1
|
S1P Increases VEGF Production in Osteoblasts and Facilitates Endothelial Progenitor Cell Angiogenesis by Inhibiting miR-16-5p Expression via the c-Src/FAK Signaling Pathway in Rheumatoid Arthritis. Cells 2021; 10:cells10082168. [PMID: 34440937 PMCID: PMC8393529 DOI: 10.3390/cells10082168] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 02/06/2023] Open
Abstract
Angiogenesis is a critical process in the formation of new capillaries and a key participant in rheumatoid arthritis (RA) pathogenesis. Vascular endothelial growth factor (VEGF) stimulation of endothelial progenitor cells (EPCs) facilitates angiogenesis and the progression of RA. Phosphorylation of sphingosine kinase 1 (SphK1) produces sphingosine-1-phosphate (S1P), which increases inflammatory cytokine production, although the role of S1P in RA angiogenesis is unclear. In this study, we evaluated the impact of S1P treatment on VEGF-dependent angiogenesis in osteoblast-like cells (MG-63 cells) and the significance of SphK1 short hairpin RNA (shRNA) on S1P production in an in vivo model. We found significantly higher levels of S1P and VEGF expression in synovial fluid from RA patients compared with those with osteoarthritis by ELISA analysis. Treating MG-63 cells with S1P increased VEGF production, while focal adhesion kinase (FAK) and Src siRNAs and inhibitors decreased VEGF production in S1P-treated MG-63 cells. Conditioned medium from S1P-treated osteoblasts significantly increased EPC tube formation and migration by inhibiting miR-16-5p synthesis via proto-oncogene tyrosine-protein kinase src (c-Src) and FAK signaling in chick chorioallantoic membrane (CAM) and Matrigel plug assays. Infection with SphK1 shRNA reduced angiogenesis, articular swelling and cartilage erosion in the ankle joints of mice with collagen-induced arthritis (CIA). S1P appears to have therapeutic potential in RA treatment.
Collapse
|
2
|
Cellular Crosstalk between Endothelial and Smooth Muscle Cells in Vascular Wall Remodeling. Int J Mol Sci 2021; 22:ijms22147284. [PMID: 34298897 PMCID: PMC8306829 DOI: 10.3390/ijms22147284] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/25/2021] [Accepted: 07/01/2021] [Indexed: 12/24/2022] Open
Abstract
Pathological vascular wall remodeling refers to the structural and functional changes of the vessel wall that occur in response to injury that eventually leads to cardiovascular disease (CVD). Vessel wall are composed of two major primary cells types, endothelial cells (EC) and vascular smooth muscle cells (VSMCs). The physiological communications between these two cell types (EC–VSMCs) are crucial in the development of the vasculature and in the homeostasis of mature vessels. Moreover, aberrant EC–VSMCs communication has been associated to the promotor of various disease states including vascular wall remodeling. Paracrine regulations by bioactive molecules, communication via direct contact (junctions) or information transfer via extracellular vesicles or extracellular matrix are main crosstalk mechanisms. Identification of the nature of this EC–VSMCs crosstalk may offer strategies to develop new insights for prevention and treatment of disease that curse with vascular remodeling. Here, we will review the molecular mechanisms underlying the interplay between EC and VSMCs. Additionally, we highlight the potential applicable methodologies of the co-culture systems to identify cellular and molecular mechanisms involved in pathological vascular wall remodeling, opening questions about the future research directions.
Collapse
|
3
|
Hu SL, Huang CC, Tseng TT, Liu SC, Tsai CH, Fong YC, Tang CH. S1P facilitates IL-1β production in osteoblasts via the JAK and STAT3 signaling pathways. ENVIRONMENTAL TOXICOLOGY 2020; 35:991-997. [PMID: 32401414 DOI: 10.1002/tox.22935] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/14/2020] [Accepted: 04/22/2020] [Indexed: 06/11/2023]
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune inflammatory disease, in which the immune system attacks synovial joint tissues. Interleukin (IL)-1β is a critical proinflammatory cytokine in RA progression. Sphingosine-1-phosphate (S1P), a platelet-derived lysophospholipid mediator, reportedly regulates osteoimmunology. Here, we investigated how S1P mediates IL-1β expression in osteoblasts. Our analysis of records from the Gene Expression Omnibus (GEO) database demonstrate higher levels of IL-1β in patients with RA compared with those with osteoarthritis. Stimulation of osteoblasts with S1P concentration dependently increased mRNA and protein expression of IL-1β. Elevations in IL-1β mRNA expression induced by S1P were reduced by the small interfering RNA (siRNA) against the S1P1 receptor. S1P also augmented JAK and STAT3 molecular cascades. We also found that JAK and STAT3 inhibitors and their siRNAs antagonized S1P-promoted IL-1β expression. Our results indicate that S1P promotes the expression of IL-1β in osteoblasts via the S1P1 receptor and the JAK and STAT3 signaling pathways.
