1
|
Meng L, Chen HM, Zhang JS, Wu YR, Xu YZ. Matricellular proteins: From cardiac homeostasis to immune regulation. Biomed Pharmacother 2024; 180:117463. [PMID: 39305814 DOI: 10.1016/j.biopha.2024.117463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/23/2024] [Accepted: 09/19/2024] [Indexed: 11/14/2024] Open
Abstract
Tissue repair after myocardial injury is a complex process involving changes in all aspects of the myocardial tissue, including the extracellular matrix (ECM). The ECM is composed of large structural proteins such as collagen and elastin and smaller proteins with major regulatory properties called matricellular proteins. Matricellular cell proteins exert their functions and elicit cellular responses by binding to structural proteins not limited to interactions with cell surface receptors, cytokines, or proteases. At the same time, matricellular proteins act as the "bridge" of information exchange between cells and ECM, maintaining the integrity of the cardiac structure and regulating the immune environment, which is a key factor in determining cardiac homeostasis. In this review, we present an overview of the identified matricellular proteins and summarize the current knowledge regarding their roles in maintaining cardiac homeostasis and regulating the immune system.
Collapse
Affiliation(s)
- Li Meng
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou 310053, China; Department of Cardiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Zhejiang 310006, China
| | - Hui-Min Chen
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou 310053, China; Department of Cardiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Zhejiang 310006, China
| | - Jia-Sheng Zhang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou 310053, China; Department of Cardiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Zhejiang 310006, China
| | - Yi-Rong Wu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Zhejiang 310006, China.
| | - Yi-Zhou Xu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Zhejiang 310006, China.
| |
Collapse
|
2
|
Qiao B, Liu X, Wang B, Wei S. The role of periostin in cardiac fibrosis. Heart Fail Rev 2024; 29:191-206. [PMID: 37870704 DOI: 10.1007/s10741-023-10361-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/10/2023] [Indexed: 10/24/2023]
Abstract
Cardiac fibrosis, which is the buildup of proteins in the connective tissues of the heart, can lead to end-stage extracellular matrix (ECM) remodeling and ultimately heart failure. Cardiac remodeling involves changes in gene expression in cardiac cells and ECM, which significantly leads to the morbidity and mortality in heart failure. However, despite extensive research, the elusive intricacies underlying cardiac fibrosis remain unidentified. Periostin, an extracellular matrix (ECM) protein of the fasciclin superfamily, acts as a scaffold for building complex architectures in the ECM, which improves intermolecular interactions and augments the mechanical properties of connective tissues. Recent research has shown that periostin not only contributes to normal ECM homeostasis in a healthy heart but also serves as a potent inducible regulator of cellular reorganization in cardiac fibrosis. Here, we reviewed the constitutive domain of periostin and its interaction with other ECM proteins. We have also discussed the critical pathophysiological functions of periostin in cardiac remodeling mechanisms, including two distinct yet potentially intertwined mechanisms. Furthermore, we will focus on the intrinsic complexities within periostin research, particularly surrounding the contentious issues observed in experimental findings.
Collapse
Affiliation(s)
- Bao Qiao
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Xuehao Liu
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Bailu Wang
- Clinical Trial Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Shujian Wei
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China.
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China.
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
3
|
Pihlström S, Määttä K, Öhman T, Mäkitie RE, Aronen M, Varjosalo M, Mäkitie O, Pekkinen M. A multi-omics study to characterize the transdifferentiation of human dermal fibroblasts to osteoblast-like cells. Front Mol Biosci 2022; 9:1032026. [PMID: 36465561 PMCID: PMC9714459 DOI: 10.3389/fmolb.2022.1032026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/26/2022] [Indexed: 09/19/2023] Open
Abstract
Background: Various skeletal disorders display defects in osteoblast development and function. An in vitro model can help to understand underlying disease mechanisms. Currently, access to appropriate starting material for in vitro osteoblastic studies is limited. Native osteoblasts and their progenitors, the bone marrow mesenchymal stem cells, (MSCs) are problematic to isolate from affected patients and challenging to expand in vitro. Human dermal fibroblasts in vitro are a promising substitute source of cells. Method: We developed an in vitro culturing technique to transdifferentiate fibroblasts into osteoblast-like cells. We obtained human fibroblasts from forearm skin biopsy and differentiated them into osteoblast-like cells with ß-glycerophosphate, ascorbic acid, and dexamethasone treatment. Osteoblastic phenotype was confirmed by staining for alkaline phosphatase (ALP), calcium and phosphate deposits (Alizarin Red, Von Kossa) and by a multi-omics approach (transcriptomic, proteomic, and phosphoproteomic analyses). Result: After 14 days of treatment, both fibroblasts and MSCs (reference cells) stained positive for ALP together with a significant increase in bone specific ALP (p = 0.04 and 0.004, respectively) compared to untreated cells. At a later time point, both cell types deposited minerals, indicating mineralization. In addition, fibroblasts and MSCs showed elevated expression of several osteogenic genes (e.g. ALPL, RUNX2, BMPs and SMADs), and decreased expression of SOX9. Ingenuity Pathways Analysis of RNA sequencing data from fibroblasts and MSCs showed that the osteoarthritis pathway was activated in both cell types (p_adj. = 0.003 and 0.004, respectively). Discussion: These data indicate that our in vitro treatment induces osteoblast-like differentiation in fibroblasts and MSCs, producing an in vitro osteoblastic cell system. This culturing system provides an alternative tool for bone biology research and skeletal tissue engineering.
Collapse
Affiliation(s)
- Sandra Pihlström
- Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kirsi Määttä
- Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tiina Öhman
- Institute of Biotechnology and Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Riikka E. Mäkitie
- Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Otorhinolaryngology—Head and Neck Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Mira Aronen
- Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
| | - Markku Varjosalo
- Institute of Biotechnology and Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Outi Mäkitie
- Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Children’s Hospital, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- Department of Molecular Medicine and Surgery and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Minna Pekkinen
- Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Children’s Hospital, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
4
|
Dorafshan S, Razmi M, Safaei S, Gentilin E, Madjd Z, Ghods R. Periostin: biology and function in cancer. Cancer Cell Int 2022; 22:315. [PMID: 36224629 PMCID: PMC9555118 DOI: 10.1186/s12935-022-02714-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 09/13/2022] [Indexed: 11/24/2022] Open
Abstract
Periostin (POSTN), a member of the matricellular protein family, is a secreted adhesion-related protein produced in the periosteum and periodontal ligaments. Matricellular proteins are a nonstructural family of extracellular matrix (ECM) proteins that regulate a wide range of biological processes in both normal and pathological conditions. Recent studies have demonstrated the key roles of these ECM proteins in the tumor microenvironment. Furthermore, periostin is an essential regulator of bone and tooth formation and maintenance, as well as cardiac development. Also, periostin interacts with multiple cell-surface receptors, especially integrins, and triggers signals that promote tumor growth. According to recent studies, these signals are implicated in cancer cell survival, epithelial-mesenchymal transition (EMT), invasion, and metastasis. In this review, we will summarize the most current data regarding periostin, its structure and isoforms, expressions, functions, and regulation in normal and cancerous tissues. Emphasis is placed on its association with cancer progression, and also future potential for periostin-targeted therapeutic approaches will be explored.
Collapse
Affiliation(s)
- Shima Dorafshan
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Mahdieh Razmi
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Sadegh Safaei
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Erica Gentilin
- Bioacoustics Research Laboratory, Department of Neurosciences, University of Padua, via G. Orus, 2b, 35129, Padua, Italy
| | - Zahra Madjd
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran.
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| | - Roya Ghods
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran.
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| |
Collapse
|
5
|
Trinh K, Julovi SM, Rogers NM. The Role of Matrix Proteins in Cardiac Pathology. Int J Mol Sci 2022; 23:ijms23031338. [PMID: 35163259 PMCID: PMC8836004 DOI: 10.3390/ijms23031338] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/15/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023] Open
Abstract
The extracellular matrix (ECM) and ECM-regulatory proteins mediate structural and cell-cell interactions that are crucial for embryonic cardiac development and postnatal homeostasis, as well as organ remodeling and repair in response to injury. These proteins possess a broad functionality that is regulated by multiple structural domains and dependent on their ability to interact with extracellular substrates and/or cell surface receptors. Several different cell types (cardiomyocytes, fibroblasts, endothelial and inflammatory cells) within the myocardium elaborate ECM proteins, and their role in cardiovascular (patho)physiology has been increasingly recognized. This has stimulated robust research dissecting the ECM protein function in human health and disease and replicating the genetic proof-of-principle. This review summarizes recent developments regarding the contribution of ECM to cardiovascular disease. The clear importance of this heterogeneous group of proteins in attenuating maladaptive repair responses provides an impetus for further investigation into these proteins as potential pharmacological targets in cardiac diseases and beyond.
Collapse
Affiliation(s)
- Katie Trinh
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; (K.T.); (S.M.J.)
- Faculty of Medicine and Health Sydney, School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Sohel M. Julovi
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; (K.T.); (S.M.J.)
- Faculty of Medicine and Health Sydney, School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Natasha M. Rogers
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; (K.T.); (S.M.J.)
- Faculty of Medicine and Health Sydney, School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia
- Renal and Transplantation Medicine, Westmead Hospital, Westmead, NSW 2145, Australia
- Correspondence:
| |
Collapse
|
6
|
Rajagopal S, Gupta A, Parveen R, Shukla N, Bhattacharya S, Naravula J, Kumar S A, Mathur P, Simlot A, Mehta S, Bihari C, Mehta S, Mishra AK, Nair BG, Medicherla KM, Reddy GB, Sreenivasulu N, Kishor PK, Suravajhala P. Vitamin K in human health and metabolism: A nutri-genomics review. Trends Food Sci Technol 2022. [DOI: 10.1016/j.tifs.2021.12.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
7
|
Opitz FV, Haeberle L, Daum A, Esposito I. Tumor Microenvironment in Pancreatic Intraepithelial Neoplasia. Cancers (Basel) 2021; 13:cancers13246188. [PMID: 34944807 PMCID: PMC8699458 DOI: 10.3390/cancers13246188] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Pancreatic ductal adenocarcinoma (PDAC) is a very aggressive neoplasm with a poor survival rate. This is mainly due to late detection, which substantially limits therapy options. A better understanding of the early phases of pancreatic carcinogenesis is fundamental for improving patient prognosis in the future. In this article, we focused on the tumor microenvironment (TME), which provides the biological niche for the development of PDAC from its most common precursor lesions, PanIN (pancreatic intraepithelial neoplasias). Abstract Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive tumors with a poor prognosis. A characteristic of PDAC is the formation of an immunosuppressive tumor microenvironment (TME) that facilitates bypassing of the immune surveillance. The TME consists of a desmoplastic stroma, largely composed of cancer-associated fibroblasts (CAFs), immunosuppressive immune cells, immunoregulatory soluble factors, neural network cells, and endothelial cells with complex interactions. PDAC develops from various precursor lesions such as pancreatic intraepithelial neoplasia (PanIN), intraductal papillary mucinous neoplasms (IPMN), mucinous cystic neoplasms (MCN), and possibly, atypical flat lesions (AFL). In this review, we focus on the composition of the TME in PanINs to reveal detailed insights into the complex restructuring of the TME at early time points in PDAC progression and to explore ways of modifying the TME to slow or even halt tumor progression.
Collapse
|
8
|
Dwyer KD, Coulombe KL. Cardiac mechanostructure: Using mechanics and anisotropy as inspiration for developing epicardial therapies in treating myocardial infarction. Bioact Mater 2021; 6:2198-2220. [PMID: 33553810 PMCID: PMC7822956 DOI: 10.1016/j.bioactmat.2020.12.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 12/14/2022] Open
Abstract
The mechanical environment and anisotropic structure of the heart modulate cardiac function at the cellular, tissue and organ levels. During myocardial infarction (MI) and subsequent healing, however, this landscape changes significantly. In order to engineer cardiac biomaterials with the appropriate properties to enhance function after MI, the changes in the myocardium induced by MI must be clearly identified. In this review, we focus on the mechanical and structural properties of the healthy and infarcted myocardium in order to gain insight about the environment in which biomaterial-based cardiac therapies are expected to perform and the functional deficiencies caused by MI that the therapy must address. From this understanding, we discuss epicardial therapies for MI inspired by the mechanics and anisotropy of the heart focusing on passive devices, which feature a biomaterials approach, and active devices, which feature robotic and cellular components. Through this review, a detailed analysis is provided in order to inspire further development and translation of epicardial therapies for MI.
Collapse
Affiliation(s)
- Kiera D. Dwyer
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, USA
| | - Kareen L.K. Coulombe
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, USA
| |
Collapse
|
9
|
Aujla PK, Kassiri Z. Diverse origins and activation of fibroblasts in cardiac fibrosis. Cell Signal 2020; 78:109869. [PMID: 33278559 DOI: 10.1016/j.cellsig.2020.109869] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 12/21/2022]
Abstract
Cardiac fibroblasts (cFBs) have emerged as a heterogenous cell population. Fibroblasts are considered the main cell source for synthesis of the extracellular matrix (ECM) and as such a dysregulation in cFB function, activity, or viability can lead to disrupted ECM structure or fibrosis. Fibrosis can be initiated in response to different injuries and stimuli, and can be reparative (beneficial) or reactive (damaging). FBs need to be activated to myofibroblasts (MyoFBs) which have augmented capacity in synthesizing ECM proteins, causing fibrosis. In addition to the resident FBs in the myocardium, a number of other cells (pericytes, fibrocytes, mesenchymal, and hematopoietic cells) can transform into MyoFBs, further driving the fibrotic response. Multiple molecules including hormones, cytokines, and growth factors stimulate this process leading to generation of activated MyoFBs. Contribution of different cell types to cFBs and MyoFBs can result in an exponential increase in the number of MyoFBs and an accelerated pro-fibrotic response. Given the diversity of the cell sources, and the array of interconnected signalling pathways that lead to formation of MyoFBs and subsequently fibrosis, identifying a single target to limit the fibrotic response in the myocardium has been challenging. This review article will delineate the importance and relevance of fibroblast heterogeneity in mediating fibrosis in different models of heart failure and will highlight important signalling pathways implicated in myofibroblast activation.
