1
|
Xu Q, Sun J, Holden CM, Neto HCF, Wang T, Zhang C, Fu Z, Joseph G, Shi R, Wang J, Leask A, Taylor WR, Lin Z. Cellular communication network factor 2 regulates smooth muscle cell transdifferentiation and lipid accumulation in atherosclerosis. Cardiovasc Res 2024:cvae215. [PMID: 39365752 DOI: 10.1093/cvr/cvae215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 07/17/2024] [Accepted: 08/06/2024] [Indexed: 10/06/2024] Open
Abstract
AIMS Accruing evidence illustrates an emerging paradigm of dynamic vascular smooth muscle cell (SMC) transdifferentiation during atherosclerosis progression. However, the molecular regulators that govern SMC phenotype diversification remain poorly defined. This study aims to elucidate the functional role and underlying mechanisms of cellular communication network factor 2 (CCN2), a matricellular protein, in regulating SMC plasticity in the context of atherosclerosis. METHODS AND RESULTS In both human and murine atherosclerosis, an up-regulation of CCN2 is observed in transdifferentiated SMCs. Using an inducible murine SMC CCN2 deletion model, we demonstrate that SMC-specific CCN2 knockout mice are hypersusceptible to atherosclerosis development as evidenced by a profound increase in lipid-rich plaques along the entire aorta. Single-cell RNA sequencing studies reveal that SMC deficiency of CCN2 positively regulates machinery involved in endoplasmic reticulum stress, endocytosis, and lipid accumulation in transdifferentiated macrophage-like SMCs during the progression of atherosclerosis, findings recapitulated in CCN2-deficient human aortic SMCs. CONCLUSION Our studies illuminate an unanticipated protective role of SMC-CCN2 against atherosclerosis. Disruption of vascular wall homeostasis resulting from vascular SMC CCN2 deficiency predisposes mice to atherosclerosis development and progression.
Collapse
Affiliation(s)
- Qian Xu
- Cardiology Division, Department of Medicine, Emory University School of Medicine, 1750 Haygood Drive, Atlanta, GA 30322, USA
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
| | - Jisheng Sun
- Cardiology Division, Department of Medicine, Emory University School of Medicine, 1750 Haygood Drive, Atlanta, GA 30322, USA
| | - Claire M Holden
- Cardiology Division, Department of Medicine, Emory University School of Medicine, 1750 Haygood Drive, Atlanta, GA 30322, USA
| | | | - Ti Wang
- Cardiology Division, Department of Medicine, Emory University School of Medicine, 1750 Haygood Drive, Atlanta, GA 30322, USA
- The Hospital Affiliated to Medical School of Yangzhou University (Taizhou People's Hospital), Yangzhou University Medical College, Jiangsu, China
| | - Chiyuan Zhang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
| | - Zuli Fu
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
| | - Giji Joseph
- Cardiology Division, Department of Medicine, Emory University School of Medicine, 1750 Haygood Drive, Atlanta, GA 30322, USA
| | - Ruizheng Shi
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
| | - Jinhu Wang
- Cardiology Division, Department of Medicine, Emory University School of Medicine, 1750 Haygood Drive, Atlanta, GA 30322, USA
| | - Andrew Leask
- College of Dentistry, University of Saskatchewan, 105 Wiggins Road, Saskatoon, SK, Canada
| | - W Robert Taylor
- Cardiology Division, Department of Medicine, Emory University School of Medicine, 1750 Haygood Drive, Atlanta, GA 30322, USA
| | - Zhiyong Lin
- Cardiology Division, Department of Medicine, Emory University School of Medicine, 1750 Haygood Drive, Atlanta, GA 30322, USA
| |
Collapse
|
2
|
Pervaiz N, Kathuria I, Aithabathula RV, Singla B. Matricellular proteins in atherosclerosis development. Matrix Biol 2023; 120:1-23. [PMID: 37086928 PMCID: PMC10225360 DOI: 10.1016/j.matbio.2023.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 04/24/2023]
Abstract
The extracellular matrix (ECM) is an intricate network composed of various multi-domain macromolecules like collagen, proteoglycans, and fibronectin, etc., that form a structurally stable composite, contributing to the mechanical properties of tissue. However, matricellular proteins are non-structural, secretory extracellular matrix proteins, which modulate various cellular functions via interacting with cell surface receptors, proteases, hormones, and cell-matrix. They play essential roles in maintaining tissue homeostasis by regulating cell differentiation, proliferation, adhesion, migration, and several signal transduction pathways. Matricellular proteins display a broad functionality regulated by their multiple structural domains and their ability to interact with different extracellular substrates and/or cell surface receptors. The expression of these proteins is low in adults, however, gets upregulated following injuries, inflammation, and during tumor growth. The marked elevation in the expression of these proteins during atherosclerosis suggests a positive association between their expression and atherosclerotic lesion formation. The role of matricellular proteins in atherosclerosis development has remained an area of research interest in the last two decades and studies revealed these proteins as important players in governing vascular function, remodeling, and plaque formation. Despite extensive research, many aspects of the matrix protein biology in atherosclerosis are still unknown and future studies are required to investigate whether targeting pathways stimulated by these proteins represent viable therapeutic approaches for patients with atherosclerotic vascular diseases. This review summarizes the characteristics of distinct matricellular proteins, discusses the available literature on the involvement of matrix proteins in the pathogenesis of atherosclerosis and suggests new avenues for future research.