Collapse
Affiliation(s)
- Sung-Lin Hu
- School of Medicine, China Medical University, Taichung, Taiwan
- Department of Family Medicine, China Medical University Hsinchu Hospital, Hsinchu, Taiwan
| | - Chien-Chung Huang
- School of Medicine, China Medical University, Taichung, Taiwan
- Division of Immunology and Rheumatology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Tzu-Ting Tseng
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Shan-Chi Liu
- Department of Medical Education and Research, China Medical University Beigang Hospital, Yunlin, Taiwan
| | - Chun-Hao Tsai
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Yi-Chin Fong
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Chih-Hsin Tang
- School of Medicine, China Medical University, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
- Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| |
Collapse
|
4
|
Hu SL, Huang CC, Tzeng TT, Liu SC, Tsai CH, Fong YC, Tang CH. S1P promotes IL-6 expression in osteoblasts through the PI3K, MEK/ERK and NF-κB signaling pathways. Int J Med Sci 2020; 17:1207-1214. [PMID: 32547316 PMCID: PMC7294913 DOI: 10.7150/ijms.44612] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/27/2020] [Indexed: 12/14/2022] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune inflammatory disease, in which the immune system attacks joint tissue. Interleukin (IL)-6 is a key proinflammatory cytokine in RA progression. Sphingosine-1-phosphate (S1P), a platelet-derived lysophospholipid mediator, reportedly regulates osteoimmunology. Here, we examined the effects of S1P on IL-6 expression in osteoblasts. Our results and records from the Gene Expression Omnibus (GEO) database demonstrate higher levels of IL-6 in patients with RA compared with those with osteoarthritis. Stimulation of osteoblasts with S1P increased mRNA and protein expression of IL-6. PI3K, MEK, ERK and NF-κB inhibitors and their small interfering RNAs (siRNAs) reduced S1P-promoted IL-6 expression. S1P also facilitated PI3K, MEK/ERK and NF-κB signaling cascades. Our results indicate that S1P promotes the expression of IL-6 in osteoblasts via the PI3K, MEK/ERK and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Sung-Lin Hu
- School of Medicine, China Medical University, Taichung, Taiwan.,Department of Family Medicine, China Medical University Hsinchu Hospital, Hsinchu, Taiwan
| | - Chien-Chung Huang
- School of Medicine, China Medical University, Taichung, Taiwan.,Division of Immunology and Rheumatology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Tzu-Ting Tzeng
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Shan-Chi Liu
- Department of Medical Education and Research, China Medical University Beigang Hospital, Yunlin, Taiwan
| | - Chun-Hao Tsai
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan.,Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Yi-Chin Fong
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan.,Department of Orthopedic Surgery, China Medical University Beigang Hospital, Yunlin, Taiwan
| | - Chih-Hsin Tang
- School of Medicine, China Medical University, Taichung, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,Chinese Medicine Research Center, China Medical University, Taichung, Taiwan.,Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| |
Collapse
|
5
|
Wang CQ, Lin CY, Huang YL, Wang SW, Wang Y, Huang BF, Lai YW, Weng SL, Fong YC, Tang CH, Lv Z. Sphingosine-1-phosphate promotes PDGF-dependent endothelial progenitor cell angiogenesis in human chondrosarcoma cells. Aging (Albany NY) 2019; 11:11040-11053. [PMID: 31809267 PMCID: PMC6932882 DOI: 10.18632/aging.102508] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 11/18/2019] [Indexed: 02/07/2023]
Abstract
The malignant bone tumors that are categorized as chondrosarcomas display a high potential for metastasis in late-stage disease. Higher-grade chondrosarcomas contain higher levels of expression of platelet-derived growth factor (PDGF) and its receptor. The phosphorylation of sphingosine by sphingosine kinase enzymes SphK1 and SphK2 generates sphingosine-1-phosphate (S1P), which inhibits human chondrosarcoma cell migration, while SphK1 overexpression suppresses lung metastasis of chondrosarcoma. We sought to determine whether S1P mediates levels of PDGF-A expression and angiogenesis in chondrosarcoma. Surprisingly, our investigations found that treatment of chondrosarcoma cells with S1P and transfecting them with SphK1 cDNA increased PDGF-A expression and induced angiogenesis of endothelial progenitor cells (EPCs). Ras, Raf, MEK, ERK and AP-1 inhibitors and their small interfering RNAs (siRNAs) inhibited S1P-induced PDGF-A expression and EPC angiogenesis. Our results indicate that S1P promotes the expression of PDGF-A in chondrosarcoma via the Ras/Raf/MEK/ERK/AP-1 signaling cascade and stimulates EPC angiogenesis.
