1
|
Shi J, Cheng L, Chen M, Fang Y, Li G, Luo X, He P, Wu H, Chen J. CSPG4 overexpression implicates higher risks of recurrence and tumorigenesis after surgical intervention of vocal fold Leukoplakia. Eur Arch Otorhinolaryngol 2025; 282:1381-1392. [PMID: 39863819 DOI: 10.1007/s00405-025-09217-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] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND Vocal fold leukoplakia (VFL), a precancerous lesion of the larynx, is characterized by white plaques on the vocal fold mucous membrane. Currently, there are no reliable biomarkers to predict the recurrence and malignant transformation of VFL. Considering chondroitin sulfate proteoglycan 4 (CSPG4) as a biomarker for malignant tumors such as laryngeal squamous cell carcinoma (LSCC), we conducted this cohort study to evaluate the prognostic influence of CSPG4 expression on VFL patients. METHODS This study included 44 patients with surgical intervention of VFL. CSPG4 expression in VFL were observed using immunohistochemistry, and the relationship between the prognosis of VFL patients and CSPG4 expression were investigated using Spearman correlation analysis and multiple Cox regression models. RESULTS During a median follow-up of 89.3 months, recurrence occurred in 19 postsurgical VFL patients, and malignant transformation occurred in 10 patients. The recurrence and malignant transformation group showed significant differences in the H-SCORE of CSPG4 (P = 0.005 and P = 0.045) compared to the the non-recurrence group and the non-malignant transformation group. And the CSPG4 expression level was positive correlation with recurrence and malignant transformation (0.306 or 0.416, spearman correlation confident). Receiver operating characteristic curve analysis indicated that CSPG4 H-SCORE has predictive value for poor prognosis (areas under the curve, 0.742 and 0.710; cut-off points, 82.5 and 177.5, respectively). Multiple Cox regression analysis revealed a statistically significant effect of lesion size (P = 0.014) and CSPG4 H-SCORE (P = 0.005) on recurrence as well as pathological type (P = 0.001) and CSPG4 H-SCORE stratified by 177.5 (P = 0.039). Our survival data indicated that higher CSPG4 expression was associated with a shorter time of recurrence (P = 0.003) and tumorigenesis (P<0.001). CONCLUSION CSPG4 overexpression indicates higher risks and shorter time of postoperative VFL recurrence and tumorigenesis. Detection of CSPG4 expression in VFL may be a novel approach to assess the outcome of VFL patients.
Collapse
Affiliation(s)
- Jiajia Shi
- ENT institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 FenYang Road, Shanghai, 200031, China
| | - Lei Cheng
- ENT institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 FenYang Road, Shanghai, 200031, China
| | - Min Chen
- ENT institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 FenYang Road, Shanghai, 200031, China
| | - Yi Fang
- ENT institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 FenYang Road, Shanghai, 200031, China
| | - Guangfei Li
- ENT institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 FenYang Road, Shanghai, 200031, China
| | - Xiaomei Luo
- Department of General Medicine, Damen Town Central Health Center, Wenzhou, 325701, Zhejiang, China
| | - Peijie He
- ENT institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 FenYang Road, Shanghai, 200031, China
| | - Haitao Wu
- ENT institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 FenYang Road, Shanghai, 200031, China.
| | - Jian Chen
- ENT institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 FenYang Road, Shanghai, 200031, China.
| |
Collapse
|
2
|
Yan X, Zhang X, Li H, Zou Y, Lu W, Zhan M, Liang Z, Zhuang H, Ran X, Ma G, Lin X, Yang H, Huang Y, Wang H, Shen L. Application of Proteomics and Machine Learning Methods to Study the Pathogenesis of Diabetic Nephropathy and Screen Urinary Biomarkers. J Proteome Res 2024; 23:3612-3625. [PMID: 38949094 DOI: 10.1021/acs.jproteome.4c00267] [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] [Indexed: 07/02/2024]
Abstract
Diabetic nephropathy (DN) has become the main cause of end-stage renal disease worldwide, causing significant health problems. Early diagnosis of the disease is quite inadequate. To screen urine biomarkers of DN and explore its potential mechanism, this study collected urine from 87 patients with type 2 diabetes mellitus (which will be classified into normal albuminuria, microalbuminuria, and macroalbuminuria groups) and 38 healthy subjects. Twelve individuals from each group were then randomly selected as the screening cohort for proteomics analysis and the rest as the validation cohort. The results showed that humoral immune response, complement activation, complement and coagulation cascades, renin-angiotensin system, and cell adhesion molecules were closely related to the progression of DN. Five overlapping proteins (KLK1, CSPG4, PLAU, SERPINA3, and ALB) were identified as potential biomarkers by machine learning methods. Among them, KLK1 and CSPG4 were positively correlated with the urinary albumin to creatinine ratio (UACR), and SERPINA3 was negatively correlated with the UACR, which were validated by enzyme-linked immunosorbent assay (ELISA). This study provides new insights into disease mechanisms and biomarkers for early diagnosis of DN.
