Cargando…

Lysophosphatidic Acid Enhanced the Angiogenic Capability of Human Chondrocytes by Regulating Gi/NF-kB-Dependent Angiogenic Factor Expression

Lysophosphatidic acid (LPA) has been found to mediate myeloid differentiation, stimulate osteogenesis, alter cell proliferation and migration, and inhibit apoptosis in chondrocytes. The effect of LPA on the angiogenic capability of chondrocytes is not clear. This study aimed to investigate its effec...

Descripción completa

Detalles Bibliográficos
Autores principales: Chuang, Yi-Wen, Chang, Wen-Ming, Chen, Kai-Hua, Hong, Chang-Zern, Chang, Pey-Jium, Hsu, Hung-Chih
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2014
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4039431/
https://www.ncbi.nlm.nih.gov/pubmed/24879414
http://dx.doi.org/10.1371/journal.pone.0095180
_version_ 1782318486534488064
author Chuang, Yi-Wen
Chang, Wen-Ming
Chen, Kai-Hua
Hong, Chang-Zern
Chang, Pey-Jium
Hsu, Hung-Chih
author_facet Chuang, Yi-Wen
Chang, Wen-Ming
Chen, Kai-Hua
Hong, Chang-Zern
Chang, Pey-Jium
Hsu, Hung-Chih
author_sort Chuang, Yi-Wen
collection PubMed
description Lysophosphatidic acid (LPA) has been found to mediate myeloid differentiation, stimulate osteogenesis, alter cell proliferation and migration, and inhibit apoptosis in chondrocytes. The effect of LPA on the angiogenic capability of chondrocytes is not clear. This study aimed to investigate its effect on the angiogenic capability of human chondrocytes and the underlying mechanism of these effects. Human chondrocyte cell line, CHON-001, commercialized human chondrocytes (HC) derived from normal human articular cartilage, and human vascular endothelial cells (HUVECs) were used as cell models in this study. The angiogenic capability of chondrocytes was determined by capillary tube formation, monolayer permeability, cell migration, and cell proliferation. An angiogenesis protein array kit was used to evaluate the secretion of angiogenic factors in conditioned medium. Angiogenin, insulin-like growth factor-binding protein 1 (IGFBP-1), interleukin (IL)-8, monocyte chemoattractant protein-1 (MCP-1), matrix metalloproteinase (MMP)-9, and vascular endothelial growth factor (VEGF) mRNA and protein expressions were evaluated by Q-RT-PCR and EIA, respectively. LPA receptor (LPAR) expression was determined by RT-PCR. Signaling pathways were clarified using inhibitors, Western blot analysis, and reporter assays. The LPA treatment promoted the angiogenic capability of CHON-001 cells and HC, resulting in enhanced HUVEC capillary tube formation, monolayer permeability, migration, and cell growth. Angiogenin, IGFBP-1, IL-8, MCP-1, MMP-9, and VEGF mRNA and protein expressions were significantly enhanced in LPA-treated chondrocytes. LPA2, 3, 4 and 6 were expressed in CHON-001 and HC cells. Pretreatment with the Gi/o type G protein inhibitor, pertussis toxin (PTX), and the NF-kB inhibitor, PDTC, significantly inhibited LPA-induced angiogenin, IGFBP-1, IL-8, MCP-1, MMP-9, and VEGF expressions in chondrocytes. The PTX pretreatment also inhibited LPA-mediated NF-kB activation, suggesting the presence of active Gi/NF-kB signaling in CHON-001 and HC cells. The effect of LPA on the angiogenesis-inducing capacity of chondrocytes may be due to the increased angiogenesis factor expression via the Gi/NF-kB signaling pathway.
format Online
Article
Text
id pubmed-4039431
institution National Center for Biotechnology Information
language English
publishDate 2014
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-40394312014-06-02 Lysophosphatidic Acid Enhanced the Angiogenic Capability of Human Chondrocytes by Regulating Gi/NF-kB-Dependent Angiogenic Factor Expression Chuang, Yi-Wen Chang, Wen-Ming Chen, Kai-Hua Hong, Chang-Zern Chang, Pey-Jium Hsu, Hung-Chih PLoS One Research Article Lysophosphatidic acid (LPA) has been found to mediate myeloid differentiation, stimulate osteogenesis, alter cell proliferation and migration, and inhibit apoptosis in chondrocytes. The effect of LPA on the angiogenic capability of chondrocytes is not clear. This study aimed to investigate its effect on the angiogenic capability of human chondrocytes and the underlying mechanism of these effects. Human chondrocyte