Collapse
Affiliation(s)
- Chao-Qun Wang
- Department of Pathology, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Chih-Yang Lin
- Department of Medicine, Mackay Medical College, New Taipei, Taiwan
| | - Yuan-Li Huang
- Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| | - Shih-Wei Wang
- Department of Medicine, Mackay Medical College, New Taipei, Taiwan.,Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yan Wang
- Department of Medical Oncology, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Bi-Fei Huang
- Department of Pathology, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Yu-Wei Lai
- Division of Urology, Taipei Hospital Renai Branch, Taipei, Taiwan.,Department of Urology, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Shun-Long Weng
- Department of Obstetrics and Gynaecology, Hsinchu MacKay Memorial Hospital, Hsinchu, Taiwan
| | - Yi-Chin Fong
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan.,Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Chih-Hsin Tang
- Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan.,Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan.,Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| | - Zhong Lv
- Department of General Surgery, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| |
Collapse
|
6
|
Xuejing D, Wenyu W, Hong W, Zhengrong Z, Li D, Jun F, Ran D, Feng L, Yan W, Xiang Z. UHPLC–MS/MS analysis of sphingosine 1‐phosphate in joint cavity dialysate and hemodialysis solution of adjuvant arthritis rats: Application to geniposide pharmacodynamic study. Biomed Chromatogr 2019; 33:e4526. [DOI: 10.1002/bmc.4526] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 02/21/2019] [Accepted: 02/26/2019] [Indexed: 11/12/2022]
Affiliation(s)
- Dai Xuejing
- Anhui University of Chinese Medicine Hefei China
- Anhui Province Key Laboratory of Xin'an Medicine, Ministry of Education Hefei China
- Anhui Province Key Laboratory of Chinese Medicinal Formula Hefei China
| | - Wang Wenyu
- Anhui University of Chinese Medicine Hefei China
- Anhui Province Key Laboratory of Xin'an Medicine, Ministry of Education Hefei China
- Anhui Province Key Laboratory of Chinese Medicinal Formula Hefei China
| | - Wu Hong
- Anhui University of Chinese Medicine Hefei China
- Anhui Province Key Laboratory of Xin'an Medicine, Ministry of Education Hefei China
| | - Zhang Zhengrong
- Anhui University of Chinese Medicine Hefei China
- Anhui Province Key Laboratory of Xin'an Medicine, Ministry of Education Hefei China
- Anhui Province Key Laboratory of Chinese Medicinal Formula Hefei China
| | - Dai Li
- Anhui University of Chinese Medicine Hefei China
- Anhui Province Key Laboratory of Xin'an Medicine, Ministry of Education Hefei China
| | - Fu Jun
- Anhui University of Chinese Medicine Hefei China
- Anhui Province Key Laboratory of Xin'an Medicine, Ministry of Education Hefei China
- Anhui Province Key Laboratory of Chinese Medicinal Formula Hefei China
| | - Deng Ran
- Anhui University of Chinese Medicine Hefei China
- Anhui Province Key Laboratory of Xin'an Medicine, Ministry of Education Hefei China
- Anhui Province Key Laboratory of Chinese Medicinal Formula Hefei China
| | - Li Feng
- Anhui University of Chinese Medicine Hefei China
- Anhui Province Key Laboratory of Xin'an Medicine, Ministry of Education Hefei China
- Anhui Province Key Laboratory of Chinese Medicinal Formula Hefei China
| | - Wang Yan
- Anhui University of Chinese Medicine Hefei China
- Anhui Province Key Laboratory of Xin'an Medicine, Ministry of Education Hefei China
- Anhui Province Key Laboratory of Chinese Medicinal Formula Hefei China
| | - Zhan Xiang
- Anhui University of Chinese Medicine Hefei China
- Anhui Province Key Laboratory of Xin'an Medicine, Ministry of Education Hefei China
- Anhui Province Key Laboratory of Chinese Medicinal Formula Hefei China
| |
Collapse
|
7
|
Tsai C, Yang D, Lin C, Chen T, Tang C, Huang Y. Sphingosine-1-phosphate suppresses chondrosarcoma metastasis by upregulation of tissue inhibitor of metalloproteinase 3 through suppressing miR-101 expression. Mol Oncol 2017; 11:1380-1398. [PMID: 28672103 PMCID: PMC5623823 DOI: 10.1002/1878-0261.12106] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 06/01/2017] [Accepted: 06/19/2017] [Indexed: 02/06/2023] Open
Abstract
Chondrosarcoma is the second most common primary malignancy form of bone cancer, exhibiting resistance to chemotherapy and radiation therapy as well as developing high metastasis ability in late‐stage tumors. Thus, understanding the metastatic processes of chondrosarcoma is considered a strategy for the treatment of this disease. Sphingosine 1‐phosphate (S1P), a bioactive sphingolipid, is produced intracellularly by sphingosine kinase (SphK) and is regarded as a second signaling molecule that regulates inflammation, proliferation, angiogenesis, and metastasis. However, the effect of S1P on chondrosarcoma remains uncertain. As demonstrated by the transwell, immunoblotting, and real‐time PCR analyses, we found that S1P inhibited cell migration and MMP‐2 expression through the upregulation of the tissue inhibitor of metalloproteinase‐3 (TIMP‐3) expression in human chondrosarcoma cells. Additionally, we also showed that microRNA (miRNA)‐101, which targets the 3′ untranslated region (3′UTR) of TIMP‐3, decreased significantly following S1P treatment. After transfection with miR‐101 mimics, the S1P‐regulated cell migration and TIMP‐3 expression were both reversed. Furthermore, we also showed that the S1P‐inhibited cell migration is mediated through the c‐Src/MEK/ERK signaling axis. Meanwhile, the in vivo study indicated that overexpression of SphK1 decreases chondrosarcoma metastasis to the lungs. Our results illustrate the clinical significance between SphK1, TIMP‐3, and miR‐101 in human chondrosarcoma patients. Taken together, our results suggest that S1P and miR‐101 may prove to be potential therapeutic targets for future chondrosarcoma treatment.