Collapse
Affiliation(s)
- Preetinder K Aujla
- Department of Physiology, Cardiovascular Research Center, University of Alberta, Edmonton, Alberta, Canada
| | - Zamaneh Kassiri
- Department of Physiology, Cardiovascular Research Center, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
10
|
Rachner TD, Göbel A, Hoffmann O, Erdmann K, Kasimir-Bauer S, Breining D, Kimmig R, Hofbauer LC, Bittner AK. High serum levels of periostin are associated with a poor survival in breast cancer. Breast Cancer Res Treat 2020; 180:515-524. [PMID: 32040688 DOI: 10.1007/s10549-020-05570-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 02/03/2020] [Indexed: 12/21/2022]
Abstract
PURPOSE Periostin is a secreted extracellular matrix protein, which was originally described in osteoblasts. It supports osteoblastic differentiation and bone formation and has been implicated in the pathogenesis of several human malignancies, including breast cancer. However, little is known about the prognostic value of serum periostin levels in breast cancer. METHODS In this study, we analyzed serum levels of periostin in a cohort of 509 primary, non-metastatic breast cancer patients. Disseminated tumor cell (DTC) status was determined using bone marrow aspirates obtained from the anterior iliac crests. Periostin levels were stratified according to several clinical parameters and Pearson correlation analyses were performed. Kaplan-Meier survival curves were assessed by using the log-rank (Mantel-Cox) test. To identify prognostic factors, multivariate Cox regression analyses were used. RESULTS Mean serum levels of periostin were 505 ± 179 pmol/l. In older patients (> 60 years), periostin serum levels were significantly increased compared to younger patients (540 ± 184 pmol/l vs. 469 ± 167 pmol/l; p < 0.0001) and age was positively correlated with periostin expression (p < 0.0001). When stratifying the cohort according to periostin serum concentrations, the overall and breast cancer-specific mortality were significantly higher in those patients with high serum periostin (above median) compared to those with low periostin during a mean follow-up of 8.5 years (17.7% vs. 11.4% breast cancer-specific death; p = 0.03; hazard ratio 1.65). Periostin was confirmed to be an independent prognostic marker for breast cancer-specific survival (p = 0.017; hazard ratio 1.79). No significant differences in serum periostin were observed when stratifying the patients according to their DTC status. CONCLUSIONS Our findings emphasize the relevance of periostin in breast cancer and reveal serum periostin as a potential marker for disease prediction, independent on the presence of micrometastases.
Collapse
Affiliation(s)
- Tilman D Rachner
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Ageing Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andy Göbel
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany. .,Center for Healthy Ageing Department of Medicine III, Technische Universität Dresden, Dresden, Germany. .,German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Oliver Hoffmann
- Department of Gynecology and Obstetrics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Kati Erdmann
- Department of Urology, Technische Universität Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Helmholtz Association / Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Sabine Kasimir-Bauer
- Department of Gynecology and Obstetrics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Dorit Breining
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Ageing Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rainer Kimmig
- Department of Gynecology and Obstetrics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Lorenz C Hofbauer
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Ageing Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ann-Kathrin Bittner
- Department of Gynecology and Obstetrics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
11
|
Fibronectin and Periostin as Prognostic Markers in Ovarian Cancer. Cells 2020; 9:cells9010149. [PMID: 31936272 PMCID: PMC7016975 DOI: 10.3390/cells9010149] [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: 12/07/2019] [Revised: 12/30/2019] [Accepted: 01/05/2020] [Indexed: 12/28/2022] Open
Abstract
Previously, based on a DNA microarray experiment, we identified a 96-gene prognostic signature associated with the shorter survival of ovarian cancer patients. We hypothesized that some differentially expressed protein-coding genes from this signature could potentially serve as prognostic markers. The present study was aimed to validate two proteins, namely fibronectin (FN1) and periostin (POSTN), in the independent set of ovarian cancer samples. Both proteins are mainly known as extracellular matrix proteins with many important functions in physiology. However, there are also indications that they are implicated in cancer, including ovarian cancer. The expression of these proteins was immunohistochemically analyzed in 108 surgical samples of advanced ovarian cancer (majority: high-grade serous) and additionally on tissue arrays representing different stages of the progression of ovarian and fallopian tube epithelial tumors, from normal epithelia, through benign tumors, to adenocarcinomas of different stages. The correlation with clinical, pathological, and molecular features was evaluated. Kaplan-Meier survival analysis and Cox-proportional hazards models were used to estimate the correlation of the expression levels these proteins with survival. We observed that the higher expression of fibronectin in the tumor stroma was highly associated with shorter overall survival (OS) (Kaplan-Meier analysis, log-rank test p = 0.003). Periostin was also associated with shorter OS (p = 0.04). When we analyzed the combined score, calculated by adding together individual scores for stromal fibronectin and periostin expression, Cox regression demonstrated that this joint FN1&POSTN score was an independent prognostic factor for OS (HR = 2.16; 95% CI: 1.02-4.60; p = 0.044). The expression of fibronectin and periostin was also associated with the source of ovarian tumor sample: metastases showed higher expression of these proteins than primary tumor samples (χ2 test, p = 0.024 and p = 0.032). Elevated expression of fibronectin and periostin was also more common in fallopian cancers than in ovarian cancers. Our results support some previous observations that fibronectin and periostin have a prognostic significance in ovarian cancer. In addition, we propose the joint FN1&POSTN score as an independent prognostic factor for OS. Based on our results, it may also be speculated that these proteins are related to tumor progression and/or may indicate fallopian-epithelial origin of the tumor.
Collapse
|
12
|
Rigoglio NN, Rabelo ACS, Borghesi J, de Sá Schiavo Matias G, Fratini P, Prazeres PHDM, Pimentel CMMM, Birbrair A, Miglino MA. The Tumor Microenvironment: Focus on Extracellular Matrix. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1245:1-38. [PMID: 32266651 DOI: 10.1007/978-3-030-40146-7_1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The extracellular matrix (ECM) regulates the development and maintains tissue homeostasis. The ECM is composed of a complex network of molecules presenting distinct biochemical properties to regulate cell growth, survival, motility, and differentiation. Among their components, proteoglycans (PGs) are considered one of the main components of ECM. Its composition, biomechanics, and anisotropy are exquisitely tuned to reflect the physiological state of the tissue. The loss of ECM's homeostasis is seen as one of the hallmarks of cancer and, typically, defines transitional events in tumor progression and metastasis. In this chapter, we discuss the types of proteoglycans and their roles in cancer. It has been observed that the amount of some ECM components is increased, while others are decreased, depending on the type of tumor. However, both conditions corroborate with tumor progression and malignancy. Therefore, ECM components have an increasingly important role in carcinogenesis and this leads us to believe that their understanding may be a key in the discovery of new anti-tumor therapies. In this book, the main ECM components will be discussed in more detail in each chapter.
Collapse
Affiliation(s)
- Nathia Nathaly Rigoglio
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Ana Carolina Silveira Rabelo
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Jessica Borghesi
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Gustavo de Sá Schiavo Matias
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Paula Fratini
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| | | | | | - Alexander Birbrair
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Maria Angelica Miglino
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
13
|
Conway SJ, McConnell R, Simmons O, Snider PL. Armadillo-like helical domain containing-4 is dynamically expressed in both the first and second heart fields. Gene Expr Patterns 2019; 34:119077. [PMID: 31655130 DOI: 10.1016/j.gep.2019.119077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/18/2019] [Accepted: 10/19/2019] [Indexed: 12/19/2022]
Abstract
Armadillo repeat and Armadillo-like helical domain containing proteins form a large family with diverse and fundamental functions in many eukaryotes. Herein we investigated the spatiotemporal expression pattern of Armadillo-like helical domain containing 4 (or Armh4) as an uncharacterized protein coding mouse gene, within the mouse embryo during the initial stages of heart morphogenesis. We found Armh4 is initially expressed in both first heart field as well as the second heart field progenitors and subsequently within predominantly their cardiomyocyte derivatives. Armh4 expression is initially cardiac-restricted in the developing embryo and is expressed in second heart field subpharyngeal mesoderm prior to cardiomyocyte differentiation, but Armh4 diminishes as the embryonic heart matures into the fetal heart. Armh4 is subsequently expressed in craniofacial structures and neural crest-derived dorsal root and trigeminal ganglia. Whereas lithium chloride-induced stimulation of Wnt/β-catenin signaling elevated Armh4 expression in both second heart field subpharyngeal mesodermal progenitors and outflow tract, right ventricle and atrial cardiomyocytes, neither a systemic loss of Islet-1 nor an absence of cardiac neural crest cells had any effect upon Armh4 expression. These results confirm that Wnt/β-catenin-responsive Armh4 is a useful specific biomarker of the FHF and SHF cardiomyocyte derivatives only.
Collapse
Affiliation(s)
- Simon J Conway
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Reagan McConnell
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; School of Biomedical Sciences, University of Ulster, Coleraine, BT52 1SA, Northern Ireland, UK
| | - Olga Simmons
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Paige L Snider
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
14
|
Seki M, Furukawa N, Koitabashi N, Obokata M, Conway SJ, Arakawa H, Kurabayashi M. Periostin-expressing cell-specific transforming growth factor-β inhibition in pulmonary artery prevents pulmonary arterial hypertension. PLoS One 2019; 14:e0220795. [PMID: 31437169 PMCID: PMC6705784 DOI: 10.1371/journal.pone.0220795] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 07/23/2019] [Indexed: 12/27/2022] Open
Abstract
Transforming growth factor beta (TGF-β) has been shown to play a critical role in pathogenesis of pulmonary arterial hypertension (PAH) although the precise role of TGF-β signaling remains uncertain. A recent report has shown that periostin (Pn) is one of the most upregulated proteins in human PAH lung compared with healthy lungs. We established type I TGF-β receptor knockout mice specifically with Pn expressing cell (Pn-Cre/Tgfb1fl/fl mice). Increases in PA pressure and pulmonary artery muscularization were induced by hypoxia of 10% oxygen for 4 weeks. Lung Pn expression was markedly induced by 4 week-hypoxia. Pn-Cre/Tgfb1fl/fl mice showed lower right ventricular pressure elevation, inhibition of PA medial thickening. Fluorescent co-immunostaining showed that Smad3 activation in Pn expressing cell is attenuated. These results suggest that TGF-β signaling in Pn expressing cell may have an important role in the pathogenesis of PAH by controlling medial thickening.
Collapse
Affiliation(s)
- Mitsuru Seki
- Department of Pediatrics, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
- Department of Pediatrics, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Nozomi Furukawa
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Norimichi Koitabashi
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
- * E-mail:
| | - Masaru Obokata
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Simon J. Conway
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Hirokazu Arakawa
- Department of Pediatrics, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Masahiko Kurabayashi
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| |
Collapse
|
15
|
Snider PL, Snider E, Simmons O, Lilly B, Conway SJ. Analysis of Uncharacterized mKiaa1211 Expression during Mouse Development and Cardiovascular Morphogenesis. J Cardiovasc Dev Dis 2019; 6:jcdd6020024. [PMID: 31234534 PMCID: PMC6617212 DOI: 10.3390/jcdd6020024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 06/13/2019] [Accepted: 06/19/2019] [Indexed: 12/24/2022] Open
Abstract
Mammalian Kiaa1211 and Kiaa1211-like are a homologous pair of uncharacterized, highly conserved genes cloned from fetal and adult brain cDNA libraries. Herein we map the in utero spatiotemporal expression of mKiaa1211 and mKiaa1211L mRNA and their expression patterns in postnatal testis, skin, gastrointestinal, and adipose progenitor tissues. Significantly, mKiaa1211 is present throughout the early stages of mouse heart development, particularly in the second heart field (SHF) lineage as it differentiates from mesenchymal cells into cardiomyocytes. We also show that mKiaa1211 is expressed within several early neuronal tissues destined to give rise to central, peripheral, and sympathetic nervous system structures. Expression profiling revealed that the paralog mKiaa1211L is not expressed during the normal developmental process and that mKiaa1211 expression was noticeably absent from most adult terminally differentiated tissues. Finally, we confirm that a previously uncharacterized CRISPR/CAS-generated mKiaa1211 mouse mutant allele is hypomorphic.
Collapse
Affiliation(s)
- Paige L Snider
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Elizabeth Snider
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Biosciences, Indiana University, Bloomington, IN 47405, USA.
| | - Olga Simmons
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Brenda Lilly
- The Heart Center, Nationwide Children's Hospital, Columbus, OH 43205, USA.
| | - Simon J Conway
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
16
|
Kuo HF, Hsieh CC, Wang SC, Chang CY, Hung CH, Kuo PL, Liu YR, Li CY, Liu PL. Simvastatin Attenuates Cardiac Fibrosis via Regulation of Cardiomyocyte-Derived Exosome Secretion. J Clin Med 2019; 8:jcm8060794. [PMID: 31167519 PMCID: PMC6617127 DOI: 10.3390/jcm8060794] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 05/24/2019] [Accepted: 06/03/2019] [Indexed: 12/25/2022] Open
Abstract
Exosome-mediated communication within the cardiac microenvironment is associated with cardiac fibrosis. Simvastatin (SIM), a potent statin, protects against cardiac fibrosis, but its mechanism of action is unclear. We investigated the inhibitory effects and underlying mechanism of simvastatin in cardiac fibrosis, by regulating exosome-mediated communication. Male Sprague-Dawley rats were treated with angiotensin (Ang) II alone, or with SIM for 28 d. Cardiac fibrosis, expressions of fibrosis-associated proteins and mRNAs, and collagen fiber arrangement and deposition were examined. Protein expressions in exosomes isolated from Ang II-treated cardiomyocytes (CMs) were evaluated using nano-ultra-performance liquid chromatographic system, combined with tandem mass spectrometry. Transformation of fibroblasts to myofibroblasts was evaluated using scanning electron and confocal microscopy, and migration assays. Our results showed that SIM attenuated in vivo expression of collagen and collagen-associated protein, as well as collagen deposition, and cardiac fibrosis. The statin also upregulated decorin and downregulated periostin in CM-derived exosomes. Furthermore, it suppressed Ang II-induced transformation of fibroblast to myofibroblast, as well as fibroblast migration. Exosome-mediated cell-cell communication within the cardiac tissue critically regulated cardiac fibrosis. Specifically, SIM regulated the release of CM exosomes, and attenuated Ang II-induced cardiac fibrosis, highlighting its potential as a novel therapy for cardiac fibrosis.