Collapse
Affiliation(s)
- Naveed Pervaiz
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, USA
| | - Ishita Kathuria
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, USA
| | - Ravi Varma Aithabathula
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, USA
| | - Bhupesh Singla
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, USA.
| |
Collapse
|
3
|
Yeger H. CCN proteins: opportunities for clinical studies-a personal perspective. J Cell Commun Signal 2023:10.1007/s12079-023-00761-y. [PMID: 37195381 DOI: 10.1007/s12079-023-00761-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 05/01/2023] [Indexed: 05/18/2023] Open
Abstract
The diverse members of the CCN family now designated as CCN1(CYR61), CCN2 (CTGF), CCN3(NOV), CCN4(WISP1), CCN5(WISP2), CCN6(WISP3) are a conserved matricellular family of proteins exhibiting a spectrum of functional properties throughout all organs in the body. Interaction with cell membrane receptors such as integrins trigger intracellular signaling pathways. Proteolytically cleaved fragments (constituting the active domains) can be transported to the nucleus and perform transcriptional relevant functional activities. Notably, as also found in other protein families some members act opposite to others creating a system of functionally relevant checks and balances. It has become apparent that these proteins are secreted into the circulation, are quantifiable, and can serve as disease biomarkers. How they might also serve as homeostatic regulators is just becoming appreciated. In this review I have attempted to highlight the most recent evidence under the subcategories of cancer and non-cancer relevant that could lead to potential therapeutic approaches or ideas that can be factored into clinical advances. I have added my own personal perspective on feasibility.
Collapse
Affiliation(s)
- Herman Yeger
- Developmental and Stem Cell Biology, Research Institute, SickKids, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
4
|
Li S, Li S. Temperal and spatial expression of CCN1, CCN3, CCN4, CCN5 and CCN6 proteins in the developing postnatal teeth. J Cell Commun Signal 2023:10.1007/s12079-023-00758-7. [PMID: 37160590 DOI: 10.1007/s12079-023-00758-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/28/2023] [Indexed: 05/11/2023] Open
Abstract
CCN proteins are matricellular proteins and are important modulators of development and function of adult organs. However, there is no literature reporting the localization of CCN proteins during postnatal tooth development and the formation of periodontium. Therefore, the aim of our study was to investigate the expression of CCN1, CCN3, CCN4, CCN5 and CCN6 in the developing postnatal teeth. Wistar rats were used at postnatal (PN) 3.5, 7, 16 and 21 days and maxillas were processed for immunohistochemistry. At PN3.5 and PN7, preameloblasts (PA), secretory ameloblasts (SA), odontoblasts (OD) and dental pulp (DP) showed moderate to strong staining for CCN1, CCN4 and CCN6 respectively. CCN5 was intensely expressed in predentin, whereas CCN5 was undetectable in PA, SA, OD and DP. At PN16 and PN21, moderate to strong reaction with CCN1, CCN4 and CCN6 was evident in OD, DP, reduced enamel epithelium (REE), osteoblasts (OB) and periodontal ligament (PDL) respectively, while CCN5 was negative to weakly expressed in REE, OD, DP, OB, PDL and osteocytes (OC). Interestingly, the expression of CCN1, CCN4 and CCN6 was initially negative at PN16 but strong at PN21 in OC. Furthermore, there was no staining for CCN3 in the tissues studied. These results demonstrated that the expression pattern of CCN1, CCN4 and CCN6 is similar and inversely correlated with that of CCN3. CCN5 exhibits a unique distribution pattern. These data indicate that CCN proteins might play regulatory roles in amelogenesis, dentinogenesis, osteogenesis and PDL homeostasis.
Collapse
Affiliation(s)
- Shubo Li
- Department of Stomatology, College of Medicine and Health, Lishui University, Lishui, 323000, Zhejiang Province, China.
- Department of Stomatology, Affliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Province, China.