Collapse
Affiliation(s)
- Chun‐Hao Tsai
- School of MedicineChina Medical UniversityTaichungTaiwan
- Department of Orthopedic SurgeryChina Medical University HospitalTaichungTaiwan
| | - Dong‐Ying Yang
- Graduate Institute of Basic Medical ScienceChina Medical UniversityTaichungTaiwan
| | - Chih‐Yang Lin
- Graduate Institute of Basic Medical ScienceChina Medical UniversityTaichungTaiwan
| | - Tsung‐Ming Chen
- Department of PharmacologyChina Medical UniversityTaichungTaiwan
- Department and Graduate Institute of AquacultureNational Kaohsiung Marine UniversityKaohsiungTaiwan
| | - Chih‐Hsin Tang
- Department of Orthopedic SurgeryChina Medical University HospitalTaichungTaiwan
- Graduate Institute of Basic Medical ScienceChina Medical UniversityTaichungTaiwan
- Department of BiotechnologyCollege of Medical and Health ScienceAsia UniversityTaichungTaiwan
| | - Yuan‐Li Huang
- Department of BiotechnologyCollege of Medical and Health ScienceAsia UniversityTaichungTaiwan
- Department of Medical ResearchChina Medical University HospitalTaichungTaiwan
| |
Collapse
|
8
|
Masciantonio MG, Lee CKS, Arpino V, Mehta S, Gill SE. The Balance Between Metalloproteinases and TIMPs: Critical Regulator of Microvascular Endothelial Cell Function in Health and Disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 147:101-131. [PMID: 28413026 DOI: 10.1016/bs.pmbts.2017.01.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Endothelial cells (EC), especially the microvascular EC (MVEC), have critical functions in health and disease. For example, healthy MVEC provide a barrier between the fluid and protein found within the blood, and the surrounding tissue. Following tissue injury or infection, the microvascular barrier is often disrupted due to activation and dysfunction of the MVEC. Multiple mechanisms promote MVEC activation and dysfunction, including stimulation by cytokines, mechanical interaction with activated leukocytes, and exposure to harmful leukocyte-derived molecules, which collectively result in a loss of MVEC barrier function. However, MVEC activation is also critical to facilitate recruitment of inflammatory cells, such as neutrophils (PMNs) and monocytes, into the injured or infected tissue. Metalloproteinases, including the matrix metalloproteinases (MMPs) and the closely related, a disintegrin and metalloproteinases (ADAMs), have been implicated in regulating both MVEC barrier function, through cleavage of adherens and tight junctions proteins between adjacent MVEC and through degradation of the extracellular matrix, as well as PMN-MVEC interaction, through shedding of cell surface PMN receptors. Moreover, the tissue inhibitors of metalloproteinases (TIMPs), which collectively inhibit most MMPs and ADAMs, are critical regulators of MVEC activation and dysfunction through their ability to inhibit metalloproteinases and thereby promote MVEC stability. However, TIMPs have been also found to modulate MVEC function through metalloproteinase-independent mechanisms, such as regulation of vascular endothelial growth factor signaling. This chapter is focused on examining the role of the metalloproteinases and TIMPs in regulation of MVEC function in both health and disease.
Collapse
Affiliation(s)
- Marcello G Masciantonio
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON, Canada; Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Christopher K S Lee
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON, Canada; Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Valerie Arpino
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON, Canada; Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Sanjay Mehta
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON, Canada
| | - Sean E Gill
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON, Canada; Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.
| |
Collapse
|
9
|
Stratman AN, Pezoa SA, Farrelly OM, Castranova D, Dye LE, Butler MG, Sidik H, Talbot WS, Weinstein BM. Interactions between mural cells and endothelial cells stabilize the developing zebrafish dorsal aorta. Development 2016; 144:115-127. [PMID: 27913637 DOI: 10.1242/dev.143131] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 11/17/2016] [Indexed: 12/13/2022]
Abstract
Mural cells (vascular smooth muscle cells and pericytes) play an essential role in the development of the vasculature, promoting vascular quiescence and long-term vessel stabilization through their interactions with endothelial cells. However, the mechanistic details of how mural cells stabilize vessels are not fully understood. We have examined the emergence and functional role of mural cells investing the dorsal aorta during early development using the zebrafish. Consistent with previous literature, our data suggest that cells ensheathing the dorsal aorta emerge from a sub-population of cells in the adjacent sclerotome. Inhibition of mural cell recruitment to the dorsal aorta through disruption of pdgfr signaling leads to a reduced vascular basement membrane, which in turn results in enhanced dorsal aorta vessel elasticity and failure to restrict aortic diameter. Our results provide direct in vivo evidence for a functional role for mural cells in patterning and stabilization of the early vasculature through production and maintenance of the vascular basement membrane to prevent abnormal aortic expansion and elasticity.