Collapse
Affiliation(s)
- Xi Yan
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Xinglai Zhang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Haiying Li
- Department of Endocrinology, Guiyang First People's Hospital, Guiyang, Guizhou 550002, China
| | - Yongdong Zou
- Center for Instrumental Analysis, Shenzhen University, Shenzhen 518071, China
| | - Wei Lu
- Department of Endocrinology, Guiyang First People's Hospital, Guiyang, Guizhou 550002, China
| | - Man Zhan
- Department of Endocrinology, Guiyang First People's Hospital, Guiyang, Guizhou 550002, China
| | - Zhiyuan Liang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Hongbin Zhuang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Xiaoqian Ran
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Guanwei Ma
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Xixiao Lin
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Hongbo Yang
- Center for Instrumental Analysis, Shenzhen University, Shenzhen 518071, China
| | - Yuhan Huang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Hanghang Wang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Liming Shen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, China
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| |
Collapse
|
3
|
Childress KO, Cencer CS, Tyska MJ, Lacy DB. Nectin-3 and shed forms of CSPG4 can serve as epithelial cell receptors for Clostridioides difficile TcdB. mBio 2023; 14:e0185723. [PMID: 37747247 PMCID: PMC10653914 DOI: 10.1128/mbio.01857-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 07/26/2023] [Indexed: 09/26/2023] Open
Abstract
IMPORTANCE Toxin B (TcdB) is a major virulence factor of Clostridioides difficile, a Gram-positive pathogen that is a leading cause of hospital-acquired diarrhea. While previous studies have established that TcdB can engage multiple cell surface receptors in vitro, little is known about how these interactions promote disease and where these receptors localize on colonic tissue. Here, we used immunofluorescence microscopy to visualize Nectin-3 and CSPG4 on tissue, revealing unexpected localization of both receptors on colonic epithelial cells. We show that Nectin-3, which was previously characterized as an adherens junction protein, is also localized to the brush border of colonocytes. Staining for CSPG4 revealed that it is present along epithelial cell junctions, suggesting that it is shed by fibroblasts along the crypt-surface axis. Collectively, our study provides new insights into how TcdB can gain access to the receptors Nectin-3 and CSPG4 to intoxicate colonic epithelial cells.
Collapse
Affiliation(s)
- Kevin O. Childress
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Caroline S. Cencer
- Department of Cell and Development Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Matthew J. Tyska
- Department of Cell and Development Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - D. Borden Lacy
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
4
|
Bagheri Varzaneh M, Zhao Y, Rozynek J, Han M, Reed DA. Disrupting mechanical homeostasis promotes matrix metalloproteinase-13 mediated processing of neuron glial antigen 2 in mandibular condylar cartilage. Eur Cell Mater 2023; 45:113-130. [PMID: 37154195 PMCID: PMC10405277 DOI: 10.22203/ecm.v045a08] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/10/2023] Open
Abstract
Post-traumatic osteoarthritis in the temporomandibular joint (TMJ OA) is associated dysfunctional cellmatrix mediated signalling resulting from changes in the pericellular microenvironment after injury. Matrix metalloproteinase (MMP)-13 is a critical enzyme in biomineralisation and the progression of OA that can both degrade the extracellular matrix and modify extracellular receptors. This study focused on MMP-13 mediated changes in a transmembrane proteoglycan, Neuron Glial antigen 2 (NG2/CSPG4). NG2/CSPG4 is a receptor for type VI collagen and a known substrate for MMP-13. In healthy articular layer chondrocytes, NG2/CSPG4 is membrane bound but becomes internalised during TMJ OA. The objective of this study was to determine if MMP-13 contributed to the cleavage and internalisation of NG2/CSPG4 during mechanical loading and OA progression. Using preclinical and clinical samples, it was shown that MMP-13 was present in a spatiotemporally consistent pattern with NG2/CSPG4 internalisation during TMJ OA. In vitro, it was illustrated that inhibiting MMP-13 prevented retention of the NG2/CSPG4 ectodomain in the extracellular matrix. Inhibiting MMP-13 promoted the accumulation of membrane-associated NG2/CSPG4 but did not affect the formation of mechanical-loading dependent variant specific fragments of the ectodomain. MMP- 13 mediated cleavage of NG2/CSPG4 is necessary to initiate clathrin-mediated internalisation of the NG2/ CSPG4 intracellular domain following mechanical loading. This mechanically sensitive MMP-13-NG2/CSPG4 axis affected the expression of key mineralisation and OA genes including bone morphogenetic protein 2, and parathyroid hormone-related protein. Together, these findings implicated MMP-13 mediated cleavage of NG2/CSPG4 in the mechanical homeostasis of mandibular condylar cartilage during the progression of degenerative arthropathies such as OA.