Collapse
Affiliation(s)
- Hsuan-Fu Kuo
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 801, Taiwan.
| | - Chong-Chao Hsieh
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Division of Cardiovascular Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Shu-Chi Wang
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Chia-Yuan Chang
- Department of Engineering Science, National Cheng Kung University, Tainan 701, Taiwan.
| | - Chih-Hsin Hung
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Pediatrics, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung 812, Taiwan.
| | - Po-Lin Kuo
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Yu-Ru Liu
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Chia-Yang Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Po-Len Liu
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
17
|
Periostin in chronic liver diseases: Current research and future perspectives. Life Sci 2019; 226:91-97. [PMID: 30978348 DOI: 10.1016/j.lfs.2019.04.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 03/26/2019] [Accepted: 04/08/2019] [Indexed: 01/06/2023]
Abstract
The liver is importantly metabolic and detoxifying organ in the body. When various pathogenic factors affect the liver, the normal physiological and biochemical functions are weakened, resulting in liver diseases. Liver fibrosis is a common pathological process of chronic liver disease. During hepatic fibrosis the changes in the components of the extracellular matrix (ECM) provide an environment that facilitates tissue remodeling. Among these ECM components, periostin, a glycoprotein that is predominantly secreted by osteoblasts and their precursors, playing an important role in bone formation, has attracted great attention. Periostin not only involves in bone metabolism, but also functions in modulating the cell fate determination, proliferation, inflammatory responses, even tumorigenesis of many other tissues and organs including liver. In different categories of liver disease patients, the serum and liver tissue levels of periostin were closely related to the decline of liver function, and the pathological stage. Numerous animal studies and experiments in vitro subsequently demonstrated that the abnormal expression of periostin resulted in metabolic disorders, liver inflammation, fibrosis and even tumorigenesis. Here we review the current progress on the role of periostin in pathologic pathways of liver system to explore whether periostin is a potential therapeutic target for the treatment of different liver diseases.
Collapse
|
18
|
Lehmann AE, Scangas GA, Bergmark RW, El Rassi E, Stankovic KM, Metson R. Periostin and Inflammatory Disease: Implications for Chronic Rhinosinusitis. Otolaryngol Head Neck Surg 2019; 160:965-973. [PMID: 30935271 DOI: 10.1177/0194599819838782] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To provide a comprehensive overview of the emerging role of periostin, an extracellular matrix protein, as a key component in the development, diagnosis, and treatment of patients with chronic rhinosinusitis. DATA SOURCES Medline database. REVIEW METHODS A state of the art review was performed targeting English-language studies investigating the role of periostin in cardiopulmonary, neoplastic, and inflammatory diseases, with emphasis on recent advances in the study of periostin in chronic rhinosinusitis. CONCLUSIONS Periostin has emerged as a novel biomarker and therapeutic target for numerous human pathologies, including cardiac, pulmonary, and neoplastic disease. The upregulation of periostin in chronic rhinosinusitis suggests the potential for similar roles among patients with sinonasal disease. IMPLICATIONS FOR PRACTICE Chronic rhinosinusitis is a widespread disease with major clinical and societal impact. A critical limitation in the current treatment of patients with chronic rhinosinusitis is the absence of clinically relevant biomarkers to guide diagnosis and treatment selection. A review of the literature supports a likely role of periostin as a biomarker of chronic rhinosinusitis, as well as a novel therapeutic target in the future treatment of patients with sinonasal disease.
Collapse
Affiliation(s)
- Ashton E Lehmann
- 1 Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, USA.,2 Department of Otolaryngology, Massachusetts Eye and Ear, Boston, Massachusetts, USA
| | - George A Scangas
- 1 Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, USA.,2 Department of Otolaryngology, Massachusetts Eye and Ear, Boston, Massachusetts, USA
| | - Regan W Bergmark
- 1 Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, USA.,2 Department of Otolaryngology, Massachusetts Eye and Ear, Boston, Massachusetts, USA
| | - Edward El Rassi
- 1 Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, USA.,2 Department of Otolaryngology, Massachusetts Eye and Ear, Boston, Massachusetts, USA
| | - Konstantina M Stankovic
- 1 Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, USA.,2 Department of Otolaryngology, Massachusetts Eye and Ear, Boston, Massachusetts, USA
| | - Ralph Metson
- 1 Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, USA.,2 Department of Otolaryngology, Massachusetts Eye and Ear, Boston, Massachusetts, USA
| |
Collapse
|
19
|
Functions of Periostin in Dental Tissues and Its Role in Periodontal Tissue Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1132:63-72. [PMID: 31037625 DOI: 10.1007/978-981-13-6657-4_7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The goal of periodontal regeneration therapy is to reliably restore teeth's supporting periodontal tissue, while aiding the formation of new connective tissue attached to the periodontal ligament (PDL) fibers and new alveolar bone. Periostin is a matricellular protein, primarily expressed in the periosteum and PDL of adult mice. Its biological functions have been extensively studied in the fields of cardiovascular physiology and oncology. Despite being initially identified in bone and dental tissue, the function of Periostin in PDL and the pathophysiology associated with alveolar bone are scarcely studied. Recently, several studies have suggested that Periostin may be an important regulator of periodontal tissue formation. By promoting collagen fibrillogenesis and the migration of fibroblasts and osteoblasts, Periostin might play a key role in the regeneration of PDL and alveolar bone after periodontal surgery. In this chapter, the implications of Periostin in periodontal tissue biology and its potential use in periodontal tissue regeneration are reviewed.
Collapse
|
20
|
An JN, Yang SH, Kim YC, Hwang JH, Park JY, Kim DK, Kim JH, Kim DW, Hur DG, Oh YK, Lim CS, Kim YS, Lee JP. Periostin induces kidney fibrosis after acute kidney injury via the p38 MAPK pathway. Am J Physiol Renal Physiol 2018; 316:F426-F437. [PMID: 30539653 DOI: 10.1152/ajprenal.00203.2018] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Periostin plays a crucial role in fibrosis, and acute kidney injury results in a high risk of progression to chronic kidney disease. Therefore, we hypothesized that periostin was involved in the progression of acute kidney injury to kidney fibrosis. Unilateral ischemia-reperfusion injury (UIRI) was induced in 7- to 8-wk-old male wild-type and periostin-null mice, and the animals were observed for 6 wk. In vitro, human kidney-2 cells and primary-cultured human tubular epithelial cells were incubated under hypoxic conditions (5% O2, 5% CO2, and 90% N2) for 5 days. The cells were also cultured with recombinant periostin (rPeriostin) and a p38 mitogen-activated protein kinase (MAPK) inhibitor in a hypoxic incubator. At 6 wk after UIRI, interstitial fibrosis/tubular atrophy was significantly alleviated in periostin-null mice compared with wild-type controls. In addition, periostin-null mice had attenuated expression of fibrosis/apoptosis markers and phosphorylated-p38 MAPK compared with wild-type controls. In vitro, hypoxic injury increased the expression of fibrosis markers, periostin, and phosphorylated-p38 MAPK, which was comparable to or substantially greater than their expression levels following treatment with recombinant transforming growth factor-β1 under normoxic conditions. Furthermore, rPeriostin treatment under hypoxic conditions enhanced fibrosis/apoptosis markers and phosphorylated-p38 MAPK. In contrast, p38 MAPK inhibition ameliorated hypoxia-induced fibrosis, and the addition of the p38 MAPK inhibitor to rPeriostin significantly ameliorated the changes induced by rPeriostin. In conclusion, periostin promotes kidney fibrosis via the p38 MAPK pathway following acute kidney injury triggered by a hypoxic or ischemic insult. Periostin ablation may protect against chronic kidney disease progression.
Collapse
Affiliation(s)
- Jung Nam An
- Department of Internal Medicine, Seoul National University Boramae Medical Center , Seoul , Korea.,Department of Critical Care Medicine, Seoul National University Boramae Medical Center , Seoul , Korea
| | - Seung Hee Yang
- Seoul National University Kidney Research Institute , Seoul , Korea
| | - Yong Chul Kim
- Department of Internal Medicine, Seoul National University Hospital , Seoul , Korea
| | - Jin Ho Hwang
- Department of Internal Medicine, Chung-Ang University Hospital , Seoul , Korea
| | - Jae Yoon Park
- Department of Internal Medicine, Dongguk University Ilsan Hospital, Goyang, Gyeonggido, Korea
| | - Dong Ki Kim
- Department of Internal Medicine, Seoul National University Hospital , Seoul , Korea.,Department of Internal Medicine, Seoul National University College of Medicine , Seoul , Korea
| | - Jin Hyuk Kim
- Department of Internal Medicine, Seoul National University Boramae Medical Center , Seoul , Korea
| | - Dae Woo Kim
- Department of Otorhinolaryngology, Seoul National University Boramae Medical Center , Seoul , Korea
| | - Dong Gu Hur
- Department of Otorhinolaryngology, Gyeongsang National University Hospital , Changwon , Korea
| | - Yun Kyu Oh
- Department of Internal Medicine, Seoul National University Boramae Medical Center , Seoul , Korea.,Department of Internal Medicine, Seoul National University College of Medicine , Seoul , Korea
| | - Chun Soo Lim
- Department of Internal Medicine, Seoul National University Boramae Medical Center , Seoul , Korea.,Department of Internal Medicine, Seoul National University College of Medicine , Seoul , Korea
| | - Yon Su Kim
- Department of Internal Medicine, Seoul National University Hospital , Seoul , Korea.,Department of Internal Medicine, Seoul National University College of Medicine , Seoul , Korea
| | - Jung Pyo Lee
- Department of Internal Medicine, Seoul National University Boramae Medical Center , Seoul , Korea.,Department of Internal Medicine, Seoul National University College of Medicine , Seoul , Korea
| |
Collapse
|
21
|
Huang YW, Chiang MF, Ho CS, Hung PL, Hsu MH, Lee TH, Chu LJ, Liu H, Tang P, Victor Ng W, Lin DS. A Transcriptome Study of Progeroid Neurocutaneous Syndrome Reveals POSTN As a New Element in Proline Metabolic Disorder. Aging Dis 2018; 9:1043-1057. [PMID: 30574417 PMCID: PMC6284769 DOI: 10.14336/ad.2018.0222] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 02/22/2018] [Indexed: 12/27/2022] Open
Abstract
Aging is a complex biological process. A study of pyrroline-5-carboxylate reductase 1 (PYCR1) deficiency, which causes a progeroid syndrome, may not only shed light on its genetic contribution to autosomal recessive cutis laxa (ARCL) but also help elucidate the functional mechanisms associated with aging. In this study, we used RNA-Seq technology to examine gene expression changes in primary skin fibroblasts from healthy controls and patients with PYCR1 mutations. Approximately 22 and 32 candidate genes were found to be up- and downregulated, respectively, in fibroblasts from patients. Among the downregulated candidates in fibroblasts with PYCR1 mutations, a strong reduction in the expression of 17 genes (53.1%) which protein products are localized in the extracellular space was detected. These proteins included several important ECM components, periostin (POSTN), elastin (ELN), and decorin (DCN); genetic mutations in these proteins are associated with different phenotypes of aging, such as cutis laxa and joint and dermal manifestations. The differential expression of ten selected extracellular space genes was further validated using quantitative RT-PCR. Ingenuity Pathway Analysis revealed that some of the affected genes may be associated with cardiovascular system development and function, dermatological diseases and conditions, and cardiovascular disease. POSTN, one of the most downregulated gene candidates in affected individuals, is a matricellular protein with pivotal functions in heart valvulogenesis, skin wound healing, and brain development. Perturbation of PYCR1 expression revealed that it is positively correlated with the POSTN levels. Taken together, POSTN might be one of the key molecules that deserves further investigation for its role in this progeroid neurocutaneous syndrome.
Collapse
Affiliation(s)
- Yu-Wen Huang
- Institute of Biotechnology in Medicine and Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming University, Taipei, Taiwan.
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan.
| | - Ming-Fu Chiang
- Department of Neurosurgery, Mackay Memorial Hospital, Taipei, Taiwan.
- Mackay Junior College of Medicine, Nursing and Management, Taipei, Taiwan.
- Graduate Institute of Injury Prevention and Control, Taipei Medical University, Taipei, Taiwan.
| | - Che-Sheng Ho
- Department of Pediatrics, Mackay Memorial Hospital, Taipei, Taiwan.
| | - Pi-Lien Hung
- Department of Pediatric Neurology, Kaohsiung Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| | - Mei-Hsin Hsu
- Department of Pediatric Neurology, Kaohsiung Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| | - Tsung-Han Lee
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan.
| | - Lichieh Julie Chu
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan.
| | - Hsuan Liu
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan.
- Department of Cell and Molecular Biology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Petrus Tang
- Molecular Regulation and Bioinformatics Laboratory and Department of Parasitology, Chang Gung University, Taoyuan, Taiwan.
| | - Wailap Victor Ng
- Institute of Biotechnology in Medicine and Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming University, Taipei, Taiwan.
- Institute of Biomedical Informatics and Center for Systems and Synthetic Biology, National Yang Ming University, Taipei, Taiwan.
- Department of Biochemistry, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Dar-Shong Lin
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan.
- Department of Pediatrics, Mackay Memorial Hospital, Taipei, Taiwan.