Collapse
Affiliation(s)
- Amber N Stratman
- Program in Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sofia A Pezoa
- Program in Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Olivia M Farrelly
- Program in Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel Castranova
- Program in Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Louis E Dye
- Microscopy & Imaging Core, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Matthew G Butler
- Program in Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Harwin Sidik
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - William S Talbot
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Brant M Weinstein
- Program in Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
10
|
Huang CH, Ciou JS, Chen ST, Kok VC, Chung Y, Tsai JJP, Kurubanjerdjit N, Huang CYF, Ng KL. Identify potential drugs for cardiovascular diseases caused by stress-induced genes in vascular smooth muscle cells. PeerJ 2016; 4:e2478. [PMID: 27703845 PMCID: PMC5045879 DOI: 10.7717/peerj.2478] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 08/23/2016] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Abnormal proliferation of vascular smooth muscle cells (VSMC) is a major cause of cardiovascular diseases (CVDs). Many studies suggest that vascular injury triggers VSMC dedifferentiation, which results in VSMC changes from a contractile to a synthetic phenotype; however, the underlying molecular mechanisms are still unclear. METHODS In this study, we examined how VSMC responds under mechanical stress by using time-course microarray data. A three-phase study was proposed to investigate the stress-induced differentially expressed genes (DEGs) in VSMC. First, DEGs were identified by using the moderated t-statistics test. Second, more DEGs were inferred by using the Gaussian Graphical Model (GGM). Finally, the topological parameters-based method and cluster analysis approach were employed to predict the last batch of DEGs. To identify the potential drugs for vascular diseases involve VSMC proliferation, the drug-gene interaction database, Connectivity Map (cMap) was employed. Success of the predictions were determined using in-vitro data, i.e. MTT and clonogenic assay. RESULTS Based on the differential expression calculation, at least 23 DEGs were found, and the findings were qualified by previous studies on VSMC. The results of gene set enrichment analysis indicated that the most often found enriched biological processes are cell-cycle-related processes. Furthermore, more stress-induced genes, well supported by literature, were found by applying graph theory to the gene association network (GAN). Finally, we showed that by processing the cMap input queries with a cluster algorithm, we achieved a substantial increase in the number of potential drugs with experimental IC50 measurements. With this novel approach, we have not only successfully identified the DEGs, but also improved the DEGs prediction by performing the topological and cluster analysis. Moreover, the findings are remarkably validated and in line with the literature. Furthermore, the cMap and DrugBank resources were used to identify potential drugs and targeted genes for vascular diseases involve VSMC proliferation. Our findings are supported by in-vitro experimental IC50, binding activity data and clinical trials. CONCLUSION This study provides a systematic strategy to discover potential drugs and target genes, by which we hope to shed light on the treatments of VSMC proliferation associated diseases.
Collapse
Affiliation(s)
- Chien-Hung Huang
- Department of Computer Science and Information Engineering, National Formosa University, Yun-Lin, Taiwan
| | - Jin-Shuei Ciou
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| | - Shun-Tsung Chen
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| | - Victor C. Kok
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
- Division of Medical Oncology, Kuang Tien General Hospital Cancer Center, Taichung, Taiwan
| | - Yi Chung
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| | - Jeffrey J. P. Tsai
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| | | | - Chi-Ying F. Huang
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Ka-Lok Ng
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
11
|
Sphingosylphosphorylcholine inhibits macrophage adhesion to vascular smooth muscle cells. Biochem Pharmacol 2016; 115:43-50. [PMID: 27402344 PMCID: PMC4981491 DOI: 10.1016/j.bcp.2016.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 07/06/2016] [Indexed: 12/16/2022]
Abstract
Inflammation in de-endothelialised arteries contributes to the development of cardiovascular diseases. The process that initiates this inflammatory response is the adhesion of monocytes/macrophages to exposed vascular smooth muscle cells, typically stimulated by cytokines such as tumour necrosis factor-α (TNF). The aim of this study was to determine the effect of the sphingolipid sphingosylphosphorylcholine (SPC) on the interaction of monocytes/macrophages with vascular smooth muscle cells. Rat aortic smooth muscle cells and rat bone marrow-derived macrophages were co-cultured using an in vitro assay following incubation with sphingolipids to assess inter-cellular adhesion. We reveal that SPC inhibits the TNF-induced adhesion of macrophages to smooth muscle cells. This anti-adhesive effect was the result of SPC-induced changes to the smooth muscle cells (but not the macrophages) and was mediated, at least partly, via the sphingosine 1-phosphate receptor subtype 2. Lipid raft domains were also required. Although SPC did not alter expression or membrane distribution of the adhesion proteins intercellular adhesion molecule-1 and vascular cellular adhesion protein-1 in smooth muscle cells, SPC preincubation inhibited the TNF-induced increase in inducible nitric oxide synthase (NOS2) resulting in a subsequent decrease in nitric oxide production. Inhibiting NOS2 activation in smooth muscle cells led to a decrease in the adhesion of macrophages to smooth muscle cells. This study has therefore delineated a novel pathway which can inhibit the interaction between macrophages and vascular smooth muscle cells via SPC-induced repression of NOS2 expression. This mechanism could represent a potential drug target in vascular disease.
