDK1. However, LEPCs with CD276 knockdown resulted in increased tube formation in vitro and angiogenesis in a Matrigel plug assay in vivo. FoxC1/C2, an upstream signal of Notch in arterial cell proliferation, and Hey1/2, which is known to promote arterial differentiation in the vasculature, were upregulated in CD276 knockdown LEPCs. In LEPCS, CD276 has a positive effect on proliferation and migration of endothelial cells, but negative effects on angiogenesis, particularly endothelial cell differentiation. Our data indicate, for therapeutic purpose, that CD276 can be used to acquire and maintain cell populations of LEPCs and blocking CD276 will promote angiogenetic differentiation. We found that CD276 (B7-H3) is enriched on the cell membrane of LEPCs. CD276 knockdown reduced proliferation and migration of LEPCs by increasing cell cycle inhibitors such as p21cip1 and pRb and decreasing pErk1/2 and pAkt but promoted angiogenesis and endothelial cell differentiation by elevating vascular endothelial growth factor-vascular endothelial growth factor receptor 1 and p-p38. Stem Cells 2019;37:382-394.
Collapse
Affiliation(s)
- YeonSung Son
- Department of Biochemistry, BK21 Plus and Research Institute for Veterinary Science, School of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Sang-Mo Kwon
- Laboratory for Vascular Medicine & Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Je-Yoel Cho
- Department of Biochemistry, BK21 Plus and Research Institute for Veterinary Science, School of Veterinary Medicine, Seoul National University, Seoul, Korea
| |
Collapse
|
147
|
Smits J, Tasev D, Andersen S, Szulcek R, Botros L, Ringgaard S, Andersen A, Vonk-Noordegraaf A, Koolwijk P, Bogaard HJ. Blood Outgrowth and Proliferation of Endothelial Colony Forming Cells are Related to Markers of Disease Severity in Patients with Pulmonary Arterial Hypertension. Int J Mol Sci 2018; 19:ijms19123763. [PMID: 30486375 PMCID: PMC6321271 DOI: 10.3390/ijms19123763] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/15/2018] [Accepted: 11/23/2018] [Indexed: 12/27/2022] Open
Abstract
In pulmonary arterial hypertension (PAH), lung-angioproliferation leads to increased pulmonary vascular resistance, while simultaneous myocardial microvessel loss contributes to right ventricular (RV) failure. Endothelial colony forming cells (ECFC) are highly proliferative, angiogenic cells that may contribute to either pulmonary vascular obstruction or to RV microvascular adaptation. We hypothesize ECFC phenotypes (outgrowth, proliferation, tube formation) are related to markers of disease severity in a prospective cohort-study of 33 PAH and 30 healthy subjects. ECFC were transplanted in pulmonary trunk banded rats with RV failure. The presence of ECFC outgrowth in PAH patients was associated with low RV ejection fraction, low central venous saturation and a shorter time to clinical worsening (5.4 months (0.6–29.2) vs. 36.5 months (7.4–63.4), p = 0.032). Functionally, PAH ECFC had higher proliferative rates compared to control in vitro, although inter-patient variability was high. ECFC proliferation was inversely related to RV end diastolic volume (R2 = 0.39, p = 0.018), but not pulmonary vascular resistance. Tube formation-ability was similar among donors. Normal and highly proliferative PAH ECFC were transplanted in pulmonary trunk banded rats. While no effect on hemodynamic measurements was observed, RV vascular density was restored. In conclusion, we found that ECFC outgrowth associates with high clinical severity in PAH, suggesting recruitment. Transplantation of highly proliferative ECFC restored myocardial vascular density in pulmonary trunk banded rats, while RV functional improvements were not observed.
Collapse
Affiliation(s)
- Josien Smits
- Amsterdam UMC, VU University Medical Center, Department of Pulmonary Diseases, Amsterdam Cardiovascular Sciences (ACS), De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands.
- Amsterdam UMC, VU University Medical Center, Department of Physiology, Amsterdam Cardiovascular Sciences (ACS), De Boelelaan 1108, 1081 HV Amsterdam, The Netherlands.
| | - Dimitar Tasev
- Amsterdam UMC, VU University Medical Center, Department of Physiology, Amsterdam Cardiovascular Sciences (ACS), De Boelelaan 1108, 1081 HV Amsterdam, The Netherlands.
| | - Stine Andersen
- Aarhus University Hospital, Department of Cardiology, Palle Juul-Jensens Boulevaard 99, 8200 Aarhus N, Denmark.
| | - Robert Szulcek
- Amsterdam UMC, VU University Medical Center, Department of Pulmonary Diseases, Amsterdam Cardiovascular Sciences (ACS), De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands.
- Amsterdam UMC, VU University Medical Center, Department of Physiology, Amsterdam Cardiovascular Sciences (ACS), De Boelelaan 1108, 1081 HV Amsterdam, The Netherlands.
| | - Liza Botros
- Amsterdam UMC, VU University Medical Center, Department of Pulmonary Diseases, Amsterdam Cardiovascular Sciences (ACS), De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands.