| | - Shufang Li
- Department of Stomatology, Affliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Province, China
| |
Collapse
|
5
|
Song Y, Li C, Luo Y, Guo J, Kang Y, Yin F, Ye L, Sun D, Yu J, Zhang X. CCN6 improves hepatic steatosis, inflammation, and fibrosis in non-alcoholic steatohepatitis. Liver Int 2023; 43:357-369. [PMID: 36156376 DOI: 10.1111/liv.15430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 07/20/2022] [Accepted: 09/20/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND AND AIMS CCN6 is a secretory protein with functions of maintaining mitochondrial homeostasis and anti-oxidative stress; and yet, whether it is involved in the pathogenesis of non-alcoholic steatohepatitis (NASH) is still obscure. We investigated the role and mechanism of CCN6 in the development of NASH. METHODS Human liver tissue samples were collected to detect the expression profile of CCN6. High-fat-high-cholesterol (HFHC) and methionine choline-deficient (MCD) diet were applied to mice to establish NASH animal models. Liver-specific overexpression of CCN6 was induced in mice by tail vein injection of adeno-associated virus (AAV), and then the effect of CCN6 on the course of NASH was observed. Free fatty acid (FFA) was applied to HepG2 cells to construct the cell model of steatosis, and the effect of CCN6 was investigated by knocking down the expression of CCN6 through small interfering RNA (siRNA) transfection. RESULTS We found that CCN6 expression was significantly downregulated in the liver of NASH. We confirmed that liver-specific overexpression of CCN6 significantly attenuated hepatic steatosis, inflammation response and fibrosis in NASH mice. Based on RNA-seq analysis, we revealed that CCN6 significantly affected the MAPK pathway. Then, by interfering with apoptosis signal-regulating kinase 1 (ASK1), we identified the ASK1/MAPK pathway pairs as the targets of CCN6 action. CONCLUSIONS CCN6 protects against hepatic steatosis, inflammation response and fibrosis by inhibiting the activation of ASK1 along with its downstream MAPK signalling. CCN6 may be a potential therapeutic target for the treatment of NASH.
Collapse
Affiliation(s)
- Yiran Song
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chenyang Li
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yuxin Luo
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jinbo Guo
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yaxing Kang
- Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Fengrong Yin
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lihong Ye
- Department of Pathology, Shijiazhuang Fifth Hospital, Shijiazhuang, China
| | - Donglei Sun
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jun Yu
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaolan Zhang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
6
|
Thankam FG, La V, Agrawal DK. Single-cell genomics illustrates heterogeneous phenotypes of myocardial fibroblasts under ischemic insults. Biochem Cell Biol 2023; 101:12-51. [PMID: 36458696 DOI: 10.1139/bcb-2022-0229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Myocardial regenerative strategies are promising where the choice of ideal cell population is crucial for successful translational applications. Herein, we explored the regenerative/repair responses of infarct zone cardiac fibroblast(s) (CF) by unveiling their phenotype heterogeneity at single-cell resolution. CF were isolated from the infarct zone of Yucatan miniswine that suffered myocardial infarction, cultured under simulated ischemic and reperfusion, and grouped into control, ischemia, and ischemia/reperfusion. The single-cell RNA sequencing analysis revealed 19 unique cell clusters suggesting distinct subpopulations. The status of gene expression (log2 fold change (log2 FC) > 2 and log2 FC < -2) was used to define the characteristics of each cluster unveiling with diverse features, including the pro-survival/cardioprotective (Clusters 1, 3, 5, 9, and 18), vasculoprotective (Clusters 2 and 5), anti-inflammatory (Clusters 4 and 17), proliferative (Clusters 4 and 5), nonproliferative (Clusters 6, 8, 11, 16, 17, and 18), proinflammatory (Cluster 6), profibrotic/pathologic (Clusters 8 and 19), antihypertrophic (Clusters 8 and 10), extracellular matrix restorative (Clusters 9 and 12), angiogenic (Cluster 16), and normal (Clusters 7 and 15) phenotypes. Further understanding of these unique phenotypes of CF will provide significant translational opportunities for myocardial regeneration and cardiac management.
Collapse
Affiliation(s)
- Finosh G Thankam
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Vy La
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
7
|
YAP Overexpression in Breast Cancer Cells Promotes Angiogenesis through Activating YAP Signaling in Vascular Endothelial Cells. Anal Cell Pathol (Amst) 2022; 2022:5942379. [PMID: 36226237 PMCID: PMC9550503 DOI: 10.1155/2022/5942379] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 08/16/2022] [Accepted: 08/31/2022] [Indexed: 11/17/2022] Open
Abstract
Purpose. The YAP signaling pathway is altered and implicated as oncogenic in human mammary cancers. However, roles of YAP signaling that regulate the breast tumor angiogenesis have remained elusive. Tumor angiogenesis is coordinated by the activation of both cancer cells and vascular endothelial cells. Whether the YAP signaling pathway can regulate the intercellular interaction between cancer cells and endothelial cells is essentially unknown. Methods. The effects of YAP on tumor angiogenesis, migration, and proliferation of vascular endothelial cells were evaluated in vitro. Expression of proteins and phosphorylating proteins involved in YAP, G13-RhoA, and PI3K/Akt signaling pathways was evaluated using the Western blotting, immunofluorescence staining, and immunohistochemistry analysis. In addition, the effects of YAP on breast cancer angiogenesis were evaluated in vivo by tumor xenograft mice. Results. We showed here that conditioned media from YAP overexpressed breast cancer cells (CM-YAP+) could promote angiogenesis, accompanied by increased tube formation, migration, and proliferation of human umbilical vein endothelial cells (HUVECs). Down regulation of YAP in HUVECs reversed CM-YAP+ induced angiogenesis. CM-YAP+ time-dependently activated YAP in HUVECs by dephosphorylating YAP and increasing nuclear translocation. We also identified that both G13-RhoA and PI3K/Akt signaling pathway were necessary for CM-YAP+ induced activation of YAP. Besides, connective tissue growth factor (CTGF) and angiopoietin-2 (ANG-2) acted as down-stream of YAP in HUVECs to promote angiogenesis. In addition, subcutaneous tumors nude mice model demonstrated that tumors overexpressed YAP revealed more neovascularization in vivo. Conclusion. YAP-YAP interaction between breast cancer cells and endothelial cells could promote tumor angiogenesis, supporting that YAP is a potential marker and target for developing novel therapeutic strategies against breast cancer.