Collapse
|
12
|
Zhang F, Xia Y, Yan W, Zhang H, Zhou F, Zhao S, Wang W, Zhu D, Xin C, Lee Y, Zhang L, He Y, Gao E, Tao L. Sphingosine 1-phosphate signaling contributes to cardiac inflammation, dysfunction, and remodeling following myocardial infarction. Am J Physiol Heart Circ Physiol 2015; 310:H250-61. [PMID: 26589326 DOI: 10.1152/ajpheart.00372.2015] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 10/15/2015] [Indexed: 01/24/2023]
Abstract
Sphingosine 1-phosphate (S1P) mediates multiple pathophysiological effects in the cardiovascular system. However, the role of S1P signaling in pathological cardiac remodeling following myocardial infarction (MI) remains controversial. In this study, we found that cardiac S1P greatly increased post-MI, accompanied with a significant upregulation of cardiac sphingosine kinase-1 (SphK1) and S1P receptor 1 (S1PR1) expression. In MI-operated mice, inhibition of S1P production by using PF543 (the SphK1 inhibitor) ameliorated cardiac remodeling and dysfunction. Conversely, interruption of S1P degradation by inhibiting S1P lyase augmented cardiac S1P accumulation and exacerbated cardiac remodeling and dysfunction. In the cardiomyocyte, S1P directly activated proinflammatory responses via a S1PR1-dependent manner. Furthermore, activation of SphK1/S1P/S1PR1 signaling attributed to β1-adrenergic receptor stimulation-induced proinflammatory responses in the cardiomyocyte. Administration of FTY720, a functional S1PR1 antagonist, obviously blocked cardiac SphK1/S1P/S1PR1 signaling, ameliorated chronic cardiac inflammation, and then improved cardiac remodeling and dysfunction in vivo post-MI. In conclusion, our results demonstrate that cardiac SphK1/S1P/S1PR1 signaling plays an important role in the regulation of proinflammatory responses in the cardiomyocyte and targeting cardiac S1P signaling is a novel therapeutic strategy to improve post-MI cardiac remodeling and dysfunction.
Collapse
Affiliation(s)
- Fuyang Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi, China
| | - Yunlong Xia
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi, China
| | - Wenjuan Yan
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi, China
| | - Haoqiang Zhang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi, China; and
| | - Fen Zhou
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi, China
| | - Shihao Zhao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi, China
| | - Wei Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi, China
| | - Di Zhu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi, China
| | - Chao Xin
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi, China
| | - Yan Lee
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi, China
| | - Ling Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi, China
| | - Yuan He
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi, China
| | - Erhe Gao
- Center for Translational Medicine, Temple University, Philadelphia, Pennsylvania
| | - Ling Tao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi, China;
| |
Collapse
|
13
|
Masuda S, Matsuura K, Anazawa M, Iwamiya T, Shimizu T, Okano T. Formation of vascular network structures within cardiac cell sheets from mouse embryonic stem cells. Regen Ther 2015; 2:6-16. [PMID: 31245454 PMCID: PMC6581788 DOI: 10.1016/j.reth.2015.10.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 09/14/2015] [Accepted: 10/02/2015] [Indexed: 01/16/2023] Open
Abstract
Bioengineered cardiac tissues represent a promising strategy for regenerative medicine. However, methods of vascularization and suitable cell sources for tissue engineering and regenerative medicine have not yet been established. In this study, we developed methods for the induction of vascular endothelial cells from mouse embryonic stem (ES) cells using three-dimensional (3D) suspension culture, and fabricated cardiac cell sheets with a pre-vascularized structure by co-culture of mouse ES cell-derived endothelial cells. After induction, isolated CD31+ cells expressed several endothelial cell marker genes and exhibited the ability to form vascular network structures similar to CD31+ cells from neonatal mouse heart. Co-culture of ES cell-derived CD31+ cells with ES cell-derived cardiomyocytes and dermal fibroblasts resulted in the formation of cardiac cell sheets with microvascular network formation. In contrast, microvascular network formation was reduced in co-cultures without cardiomyocytes, suggesting that cardiomyocytes within the cell sheet might enhance vascular endothelial cell sprouting. Polymerase chain reaction array analysis revealed that the expression levels of several angiogenesis-related genes, including fibroblast growth factor 1 (FGF1), were up-regulated in co-culture with cardiomyocytes compared with cultures without cardiomyocytes. The microvascular network in the cardiac sheets was attenuated by treatment with anti-FGF1 antibody. These results indicate that 3D suspension culture methods may be used to prepare functional vascular endothelial cells from mouse ES cells, and that cardiomyocyte-mediated paracrine effects might be important for fabricating pre-vascularized cardiac cell sheets. We developed 3D culture to induce vascular endothelial cells from mouse ES cells. Pre-vascularized cardiac sheets from mouse ES cell-derived cells were prepared. ES cell-derived cardiomyocytes promote vascular network formation via secreted FGF1.