- Department of Medicine, Mackay Medical College, New Taipei, Taiwan
| |
Collapse
|
22
|
Liu Q, Huang P, Guo SJ. [Progress relationship between periostin and periodontitis]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2018; 36:681-685. [PMID: 30593118 DOI: 10.7518/hxkq.2018.06.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Periostin, a kind of matricellular protein highly expressed in periodontal ligament and periosteum, is an important regulator of the integrity of periodontal ligament and periodontitis processes. Periostin has been shown to play a positive role in the recovery of periodontitis. This paper reviews relevant literature about the role of periostin in periodontal tissue and periodontitis.
Collapse
Affiliation(s)
- Qian Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ping Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Shu-Juan Guo
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
23
|
González-González L, Alonso J. Periostin: A Matricellular Protein With Multiple Functions in Cancer Development and Progression. Front Oncol 2018; 8:225. [PMID: 29946533 PMCID: PMC6005831 DOI: 10.3389/fonc.2018.00225] [Citation(s) in RCA: 186] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 05/30/2018] [Indexed: 01/19/2023] Open
Abstract
Tumor microenvironment is considered nowadays as one of the main players in cancer development and progression. Tumor microenvironment is highly complex and consists of non-tumor cells (i.e., cancer-associated fibroblast, endothelial cells, or infiltrating leukocytes) and a large list of extracellular matrix proteins and soluble factors. The way that microenvironment components interact among them and with the tumor cells is very complex and only partially understood. However, it is now clear that these interactions govern and modulate many of the cancer hallmarks such as cell proliferation, the resistance to death, the differentiation state of tumor cells, their ability to migrate and metastasize, and the immune response against tumor cells. One of the microenvironment components that have emerged in the last years with strength is a heterogeneous group of multifaceted proteins grouped under the name of matricellular proteins. Matricellular proteins are a family of non-structural matrix proteins that regulate a variety of biological processes in normal and pathological situations. Many components of this family such as periostin (POSTN), osteopontin (SPP1), or the CNN family of proteins have been shown to regulate key aspect of tumor biology, including proliferation, invasion, matrix remodeling, and dissemination to pre-metastatic niches in distant organs. Matricellular proteins can be produced by tumor cells themselves or by tumor-associated cells, and their synthesis can be affected by intrinsic and/or extrinsic tumor cell factors. In this review, we will focus on the role of POSTN in the development and progression of cancer. We will describe their functions in normal tissues and the mechanisms involved in their regulation. We will analyze the tumors in which their expression is altered and their usefulness as a biomarker of tumor progression. Finally, we will speculate about future directions for research and therapeutic approaches targeting POSTN.
Collapse
Affiliation(s)
- Laura González-González
- Unidad de Tumores Sólidos Infantiles, Área de Genética Humana, Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Alonso
- Unidad de Tumores Sólidos Infantiles, Área de Genética Humana, Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
24
|
Wen L, Chen J, Duan L, Li S. Vitamin K‑dependent proteins involved in bone and cardiovascular health (Review). Mol Med Rep 2018; 18:3-15. [PMID: 29749440 PMCID: PMC6059683 DOI: 10.3892/mmr.2018.8940] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 02/13/2018] [Indexed: 12/19/2022] Open
Abstract
In postmenopausal women and elderly men, bone density decreases with age and vascular calcification is aggravated. This condition is closely associated with vitamin K2 deficiency. A total of 17 different vitamin K-dependent proteins have been identified to date. Vitamin K-dependent proteins are located within the bone, heart and blood vessels. For instance, carboxylated osteocalcin is beneficial for bone and aids the deposition of calcium into the bone matrix. Carboxylated matrix Gla protein effectively protects blood vessels and may prevent calcification within the vascular wall. Furthermore, carboxylated Gla-rich protein has been reported to act as an inhibitor in the calcification of the cardiovascular system, while growth arrest-specific protein-6 protects endothelial cells and vascular smooth muscle cells, resists apoptosis and inhibits the calcification of blood vessels by inhibiting the apoptosis of vascular smooth muscle cells. In addition, periostin may promote the differentiation, aggregation, adhesion and proliferation of osteoblasts. Periostin also occurs in the heart and may be associated with the reconstruction of heart function. These vitamin K-dependent proteins may exert their functions following γ-carboxylation with vitamin K, and different vitamin K-dependent proteins may exhibit synergistic effects or antagonistic effects on each other. In the cardiovascular system with vitamin K antagonist supplement or vitamin K deficiency, calcification occurs in the endothelium of blood vessels and vascular smooth muscle cells are transformed into osteoblast-like cells, a phenomenon that resembles bone growth. Both the bone and cardiovascular system are closely associated during embryonic development. Thus, the present study hypothesized that embryonic developmental position and tissue calcification may have a certain association for the bone and the cardiovascular system. This review describes and briefly discusses several important vitamin K-dependent proteins that serve an important role in bone and the cardiovascular system. The results of the review suggest that the vascular calcification and osteogenic differentiation of vascular smooth muscle cells may be associated with the location of the bone and cardiovascular system during embryonic development.
Collapse
Affiliation(s)
- Lianpu Wen
- Department of Physiology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Jiepeng Chen
- Sungen Bioscience Co., Ltd., Shantou, Guangdong 515000, P.R. China
| | - Lili Duan
- Sungen Bioscience Co., Ltd., Shantou, Guangdong 515000, P.R. China
| | - Shuzhuang Li
- Department of Physiology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| |
Collapse
|
25
|
Olsan EE, West JD, Torres JA, Doerr N, Weimbs T. Identification of targets of IL-13 and STAT6 signaling in polycystic kidney disease. Am J Physiol Renal Physiol 2018. [PMID: 29513071 DOI: 10.1152/ajprenal.00346.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a life-threatening, highly prevalent monogenic disease caused by mutations in polycystin-1 (PC1) in 85% of patients. We have previously identified a COOH-terminal cleavage fragment of PC1, PC1-p30, which interacts with the transcription factor STAT6 to promote transcription. STAT6 is aberrantly active in PKD mouse models and human ADPKD, and genetic removal or pharmacological inhibition of STAT6 attenuates disease progression. High levels of IL-13, a STAT6-activating cytokine, are found in the cyst fluid of PKD mouse models and increased IL-13 receptors in ADPKD patient tissue, suggesting that a positive feedback loop exists between IL-13 and STAT6 is activated in cystic epithelial cells and contributes to disease progression. In this study, we aimed to identify genes aberrantly regulated by STAT6 to better understand how increased IL-13/STAT6 signaling may contribute to PKD progression. We demonstrate that the expression of periostin, galectin-3, and IL-24 is upregulated in various forms of PKD and that their aberrant regulation is mediated by IL-13 and STAT6 activity. Periostin and galectin-3 have previously been implicated in PKD progression. We support these findings by showing that periostin expression is increased after IL-13 treatment in kidney epithelial cells, that galectin-3 expression is increased after injecting IL-13 in vivo and that IL-24 expression is upregulated by both IL-13 treatment and PC1-p30 overexpression in mouse and human kidney cells. Overall, these findings provide insight into the possible mechanisms by which increased IL-13/STAT6 signaling contributes to PKD progression and suggest potential therapeutic targets.
Collapse
Affiliation(s)
- Erin E Olsan
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara , Santa Barbara, California.,Neuroscience Research Institute, University of California Santa Barbara , Santa Barbara, California
| | - Jonathan D West
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara , Santa Barbara, California.,Neuroscience Research Institute, University of California Santa Barbara , Santa Barbara, California
| | - Jacob A Torres
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara , Santa Barbara, California.,Neuroscience Research Institute, University of California Santa Barbara , Santa Barbara, California
| | - Nicholas Doerr
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara , Santa Barbara, California.,Neuroscience Research Institute, University of California Santa Barbara , Santa Barbara, California
| | - Thomas Weimbs
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara , Santa Barbara, California.,Neuroscience Research Institute, University of California Santa Barbara , Santa Barbara, California
| |
Collapse
|
26
|
Wang Q, Chen Z, Huang X, Chen L, Chen B, Zhu Y, Cao S, Liao W, Bin J, Kitakaze M, Liao Y. Olmesartan attenuates pressure-overload- or post-infarction-induced cardiac remodeling in mice. Oncotarget 2017; 9:24601-24618. [PMID: 29872491 PMCID: PMC5973849 DOI: 10.18632/oncotarget.23628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 12/11/2017] [Indexed: 01/10/2023] Open
Abstract
Either angiotensin converting enzyme inhibitor (ACEI) or angiotensin receptor 1 blocker (ARB) attenuates cardiac remodeling. However, the overall molecular modulation of the reversing remodeling process in response to the ACEI or ARB treatment is not yet well determined. In this study, we examined whether gene expressions are modulated by ACEI (temocapril), ARB (olmesartan) or both in a murine model with transverse aortic constriction (TAC) and confirm whether periostin is a target gene of olmesartan in mice with myocardial infarction (MI). We detected 109 genes that were significantly up-regulated in TAC mice and a majority of these were down-regulated in response to temocapril, olmesartan or their combination which significantly attenuated cardiac remodeling at one or four weeks. Real-time RT-PCR demonstrated that olmesartan, temocapril or their combination down-regulated the expression of periostin. In MI mice treated with olmesartan for 4 weeks, the left ventricular end-diastolic and systolic dimensions measured with echocardiography were lower, whereas maximum rate of rise and fall rate of LV pressure (±dp/dt max) were greater, and Azan-staining cardiac fibrotic area was smaller. Furthermore, periostin was upregulated in response to MI, whereas olmesartan blocked this upregulation. Post-MI fibrosis was smaller in periostin knockout adult mice than in wildtype mice, while glycogen synthase kinase 3β was increased and cyclin D1 was decreased in periostin knockout mice. These findings indicate that periostin is a target gene of ARB and olmesartan reverses cardiac remodeling at least partially through the downregulation of periostin.
Collapse
Affiliation(s)
- Qiancheng Wang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Department of Cardiology, Jiaozuo People's Hospital of Henan Province, Jiaozuo 454000, China
| | - Zhenhuan Chen
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiaobo Huang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Lin Chen
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Baihe Chen
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yingqi Zhu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Shiping Cao
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jianping Bin
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Masafumi Kitakaze
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Cardiovascular Division of the Department of Medicine, National Cerebral and Cardiovascular Center, Suita, Osaka 565-8565, Japan
| | - Yulin Liao
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
27
|
Du J, Li M. Functions of Periostin in dental tissues and its role in periodontal tissues' regeneration. Cell Mol Life Sci 2017; 74:4279-4286. [PMID: 28889194 PMCID: PMC11107504 DOI: 10.1007/s00018-017-2645-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 09/04/2017] [Indexed: 02/08/2023]
Abstract
The goal of periodontal regenerative therapy is to predictably restore the tooth's supporting periodontal tissues and form a new connective tissue attachment of periodontal ligament (PDL) fibers and new alveolar bone. Periostin is a matricellular protein so named for its expression primarily in the periosteum and PDL of adult mice. Its biological functions have been widely studied in areas such as cardiovascular physiology and oncology. Despite being initially identified in the dental tissues and bone, investigations of Periostin functions in PDL and alveolar-bone-related physiopathology are less abundant. Recently, several studies have suggested that Periostin may be an important regulator of periodontal tissue formation. By promoting collagen fibrillogenesis and the migration of fibroblasts and osteoblasts, Periostin might play a pivotal part in regeneration of the PDL and alveolar bone following periodontal surgery. The aim of this article is to provide an extensive review of the implications of Periostin in periodontal tissue biology and its potential use in periodontal tissue regeneration.
Collapse
Affiliation(s)
- Juan Du
- Department of Bone Metabolism, School of Stomatology, Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Shandong University, Jinan, 250012, China
| | - Minqi Li
- Department of Bone Metabolism, School of Stomatology, Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Shandong University, Jinan, 250012, China.
| |
Collapse
|
28
|
Kudo A. Introductory review: periostin-gene and protein structure. Cell Mol Life Sci 2017; 74:4259-4268. [PMID: 28884327 PMCID: PMC11107487 DOI: 10.1007/s00018-017-2643-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 09/04/2017] [Indexed: 01/08/2023]
Abstract
Although many studies have described the role of periostin in various diseases, the function of the periostin protein structures derived from alternative splicing and proteinase cleavage at the C-terminal remain unknown. Further experiments revealing the protein structures that are highly related to diseases are essential to understand the function of periostin in depth, which would accelerate its clinical application by establishing new approaches for curing intractable diseases. Furthermore, this understanding would enhance our knowledge of novel functions of periostin related to stemness and response to mechanical stress.
Collapse
Affiliation(s)
- Akira Kudo
- International Frontier, Tokyo Institute of Technology, S3-8, 2-12-1 Oookayama, Meguro-ku, Tokyo, 152-8550, Japan.
- Department of Pharmacology, School of Dentistry, Showa University, Tokyo, 142-8555, Japan.
| |
Collapse
|
29
|
Landry NM, Cohen S, Dixon IMC. Periostin in cardiovascular disease and development: a tale of two distinct roles. Basic Res Cardiol 2017; 113:1. [PMID: 29101484 DOI: 10.1007/s00395-017-0659-5] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 10/12/2017] [Indexed: 12/18/2022]
Abstract
Tissue development and homeostasis are dependent upon the concerted synthesis, maintenance, and degradation of extracellular matrix (ECM) molecules. Cardiac fibrosis is now recognized as a primary contributor to incidence of heart failure, particularly heart failure with preserved ejection fraction, wherein cardiac filling in diastole is compromised. Periostin is a cell-associated protein involved in cell fate determination, proliferation, tumorigenesis, and inflammatory responses. As a non-structural component of the ECM, secreted 90 kDa periostin is emerging as an important matricellular factor in cardiac mesenchymal tissue development. In addition, periostin's role as a mediator in cell-matrix crosstalk has also garnered attention for its association with fibroproliferative diseases in the myocardium, and for its association with TGF-β/BMP signaling. This review summarizes the phylogenetic history of periostin, its role in cardiac development, and the major signaling pathways influencing its expression in cardiovascular pathology. Further, we provide a synthesis of the current literature to distinguish the multiple roles of periostin in cardiac health, development and disease. As periostin may be targeted for therapeutic treatment of cardiac fibrosis, these insights may shed light on the putative timing for application of periostin-specific therapies.