Collapse
Affiliation(s)
| | | | | | | | - D A Reed
- 801 South Paulina Street, Room 431, Chicago, IL 60612,
| |
Collapse
|
5
|
do Valle IB, Oliveira SR, da Silva JM, Peterle GT, Có ACG, Sousa-Neto SS, Mendonça EF, de Arruda JAA, Gomes NA, da Silva G, Leopoldino AM, Macari S, Birbrair A, von Zeidler SV, Diniz IMA, Silva TA. The participation of tumor residing pericytes in oral squamous cell carcinoma. Sci Rep 2023; 13:5460. [PMID: 37015965 PMCID: PMC10073133 DOI: 10.1038/s41598-023-32528-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 03/29/2023] [Indexed: 04/06/2023] Open
Abstract
Pericytes are perivascular cells related to vessel structure and angiogenesis that can interact with neoplastic cells, interfering with cancer progression and outcomes. This study focused on the characterization of pericytes in oral squamous cell carcinoma (OSCC) using clinical samples and a transgenic mouse model of oral carcinogenesis. Nestin-/NG2+ (type-1) and nestin+/NG2+ (type-2) pericytes were analyzed by direct fluorescence after induction of oral carcinogenesis (4-nitroquinoline-1-oxide). Gene expression of neuron glial antigen-2 (NG2), platelet-derived growth factor receptor beta (PDGFR-β), and cluster of differentiation 31 (CD31) was examined in human OSCC tissues. The protein expression of von Willebrand factor and NG2 was assessed in oral leukoplakia (i.e., oral potentially malignant disorders) and OSCC samples. Additionally, clinicopathological aspects and survival data were correlated and validated by bioinformatics using The Cancer Genome Atlas (TCGA). Induction of carcinogenesis in mice produced an increase in both NG2+ pericyte subsets. In human OSCC, advanced-stage tumors showed a significant reduction in CD31 mRNA and von Willebrand factor-positive vessels. Low PDGFR-β expression was related to a shorter disease-free survival time, while NG2 mRNA overexpression was associated with a reduction in overall survival, consistent with the TCGA data. Herein, oral carcinogenesis resulted in an increase in NG2+ pericytes, which negatively affected survival outcomes.
Collapse
Affiliation(s)
- Isabella Bittencourt do Valle
- Department of Oral Surgery, Pathology and Clinical Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, room 3105, Belo Horizonte, Minas Gerais, CEP: 31.270-901, Brazil
| | - Sicília Rezende Oliveira
- Department of Oral Surgery, Pathology and Clinical Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, room 3105, Belo Horizonte, Minas Gerais, CEP: 31.270-901, Brazil
| | - Janine Mayra da Silva
- Department of Oral Surgery, Pathology and Clinical Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, room 3105, Belo Horizonte, Minas Gerais, CEP: 31.270-901, Brazil
| | - Gabriela Tonini Peterle
- Biotechnology Post-graduation Program, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Anna Clara Gregório Có
- Biotechnology Post-graduation Program, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Sebastião Silvério Sousa-Neto
- Department of Stomatology (Oral Pathology), School of Dentistry, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Elismauro Francisco Mendonça
- Department of Stomatology (Oral Pathology), School of Dentistry, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - José Alcides Almeida de Arruda
- Department of Oral Surgery, Pathology and Clinical Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, room 3105, Belo Horizonte, Minas Gerais, CEP: 31.270-901, Brazil
| | - Natália Aparecida Gomes
- Department of Restorative Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Gabriel da Silva
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Andréia Machado Leopoldino
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Soraia Macari
- Department of Restorative Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Alexander Birbrair
- Department of Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Sandra Ventorin von Zeidler
- Biotechnology Post-graduation Program, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Ivana Márcia Alves Diniz
- Department of Restorative Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Tarcília Aparecida Silva
- Department of Oral Surgery, Pathology and Clinical Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, room 3105, Belo Horizonte, Minas Gerais, CEP: 31.270-901, Brazil.
| |
Collapse
|
6
|
Mead TJ, Bhutada S, Martin DR, Apte SS. Proteolysis: a key post-translational modification regulating proteoglycans. Am J Physiol Cell Physiol 2022; 323:C651-C665. [PMID: 35785985 PMCID: PMC9448339 DOI: 10.1152/ajpcell.00215.2022] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/28/2022] [Accepted: 06/28/2022] [Indexed: 11/22/2022]
Abstract
Proteoglycans are composite molecules comprising a protein backbone, i.e., the core protein, with covalently attached glycosaminoglycan chains of distinct chemical types. Most proteoglycans are secreted or attached to the cell membrane. Their specialized structures, binding properties, and biophysical attributes underlie diverse biological roles, which include modulation of tissue mechanics, cell adhesion, and the sequestration and regulated release of morphogens, growth factors, and cytokines. As an irreversible post-translational modification, proteolysis has a profound impact on proteoglycan function, abundance, and localization. Proteolysis is required for molecular maturation of some proteoglycans, clearance of extracellular matrix proteoglycans during tissue remodeling, generation of bioactive fragments from proteoglycans, and ectodomain shedding of cell-surface proteoglycans. Genetic evidence shows that proteoglycan core protein proteolysis is essential for diverse morphogenetic events during embryonic development. In contrast, dysregulated proteoglycan proteolysis contributes to osteoarthritis, cardiovascular disorders, cancer, and inflammation. Proteolytic fragments of perlecan, versican, aggrecan, brevican, collagen XVIII, and other proteoglycans are associated with independent biological activities as so-called matrikines. Yet, proteoglycan proteolysis has been investigated to only a limited extent to date. Here, we review the actions of proteases on proteoglycans and illustrate their functional impact with several examples. We discuss the applications and limitations of strategies used to define cleavage sites in proteoglycans and explain how proteoglycanome-wide proteolytic mapping, which is desirable to fully understand the impact of proteolysis on proteoglycans, can be facilitated by integrating classical proteoglycan isolation methods with mass spectrometry-based proteomics.