Collapse
Key Words
- 3D, three-dimensional
- Acvrl1, activin receptor like 1
- CXCR4, chemokine receptor type 4
- Cardiac cell sheet
- Col4a3, collagen type IV alpha 3
- Cxcl5, chemokine (C-X-C motif) ligand 5
- Dll4, delta-like ligand 4
- ES cell, embryonic stem cell
- EYFP, enhanced yellow fluorescent protein
- Efnb2, ephrin-B2
- Egf, epidermal growth factor
- Endothelial cell differentiation
- Ephb4, ephrin type-B receptor 4
- FBS, fetal bovine serum
- FGF1, fibroblast growth factor 1
- Fibroblast growth factor 1
- Flk1, fetal liver kinase 1
- Flt1, fms-related tyrosine kinase 1
- Gusb, glucuronidase, beta
- KDR, kinase insert domain receptor
- LIF, leukemia inhibitory factor
- MACS, magnetic-activated cell sorting
- Mdk, midkine
- Microvascular network formation
- Mouse embryonic stem cells
- NEAA, non-essential amino acids
- Notch1, Notch homolog 1
- Nr2f2, nuclear receptor subfamily 2, group f, member 2
- PCR, polymerase chain reaction
- Pecam1, platelet/endothelial adhesion molecule 1
- Pou5f1, POU class 5 homeobox 1
- SMA, smooth muscle actin
- TIMP2, tissue inhibitor of metalloproteinase 2
- Tgfa, transforming growth factor, alpha
- Three-dimensional suspension culture
- Tymp, thymidine phosphorylase
- VE-cadherin, vascular endothelial cadherin
- VEGF, vascular endothelial growth factor
- eNOS, endothelial nitric oxide synthase
- iPS cell, inducible pluripotent stem cell
- qRT-PCR, quantitative real-time PCR
Collapse
Affiliation(s)
- Shinako Masuda
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
| | - Katsuhisa Matsuura
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
| | - Mie Anazawa
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
| | - Takahiro Iwamiya
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
| | - Teruo Okano
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
| |
Collapse
|
14
|
Measuring dynamic cell-material interactions and remodeling during 3D human mesenchymal stem cell migration in hydrogels. Proc Natl Acad Sci U S A 2015; 112:E3757-64. [PMID: 26150508 DOI: 10.1073/pnas.1511304112] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Biomaterials that mimic aspects of the extracellular matrix by presenting a 3D microenvironment that cells can locally degrade and remodel are finding increased applications as wound-healing matrices, tissue engineering scaffolds, and even substrates for stem cell expansion. In vivo, cells do not simply reside in a static microenvironment, but instead, they dynamically reengineer their surroundings. For example, cells secrete proteases that degrade extracellular components, attach to the matrix through adhesive sites, and can exert traction forces on the local matrix, causing its spatial reorganization. Although biomaterials scaffolds provide initially well-defined microenvironments for 3D culture of cells, less is known about the changes that occur over time, especially local matrix remodeling that can play an integral role in directing cell behavior. Here, we use microrheology as a quantitative tool to characterize dynamic cellular remodeling of peptide-functionalized poly(ethylene glycol) (PEG) hydrogels that degrade in response to cell-secreted matrix metalloproteinases (MMPs). This technique allows measurement of spatial changes in material properties during migration of encapsulated cells and has a sensitivity that identifies regions where cells simply adhere to the matrix, as well as the extent of local cell remodeling of the material through MMP-mediated degradation. Collectively, these microrheological measurements provide insight into microscopic, cellular manipulation of the pericellular region that gives rise to macroscopic tracks created in scaffolds by migrating cells. This quantitative and predictable information should benefit the design of improved biomaterial scaffolds for medically relevant applications.
Collapse
|
15
|
Lilly B. We have contact: endothelial cell-smooth muscle cell interactions. Physiology (Bethesda) 2015; 29:234-41. [PMID: 24985327 DOI: 10.1152/physiol.00047.2013] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Blood vessels are composed of two primary cell types, endothelial cells and smooth muscle cells, each providing a unique contribution to vessel function. Signaling between these two cell types is essential for maintaining tone in mature vessels, and their communication is critical during development, and for repair and remodeling associated with blood vessel growth. This review will highlight the pathways that endothelial cells and smooth muscle cells utilize to communicate during vessel formation and discuss how disruptions in these pathways contribute to disease.
Collapse
Affiliation(s)
- Brenda Lilly
- Department of Pediatrics, Nationwide Children's Hospital, The Heart Center, The Ohio State University, Columbus, Ohio
| |
Collapse
|
16
|
de Faria Poloni J, Bonatto D. Systems Chemo-Biology and Transcriptomic Meta-Analysis Reveal the Molecular Roles of Bioactive Lipids in Cardiomyocyte Differentiation. J Cell Biochem 2015; 116:2018-31. [PMID: 25752681 DOI: 10.1002/jcb.25156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 03/03/2015] [Indexed: 11/12/2022]
Abstract
Lipids, which are essential constituents of biological membranes, play structural and functional roles in the cell. In recent years, certain lipids have been identified as regulatory signaling molecules and have been termed "bioactive lipids". Subsequently, the importance of bioactive lipids in stem cell differentiation and cardiogenesis has gained increasing recognition. Therefore, the aim of this study was to identify the biological processes underlying murine cardiac differentiation and the mechanisms by which bioactive lipids affect these processes. For this purpose, a transcriptomic meta-analysis of microarray and RNA-seq data from murine stem cells undergoing cardiogenic differentiation was performed. The differentially expressed genes identified via this meta-analysis, as well as bioactive lipids, were evaluated using systems chemo-biology tools. These data indicated that bioactive lipids are associated with the regulation of cell motility, cell adhesion, cytoskeletal rearrangement, and gene expression. Moreover, bioactive lipids integrate the signaling pathways involved in cell migration, the secretion and remodeling of extracellular matrix components, and the establishment of the cardiac phenotype. In conclusion, this study provides new insights into the contribution of bioactive lipids to the induction of cellular responses to various stimuli, which may originate from the extracellular environment and morphogens, and the manner in which this contribution directly affects murine heart morphogenesis.