Collapse
Affiliation(s)
- Natalie M Landry
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Max Rady College of Medicine, Institute of Cardiovascular Sciences, University of Manitoba, Winnipeg, Canada
| | - Smadar Cohen
- Regenerative Medicine and Stem Cell Research Center, Ilse Katz Institute for Nanoscale Science and Technology, Beersheba, Israel.,Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Ian M C Dixon
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Max Rady College of Medicine, Institute of Cardiovascular Sciences, University of Manitoba, Winnipeg, Canada. .,Laboratory of Molecular Cardiology, St. Boniface Hospital Albrechtsen Research Centre, R3010-351 Taché Avenue, Winnipeg, MB R2H 2A6, Canada.
| |
Collapse
|
30
|
Um JE, Park JT, Nam BY, Lee JP, Jung JH, Kim Y, Kim S, Park J, Wu M, Han SH, Yoo TH, Kang SW. Periostin-binding DNA aptamer treatment attenuates renal fibrosis under diabetic conditions. Sci Rep 2017; 7:8490. [PMID: 28819200 PMCID: PMC5561139 DOI: 10.1038/s41598-017-09238-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 07/17/2017] [Indexed: 02/07/2023] Open
Abstract
Diabetic nephropathy, the major cause of chronic kidney disease, is associated with progressive renal fibrosis. Recently, accumulation of periostin, an extracellular matrix protein, was shown to augment renal fibrosis. Aptamers have higher binding affinities without developing the common side effects of antibodies. Thus, we evaluated the effect of periostin inhibition by an aptamer-based inhibitor on renal fibrosis under diabetic conditions. In vitro, transforming growth factor-β1 (TGF-β1) treatment significantly upregulated periostin, fibronectin, and type I collagen mRNA and protein expressions in inner medullary collecting duct (IMCD) cells. These increases were attenuated significantly in periostin-binding DNA aptamer (PA)-treated IMCD cells exposed to TGF-β1. In vivo, PA treatment attenuated the increased blood urea nitrogen levels in the diabetic mice significantly. Fibronectin and type I collagen mRNA and protein expressions increased significantly in the kidneys of diabetic mice: PA administration abrogated these increases significantly. Immunohistochemistry and Sirius Red staining also revealed that fibronectin expression was significantly higher and tubulointersititial fibrosis was significantly worse in diabetic mice kidneys compared with control mice. These changes were ameliorated by PA treatment. These findings suggested that inhibition of periostin using a DNA aptamer could be a potential therapeutic strategy against renal fibrosis in diabetic nephropathy.
Collapse
Affiliation(s)
- Jae Eun Um
- Severance Biomedical Science Institute, College of Medicine, Yonsei University, Seoul, Korea
| | - Jung Tak Park
- Department of Internal Medicine, College of Medicine, Severance Biomedical Science Institute, Brain Korea 21 PLUS, Institute of Kidney Disease Research, Yonsei University, Seoul, Korea
| | - Bo Young Nam
- Severance Biomedical Science Institute, College of Medicine, Yonsei University, Seoul, Korea
| | - Jung Pyo Lee
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Jong Ha Jung
- Aptamer Sciences Inc., POSTECH Biotech Center, Pohang, Gyeongbuk, Korea
| | - Youndong Kim
- Aptamer Sciences Inc., POSTECH Biotech Center, Pohang, Gyeongbuk, Korea
| | - Seonghun Kim
- Department of Internal Medicine, College of Medicine, Severance Biomedical Science Institute, Brain Korea 21 PLUS, Institute of Kidney Disease Research, Yonsei University, Seoul, Korea
| | - Jimin Park
- Department of Internal Medicine, College of Medicine, Severance Biomedical Science Institute, Brain Korea 21 PLUS, Institute of Kidney Disease Research, Yonsei University, Seoul, Korea
| | - Meiyan Wu
- Department of Internal Medicine, College of Medicine, Severance Biomedical Science Institute, Brain Korea 21 PLUS, Institute of Kidney Disease Research, Yonsei University, Seoul, Korea
| | - Seung Hyeok Han
- Department of Internal Medicine, College of Medicine, Severance Biomedical Science Institute, Brain Korea 21 PLUS, Institute of Kidney Disease Research, Yonsei University, Seoul, Korea
| | - Tae-Hyun Yoo
- Department of Internal Medicine, College of Medicine, Severance Biomedical Science Institute, Brain Korea 21 PLUS, Institute of Kidney Disease Research, Yonsei University, Seoul, Korea
| | - Shin-Wook Kang
- Department of Internal Medicine, College of Medicine, Severance Biomedical Science Institute, Brain Korea 21 PLUS, Institute of Kidney Disease Research, Yonsei University, Seoul, Korea.
| |
Collapse
|
31
|
Yan J, Liu HJ, Li H, Chen L, Bian YQ, Zhao B, Han HX, Han SZ, Han LR, Wang DW, Yang XF. Circulating periostin levels increase in association with bone density loss and healing progression during the early phase of hip fracture in Chinese older women. Osteoporos Int 2017; 28:2335-2341. [PMID: 28382553 DOI: 10.1007/s00198-017-4034-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 03/30/2017] [Indexed: 11/26/2022]
Abstract
UNLABELLED The present study shows that hip fracture women had higher serum periostin (sPostn) levels. The elevation of sPostn is associated with bone density loss, yet fracture itself may even increase sPostn levels during early healing phase. INTRODUCTION The study aims to quantify the associations of sPostn levels with bone density loss and the possible effect on the fracture healing. METHODS This study enrolled 261 older women with osteoporotic hip fracture and 106 age-matched women without fracture serving as controls. Clinical features, bone mineral density (BMD), and bone turnover markers including sPostn level were measured after fracture within 2 days. Follow-up sPostn levels during 1 year after 2 days were available for 128 patients. RESULTS Initial levels of sPostn after fracture were significantly higher in patients than controls. sPostn was correlated with BMD of femoral neck (r = -0.529, P < 0.001), β-isomerized C-terminal crosslinking of type I collagen (β-CTX) (r = 0.403, P = 0.008), and N-terminal procollagen of type I collagen (PINP) (r = 0.236, P = 0.042) in the entire cohort. After multivariate analysis, sPostn remained as an independent risk factor for femoral neck BMD, which explained 19.1% of the variance in BMD. sPostn sampled within 7 days after fracture were acutely increasing from day 2 and then decreased and maintained at slightly high levels at 360 days. The percentage changes of sPostn positively correlated with the variation in β-CTX (r = 0.396, P = 0.002) and PINP (r = 0.180, P = 0.033) at day 7 after fracture. CONCLUSIONS High sPostn levels were an independent predictor of femoral neck BMD in older women presenting with an acute hip fracture. Increased sPostn levels during early healing phase may imply that Postn play a role in bone repair.
Collapse
Affiliation(s)
- J Yan
- Department of Orthopaedic Surgery, Liaocheng People's Hospital, Liaocheng, Shandong, China
| | - H J Liu
- Department of Endocrinology, Liaocheng People's Hospital, Liaocheng, Shandong, China
| | - H Li
- Department of Orthopaedic Surgery, Liaocheng People's Hospital, Liaocheng, Shandong, China
| | - L Chen
- Department of Orthopaedic Surgery, Liaocheng People's Hospital, Liaocheng, Shandong, China
| | - Y Q Bian
- Department of Orthopaedic Surgery, Liaocheng People's Hospital, Liaocheng, Shandong, China
| | - B Zhao
- Department of Orthopaedic Surgery, Liaocheng People's Hospital, Liaocheng, Shandong, China
| | - H X Han
- Second Department of Clinical Medicine, Medical University of Anhui, Hefei, Anhui, China
| | - S Z Han
- Department of Orthopaedic Surgery, Liaocheng People's Hospital, Liaocheng, Shandong, China
| | - L R Han
- Department of Orthopaedic Surgery, Liaocheng People's Hospital, Liaocheng, Shandong, China.
| | - D W Wang
- Department of Orthopaedic Surgery, Liaocheng People's Hospital, Liaocheng, Shandong, China.
| | - X F Yang
- Department of Orthopaedic Surgery, Liaocheng People's Hospital, Liaocheng, Shandong, China
| |
Collapse
|
32
|
Tavares ALP, Brown JA, Ulrich EC, Dvorak K, Runyan RB. Runx2-I is an Early Regulator of Epithelial-Mesenchymal Cell Transition in the Chick Embryo. Dev Dyn 2017. [PMID: 28631378 DOI: 10.1002/dvdy.24539] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Although normally linked to bone and cartilage development, the Runt-related transcription factor, RUNX2, was reported in the mouse heart during development of the valves. We examined RUNX2 expression and function in the developing avian heart as it related to the epithelial-mesenchymal transition (EMT) in the atrioventricular canal. EMT can be separated into an activation stage involving hypertrophy and cell separation and an invasion stage where cells invade the extracellular matrix. The localization and activity of RUNX2 was explored in relation to these steps in the heart. As RUNX2 was also reported in cancer tissues, we examined its expression in the progression of esophageal cancer in staged tissues. RESULTS A specific isoform, RUNX2-I, is present and required for EMT by endothelia of the atrioventricular canal. Knockdown of RUNX2-I inhibits the cell-cell separation that is characteristic of initial activation of EMT. Loss of RUNX2-I altered expression of EMT markers to a greater extent during activation than during subsequent cell invasion. Transforming growth factor beta 2 (TGFβ2) mediates activation during cardiac endothelial EMT. Consistent with a role in activation, RUNX2-I is regulated by TGFβ2 and its activity is independent of similarly expressed Snai2 in regulation of EMT. Examination of RUNX2 expression in esophageal cancer showed its upregulation concomitant with the development of dysplasia and continued expression in adenocarcinoma. CONCLUSIONS These data introduce the RUNX2-I isoform as a critical early transcription factor mediating EMT in the developing heart after induction by TGFβ2. Its expression in tumor tissue suggests a similar role for RUNX2 in the EMT of metastasis. Developmental Dynamics 247:542-554, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Andre L P Tavares
- Department of Craniofacial Biology, School of Dentistry, University of Colorado Anschutz Medical Campus, Denver, Colorado.,Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona
| | - Jessie A Brown
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona
| | - Emily C Ulrich
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona
| | - Katerina Dvorak
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona
| | - Raymond B Runyan
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona
| |
Collapse
|
33
|
Sawafuji R, Cappellini E, Nagaoka T, Fotakis AK, Jersie-Christensen RR, Olsen JV, Hirata K, Ueda S. Proteomic profiling of archaeological human bone. ROYAL SOCIETY OPEN SCIENCE 2017; 4:161004. [PMID: 28680659 PMCID: PMC5493901 DOI: 10.1098/rsos.161004] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 05/09/2017] [Indexed: 05/18/2023]
Abstract
Ancient protein analysis provides clues to human life and diseases from ancient times. Here, we performed shotgun proteomics of human archeological bones for the first time, using rib bones from the Hitotsubashi site (AD 1657-1683) in Tokyo, called Edo in ancient times. The output data obtained were analysed using Gene Ontology and label-free quantification. We detected leucocyte-derived proteins, possibly originating from the bone marrow of the rib. Particularly prevalent and relatively high expression of eosinophil peroxidase suggests the influence of infectious diseases. This scenario is plausible, considering the overcrowding and unhygienic living conditions of the Edo city described in the historical literature. We also observed age-dependent differences in proteome profiles, particularly for proteins involved in developmental processes. Among them, alpha-2-HS-glycoprotein demonstrated a strong negative correlation with age. These results suggest that analysis of ancient proteins could provide a useful indicator of stress, disease, starvation, obesity and other kinds of physiological and pathological information.
Collapse
Affiliation(s)
- Rikai Sawafuji
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Department of Human Biology and Anatomy, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa 903-0215, Japan
| | - Enrico Cappellini
- Centre for GeoGenetics, Natural History Museum of Denmark, University of Copenhagen, ster Voldgade 5–7, 1350 Copenhagen, Denmark
| | - Tomohito Nagaoka
- Department of Anatomy, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa 216-8511, Japan
| | - Anna K. Fotakis
- Centre for GeoGenetics, Natural History Museum of Denmark, University of Copenhagen, ster Voldgade 5–7, 1350 Copenhagen, Denmark
| | - Rosa Rakownikow Jersie-Christensen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3b, 2200 Copenhagen, Denmark
| | - Jesper V. Olsen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3b, 2200 Copenhagen, Denmark
| | - Kazuaki Hirata
- Department of Anatomy, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa 216-8511, Japan
| | - Shintaroh Ueda
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- School of Medicine, Hangzhou Normal University, No.58, Haishu Road, Cangqian, Yuhang District, Hangzhou, Zhejiang 311121, People’s Republic of China
- Author for correspondence: Shintaroh Ueda e-mail:
| |
Collapse
|
34
|
Prakoura N, Kavvadas P, Kormann R, Dussaule JC, Chadjichristos CE, Chatziantoniou C. NF κB-Induced Periostin Activates Integrin- β3 Signaling to Promote Renal Injury in GN. J Am Soc Nephrol 2016; 28:1475-1490. [PMID: 27920156 DOI: 10.1681/asn.2016070709] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 10/31/2016] [Indexed: 12/31/2022] Open
Abstract
De novo expression in the kidney of periostin, a protein involved in odontogenesis and osteogenesis, has been suggested as a biomarker of renal disease. In this study, we investigated the mechanism(s) of induction and the role of periostin in renal disease. Using a combination of bioinformatics, reporter assay, and chromatin immunoprecipitation analyses, we found that NFκB and other proinflammatory transcription factors induce periostin expression in vitro and that binding of these factors on the periostin promoter is enriched in glomeruli during experimental GN. Mice lacking expression of periostin displayed preserved renal function and structure during GN. Furthermore, delayed administration of periostin antisense oligonucleotides in wild-type animals with GN reversed already established proteinuria, diminished tissue inflammation, and improved renal structure. Lack of periostin expression also blunted the de novo renal expression of integrin-β3 and phosphorylation of focal adhesion kinase and AKT, known mediators of integrin-β3 signaling that affect cell motility and survival, observed during GN in wild-type animals. In vitro, recombinant periostin increased the expression of integrin-β3 and the concomitant phosphorylation of focal adhesion kinase and AKT in podocytes. Notably, periostin and integrin-β3 were highly colocalized in biopsy specimens from patients with inflammatory GN. These results demonstrate that interplay between periostin and renal inflammation orchestrates inflammatory and fibrotic responses, driving podocyte damage through downstream activation of integrin-β3 signaling. Targeting periostin may be a novel therapeutic strategy for treating CKD.