cell line, CHON-001, commercialized human chondrocytes (HC) derived from normal human articular cartilage, and human vascular endothelial cells (HUVECs) were used as cell models in this study. The angiogenic capability of chondrocytes was determined by capillary tube formation, monolayer permeability, cell migration, and cell proliferation. An angiogenesis protein array kit was used to evaluate the secretion of angiogenic factors in conditioned medium. Angiogenin, insulin-like growth factor-binding protein 1 (IGFBP-1), interleukin (IL)-8, monocyte chemoattractant protein-1 (MCP-1), matrix metalloproteinase (MMP)-9, and vascular endothelial growth factor (VEGF) mRNA and protein expressions were evaluated by Q-RT-PCR and EIA, respectively. LPA receptor (LPAR) expression was determined by RT-PCR. Signaling pathways were clarified using inhibitors, Western blot analysis, and reporter assays. The LPA treatment promoted the angiogenic capability of CHON-001 cells and HC, resulting in enhanced HUVEC capillary tube formation, monolayer permeability, migration, and cell growth. Angiogenin, IGFBP-1, IL-8, MCP-1, MMP-9, and VEGF mRNA and protein expressions were significantly enhanced in LPA-treated chondrocytes. LPA2, 3, 4 and 6 were expressed in CHON-001 and HC cells. Pretreatment with the Gi/o type G protein inhibitor, pertussis toxin (PTX), and the NF-kB inhibitor, PDTC, significantly inhibited LPA-induced angiogenin, IGFBP-1, IL-8, MCP-1, MMP-9, and VEGF expressions in chondrocytes. The PTX pretreatment also inhibited LPA-mediated NF-kB activation, suggesting the presence of active Gi/NF-kB signaling in CHON-001 and HC cells. The effect of LPA on the angiogenesis-inducing capacity of chondrocytes may be due to the increased angiogenesis factor expression via the Gi/NF-kB signaling pathway. Public Library of Science 2014-05-30 /pmc/articles/PMC4039431/ /pubmed/24879414 http://dx.doi.org/10.1371/journal.pone.0095180 Text en © 2014 Chuang et al http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are properly credited.
spellingShingle Research Article
Chuang, Yi-Wen
Chang, Wen-Ming
Chen, Kai-Hua
Hong, Chang-Zern
Chang, Pey-Jium
Hsu, Hung-Chih
Lysophosphatidic Acid Enhanced the Angiogenic Capability of Human Chondrocytes by Regulating Gi/NF-kB-Dependent Angiogenic Factor Expression
title Lysophosphatidic Acid Enhanced the Angiogenic Capability of Human Chondrocytes by Regulating Gi/NF-kB-Dependent Angiogenic Factor Expression
title_full Lysophosphatidic Acid Enhanced the Angiogenic Capability of Human Chondrocytes by Regulating Gi/NF-kB-Dependent Angiogenic Factor Expression
title_fullStr Lysophosphatidic Acid Enhanced the Angiogenic Capability of Human Chondrocytes by Regulating Gi/NF-kB-Dependent Angiogenic Factor Expression
title_full_unstemmed Lysophosphatidic Acid Enhanced the Angiogenic Capability of Human Chondrocytes by Regulating Gi/NF-kB-Dependent Angiogenic Factor Expression
title_short Lysophosphatidic Acid Enhanced the Angiogenic Capability of Human Chondrocytes by Regulating Gi/NF-kB-Dependent Angiogenic Factor Expression
title_sort lysophosphatidic acid enhanced the angiogenic capability of human chondrocytes by regulating gi/nf-kb-dependent angiogenic factor expression
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4039431/
https://www.ncbi.nlm.nih.gov/pubmed/24879414
http://dx.doi.org/10.1371/journal.pone.0095180
work_keys_str_mv AT chuangyiwen lysophosphatidicacidenhancedtheangiogeniccapabilityofhumanchondrocytesbyregulatingginfkbdependentangiogenicfactorexpression
AT changwenming lysophosphatidicacidenhancedtheangiogeniccapabilityofhumanchondrocytesbyregulatingginfkbdependentangiogenicfactorexpression
AT chenkaihua lysophosphatidicacidenhancedtheangiogeniccapabilityofhumanchondrocytesbyregulatingginfkbdependentangiogenicfactorexpression
AT hongchangzern lysophosphatidicacidenhancedtheangiogeniccapabilityofhumanchondrocytesbyregulatingginfkbdependentangiogenicfactorexpression
AT changpeyjium lysophosphatidicacidenhancedtheangiogeniccapabilityofhumanchondrocytesbyregulatingginfkbdependentangiogenicfactorexpression
AT hsuhungchih lysophosphatidicacidenhancedtheangiogeniccapabilityofhumanchondrocytesbyregulatingginfkbdependentangiogenicfactorexpression