Collapse
Affiliation(s)
- Timothy J Mead
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Sumit Bhutada
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Daniel R Martin
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Suneel S Apte
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| |
Collapse
|
7
|
Zhang H, Wu Z, Hu D, Yan M, Sun J, Lai J, Bai L. Immunotherapeutic Targeting of NG2/CSPG4 in Solid Organ Cancers. Vaccines (Basel) 2022; 10:vaccines10071023. [PMID: 35891187 PMCID: PMC9321363 DOI: 10.3390/vaccines10071023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/22/2022] [Accepted: 06/22/2022] [Indexed: 12/10/2022] Open
Abstract
Neuro-glia antigen 2/chondroitin sulfate proteoglycan 4 (NG2/CSPG4, also called MCSP, HMW-MAA, MSK16, MCSPG, MEL-CSPG, or gp240) is a large cell-surface antigen and an unusual cell membrane integral glycoprotein frequently expressed on undifferentiated precursor cells in multiple solid organ cancers, including cancers of the liver, pancreas, lungs, and kidneys. It is a valuable molecule involved in cancer cell adhesion, invasion, spreading, angiogenesis, complement inhibition, and signaling. Although the biological significance underlying NG2/CSPG4 proteoglycan involvement in cancer progression needs to be better defined, based on the current evidence, NG2/CSPG4+ cells, such as pericytes (PCs, NG2+/CD146+/PDGFR-β+) and cancer stem cells (CSCs), are closely associated with the liver malignancy, hepatocellular carcinoma (HCC), pancreatic malignancy, and pancreatic ductal adenocarcinoma (PDAC) as well as poor prognoses. Importantly, with a unique method, we successfully purified NG2/CSPG4-expressing cells from human HCC and PDAC vasculature tissue blocks (by core needle biopsy). The cells appeared to be spheres that stably expanded in cultures. As such, these cells have the potential to be used as sources of target antigens. Herein, we provide new information on the possibilities of frequently selecting NG2/CSPG4 as a solid organ cancer biomarker or exploiting expressing cells such as CSCs, or the PG/chondroitin sulfate chain of NG2/CSPG4 on the cell membrane as specific antigens for the development of antibody- and vaccine-based immunotherapeutic approaches to treat these cancers.
Collapse
Affiliation(s)
- Hongyu Zhang
- Hepatobiliary Institute, Southwest Hospital, Army Medical University, Chongqing 400038, China; (H.Z.); (Z.W.); (D.H.); (M.Y.); (J.S.); (J.L.)
| | - Zhenyu Wu
- Hepatobiliary Institute, Southwest Hospital, Army Medical University, Chongqing 400038, China; (H.Z.); (Z.W.); (D.H.); (M.Y.); (J.S.); (J.L.)
| | - Deyu Hu
- Hepatobiliary Institute, Southwest Hospital, Army Medical University, Chongqing 400038, China; (H.Z.); (Z.W.); (D.H.); (M.Y.); (J.S.); (J.L.)
- Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Min Yan
- Hepatobiliary Institute, Southwest Hospital, Army Medical University, Chongqing 400038, China; (H.Z.); (Z.W.); (D.H.); (M.Y.); (J.S.); (J.L.)
- Department of Nuclear Medicine, The First Affiliated Hospital, Shanxi Medical University, Taiyuan 030000, China
| | - Jing Sun
- Hepatobiliary Institute, Southwest Hospital, Army Medical University, Chongqing 400038, China; (H.Z.); (Z.W.); (D.H.); (M.Y.); (J.S.); (J.L.)
| | - Jiejuan Lai
- Hepatobiliary Institute, Southwest Hospital, Army Medical University, Chongqing 400038, China; (H.Z.); (Z.W.); (D.H.); (M.Y.); (J.S.); (J.L.)
| | - Lianhua Bai
- Hepatobiliary Institute, Southwest Hospital, Army Medical University, Chongqing 400038, China; (H.Z.); (Z.W.); (D.H.); (M.Y.); (J.S.); (J.L.)