Collapse
Affiliation(s)
- Niki Prakoura
- Institut National de la Santé Et de la Recherche Médicale, Unité Mixte de Recherche Scientifique 1155, Tenon Hospital, Paris, France
| | - Panagiotis Kavvadas
- Institut National de la Santé Et de la Recherche Médicale, Unité Mixte de Recherche Scientifique 1155, Tenon Hospital, Paris, France
| | - Raphaёl Kormann
- Institut National de la Santé Et de la Recherche Médicale, Unité Mixte de Recherche Scientifique 1155, Tenon Hospital, Paris, France.,Sorbonne Université, Université Pierre-et-Marie-Curie Paris 6, Paris, France; and
| | - Jean-Claude Dussaule
- Institut National de la Santé Et de la Recherche Médicale, Unité Mixte de Recherche Scientifique 1155, Tenon Hospital, Paris, France.,Sorbonne Université, Université Pierre-et-Marie-Curie Paris 6, Paris, France; and.,Department of Physiology, Saint-Antoine Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Christos E Chadjichristos
- Institut National de la Santé Et de la Recherche Médicale, Unité Mixte de Recherche Scientifique 1155, Tenon Hospital, Paris, France.,Sorbonne Université, Université Pierre-et-Marie-Curie Paris 6, Paris, France; and
| | - Christos Chatziantoniou
- Institut National de la Santé Et de la Recherche Médicale, Unité Mixte de Recherche Scientifique 1155, Tenon Hospital, Paris, France; .,Sorbonne Université, Université Pierre-et-Marie-Curie Paris 6, Paris, France; and
| |
Collapse
|
35
|
Ricciardelli C, Lokman NA, Ween MP, Oehler MK. WOMEN IN CANCER THEMATIC REVIEW: Ovarian cancer-peritoneal cell interactions promote extracellular matrix processing. Endocr Relat Cancer 2016; 23:T155-T168. [PMID: 27578826 DOI: 10.1530/erc-16-0320] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 08/30/2016] [Indexed: 12/13/2022]
Abstract
Ovarian cancer has a distinct tendency for metastasising via shedding of cancerous cells into the peritoneal cavity and implanting onto the peritoneum that lines the pelvic organs. Once ovarian cancer cells adhere to the peritoneal cells, they migrate through the peritoneal layer and invade the local organs. Alterations in the extracellular environment are critical for tumour initiation, progression and intra-peritoneal dissemination. To increase our understanding of the molecular mechanisms involved in ovarian cancer metastasis and to identify novel therapeutic targets, we recently studied the interaction of ovarian cancer and peritoneal cells using a proteomic approach. We identified several extracellular matrix (ECM) proteins including, fibronectin, TGFBI, periostin, annexin A2 and PAI-1 that were processed as a result of the ovarian cancer-peritoneal cell interaction. This review focuses on the functional role of these proteins in ovarian cancer metastasis. Our findings together with published literature support the notion that ECM processing via the plasminogen-plasmin pathway promotes the colonisation and attachment of ovarian cancer cells to the peritoneum and actively contributes to the early steps of ovarian cancer metastasis.
Collapse
Affiliation(s)
- C Ricciardelli
- Discipline of Obstetrics and GynaecologyAdelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - N A Lokman
- Discipline of Obstetrics and GynaecologyAdelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - M P Ween
- Lung Research LaboratoryHanson Institute, Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - M K Oehler
- Discipline of Obstetrics and GynaecologyAdelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
- Department of Gynaecological OncologyRoyal Adelaide Hospital, Adelaide, South Australia, Australia
| |
Collapse
|
36
|
Van Malderen S, Wijchers S, Akca F, Caliskan K, Szili-Torok T. Mismatch between the origin of premature ventricular complexes and the noncompacted myocardium in patients with noncompaction cardiomyopathy patients: involvement of the conduction system? Ann Noninvasive Electrocardiol 2016; 22. [PMID: 27568851 DOI: 10.1111/anec.12394] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Noncompaction cardiomyopathy (NCCM) is considered to be the result of an arrest in the normal myocardial embryogenesis. The histological, developmental, and electrophysiological explanation of ventricular arrhythmias in NCCM is still unknown. The aim of this study was to determine the origin of premature ventricular contractions (PVCs) in NCCM and to identify any predominant arrhythmic foci. METHODS Retrospective data from our NCCM registry including 101 patients were analyzed. A total number of 2069 electrocardiograms (ECGs) were studied to determine the origin of PVCs. Echocardiographic data were analyzed in patients with PVCs in all 12 leads. Segments affected by noncompaction (NC) were compared with the origin of PVCs. RESULTS PVCs were documented in 250 ECGs from 55 (54%) patients. Thirty-five ECGs recorded PVCs on all 12 leads and the origin of 20 types of PVCs could be determined. Ninety-five percent of PVCs did not originate from left ventricular NC myocardial areas and two PVCs (10%) had a true myocardial origin. All other PVCs originated from structures such as the outflow tracts (8/20), the fascicles (7/20), especially the posteromedial fascicle (6/20), and the mitral and tricuspid annulus (3/20). CONCLUSIONS Our data suggest that PVCs in NCCM mainly originate from the conduction system and related myocardium.
Collapse
Affiliation(s)
- Sophie Van Malderen
- Department of Clinical Electrophysiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Sip Wijchers
- Department of Clinical Electrophysiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ferdi Akca
- Department of Clinical Electrophysiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Kadir Caliskan
- Department of Heart Failure/Heart Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Tamas Szili-Torok
- Department of Clinical Electrophysiology, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
37
|
Walker JT, McLeod K, Kim S, Conway SJ, Hamilton DW. Periostin as a multifunctional modulator of the wound healing response. Cell Tissue Res 2016; 365:453-65. [PMID: 27234502 DOI: 10.1007/s00441-016-2426-6] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 05/04/2016] [Indexed: 12/23/2022]
Abstract
During tissue healing, the dynamic and temporal alterations required for effective repair occur in the structure and composition of the extracellular matrix (ECM). Matricellular proteins (MPs) are a group of diverse non-structural ECM components that bind cell surface receptors mediating interactions between the cell and its microenviroment, effectively regulating adhesion, migration, proliferation, signaling, and cell phenotype. Periostin (Postn), a pro-fibrogenic secreted glycoprotein, is defined as an MP based on its expression pattern and regulatory roles during development and healing and in disease processes. Postn consists of a typical signal sequence, an EMI domain responsible for binding to fibronectin, four tandem fasciclin-like domains that are responsible for integrin binding, and a C-terminal region in which multiple splice variants originate. This review focuses specifically on the role of Postn in wound healing and remodeling, an area of intense research during the last 10 years, particularly as related to skin healing and myocardium post-infarction. Postn interacts with cells through various integrin pairs and is an essential downstream effector of transforming growth factor-β superfamily signaling. Across various tissues, Postn is associated with the pro-fibrogenic process: specifically, the transition of fibroblasts to myofibroblasts, collagen fibrillogenesis, and ECM synthesis. Although the complexity of Postn as a modulator of cell behavior in tissue healing is only beginning to be elucidated, its expression is clearly a defining event in moving wound healing through the proliferative and remodeling phases.
Collapse
Affiliation(s)
- John T Walker
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond Street, London, ON, Canada, N6A 5C1
| | - Karrington McLeod
- Graduate Program in Biomedical Engineering, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond Street, London, ON, Canada, N6A 5C1
| | - Shawna Kim
- Division of Oral Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond Street, London, ON, Canada, N6A 5C1
| | - Simon J Conway
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Douglas W Hamilton
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond Street, London, ON, Canada, N6A 5C1.
- Graduate Program in Biomedical Engineering, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond Street, London, ON, Canada, N6A 5C1.
- Division of Oral Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond Street, London, ON, Canada, N6A 5C1.
| |
Collapse
|
38
|
Kaur H, Takefuji M, Ngai CY, Carvalho J, Bayer J, Wietelmann A, Poetsch A, Hoelper S, Conway SJ, Möllmann H, Looso M, Troidl C, Offermanns S, Wettschureck N. Targeted Ablation of Periostin-Expressing Activated Fibroblasts Prevents Adverse Cardiac Remodeling in Mice. Circ Res 2016; 118:1906-17. [PMID: 27140435 DOI: 10.1161/circresaha.116.308643] [Citation(s) in RCA: 187] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
RATIONALE Activated cardiac fibroblasts (CF) are crucial players in the cardiac damage response; excess fibrosis, however, may result in myocardial stiffening and heart failure development. Inhibition of activated CF has been suggested as a therapeutic strategy in cardiac disease, but whether this truly improves cardiac function is unclear. OBJECTIVE To study the effect of CF ablation on cardiac remodeling. METHODS AND RESULTS We characterized subgroups of murine CF by single-cell expression analysis and identified periostin as the marker showing the highest correlation to an activated CF phenotype. We generated bacterial artificial chromosome-transgenic mice allowing tamoxifen-inducible Cre expression in periostin-positive cells as well as their diphtheria toxin-mediated ablation. In the healthy heart, periostin expression was restricted to valvular fibroblasts; ablation of this population did not affect cardiac function. After chronic angiotensin II exposure, ablation of activated CF resulted in significantly reduced cardiac fibrosis and improved cardiac function. After myocardial infarction, ablation of periostin-expressing CF resulted in reduced fibrosis without compromising scar stability, and cardiac function was significantly improved. Single-cell transcriptional analysis revealed reduced CF activation but increased expression of prohypertrophic factors in cardiac macrophages and cardiomyocytes, resulting in localized cardiomyocyte hypertrophy. CONCLUSIONS Modulation of the activated CF population is a promising approach to prevent adverse cardiac remodeling in response to angiotensin II and after myocardial infarction.
Collapse
Affiliation(s)
- Harmandeep Kaur
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.)
| | - Mikito Takefuji
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.)
| | - C Y Ngai
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.)
| | - Jorge Carvalho
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.)
| | - Julia Bayer
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.)
| | - Astrid Wietelmann
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.)
| | - Ansgar Poetsch
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.)
| | - Soraya Hoelper
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.)
| | - Simon J Conway
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.)
| | - Helge Möllmann
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.)
| | - Mario Looso
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.)
| | - Christian Troidl
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.)
| | - Stefan Offermanns
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.)
| | - Nina Wettschureck
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.).
| |
Collapse
|
39
|
Danziger J, Young RL, Shea MK, Tracy RP, Ix JH, Jenny NS, Mukamal KJ. Vitamin K-Dependent Protein Activity and Incident Ischemic Cardiovascular Disease: The Multi-Ethnic Study of Atherosclerosis. Arterioscler Thromb Vasc Biol 2016; 36:1037-42. [PMID: 27034472 PMCID: PMC5844474 DOI: 10.1161/atvbaha.116.307273] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 03/14/2016] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Vitamin K-dependent proteins (VKDPs), which require post-translational modification to achieve biological activity, seem to contribute to thrombus formation, vascular calcification, and vessel stiffness. Whether VKDP activity is prospectively associated with incident cardiovascular disease has not been studied. APPROACH AND RESULTS VKDP activity was determined by measuring circulating des-γ-carboxy prothrombin concentrations in a random sample of 709 multiethnic adults free of cardiovascular disease drawn from the Multi-Ethnic Study of Atherosclerosis (MESA). Lower des-γ-carboxy prothrombin concentrations reflect greater VKDP activity. Subjects were followed up for the risk of ischemic cardiovascular disease (coronary heart disease, stroke, and fatal cardiovascular disease) for 11.0 years of follow-up. A total of 75 first ischemic CVD events occurred during follow-up. The incidence of ischemic cardiovascular disease increased progressively across des-γ-carboxy prothrombin quartiles, with event rates of 5.9 and 11.7 per 1000 person-years in the lowest and highest quartiles. In analyses adjusted for traditional cardiovascular risk factors and measures of vitamin K intake, a doubling of des-γ-carboxy prothrombin concentration was associated with a 1.53 (95% confidence interval, 1.09-2.13; P=0.008) higher risk of incident ischemic cardiovascular disease. The association was consistent across strata of participants with diabetes mellitus, hypertension, renal impairment, and low vitamin K nutritional intake. CONCLUSIONS In this sample of middle-aged and older adults, VKDP activity was associated with incident ischemic cardiovascular events. Further studies to understand the role of this large class of proteins in cardiovascular disease are warranted.