Collapse
|
8
|
Pathak AS, Rojas M, Stouffer GA. Expression of Cyr61 in ApoE -/- mice with chronic unilateral renal artery ligation. Sci Rep 2021; 11:3606. [PMID: 33574403 PMCID: PMC7878479 DOI: 10.1038/s41598-021-81646-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 01/06/2021] [Indexed: 11/09/2022] Open
Abstract
Cyr61 is a member of the CCN family of proteins that is expressed in atherosclerotic lesions and regulated by angiotensin II. It is unknown whether renal artery stenosis (RAS) increases Cyr61 expression. Male ApoE−/− mice were randomized to surgically induced RAS, RAS + treatment with either irbesartan, aliskiren or amlodipine or sham-surgery. RAS resulted in increased plasma angiotensin II levels, a mild, sustained increase in systolic blood pressure and increased aortic lipid deposition compared to sham-surgery. Surgically induced RAS led to the formation of atheroma in the infrarenal aorta and there was consistent and intense staining for Cyr61 within the atheroma. Treatment with irbesartan, aliskiren and amlodipine were associated with decreased aortic lipid deposition and decreased staining for Cyr61 in aortic atheroma. Serum levels of Cyr61 were not increased in mice or humans with RAS. In summary, Cyr61 expression in aortic atheroma but not serum is increased by RAS in ApoE−/− mice and is reduced by agents that lower blood pressure.
Collapse
Affiliation(s)
- Alokkumar S Pathak
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Mauricio Rojas
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - George A Stouffer
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA. .,Division of Cardiology, University of North Carolina, Chapel Hill, NC, 27599-7075, USA.
| |
Collapse
|
9
|
Liao X, Bu Y, Xu Z, Jia F, Chang F, Liang J, Jia Q, Lv Y. WISP1 Predicts Clinical Prognosis and Is Associated With Tumor Purity, Immunocyte Infiltration, and Macrophage M2 Polarization in Pan-Cancer. Front Genet 2020; 11:502. [PMID: 32523603 PMCID: PMC7261883 DOI: 10.3389/fgene.2020.00502] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 04/23/2020] [Indexed: 12/30/2022] Open
Abstract
Cancer is becoming the leading cause of death and a major public health problem. Although many advanced treatment strategies are currently in use, the general prognosis of cancer patients remains dismal due to the high frequency of recurrence, metastasis. The identification of effective biomarkers is important for predicting survival of cancer patients and improving treatment efficacy. In this study, we comprehensively analyzed WNT1-inducible-signaling pathway protein 1 (WISP1) expression and explored its correlation with prognosis in pan-cancer using tumor IMmune Estimation Resource (TIMER) and Gene Expression Profiling Interactive Analysis 2 (GEPIA2). We also examined correlations between WISP1 and immunocyte infiltration using TIMER. We identified genes co-expressed with WISP1 using the LinkedOmics database and analyzed associated gene ontology using Metascape. Finally, we constructed protein-protein interaction networks and examined correlations between genes co-expressed with WISP1 and immunocyte infiltration in pan-cancer. WISP1 level differed between human pan-cancer tissues and normal tissues, indicating its potential as a prognostic biomarker. WISP1 expression was correlated with tumor purity and immunocyte infiltration, especially monocyte-macrophage trafficking and M2 polarization. Genes co-expressed with WISP1 were mainly associated with extracellular matrix organization, with collagen members COL6A3, COL5A1, and COL8A1 being key genes correlated with macrophage infiltration and M2 polarization in pan-cancer. Conversely, in certain types of cancer with better prognoses, WISP1 was associated with low M2 macrophage infiltration. These results suggest that WISP1 affect clinical prognosis through associations with tumor purity, immune cell infiltration, and macrophage M2 polarization in pan-cancer, with collagen member proteins may serving as effector molecules of WISP1.