- Bioengineering College, Chongqing University, Chongqing 400044, China
- Department of Nuclear Medicine, The First Affiliated Hospital, Shanxi Medical University, Taiyuan 030000, China
- Correspondence: ; Tel.: +86-23-68765709; Fax: +86-2365462170
| |
Collapse
|
8
|
Chelyshev YA, Kabdesh IM, Mukhamedshina YO. Extracellular Matrix in Neural Plasticity and Regeneration. Cell Mol Neurobiol 2022; 42:647-664. [PMID: 33128689 PMCID: PMC11441266 DOI: 10.1007/s10571-020-00986-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/22/2020] [Indexed: 12/19/2022]
Abstract
The extracellular matrix (ECM) is a fundamental component of biological tissues. The ECM in the central nervous system (CNS) is unique in both composition and function. Functions such as learning, memory, synaptogenesis, and plasticity are regulated by numerous ECM molecules. The neural ECM acts as a non-specific physical barrier that modulates neuronal plasticity and axon regeneration. There are two specialized types of ECM in the CNS, diffuse perisynaptic ECM and condensed ECM, which selectively surround the perikaryon and initial part of dendritic trees in subtypes of neurons, forming perineuronal nets. This review presents the current knowledge about the role of important neuronal ECM molecules in maintaining the basic functions of a neuron, including electrogenesis and the ability to form neural circuits. The review mainly focuses on the role of ECM components that participate in the control of key events such as cell survival, axonal growth, and synaptic remodeling. Particular attention is drawn to the numerous molecular partners of the main ECM components. These regulatory molecules are integrated into the cell membrane or disposed into the matrix itself in solid or soluble form. The interaction of the main matrix components with molecular partners seems essential in molecular mechanisms controlling neuronal functions. Special attention is paid to the chondroitin sulfate proteoglycan 4, type 1 transmembrane protein, neural-glial antigen 2 (NG2/CSPG4), whose cleaved extracellular domain is such a molecular partner that it not only acts directly on neural and vascular cells, but also exerts its influence indirectly by binding to resident ECM molecules.
Collapse
Affiliation(s)
- Yurii A Chelyshev
- Department of Histology, Cytology and Embryology, Kazan State Medical University, Kazan, Russia
| | - Ilyas M Kabdesh
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kremlevskaya St 18, Kazan, Tatarstan, Russia, 420008
| | - Yana O Mukhamedshina
- Department of Histology, Cytology and Embryology, Kazan State Medical University, Kazan, Russia.
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kremlevskaya St 18, Kazan, Tatarstan, Russia, 420008.
| |
Collapse
|
9
|
Reed DA, Zhao Y, Bagheri Varzaneh M, Shin JS, Rozynek J, Miloro M, Han M. NG2/CSPG4 regulates cartilage degeneration during TMJ osteoarthritis. FRONTIERS IN DENTAL MEDICINE 2022; 3:1004942. [PMID: 36685663 PMCID: PMC9850834 DOI: 10.3389/fdmed.2022.1004942] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Changes in the mechanical homeostasis of the temporomandibular joint (TMJ) can lead to the initiation and progression of degenerative arthropathies such as osteoarthritis (OA). Cells sense and engage with their mechanical microenvironment through interactions with the extracellular matrix. In the mandibular condylar cartilage, the pericellular microenvironment is composed of type VI collagen. NG2/CSPG4 is a transmembrane proteoglycan that binds with type VI collagen, and has been implicated in the cell stress response through mechanical loading-sensitive signaling networks including ERK 1/2. The objective of this study is to define the role of NG2/CSPG4 in the initiation and progression of TMJ OA and to determine if NG2/CSPG4 engages ERK 1/2 in a mechanical loading dependent manner. In vivo, we induced TMJ OA in control and NG2/CSPG4 knockout mice using a surgical destabilization approach. In control mice, NG2/CSPG4 is depleted during the early stages of TMJ OA and NG2/CSPG4 knockout mice have more severe cartilage degeneration, elevated expression of key OA proteases, and suppression of OA matrix synthesis genes. In vitro, we characterized the transcriptome and protein from control and NG2/CSPG4 knockout cells and found significant dysregulation of the ERK 1/2 signaling axis. To characterize the mechanobiological response of NG2/CSPG4, we applied mechanical loads on cell-agarose-collagen scaffolds using a compression bioreactor and illustrate that NG2/CSPG4 knockout cells fail to mechanically activate ERK 1/2 and are associated with changes in the expression of the same key OA biomarkers measured in vivo. Together, these findings implicate NG2/CSPG4 in the mechanical homeostasis of TMJ cartilage and in the progression of degenerative arthropathies including OA.
Collapse
Affiliation(s)
- David A. Reed
- Department of Oral Biology, University of Illinois Chicago, Chicago, IL, United States,,CORRESPONDENCE: David A. Reed,
| | - Yan Zhao
- Department of Oral Biology, University of Illinois Chicago, Chicago, IL, United States
| | - Mina Bagheri Varzaneh
- Department of Oral Biology, University of Illinois Chicago, Chicago, IL, United States
| | - Jun Soo Shin
- Department of Oral Biology, University of Illinois Chicago, Chicago, IL, United States
| | - Jacob Rozynek
- Department of Oral Biology, University of Illinois Chicago, Chicago, IL, United States
| | - Michael Miloro
- Department of Oral and Maxillofacial Surgery, University of Illinois Chicago, Chicago, IL, United States
| | - Michael Han
- Department of Oral and Maxillofacial Surgery, University of Illinois Chicago, Chicago, IL, United States
| |
Collapse
|
10
|
Identification of a germline CSPG4 variation in a family with neurofibromatosis type 1-like phenotype. Cell Death Dis 2021; 12:765. [PMID: 34344877 PMCID: PMC8333038 DOI: 10.1038/s41419-021-04056-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 11/28/2022]
Abstract
Neurofibromatosis type 1 (NF1), an autosomal dominant and multisystem disorder, is generally considered to be caused by NF1 inactivation. However, there are also numerous studies showing that Neurofibromatosis type 1-like phenotype can be caused by the abnormalities in the other genes. Through targeted parallel sequencing, whole-exome sequencing, de novo genomic sequencing, and RNA isoform sequencing, we identified a germline V2097M variation in CSPG4 gene probably increased susceptibility to a NF1-like phenotype family. Besides, a series of in vitro functional studies revealed that this variant promoted cell proliferation by activating the MAPK/ERK signaling pathway via hindering ectodomain cleavage of CSPG4. Our data demonstrate that a germline variation in the CSPG4 gene might be a high risk to cause NF1-like phenotype. To our knowledge, this is the first report of mutations in the CSPG4 gene in human diseases.