Collapse
Affiliation(s)
- John Danziger
- From the Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (J.D., K.J.M.); Department of Biostatistics, University of Washington, Seattle (R.L.Y.); Human Nutrition Research Center on Aging, Tufts University, Boston, MA (K.M.S.); College of Medicine (R.P.T.) and Department of Pathology (N.S.J.), University of Vermont, Burlington; and Nephrology Section, Veterans Affairs San Diego Healthcare System, CA (J.H.I.).
| | - Rebekah L Young
- From the Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (J.D., K.J.M.); Department of Biostatistics, University of Washington, Seattle (R.L.Y.); Human Nutrition Research Center on Aging, Tufts University, Boston, MA (K.M.S.); College of Medicine (R.P.T.) and Department of Pathology (N.S.J.), University of Vermont, Burlington; and Nephrology Section, Veterans Affairs San Diego Healthcare System, CA (J.H.I.)
| | - M Kyla Shea
- From the Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (J.D., K.J.M.); Department of Biostatistics, University of Washington, Seattle (R.L.Y.); Human Nutrition Research Center on Aging, Tufts University, Boston, MA (K.M.S.); College of Medicine (R.P.T.) and Department of Pathology (N.S.J.), University of Vermont, Burlington; and Nephrology Section, Veterans Affairs San Diego Healthcare System, CA (J.H.I.)
| | - Russell P Tracy
- From the Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (J.D., K.J.M.); Department of Biostatistics, University of Washington, Seattle (R.L.Y.); Human Nutrition Research Center on Aging, Tufts University, Boston, MA (K.M.S.); College of Medicine (R.P.T.) and Department of Pathology (N.S.J.), University of Vermont, Burlington; and Nephrology Section, Veterans Affairs San Diego Healthcare System, CA (J.H.I.)
| | - Joachim H Ix
- From the Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (J.D., K.J.M.); Department of Biostatistics, University of Washington, Seattle (R.L.Y.); Human Nutrition Research Center on Aging, Tufts University, Boston, MA (K.M.S.); College of Medicine (R.P.T.) and Department of Pathology (N.S.J.), University of Vermont, Burlington; and Nephrology Section, Veterans Affairs San Diego Healthcare System, CA (J.H.I.)
| | - Nancy S Jenny
- From the Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (J.D., K.J.M.); Department of Biostatistics, University of Washington, Seattle (R.L.Y.); Human Nutrition Research Center on Aging, Tufts University, Boston, MA (K.M.S.); College of Medicine (R.P.T.) and Department of Pathology (N.S.J.), University of Vermont, Burlington; and Nephrology Section, Veterans Affairs San Diego Healthcare System, CA (J.H.I.)
| | - Kenneth J Mukamal
- From the Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (J.D., K.J.M.); Department of Biostatistics, University of Washington, Seattle (R.L.Y.); Human Nutrition Research Center on Aging, Tufts University, Boston, MA (K.M.S.); College of Medicine (R.P.T.) and Department of Pathology (N.S.J.), University of Vermont, Burlington; and Nephrology Section, Veterans Affairs San Diego Healthcare System, CA (J.H.I.)
| |
Collapse
|
40
|
Abstract
Myocardial fibrosis is a significant global health problem associated with nearly all forms of heart disease. Cardiac fibroblasts comprise an essential cell type in the heart that is responsible for the homeostasis of the extracellular matrix; however, upon injury, these cells transform to a myofibroblast phenotype and contribute to cardiac fibrosis. This remodeling involves pathological changes that include chamber dilation, cardiomyocyte hypertrophy and apoptosis, and ultimately leads to the progression to heart failure. Despite the critical importance of fibrosis in cardiovascular disease, our limited understanding of the cardiac fibroblast impedes the development of potential therapies that effectively target this cell type and its pathological contribution to disease progression. This review summarizes current knowledge regarding the origins and roles of fibroblasts, mediators and signaling pathways known to influence fibroblast function after myocardial injury, as well as novel therapeutic strategies under investigation to attenuate cardiac fibrosis.
Collapse
Affiliation(s)
- Joshua G Travers
- From the Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, OH
| | - Fadia A Kamal
- From the Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, OH
| | - Jeffrey Robbins
- From the Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, OH
| | - Katherine E Yutzey
- From the Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, OH
| | - Burns C Blaxall
- From the Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, OH.
| |
Collapse
|
41
|
Stanzel S, Stubbusch J, Pataskar A, Howard MJ, Deller T, Ernsberger U, Tiwari VK, Rohrer H, Tsarovina K. Distinct roles of hand2 in developing and adult autonomic neurons. Dev Neurobiol 2016; 76:1111-24. [PMID: 26818017 DOI: 10.1002/dneu.22378] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 01/05/2016] [Accepted: 01/07/2016] [Indexed: 11/08/2022]
Abstract
The bHLH transcription factor Hand2 is essential for the acquisition and maintenance of noradrenergic properties of embryonic sympathetic neurons and controls neuroblast proliferation. Hand2 is also expressed in embryonic and postnatal parasympathetic ganglia and remains expressed in sympathetic neurons up to the adult stage. Here, we address its function in developing parasympathetic and adult sympathetic neurons. We conditionally deleted Hand2 in the parasympathetic sphenopalatine ganglion by crossing a line of floxed Hand2 mice with DbhiCre transgenic mice, taking advantage of the transient Dbh expression in parasympathetic ganglia. Hand2 elimination does not affect Dbh expression and sphenopalatine ganglion size at E12.5 and E16.5, in contrast to sympathetic ganglia. These findings demonstrate different functions for Hand2 in the parasympathetic and sympathetic lineage. Our previous Hand2 knockdown in postmitotic, differentiated chick sympathetic neurons resulted in decreased expression of noradrenergic marker genes but it was unclear whether Hand2 is required for maintaining noradrenergic neuron identity in adult animals. We now show that Hand2 elimination in adult Dbh-expressing sympathetic neurons does not decrease the expression of Th and Dbh, in contrast to the situation during development. However, gene expression profiling of adult sympathetic neurons identified 75 Hand2-dependent target genes. Interestingly, a notable proportion of down-regulated genes (15%) encode for proteins with synaptic and neurotransmission functions. These results demonstrate a change in Hand2 target genes during maturation of sympathetic neurons. Whereas Hand2 controls genes regulating noradrenergic differentiation during development, Hand2 seems to be involved in the regulation of genes controlling neurotransmission in adult sympathetic neurons. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 76: 1111-1124, 2016.
Collapse
Affiliation(s)
- Sabine Stanzel
- Developmental Neurobiology, Max-Planck-Institute for Brain Research, Max-von-Laue-Str. 4, Frankfurt/M, 60438, Germany
| | - Jutta Stubbusch
- Developmental Neurobiology, Max-Planck-Institute for Brain Research, Max-von-Laue-Str. 4, Frankfurt/M, 60438, Germany
| | - Abhijeet Pataskar
- Institute of Molecular Biology (IMB) Boehringer Ingelheim Foundation, Ackermannweg 4, Mainz, 55128, Germany
| | - Marthe J Howard
- Department of Neurosciences and Program in Neurosciences and Neurological Disorders, University of Toledo Health Sciences Campus, Toledo, Ohio, 43614
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Goethe University Frankfurt/M, Theodor-Stern-Kai 7, Frankfurt/M, 60590, Germany
| | - Uwe Ernsberger
- Developmental Neurobiology, Max-Planck-Institute for Brain Research, Max-von-Laue-Str. 4, Frankfurt/M, 60438, Germany.,Institute of Clinical Neuroanatomy, Goethe University Frankfurt/M, Theodor-Stern-Kai 7, Frankfurt/M, 60590, Germany.,Ernst-Strüngmann-Institute, Deutschordenstr. 46, Frankfurt/M, 60528, Germany
| | - Vijay K Tiwari
- Institute of Molecular Biology (IMB) Boehringer Ingelheim Foundation, Ackermannweg 4, Mainz, 55128, Germany
| | - Hermann Rohrer
- Developmental Neurobiology, Max-Planck-Institute for Brain Research, Max-von-Laue-Str. 4, Frankfurt/M, 60438, Germany.,Institute of Clinical Neuroanatomy, Goethe University Frankfurt/M, Theodor-Stern-Kai 7, Frankfurt/M, 60590, Germany.,Ernst-Strüngmann-Institute, Deutschordenstr. 46, Frankfurt/M, 60528, Germany
| | - Konstantina Tsarovina
- Developmental Neurobiology, Max-Planck-Institute for Brain Research, Max-von-Laue-Str. 4, Frankfurt/M, 60438, Germany
| |
Collapse
|
42
|
Extracellular matrix-mediated cellular communication in the heart. J Mol Cell Cardiol 2016; 91:228-37. [PMID: 26778458 DOI: 10.1016/j.yjmcc.2016.01.011] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 01/10/2016] [Accepted: 01/11/2016] [Indexed: 01/13/2023]
Abstract
The extracellular matrix (ECM) is a complex and dynamic scaffold that maintains tissue structure and dynamics. However, the view of the ECM as an inert architectural support has been increasingly challenged. The ECM is a vibrant meshwork, a crucial organizer of cellular microenvironments. It plays a direct role in cellular interactions regulating cell growth, survival, spreading, proliferation, differentiation and migration through the intricate relationship among cellular and acellular tissue components. This complex interrelationship preserves cardiac function during homeostasis; however it is also responsible for pathologic remodeling following myocardial injury. Therefore, enhancing our understanding of this cross-talk may provide mechanistic insights into the pathogenesis of heart failure and suggest new approaches to novel, targeted pharmacologic therapies. This review explores the implications of ECM-cell interactions in myocardial cell behavior and cardiac function at baseline and following myocardial injury.
Collapse
|
43
|
Martin-Rojas T, Mourino-Alvarez L, Alonso-Orgaz S, Rosello-Lleti E, Calvo E, Lopez-Almodovar LF, Rivera M, Padial LR, Lopez JA, de la Cuesta F, Barderas MG. iTRAQ proteomic analysis of extracellular matrix remodeling in aortic valve disease. Sci Rep 2015; 5:17290. [PMID: 26620461 PMCID: PMC4664895 DOI: 10.1038/srep17290] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 10/28/2015] [Indexed: 02/08/2023] Open
Abstract
Degenerative aortic stenosis (AS) is the most common worldwide cause of valve replacement. The aortic valve is a thin, complex, layered connective tissue with compartmentalized extracellular matrix (ECM) produced by specialized cell types, which directs blood flow in one direction through the heart. There is evidence suggesting remodeling of such ECM during aortic stenosis development. Thus, a better characterization of the role of ECM proteins in this disease would increase our understanding of the underlying molecular mechanisms. Aortic valve samples were collected from 18 patients which underwent aortic valve replacement (50% males, mean age of 74 years) and 18 normal control valves were obtained from necropsies (40% males, mean age of 69 years). The proteome of the samples was analyzed by 2D-LC MS/MS iTRAQ methodology. The results showed an altered expression of 13 ECM proteins of which 3 (biglycan, periostin, prolargin) were validated by Western blotting and/or SRM analyses. These findings are substantiated by our previous results demonstrating differential ECM protein expression. The present study has demonstrated a differential ECM protein pattern in individuals with AS, therefore supporting previous evidence of a dynamic ECM remodeling in human aortic valves during AS development.
Collapse
Affiliation(s)
- Tatiana Martin-Rojas
- Department of Vascular Physiopathology, Hospital Nacional de Parapléjicos, SESCAM, Toledo, Spain
| | - Laura Mourino-Alvarez
- Department of Vascular Physiopathology, Hospital Nacional de Parapléjicos, SESCAM, Toledo, Spain
| | - Sergio Alonso-Orgaz
- Department of Vascular Physiopathology, Hospital Nacional de Parapléjicos, SESCAM, Toledo, Spain
| | - Esther Rosello-Lleti
- Cardiocirculatory Unit, Health Research Institute, Hospital La Fe, Valencia, Spain
| | | | | | - Miguel Rivera
- Cardiocirculatory Unit, Health Research Institute, Hospital La Fe, Valencia, Spain
| | - Luis R Padial
- Department of Cardiology, Hospital Virgen de la Salud, SESCAM, Toledo, Spain
| | | | - Fernando de la Cuesta
- Department of Vascular Physiopathology, Hospital Nacional de Parapléjicos, SESCAM, Toledo, Spain
| | - Maria G Barderas
- Department of Vascular Physiopathology, Hospital Nacional de Parapléjicos, SESCAM, Toledo, Spain
| |
Collapse
|
44
|
Schwanekamp JA, Lorts A, Vagnozzi RJ, Vanhoutte D, Molkentin JD. Deletion of Periostin Protects Against Atherosclerosis in Mice by Altering Inflammation and Extracellular Matrix Remodeling. Arterioscler Thromb Vasc Biol 2015; 36:60-8. [PMID: 26564821 DOI: 10.1161/atvbaha.115.306397] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 10/29/2015] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Periostin is a secreted protein that can alter extracellular matrix remodeling in response to tissue injury. However, the functional role of periostin in the development of atherosclerotic plaques has yet to be described despite its observed induction in diseased vessels and presence in the serum. APPROACH AND RESULTS Hyperlipidemic, apolipoprotein E-null mice (ApoE(-/) (-)) were crossed with periostin (Postn(-/-)) gene-deleted mice and placed on a high-fat diet for 6 or 14 weeks to induce atherosclerosis. En face analysis of aortas showed significantly decreased lesion areas of ApoE(-/-) Postn(-/-) mice compared with ApoE(-/-) mice, as well as a reduced inflammatory response with less macrophage content. Moreover, diseased aortas from ApoE(-/-) Postn(-/-) mice displayed a disorganized extracellular matrix with less collagen cross linking and smaller fibrotic caps, as well as increased matrix metalloproteinase-2, metalloproteinase-13, and procollagen-lysine, 2-oxoglutarate 5-dioxygenase-1 mRNA expression. Furthermore, the loss of periostin was associated with a switch in vascular smooth muscle cells toward a more proliferative and synthetic phenotype. Mechanistically, the loss of periostin reduced macrophage recruitment by transforming growth factor-β in cellular migration assays. CONCLUSIONS These are the first genetic data detailing the function of periostin as a regulator of atherosclerotic lesion formation and progression. The data suggest that periostin could be a therapeutic target for atherosclerotic plaque formation through modulation of the immune response and extracellular matrix remodeling.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/immunology
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Aortic Diseases/genetics
- Aortic Diseases/immunology
- Aortic Diseases/metabolism
- Aortic Diseases/pathology
- Aortic Diseases/prevention & control
- Apolipoproteins E/deficiency
- Apolipoproteins E/genetics
- Atherosclerosis/genetics
- Atherosclerosis/immunology
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/prevention & control
- Cell Adhesion Molecules/deficiency
- Cell Adhesion Molecules/genetics
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- Collagen/metabolism
- Diet, High-Fat
- Disease Models, Animal
- Disease Progression
- Extracellular Matrix/metabolism
- Gene Deletion
- Gene Expression Regulation
- Inflammation/genetics
- Inflammation/immunology
- Inflammation/metabolism
- Inflammation/pathology
- Inflammation/prevention & control
- Macrophages/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Plaque, Atherosclerotic
- RNA, Messenger/metabolism
- Signal Transduction
- Time Factors
- Vascular Remodeling
Collapse
Affiliation(s)
- Jennifer A Schwanekamp
- From the Department of Pediatrics (J.A.S., A.L., R.J.V., D.V., J.D.M.) and Howard Hughes Medical Institute (J.D.M.), Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH
| | - Angela Lorts
- From the Department of Pediatrics (J.A.S., A.L., R.J.V., D.V., J.D.M.) and Howard Hughes Medical Institute (J.D.M.), Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH
| | - Ronald J Vagnozzi
- From the Department of Pediatrics (J.A.S., A.L., R.J.V., D.V., J.D.M.) and Howard Hughes Medical Institute (J.D.M.), Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH
| | - Davy Vanhoutte
- From the Department of Pediatrics (J.A.S., A.L., R.J.V., D.V., J.D.M.) and Howard Hughes Medical Institute (J.D.M.), Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH
| | - Jeffery D Molkentin
- From the Department of Pediatrics (J.A.S., A.L., R.J.V., D.V., J.D.M.) and Howard Hughes Medical Institute (J.D.M.), Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH.