Collapse
Affiliation(s)
- Xia Liao
- Department of Nutrition, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yang Bu
- Department of Hepatobiliary Surgery, General Hospital, Ningxia Medical University, Yinchuan, China
| | - Zihan Xu
- Department of Burns and Plastic Surgery, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Fengan Jia
- Metabolite Research Center, Shaanxi Institute of Microbiology, Xi'an, China
| | - Fan Chang
- Metabolite Research Center, Shaanxi Institute of Microbiology, Xi'an, China
| | - Junrong Liang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Qingan Jia
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yi Lv
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
10
|
Higher Serum CCN3 Is Associated with Disease Activity and Inflammatory Markers in Rheumatoid Arthritis. J Immunol Res 2020; 2020:3891425. [PMID: 32455138 PMCID: PMC7232667 DOI: 10.1155/2020/3891425] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 03/30/2020] [Indexed: 01/18/2023] Open
Abstract
Nephroblastoma overexpressed protein (NOV/CCN3), the early discovered member of the CCN family, has recently been suggested to be involved in a number of inflammatory processes, including wound healing, alveolar epithelial cell inflammation, cancer metastasis, and macrophage foam cell formation. However, the role of CCN3 in rheumatoid arthritis (RA), a classic autoimmune and inflammatory disease, remains elusive. RA is a chronic systemic autoimmune disease that eventually leads to cartilage and bone destruction and joint dysfunction. In this study, we investigated the potential of serum CCN3 as a biomarker for RA. The serum levels of CCN3 were measured by ELISA. The clinical and laboratory parameters were collected from a clinical record system, and disease activity was determined by joint disease activity score 28 (DAS28). Our results showed that the serum levels of CCN3 were significantly increased in RA patients compared to healthy controls. Furthermore, the CCN3 level was positively correlated with DAS28 (CRP), DAS28 (ESR), and the level of anti-CCP Ab, an autoantibody highly specific for RA. Furthermore, CCN3 showed a positive correlation with inflammatory cytokine IL-6, while no significant correlation with TNF-α was observed. These data suggest that CCN3 plays an important role in the development of RA and might be a potential disease activity biomarker for RA.
Collapse
|
11
|
Meyers CA, Xu J, Asatrian G, Ding C, Shen J, Broderick K, Ting K, Soo C, Peault B, James AW. WISP-1 drives bone formation at the expense of fat formation in human perivascular stem cells. Sci Rep 2018; 8:15618. [PMID: 30353078 PMCID: PMC6199241 DOI: 10.1038/s41598-018-34143-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 10/08/2018] [Indexed: 01/21/2023] Open
Abstract
The vascular wall within adipose tissue is a source of mesenchymal progenitors, referred to as perivascular stem/stromal cells (PSC). PSC are isolated via fluorescence activated cell sorting (FACS), and defined as a bipartite population of pericytes and adventitial progenitor cells (APCs). Those factors that promote the differentiation of PSC into bone or fat cell types are not well understood. Here, we observed high expression of WISP-1 among human PSC in vivo, after purification, and upon transplantation in a bone defect. Next, modulation of WISP-1 expression was performed, using WISP-1 overexpression, WISP-1 protein, or WISP-1 siRNA. Results demonstrated that WISP-1 is expressed in the perivascular niche, and high expression is maintained after purification of PSC, and upon transplantation in a bone microenvironment. In vitro studies demonstrate that WISP-1 has pro-osteogenic/anti-adipocytic effects in human PSC, and that regulation of BMP signaling activity may underlie these effects. In summary, our results demonstrate the importance of the matricellular protein WISP-1 in regulation of the differentiation of human stem cell types within the perivascular niche. WISP-1 signaling upregulation may be of future benefit in cell therapy mediated bone tissue engineering, for the healing of bone defects or other orthopaedic applications.
Collapse
Affiliation(s)
- Carolyn A Meyers
- Department of Pathology, Johns Hopkins University, Baltimore, 21205, United States
| | - Jiajia Xu
- Department of Pathology, Johns Hopkins University, Baltimore, 21205, United States
| | - Greg Asatrian
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA, California, Los Angeles, 90095, United States
| | - Catherine Ding
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA, California, Los Angeles, 90095, United States
| | - Jia Shen
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA, California, Los Angeles, 90095, United States
| | - Kristen Broderick
- Department of Plastic Surgery, Johns Hopkins University, 21205, Baltimore, United States
| | - Kang Ting
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA, California, Los Angeles, 90095, United States
| | - Chia Soo
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, California, Los Angeles, 90095, United States
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, California, Los Angeles, 90095, United States
| | - Bruno Peault
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, California, Los Angeles, 90095, United States
- Center For Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, 21205, United States.
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, California, Los Angeles, 90095, United States.