Collapse
|
11
|
Kabdesh IM, Arkhipova SS, Mukhamedshina YO, James V, Rizvanov AA, Chelyshev YA. The Function of NG2/CSPG4-expressing Cells in the Rat Spinal Cord Injury: An Immunoelectron Microscopy Study. Neuroscience 2021; 467:142-149. [PMID: 34102261 DOI: 10.1016/j.neuroscience.2021.05.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/24/2021] [Accepted: 05/27/2021] [Indexed: 10/21/2022]
Abstract
Emerging evidence supports an increased role for NG2/CSPG4-expressing cells in the process of neuroregeneration and synaptic plasticity, due to the increased production of multifunctional chondroitin sulfate proteoglycan (NG2/CSPG4). However, the response of NG2/CSPG4-expressing cells in spinal cord injury (SCI) remains to be elcudiated. Expression and distribution of NG2/CSPG4-expressing cells were studied by immunoelectron microscopy in the ventral horns (VH) of an intact and injured rat spinal cord. In the intact spinal cord, NG2/CSPG4 expression was detected on the cell membrane and in the cytoplasm of NG2 glia and was absent in neurons. Large amounts of NG2/CSPG4 were found on myelin membranes. The ability of intact astrocytes to produce NG2/CSPG4 was shown, although to a lesser extent than oligodendrocytes and NG2 glia. At 7 days after SCI at the Th8 level in the reactive glial zone of VH, the expression of NG2/CSPG4 sharply increased in NG2 glia at a distance of 3-5 mm and in reactive astrocytes were observed at all investigated distances caudally from the epicenter of injury. The obtained results indicate the presence of NG2/CSPG4-positive astrocytes in the intact spinal cord, and in the case of damage, an increase in the ability of reactive astrocytes to produce NG2/CSPG4. SCI leads to increased expression of NG2/CSPG4 by NG2 glia in the early stages after injury, which decreases with distance from the epicenter of the injury, as well as at later stages.
Collapse
Affiliation(s)
| | | | - Yana O Mukhamedshina
- Kazan (Volga Region) Federal University, Kazan, Russia; Kazan State Medical University, Kazan, Russia.
| | | | | | - Yuri A Chelyshev
- Kazan (Volga Region) Federal University, Kazan, Russia; Kazan State Medical University, Kazan, Russia
| |
Collapse
|
12
|
Girolamo F, de Trizio I, Errede M, Longo G, d'Amati A, Virgintino D. Neural crest cell-derived pericytes act as pro-angiogenic cells in human neocortex development and gliomas. Fluids Barriers CNS 2021; 18:14. [PMID: 33743764 PMCID: PMC7980348 DOI: 10.1186/s12987-021-00242-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 02/13/2021] [Indexed: 02/07/2023] Open
Abstract
Central nervous system diseases involving the parenchymal microvessels are frequently associated with a ‘microvasculopathy’, which includes different levels of neurovascular unit (NVU) dysfunction, including blood–brain barrier alterations. To contribute to the understanding of NVU responses to pathological noxae, we have focused on one of its cellular components, the microvascular pericytes, highlighting unique features of brain pericytes with the aid of the analyses carried out during vascularization of human developing neocortex and in human gliomas. Thanks to their position, centred within the endothelial/glial partition of the vessel basal lamina and therefore inserted between endothelial cells and the perivascular and vessel-associated components (astrocytes, oligodendrocyte precursor cells (OPCs)/NG2-glia, microglia, macrophages, nerve terminals), pericytes fulfil a central role within the microvessel NVU. Indeed, at this critical site, pericytes have a number of direct and extracellular matrix molecule- and soluble factor-mediated functions, displaying marked phenotypical and functional heterogeneity and carrying out multitasking services. This pericytes heterogeneity is primarily linked to their position in specific tissue and organ microenvironments and, most importantly, to their ontogeny. During ontogenesis, pericyte subtypes belong to two main embryonic germ layers, mesoderm and (neuro)ectoderm, and are therefore expected to be found in organs ontogenetically different, nonetheless, pericytes of different origin may converge and colonize neighbouring areas of the same organ/apparatus. Here, we provide a brief overview of the unusual roles played by forebrain pericytes in the processes of angiogenesis and barriergenesis by virtue of their origin from midbrain neural crest stem cells. A better knowledge of the ontogenetic subpopulations may support the understanding of specific interactions and mechanisms involved in pericyte function/dysfunction, including normal and pathological angiogenesis, thereby offering an alternative perspective on cell subtype-specific therapeutic approaches. ![]()
Collapse
Affiliation(s)
- Francesco Girolamo
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy.