| |
Collapse
|
45
|
Guan J, Liu WQ, Xing MQ, Shi Y, Tan XY, Jiang CQ, Dai HY. Elevated expression of periostin in diabetic cardiomyopathy and the effect of valsartan. BMC Cardiovasc Disord 2015; 15:90. [PMID: 26281830 PMCID: PMC4539668 DOI: 10.1186/s12872-015-0084-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 08/10/2015] [Indexed: 12/13/2022] Open
Abstract
Background Periostin, an extracellular matrix protein, plays a significant role in adverse cardiac remodeling. However, no report has documented the function of periostin in left ventricular remodeling of streptozototin (STZ)-induced diabetic rats. The aim of the present study was to observe the expression of periostin in Wistar rat’s myocardium of diabetic cardiomyopathy (DCM) and the effect of valsartan on it. Methods Immunohistochemistry, real-time polymerase chain reaction, and Western blot analysis were used to determine the degree of expression and location of periostin, transforming growth factor (TGF)-β1, TGF-β1 type II receptor (TGF-β1 R II), and Type I and III collagens in the myocardium of STZ-induced diabetic rats. Results Periostin, TGF-β1, TGF-β1 R II, and Type I and III collagens were significantly increased in the myocardium of diabetic rats compared with control group on both messenger ribonucleic acid and protein levels. In addition, diabetic rats treated with valsartan could have reduced expression of periostin and improved cardiac remodeling of DCM. Conclusions Periostin may play a crucial role in cardiac remodeling and myocardial interstitial fibrosis process of DCM and it could be one of the important mechanisms for valsartan to improve the ventricular remodeling of DCM.
Collapse
Affiliation(s)
- Jun Guan
- Department of Cardiology, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Wen-Qi Liu
- Department of Cardiology, Qingdao Municipal Hospital, Qingdao, Shandong, China.,Qingdao University Medical College, Qingdao, Shandong, China.,Key Laboratory of cellular transplantation , Chinese Ministry of Public Health, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Ming-Qing Xing
- Department of Clinical laboratory, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Yue Shi
- Qingdao University Medical College, Qingdao, Shandong, China
| | - Xue-Ying Tan
- Qingdao University Medical College, Qingdao, Shandong, China.,Key Laboratory of cellular transplantation , Chinese Ministry of Public Health, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Chang-Qing Jiang
- Department of pathology department, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Hong-Yan Dai
- Department of Cardiology, Qingdao Municipal Hospital, Qingdao, Shandong, China.
| |
Collapse
|
46
|
Hong L, Shejiao D, Fenrong C, Gang Z, Lei D. Periostin down-regulation attenuates the pro-fibrogenic response of hepatic stellate cells induced by TGF-β1. J Cell Mol Med 2015; 19:2462-8. [PMID: 26249143 PMCID: PMC4594687 DOI: 10.1111/jcmm.12636] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 05/20/2015] [Indexed: 01/18/2023] Open
Abstract
Liver fibrosis is characterized by an exacerbated accumulation of deposition of the extracellular matrix (ECM), and the activation of hepatic stellate cells (HSC) plays a pivotal role in the development of liver fibrosis. Periostin has been shown to regulate cell adhesion, proliferation, migration and apoptosis; however, the involvement of periostin and its role in transforming growth factor (TGF)-β1-induced HSC activation remains unclear. We used RT-PCR and Western blot to evaluate the expression level of periostin in hepatic fibrosis tissues and HSCs, respectively. Cell proliferation was determined using the Cell Proliferation ELISA BrdU kit, cell cycle was analysed by flow cytometry. The expression of α-smooth muscle actin (α-SMA), collagen I, TGF-β1, p-Smad2 and p-Smad3 were determined by western blot. Our study found that periostin was up-regulated in liver fibrotic tissues and activated HSCs. In addition, siRNA-periostin suppressed TGF-β1-induced HSC proliferation. The HSC transfected with siRNA-periostin significantly inhibited TGF-β1-induced expression levels of α-SMA and collagen I. Furthermore, TGF-β1 stimulated the expression of periostin, and siRNA-periostin attenuated TGF-β1-induced Smad2/3 activation in HSCs. These results suggest that periostin may function as a novel regulator to modulate HSC activation, potentially by promoting the TGF-β1/Smad signalling pathway, and propose a strategy to target periostin for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Li Hong
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Dai Shejiao
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Chen Fenrong
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhao Gang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Dong Lei
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
47
|
Sonnenberg-Riethmacher E, Miehe M, Riethmacher D. Promotion of periostin expression contributes to the migration of Schwann cells. J Cell Sci 2015; 128:3345-55. [PMID: 26187852 DOI: 10.1242/jcs.174177] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 06/25/2015] [Indexed: 12/15/2022] Open
Abstract
Neuregulin ligands and their ErbB receptors are important for the development of Schwann cells, the glial cells of the peripheral nervous system (PNS). ErbB3 deficiency is characterized by a complete loss of Schwann cells along axons of the peripheral nerves, impaired fasciculation and neuronal cell death. We performed comparative gene expression analysis of dorsal root ganglia (DRG) explant cultures from ErbB3-deficient and wild-type mice in order to identify genes that are involved in Schwann cell development and migration. The extracellular matrix (ECM) gene periostin was found to exhibit the most prominent down regulation in ErbB3-deficient DRG. Expression analysis revealed that the periostin-expressing cell population in the PNS corresponds to Schwann cell precursors and Schwann cells, and is particularly high in migratory Schwann cells. Furthermore, stimulation of Schwann cells with neuregulin-1 (NRG1) or transforming growth factor β (TGFβ-1) resulted in an upregulation of periostin expression. Interestingly, DRG explant cultures of periostin-deficient mice revealed a significant reduction of the number of migrating Schwann cells. These data demonstrate that the expression of periostin is stimulated by ErbB ligand NRG1 and influences the migration of Schwann cell precursors.
Collapse
Affiliation(s)
- Eva Sonnenberg-Riethmacher
- Human Development and Health, University of Southampton, School of Medicine, Tremona Road, Southampton SO16 6YD, UK Center for Molecular Neurobiology, University of Hamburg, Falkenried 94, Hamburg 20251, Germany
| | - Michaela Miehe
- Center for Molecular Neurobiology, University of Hamburg, Falkenried 94, Hamburg 20251, Germany Institut for Immunological Engineering, University of Aarhus, Gustav Wieds Vej 10, Aarhus C 8000, Denmark
| | - Dieter Riethmacher
- Human Development and Health, University of Southampton, School of Medicine, Tremona Road, Southampton SO16 6YD, UK Center for Molecular Neurobiology, University of Hamburg, Falkenried 94, Hamburg 20251, Germany
| |
Collapse
|
48
|
Cardiac fibroblasts: from development to heart failure. J Mol Med (Berl) 2015; 93:823-30. [PMID: 26169532 DOI: 10.1007/s00109-015-1314-y] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 06/22/2015] [Accepted: 06/24/2015] [Indexed: 01/18/2023]
Abstract
Cardiac fibroblasts are a major cell population of the heart and are characterized by their capacity to produce extracellular matrix (ECM). In hearts subjected to pressure overload, excessive fibroblast accumulation is responsible for fibrosis of the myocardium, a major clinical issue. Hence, understanding mechanisms generating fibroblasts in this context has become a key question in the cardiovascular field. Recent studies now point to the activation of resident fibroblasts as the underlying cause of fibrosis. However, de novo generation of fibroblasts from endothelium and circulating hematopoietic cells has also been proposed to significantly contribute to fibrosis. Here, we discuss the latest findings on fibroblast origins, with a particular emphasis on the pressure overload model, and the implication of these findings for the development of anti-fibrotic therapies that are currently lacking.
Collapse
|
49
|
Endocardial Brg1 disruption illustrates the developmental origins of semilunar valve disease. Dev Biol 2015; 407:158-72. [PMID: 26100917 DOI: 10.1016/j.ydbio.2015.06.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 06/12/2015] [Accepted: 06/13/2015] [Indexed: 11/24/2022]
Abstract
The formation of intricately organized aortic and pulmonic valves from primitive endocardial cushions of the outflow tract is a remarkable accomplishment of embryonic development. While not always initially pathologic, developmental semilunar valve (SLV) defects, including bicuspid aortic valve, frequently progress to a disease state in adults requiring valve replacement surgery. Disrupted embryonic growth, differentiation, and patterning events that "trigger" SLV disease are coordinated by gene expression changes in endocardial, myocardial, and cushion mesenchymal cells. We explored roles of chromatin regulation in valve gene regulatory networks by conditional inactivation of the Brg1-associated factor (BAF) chromatin remodeling complex in the endocardial lineage. Endocardial Brg1-deficient mouse embryos develop thickened and disorganized SLV cusps that frequently become bicuspid and myxomatous, including in surviving adults. These SLV disease-like phenotypes originate from deficient endocardial-to-mesenchymal transformation (EMT) in the proximal outflow tract (pOFT) cushions. The missing cells are replaced by compensating neural crest or other non-EMT-derived mesenchyme. However, these cells are incompetent to fully pattern the valve interstitium into distinct regions with specialized extracellular matrices. Transcriptomics reveal genes that may promote growth and patterning of SLVs and/or serve as disease-state biomarkers. Mechanistic studies of SLV disease genes should distinguish between disease origins and progression; the latter may reflect secondary responses to a disrupted developmental system.
Collapse
|
50
|
Matsuzawa M, Arai C, Nomura Y, Murata T, Yamakoshi Y, Oida S, Hanada N, Nakamura Y. Periostin of human periodontal ligament fibroblasts promotes migration of human mesenchymal stem cell through the αvβ3 integrin/FAK/PI3K/Akt pathway. J Periodontal Res 2015; 50:855-63. [PMID: 25900259 DOI: 10.1111/jre.12277] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2015] [Indexed: 01/06/2023]
Abstract
BACKGROUND AND OBJECTIVE The periodontal ligament (PDL) is characterized by rapid turnover, high remodeling capacity and high inherent regenerative potential compared with other connective tissues. Periostin, which is highly expressed in the fibroblasts in the PDL, has been widely discussed in relation to collagen fibrillogenesis in the PDL. Recently, several reports have indicated periostin in cell migration. The aim of this study was to examine whether human PDL fibroblasts (hPDLFs) with high levels of periostin expression promote the migration of human bone marrow mesenchymal stem cells (hMSCs). MATERIAL AND METHODS The migration of hMSCs was examined by transwell chamber migration assay under different conditions: medium alone, hPDLFs, human dermal fibroblasts, recombinant periostin, integrin αvβ3 blocking antibody (anti-CD51/61 antibody) and inhibitors of FAK (PF431396) and PI3K (LY294002). Phosphorylation of FAK and Akt in hMSCs under stimulation of periostin was examined by western blotting. RESULTS The migration assay revealed that the number of migrated hMSCs by hPDLFs was significantly larger than those by dermal fibroblasts, periostin small interfering RNA hPDLFs and medium alone. Furthermore, recombinant periostin also strongly induced hMSC migration. The addition of anti-CD51/61 antibody, PF431396 and LY294002 caused a significant reduction in the number of migrated hMSCs respectively. The anti-CD51/61 antibody inhibited both FAK and Akt phosphorylations under periostin stimulation. PF431396 inhibited both FAK and Akt phosphorylations. LY294002 inhibited only Akt phosphorylation, and FAK phosphorylation was not influenced under periostin stimulation. CONCLUSION Periostin expression in hPDLFs promotes the migration of hMSCs through the αvβ3 integrin/FAK/PI3K/Akt pathway in vitro.
Collapse
Affiliation(s)
- M Matsuzawa
- Department of Orthodontics, Tsurumi University School of Dental Medicine, Tsurumi-ku, Yokohama, Japan
| | - C Arai
- Department of Orthodontics, Tsurumi University School of Dental Medicine, Tsurumi-ku, Yokohama, Japan
| | - Y Nomura
- Department of Translation Research, Tsurumi University School of Dental Medicine, Tsurumi-ku, Yokohama, Japan
| | - T Murata
- Department of Translation Research, Tsurumi University School of Dental Medicine, Tsurumi-ku, Yokohama, Japan
| | - Y Yamakoshi
- Department of Biochemistry and Molecular Biology, Tsurumi University School of Dental Medicine, Tsurumi-ku, Yokohama, Japan
| | - S Oida
- Department of Biochemistry and Molecular Biology, Tsurumi University School of Dental Medicine, Tsurumi-ku, Yokohama, Japan
| | - N Hanada
- Department of Translation Research, Tsurumi University School of Dental Medicine, Tsurumi-ku, Yokohama, Japan
| | - Y Nakamura
- Department of Orthodontics, Tsurumi University School of Dental Medicine, Tsurumi-ku, Yokohama, Japan
| |
Collapse
|