| |
Collapse
|
12
|
Chai KX, Chen YQ, Fan PL, Yang J, Yuan X. STROBE: The correlation of Cyr61, CTGF, and VEGF with polymyositis/dermatomyositis. Medicine (Baltimore) 2018; 97:e11775. [PMID: 30142763 PMCID: PMC6112880 DOI: 10.1097/md.0000000000011775] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
This study aims to explore the roles of cysteine-rich protein 61 (Cyr61/CCN1), connective tissue growth factor (CTGF/CCN2) and vascular endothelial growth factor (VEGF) in the vascular process of polymyositis (PM)/dermatomyositis (DM).Real-time quantitative polymerase chain reaction was used to determine the mRNA expression of Cyr61, CTGF, and VEGF in muscle tissues of initially treated PM/DM patients and controls. Enzyme-linked immunosorbent assay (ELISA) was used to determine the serum levels of Cyr61, CTGF, and VEGF of initially treated PM/DM patients before and after treatment. Data were statistically analyzed using statistical software SPSS 17.0.The mRNA expression levels of Cyr61, CTGF, and VEGF in muscle tissues were higher in the PM and DM groups than in the control group (P < .05). Differences in the mRNA expression levels of Cyr61, CTGF, and VEGF in muscle tissues between the PM and DM groups were not statistically significant (P > .05). Before treatment, the serum levels of Cyr61, CTGF, and VEGF were higher in the PM and DM groups than in the control group (P < .05). Furthermore, in the PM and DM groups, the expression levels of Cyr61, CTGF, and VEGF in serum at 6 months after treatment were lower than those before treatment (P < .05).Cyr61, CTGF, and VEGF are involved in the pathogenesis of PM/DM. These may be involved in the pathogenesis mainly by affecting the formation of blood vessels and promoting inflammatory response. This suggests that microvascular lesions play an important role in the immune pathogenesis of inflammatory myopathy PM/DM.
Collapse
Affiliation(s)
- Ke-Xia Chai
- Department of Rheumatism and Immunology, Affiliated Hospital of Qinghai University, Xining
| | - Yu-Qi Chen
- Department of Rheumatism and Immunology, The People's Hospital of SND, Suzhou
| | - Pei-Lin Fan
- Department of Medical Imaging Technology, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, China
| | - Jie Yang
- Department of Rheumatism and Immunology, Affiliated Hospital of Qinghai University, Xining
| | - Xia Yuan
- Department of Rheumatism and Immunology, Affiliated Hospital of Qinghai University, Xining
| |
Collapse
|
13
|
Abstract
The extracellular matrix (ECM) has central roles in tissue integrity and remodeling throughout the life span of animals. While collagens are the most abundant structural components of ECM in most tissues, tissue-specific molecular complexity is contributed by ECM glycoproteins. The matricellular glycoproteins are categorized primarily according to functional criteria and represented predominantly by the thrombospondin, tenascin, SPARC/osteonectin, and CCN families. These proteins do not self-assemble into ECM fibrils; nevertheless, they shape ECM properties through interactions with structural ECM proteins, growth factors, and cells. Matricellular proteins also promote cell migration or morphological changes through adhesion-modulating or counter-adhesive actions on cell-ECM adhesions, intracellular signaling, and the actin cytoskeleton. Typically, matricellular proteins are most highly expressed during embryonic development. In adult tissues, expression is more limited unless activated by cues for dynamic tissue remodeling and cell motility, such as occur during inflammatory response and wound repair. Many insights in the complex roles of matricellular proteins have been obtained from studies of gene knockout mice. However, with the exception of chordate-specific tenascins, these are highly conserved proteins that are encoded in many animal phyla. This review will consider the increasing body of research on matricellular proteins in nonmammalian animal models. These models provide better access to the very earliest stages of embryonic development and opportunities to study biological processes such as limb and organ regeneration. In aggregate, this research is expanding concepts of the functions and mechanisms of action of matricellular proteins.
Collapse
Affiliation(s)
- Josephine C Adams
- School of Biochemistry, University of Bristol, Bristol, United Kingdom.
| |
Collapse
|
14
|
Grünberg JR, Elvin J, Paul A, Hedjazifar S, Hammarstedt A, Smith U. CCN5/WISP2 and metabolic diseases. J Cell Commun Signal 2018; 12:309-318. [PMID: 29247377 PMCID: PMC5842198 DOI: 10.1007/s12079-017-0437-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 11/17/2017] [Indexed: 12/27/2022] Open
Abstract
Obesity and type 2 diabetes increase worldwide at an epidemic rate. It is expected that by the year 2030 around 500 million people will have diabetes; predominantly type 2 diabetes. The CCN family of proteins has become of interest in both metabolic and other common human diseases because of their effects on mesenchymal stem cell (MSCs) proliferation and differentiation as well as being important regulators of fibrosis. We here review current knowledge of the WNT1 inducible signaling pathway protein 2 (CCN5/WISP2). It has been shown to be an important regulator of both these processes through effects on both the canonical WNT and the TGFβ pathways. It is also under normal regulation by the adipogenic commitment factor BMP4, in contrast to conventional canonical WNT ligands, and allows MSCs to undergo normal adipose cell differentiation. CCN5/WISP2 is highly expressed in, and secreted by, MSCs and is an important regulator of MSCs growth. In a transgenic mouse model overexpressing CCN5/WISP2 in the adipose tissue, we have shown that it is secreted and circulating in the blood, the mice develop hypercellular white and brown adipose tissue, have increased lean body mass and enlarged hypercellular hearts. Obese transgenic mice had improved insulin sensitivity. Interestingly, the anti-fibrotic effect of CCN5/WISP2 is protective against heart failure by inhibition of the TGFβ pathway. Understanding how CCN5/WISP2 is regulated and signals is important and may be useful for developing new treatment strategies in obesity and metabolic diseases and it can also be a target in regenerative medicine.