| | - Ignazio de Trizio
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy.,Intensive Care Unit, Department of Intensive Care, Regional Hospital of Lugano, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Mariella Errede
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| | - Giovanna Longo
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Molecular Biology Unit, University of Bari School of Medicine, Bari, Italy
| | - Antonio d'Amati
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy.,Department of Emergency and Organ Transplantation, Pathology Section, University of Bari School of Medicine, Bari, Italy
| | - Daniela Virgintino
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| |
Collapse
|
13
|
Girolamo F, de Trizio I, Errede M, Longo G, d’Amati A, Virgintino D. Neural crest cell-derived pericytes act as pro-angiogenic cells in human neocortex development and gliomas. Fluids Barriers CNS 2021. [DOI: 10.1186/s12987-021-00242-7 union select null--] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
AbstractCentral nervous system diseases involving the parenchymal microvessels are frequently associated with a ‘microvasculopathy’, which includes different levels of neurovascular unit (NVU) dysfunction, including blood–brain barrier alterations. To contribute to the understanding of NVU responses to pathological noxae, we have focused on one of its cellular components, the microvascular pericytes, highlighting unique features of brain pericytes with the aid of the analyses carried out during vascularization of human developing neocortex and in human gliomas. Thanks to their position, centred within the endothelial/glial partition of the vessel basal lamina and therefore inserted between endothelial cells and the perivascular and vessel-associated components (astrocytes, oligodendrocyte precursor cells (OPCs)/NG2-glia, microglia, macrophages, nerve terminals), pericytes fulfil a central role within the microvessel NVU. Indeed, at this critical site, pericytes have a number of direct and extracellular matrix molecule- and soluble factor-mediated functions, displaying marked phenotypical and functional heterogeneity and carrying out multitasking services. This pericytes heterogeneity is primarily linked to their position in specific tissue and organ microenvironments and, most importantly, to their ontogeny. During ontogenesis, pericyte subtypes belong to two main embryonic germ layers, mesoderm and (neuro)ectoderm, and are therefore expected to be found in organs ontogenetically different, nonetheless, pericytes of different origin may converge and colonize neighbouring areas of the same organ/apparatus. Here, we provide a brief overview of the unusual roles played by forebrain pericytes in the processes of angiogenesis and barriergenesis by virtue of their origin from midbrain neural crest stem cells. A better knowledge of the ontogenetic subpopulations may support the understanding of specific interactions and mechanisms involved in pericyte function/dysfunction, including normal and pathological angiogenesis, thereby offering an alternative perspective on cell subtype-specific therapeutic approaches.
Collapse
|
14
|
iPS-Derived Early Oligodendrocyte Progenitor Cells from SPMS Patients Reveal Deficient In Vitro Cell Migration Stimulation. Cells 2020; 9:cells9081803. [PMID: 32751289 PMCID: PMC7463559 DOI: 10.3390/cells9081803] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/22/2020] [Accepted: 07/28/2020] [Indexed: 12/18/2022] Open
Abstract
The most challenging aspect of secondary progressive multiple sclerosis (SPMS) is the lack of efficient regenerative response for remyelination, which is carried out by the endogenous population of adult oligoprogenitor cells (OPCs) after proper activation. OPCs must proliferate and migrate to the lesion and then differentiate into mature oligodendrocytes. To investigate the OPC cellular component in SPMS, we developed induced pluripotent stem cells (iPSCs) from SPMS-affected donors and age-matched controls (CT). We confirmed their efficient and similar OPC differentiation capacity, although we reported SPMS-OPCs were transcriptionally distinguishable from their CT counterparts. Analysis of OPC-generated conditioned media (CM) also evinced differences in protein secretion. We further confirmed SPMS-OPC CM presented a deficient capacity to stimulate OPC in vitro migration that can be compensated by exogenous addition of specific components. Our results provide an SPMS-OPC cellular model and encouraging venues to study potential cell communication deficiencies in the progressive form of multiple sclerosis (MS) for future treatment strategies.