- Amsterdam UMC, VU University Medical Center, Department of Physiology, Amsterdam Cardiovascular Sciences (ACS), De Boelelaan 1108, 1081 HV Amsterdam, The Netherlands.
| | - Steffen Ringgaard
- Aarhus University Hospital, MR Centre, Palle Juul-Jensens Boulevaard 99, 8200 Aarhus N, Denmark.
| | - Asger Andersen
- Aarhus University Hospital, Department of Cardiology, Palle Juul-Jensens Boulevaard 99, 8200 Aarhus N, Denmark.
| | - Anton Vonk-Noordegraaf
- Amsterdam UMC, VU University Medical Center, Department of Pulmonary Diseases, Amsterdam Cardiovascular Sciences (ACS), De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands.
| | - Pieter Koolwijk
- Amsterdam UMC, VU University Medical Center, Department of Physiology, Amsterdam Cardiovascular Sciences (ACS), De Boelelaan 1108, 1081 HV Amsterdam, The Netherlands.
| | - Harm Jan Bogaard
- Amsterdam UMC, VU University Medical Center, Department of Pulmonary Diseases, Amsterdam Cardiovascular Sciences (ACS), De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands.
| |
Collapse
|
148
|
Insights into Endothelial Progenitor Cells: Origin, Classification, Potentials, and Prospects. Stem Cells Int 2018; 2018:9847015. [PMID: 30581475 PMCID: PMC6276490 DOI: 10.1155/2018/9847015] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 08/27/2018] [Accepted: 09/18/2018] [Indexed: 02/07/2023] Open
Abstract
With the discovery of endothelial progenitor cells (EPCs) in the late 1990s, a paradigm shift in the concept of neoangiogenesis occurred. The identification of circulating EPCs in peripheral blood marked the beginning of a new era with enormous potential in the rapidly transforming regenerative field. Overwhelmed with the revelation, researchers across the globe focused on isolating, defining, and interpreting the role of EPCs in various physiological and pathological conditions. Consequently, controversies emerged regarding the isolation techniques and classification of EPCs. Nevertheless, the potential of using EPCs in tissue engineering as an angiogenic source has been extensively explored. Concomitantly, the impact of EPCs on various diseases, such as diabetes, cancer, and cardiovascular diseases, has been studied. Within the limitations of the current knowledge, this review attempts to delineate the concept of EPCs in a sequential manner from the speculative history to a definitive presence (origin, sources of EPCs, isolation, and identification) and significance of these EPCs. Additionally, this review is aimed at serving as a guide for investigators, identifying potential research gaps, and summarizing our current and future prospects regarding EPCs.
Collapse
|
149
|
Lam YT, Lecce L, Yuen GSC, Wise SG, Handelsman DJ, Ng MKC. Androgen action augments ischemia-induced, bone marrow progenitor cell-mediated vasculogenesis. Int J Biol Sci 2018; 14:1985-1992. [PMID: 30585262 PMCID: PMC6299365 DOI: 10.7150/ijbs.27378] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 09/05/2018] [Indexed: 02/04/2023] Open
Abstract
Bone marrow-derived progenitor cell-mediated vasculogenesis is a key process for vascular repair and regeneration. However, the role of androgens in the mechanism of ischemia-induced vasculogenesis remains unclear. In this study, a gender-mismatch murine bone marrow transplant model was used to allow tissue tracking of transplanted cells. Bone marrow from 2-month-old male mice was transplanted into irradiated age-matched female recipients. Following the transplantation, ovariectomized female recipients were subjected to unilateral hindlimb ischemia and immediately implanted with either dihydrotestosterone (DHT) or placebo pellets. Laser Doppler perfusion imaging revealed that DHT significantly augmented blood flow recovery, with increased capillary density compared to placebo-treated female recipient controls. Flow cytometry analysis showed that DHT modulated vasculogenesis by increasing Sca1+/CXC4+ progenitor cell production in bone marrow and spleen and enhancing cell mobilization in circulating blood following hindlimb ischemia. Bone marrow cell homing was examined by detecting expression levels of male-specific SRY gene in the ischemic female tissues. DHT treatment promoted bone marrow cell homing to ischemic tissue shown by significantly higher SRY expression compared to placebo-treated females as well as reduced apoptotic features in DHT-treated females, including increased Bcl-2 expression, reduced Bax levels and decreased TUNEL staining. In conclusion, the gender-mismatched bone marrow transplant study shows that androgens directly enhance bone marrow cell-mediated vasculogenesis that contributes to ischemia-induced neovascularization.
Collapse
Affiliation(s)
- Yuen Ting Lam
- The Heart Research Institute, Newtown, Sydney NSW, 2042 Australia.,Sydney Medical School, The University of Sydney, NSW 2006 Australia
| | - Laura Lecce
- The Heart Research Institute, Newtown, Sydney NSW, 2042 Australia.,Sydney Medical School, The University of Sydney, NSW 2006 Australia
| | - Gloria S C Yuen
- The Heart Research Institute, Newtown, Sydney NSW, 2042 Australia.,Sydney Medical School, The University of Sydney, NSW 2006 Australia
| | - Steven G Wise
- The Heart Research Institute, Newtown, Sydney NSW, 2042 Australia.,Sydney Medical School, The University of Sydney, NSW 2006 Australia
| | - David J Handelsman
- ANZAC Research Institute, The University of Sydney, Concord Hospital NSW 2139 Australia
| | - Martin K C Ng
- The Heart Research Institute, Newtown, Sydney NSW, 2042 Australia.,Sydney Medical School, The University of Sydney, NSW 2006 Australia.,Royal Prince Alfred Hospital, Camperdown NSW Australia 2050
| |
Collapse
|
150
|
Wang L, Neumann M, Fu T, Li W, Cheng X, Su BL. Porous and responsive hydrogels for cell therapy. Curr Opin Colloid Interface Sci 2018. [DOI: 10.1016/j.cocis.2018.10.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|