Collapse
Affiliation(s)
- Joice de Faria Poloni
- Centro de Biotecnologia da Universidade Federal do Rio Grande do Sul, Departamento de Biologia Molecular e Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Diego Bonatto
- Centro de Biotecnologia da Universidade Federal do Rio Grande do Sul, Departamento de Biologia Molecular e Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
17
|
McCollum CW, Hans C, Shah S, Merchant FA, Gustafsson JÅ, Bondesson M. Embryonic exposure to sodium arsenite perturbs vascular development in zebrafish. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2014; 152:152-163. [PMID: 24768856 DOI: 10.1016/j.aquatox.2014.04.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 04/02/2014] [Accepted: 04/05/2014] [Indexed: 06/03/2023]
Abstract
Exposure to arsenic in its inorganic form, arsenite, causes adverse effects to many different organs and tissues. Here, we have investigated arsenite-induced adverse effects on vascular tissues in the model organism zebrafish, Danio rerio. Zebrafish embryos were exposed to arsenite at different exposure windows and the susceptibility to vascular tissue damage was recorded at 72hours post fertilization (hpf). Intersegmental vessel sprouting and growth was most perturbed by exposure to arsenite during the 24-48hpf window, while disruption in the condensation of the caudal vein plexus was more often observed at the 48-72hpf exposure window, reflecting when these structures develop during normal embryogenesis. The vascular growth rate was decreased by arsenite exposure, and deviated from that of control embryos at around 24-26.5hpf. We further mapped changes in expression of key regulators of angiogenesis and vasculogenesis. Downregulation of vascular endothelial growth factor receptor 1/fms-related tyrosine kinase 1 (vegfr1/flt1) expression was evident already at 24hpf, coinciding with the decreased vascular growth rate. At later time points, matrix metalloproteinase 9 (mmp9) expression was upregulated, suggesting that arsenite affects the composition of the extracellular matrix. In total, the expression of eight key factors involved in different aspects of vascularization was significantly altered by arsenic exposure. In conclusion, our results show that arsenite is a potent vascular disruptor in the developing zebrafish embryo, a finding that calls for an evaluation of arsenite as a developmental vascular toxicant in mammalian model systems.
Collapse
Affiliation(s)
- Catherine W McCollum
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA.
| | - Charu Hans
- Department of Computer Science, University of Houston, Houston, TX 77204, USA
| | - Shishir Shah
- Department of Computer Science, University of Houston, Houston, TX 77204, USA
| | - Fatima A Merchant
- Department of Computer Science, University of Houston, Houston, TX 77204, USA; Department of Engineering Technology, University of Houston, Houston, TX 77204, USA
| | - Jan-Åke Gustafsson
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Maria Bondesson
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| |
Collapse
|
18
|
Joo HJ, Seo HR, Jeong HE, Choi SC, Park JH, Yu CW, Hong SJ, Chung S, Lim DS. Smooth muscle progenitor cells from peripheral blood promote the neovascularization of endothelial colony-forming cells. Biochem Biophys Res Commun 2014; 449:405-11. [PMID: 24858689 DOI: 10.1016/j.bbrc.2014.05.061] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 05/15/2014] [Indexed: 11/24/2022]
Abstract
Proangiogenic cell therapy using autologous progenitors is a promising strategy for treating ischemic disease. Considering that neovascularization is a harmonized cellular process that involves both endothelial cells and vascular smooth muscle cells, peripheral blood-originating endothelial colony-forming cells (ECFCs) and smooth muscle progenitor cells (SMPCs), which are similar to mature endothelial cells and vascular smooth muscle cells, could be attractive cellular candidates to achieve therapeutic neovascularization. We successfully induced populations of two different vascular progenitor cells (ECFCs and SMPCs) from adult peripheral blood. Both progenitor cell types expressed endothelial-specific or smooth muscle-specific genes and markers, respectively. In a protein array focused on angiogenic cytokines, SMPCs demonstrated significantly higher expression of bFGF, EGF, TIMP2, ENA78, and TIMP1 compared to ECFCs. Conditioned medium from SMPCs and co-culture with SMPCs revealed that SMPCs promoted cell proliferation, migration, and the in vitro angiogenesis of ECFCs. Finally, co-transplantation of ECFCs and SMPCs induced robust in vivo neovascularization, as well as improved blood perfusion and tissue repair, in a mouse ischemic hindlimb model. Taken together, we have provided the first evidence of a cell therapy strategy for therapeutic neovascularization using two different types of autologous progenitors (ECFCs and SMPCs) derived from adult peripheral blood.
Collapse
Affiliation(s)
- Hyung Joon Joo
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Ha-Rim Seo
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Hyo Eun Jeong
- Department of Mechanical Engineering, Korea University, Seoul, Republic of Korea
| | - Seung-Cheol Choi
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Jae Hyung Park
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Cheol Woong Yu
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Soon Jun Hong
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Seok Chung
- Department of Mechanical Engineering, Korea University, Seoul, Republic of Korea
| | - Do-Sun Lim
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, Seoul, Republic of Korea.
| |
Collapse
|
19
|
Sphingosine-1-phosphate-induced Flk-1 transactivation stimulates mouse embryonic stem cell proliferation through S1P1/S1P3-dependent β-arrestin/c-Src pathways. Stem Cell Res 2014; 12:69-85. [DOI: 10.1016/j.scr.2013.08.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 08/08/2013] [Accepted: 08/29/2013] [Indexed: 01/21/2023] Open
|