Collapse
Affiliation(s)
- John R Grünberg
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK.
| | - Johannes Elvin
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy, University of Gothenburg, 405 30, Gothenburg, Sweden
| | - Alexandra Paul
- Department of Biology and Biological Engineering, Chalmers University of Technology, 41296, Gothenburg, Sweden
| | - Shahram Hedjazifar
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy, University of Gothenburg, 405 30, Gothenburg, Sweden
| | - Ann Hammarstedt
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy, University of Gothenburg, 405 30, Gothenburg, Sweden
| | - Ulf Smith
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy, University of Gothenburg, 405 30, Gothenburg, Sweden
| |
Collapse
|
15
|
Wu W, Hu X, Zhou X, Klenotic PA, Zhou Q, Lin Z. Myeloid deficiency of CCN3 exacerbates liver injury in a mouse model of nonalcoholic fatty liver disease. J Cell Commun Signal 2017; 12:389-399. [PMID: 29214510 DOI: 10.1007/s12079-017-0432-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 10/28/2017] [Indexed: 11/28/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a condition in which fat accumulates in the liver of patients without a prior history of alcohol abuse. The most severe form, nonalcoholic steatohepatitis (NASH), often leads to hepatic fibrosis and cirrhosis with ensuing complications. To date, there is no pharmacologic treatment for NASH. The biological effects of CCN3, specifically its role in the regulation of inflammation, reactive oxygen species production and angiogenesis, have been recently established. Additional data suggests that CCN3 is associated with the development of tumors in the liver yet may be protective of liver fibrogenesis. Currently, the role of CCN3 in NAFLD/NASH remains unexplored. Therefore, the objective of our investigation was to decipher the role of myeloid-deficient CCN3 in the pathogenesis of NASH and the underlying mechanisms of CCN3 in modulation of hepatic function. Wild type and myeloid CCN3-deficient mice were fed a methionine- and choline-deficient diet to induced NASH. Increased lipid, cholesterol, and cholesterol ester accumulation was observed in myeloid CCN3-deficient mice when compared to the control group. This disease state was associated with alterations of key genes involved in lipid synthesis, β-oxidation and lipid uptake. Additionally, the levels of important molecules critical for inflammation, ROS generation, ER stress and liver injury were significantly elevated; as was the observed severity of hepatic apoptosis and necroptosis. Therefore, CCN3 is critical for protection from hepatic apoptosis and necroptosis in our induced NASH model and our findings suggest that CCN3 can be exploited as a therapeutic target for the treatment of NASH.
Collapse
Affiliation(s)
- Wenconghui Wu
- Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Harrington Heart and Vascular Institute, University Hospitals Case Medical Center, 2103 Cornell Road, Room 4-541, Cleveland, OH, 44106, USA.,Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei Province, 430030, China.,Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan, China
| | - Xingjian Hu
- Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Harrington Heart and Vascular Institute, University Hospitals Case Medical Center, 2103 Cornell Road, Room 4-541, Cleveland, OH, 44106, USA.,Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xianming Zhou
- Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Harrington Heart and Vascular Institute, University Hospitals Case Medical Center, 2103 Cornell Road, Room 4-541, Cleveland, OH, 44106, USA.,Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Philip A Klenotic
- Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Harrington Heart and Vascular Institute, University Hospitals Case Medical Center, 2103 Cornell Road, Room 4-541, Cleveland, OH, 44106, USA
| | - Qi Zhou
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei Province, 430030, China
| | - Zhiyong Lin
- Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Harrington Heart and Vascular Institute, University Hospitals Case Medical Center, 2103 Cornell Road, Room 4-541, Cleveland, OH, 44106, USA.
| |
Collapse
|
16
|
Labrousse-Arias D, Martínez-Ruiz A, Calzada MJ. Hypoxia and Redox Signaling on Extracellular Matrix Remodeling: From Mechanisms to Pathological Implications. Antioxid Redox Signal 2017; 27:802-822. [PMID: 28715969 DOI: 10.1089/ars.2017.7275] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
SIGNIFICANCE The extracellular matrix (ECM) is an essential modulator of cell behavior that influences tissue organization. It has a strong relevance in homeostasis and translational implications for human disease. In addition to ECM structural proteins, matricellular proteins are important regulators of the ECM that are involved in a myriad of different pathologies. Recent Advances: Biochemical studies, animal models, and study of human diseases have contributed to the knowledge of molecular mechanisms involved in remodeling of the ECM, both in homeostasis and disease. Some of them might help in the development of new therapeutic strategies. This review aims to review what is known about some of the most studied matricellular proteins and their regulation by hypoxia and redox signaling, as well as the pathological implications of such regulation. CRITICAL ISSUES Matricellular proteins have complex regulatory functions and are modulated by hypoxia and redox signaling through diverse mechanisms, in some cases with controversial effects that can be cell or tissue specific and context dependent. Therefore, a better understanding of these regulatory processes would be of great benefit and will open new avenues of considerable therapeutic potential. FUTURE DIRECTIONS Characterizing the specific molecular mechanisms that modulate matricellular proteins in pathological processes that involve hypoxia and redox signaling warrants additional consideration to harness the potential therapeutic value of these regulatory proteins. Antioxid. Redox Signal. 27, 802-822.
Collapse
Affiliation(s)
- David Labrousse-Arias
- 1 Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP) , Madrid, Spain
| | - Antonio Martínez-Ruiz
- 1 Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP) , Madrid, Spain .,2 Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV) , Madrid, Spain
| | - María J Calzada
- 1 Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP) , Madrid, Spain .,3 Departmento de Medicina, Universidad Autónoma de Madrid , Madrid, Spain
| |
Collapse
|
17
|
Abstract
Tumours display considerable variation in the patterning and properties of angiogenic blood vessels, as well as in their responses to anti-angiogenic therapy. Angiogenic programming of neoplastic tissue is a multidimensional process regulated by cancer cells in concert with a variety of tumour-associated stromal cells and their bioactive products, which encompass cytokines and growth factors, the extracellular matrix and secreted microvesicles. In this Review, we discuss the extrinsic regulation of angiogenesis by the tumour microenvironment, highlighting potential vulnerabilities that could be targeted to improve the applicability and reach of anti-angiogenic cancer therapies.
Collapse
Affiliation(s)
- Michele De Palma
- The Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Daniela Biziato
- The Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Tatiana V Petrova
- Department of Fundamental Oncology, Ludwig Institute for Cancer Research and Division of Experimental Pathology, University of Lausanne and University of Lausanne Hospital, 1066 Lausanne, Switzerland
| |
Collapse
|
18
|
Siqueira M, Francis D, Gisbert D, Gomes FCA, Stipursky J. Radial Glia Cells Control Angiogenesis in the Developing Cerebral Cortex Through TGF-β1 Signaling. Mol Neurobiol 2017; 55:3660-3675. [PMID: 28523566 DOI: 10.1007/s12035-017-0557-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 04/12/2017] [Indexed: 10/19/2022]
Abstract
Neuroangiogenesis in the developing central nervous system is controlled by interactions between endothelial cells (ECs) and radial glia (RG) neural stem cells, although RG-derived molecules implicated in these events are not fully known. Here, we investigated the role of RG-secreted TGF-β1, in angiogenesis in the developing cerebral cortex. By isolation of murine microcapillary brain endothelial cells (MBECs), we demonstrate that conditioned medium from RG cultures (RG-CM) promoted MBEC migration and formation of vessel-like structures in vitro, in a TGF-β1-dependent manner. These events were followed by endothelial regulation of GPR124 and BAI-1 gene expression by RG-CM. Proteome profile of RG-CM identified angiogenesis-related molecules IGFBP2/3, osteopontin, endostatin, SDF1, fractalkine, TIMP1/4, Ang-1, pentraxin3, and Cyr61, some of them modulated by TGF-β1 induction. In vivo gain and loss of function assays targeting RG cells demonstrates a specific TGF-β1-dependent control of blood vessels branching in the cerebral cortex. Together, our results point to TGF-β1 signaling pathway as a potential mediator of the RG-EC interactions and shed light to the key role of RG in paving the brain vascular network.
Collapse
Affiliation(s)
- Michele Siqueira
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Daniel Francis
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Diego Gisbert
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Joice Stipursky
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil. .,Laboratório de Neurobiologia Celular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro - Centro de Ciências da Saúde, Bloco F, Sala F15, Ilha do Fundão, Rio de Janeiro, RJ, 21949-902, Brazil.
| |
Collapse
|
19
|
Extracellular Matrix, a Hard Player in Angiogenesis. Int J Mol Sci 2016; 17:ijms17111822. [PMID: 27809279 PMCID: PMC5133823 DOI: 10.3390/ijms17111822] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 09/30/2016] [Accepted: 10/21/2016] [Indexed: 12/11/2022] Open
Abstract
The extracellular matrix (ECM) is a complex network of proteins, glycoproteins, proteoglycans, and polysaccharides. Through multiple interactions with each other and the cell surface receptors, not only the ECM determines the physical and mechanical properties of the tissues, but also profoundly influences cell behavior and many physiological and pathological processes. One of the functions that have been extensively explored is its impingement on angiogenesis. The strong impact of the ECM in this context is both direct and indirect by virtue of its ability to interact and/or store several growth factors and cytokines. The aim of this review is to provide some examples of the complex molecular mechanisms that are elicited by these molecules in promoting or weakening the angiogenic processes. The scenario is intricate, since matrix remodeling often generates fragments displaying opposite effects compared to those exerted by the whole molecules. Thus, the balance will tilt towards angiogenesis or angiostasis depending on the relative expression of pro- or anti-angiogenetic molecules/fragments composing the matrix of a given tissue. One of the vital aspects of this field of research is that, for its endogenous nature, the ECM can be viewed as a reservoir to draw from for the development of new more efficacious therapies to treat angiogenesis-dependent pathologies.
Collapse
|