Collapse
|
15
|
de Vrij FM, Bouwkamp CG, Gunhanlar N, Shpak G, Lendemeijer B, Baghdadi M, Gopalakrishna S, Ghazvini M, Li TM, Quadri M, Olgiati S, Breedveld GJ, Coesmans M, Mientjes E, de Wit T, Verheijen FW, Beverloo HB, Cohen D, Kok RM, Bakker PR, Nijburg A, Spijker AT, Haffmans PMJ, Hoencamp E, Bergink V, Vorstman JA, Wu T, Olde Loohuis LM, Amin N, Langen CD, Hofman A, Hoogendijk WJ, van Duijn CM, Ikram MA, Vernooij MW, Tiemeier H, Uitterlinden AG, Elgersma Y, Distel B, Gribnau J, White T, Bonifati V, Kushner SA. Candidate CSPG4 mutations and induced pluripotent stem cell modeling implicate oligodendrocyte progenitor cell dysfunction in familial schizophrenia. Mol Psychiatry 2019; 24:757-771. [PMID: 29302076 PMCID: PMC6755981 DOI: 10.1038/s41380-017-0004-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 09/24/2017] [Accepted: 11/01/2017] [Indexed: 01/09/2023]
Abstract
Schizophrenia is highly heritable, yet its underlying pathophysiology remains largely unknown. Among the most well-replicated findings in neurobiological studies of schizophrenia are deficits in myelination and white matter integrity; however, direct etiological genetic and cellular evidence has thus far been lacking. Here, we implement a family-based approach for genetic discovery in schizophrenia combined with functional analysis using induced pluripotent stem cells (iPSCs). We observed familial segregation of two rare missense mutations in Chondroitin Sulfate Proteoglycan 4 (CSPG4) (c.391G > A [p.A131T], MAF 7.79 × 10-5 and c.2702T > G [p.V901G], MAF 2.51 × 10-3). The CSPG4A131T mutation was absent from the Swedish Schizophrenia Exome Sequencing Study (2536 cases, 2543 controls), while the CSPG4V901G mutation was nominally enriched in cases (11 cases vs. 3 controls, P = 0.026, OR 3.77, 95% CI 1.05-13.52). CSPG4/NG2 is a hallmark protein of oligodendrocyte progenitor cells (OPCs). iPSC-derived OPCs from CSPG4A131T mutation carriers exhibited abnormal post-translational processing (P = 0.029), subcellular localization of mutant NG2 (P = 0.007), as well as aberrant cellular morphology (P = 3.0 × 10-8), viability (P = 8.9 × 10-7), and myelination potential (P = 0.038). Moreover, transfection of healthy non-carrier sibling OPCs confirmed a pathogenic effect on cell survival of both the CSPG4A131T (P = 0.006) and CSPG4V901G (P = 3.4 × 10-4) mutations. Finally, in vivo diffusion tensor imaging of CSPG4A131T mutation carriers demonstrated a reduction of brain white matter integrity compared to unaffected sibling and matched general population controls (P = 2.2 × 10-5). Together, our findings provide a convergence of genetic and functional evidence to implicate OPC dysfunction as a candidate pathophysiological mechanism of familial schizophrenia.
Collapse
Affiliation(s)
- Femke M de Vrij
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Christian G Bouwkamp
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Nilhan Gunhanlar
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Guy Shpak
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Bas Lendemeijer
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Maarouf Baghdadi
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Mehrnaz Ghazvini
- Department of Developmental Biology, and Erasmus MC iPS Facility, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Tracy M Li
- Department of Developmental Biology, and Erasmus MC iPS Facility, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marialuisa Quadri
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Simone Olgiati
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Guido J Breedveld
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Michiel Coesmans
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
- Delta Psychiatric Center, Poortugaal, The Netherlands
| | - Edwin Mientjes
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ton de Wit
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Frans W Verheijen
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - H Berna Beverloo
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Dan Cohen
- Mental Health Care Organization North-Holland North, Heerhugowaard, The Netherlands
| | - Rob M Kok
- Parnassia Psychiatric Institute, The Hague, The Netherlands
| | - P Roberto Bakker
- Department of Psychiatry and Psychology, School of Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
- Psychiatric Center GGZ Centraal, Amersfoort, The Netherlands
| | - Aviva Nijburg
- Parnassia Psychiatric Institute, The Hague, The Netherlands
| | | | - P M Judith Haffmans
- Faculty of Social and Behavioral Sciences Clinical, Health and Neuro Psychology, Department of Affective Disorders, PsyQ, Leiden University, Leiden, The Netherlands
| | - Erik Hoencamp
- Parnassia Psychiatric Institute, The Hague, The Netherlands
- Institute of Psychology, Leiden University, Leiden, The Netherlands
| | - Veerle Bergink
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jacob A Vorstman
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Psychiatry, The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Timothy Wu
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Loes M Olde Loohuis
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Najaf Amin
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Carolyn D Langen
- Department of Radiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Medical Informatics, Erasmus Medical Center, Rotterdam, The Netherlands
- Biomedical Imaging Group Rotterdam, Departments of Radiology & Medical Informatics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Witte J Hoogendijk
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - M Arfan Ikram
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Radiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Meike W Vernooij
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Radiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Henning Tiemeier
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Medical Informatics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - André G Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ype Elgersma
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ben Distel
- Department of Medical Biochemistry, Academic Medical Centre, Amsterdam, The Netherlands
| | - Joost Gribnau
- Department of Developmental Biology, and Erasmus MC iPS Facility, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Tonya White
- Department of Medical Informatics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Vincenzo Bonifati
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Steven A Kushner
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
16
|
Tamburini E, Dallatomasina A, Quartararo J, Cortelazzi B, Mangieri D, Lazzaretti M, Perris R. Structural deciphering of the NG2/CSPG4 proteoglycan multifunctionality. FASEB J 2018; 33:3112-3128. [PMID: 30550356 DOI: 10.1096/fj.201801670r] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The chondroitin sulfate proteoglycan 4 ( CSPG4) gene encodes a transmembrane proteoglycan (PG) constituting the largest and most structurally complex macromolecule of the human surfaceome. Its transcript shows an extensive evolutionary conservation and, due to the elaborated intracellular processing of the translated protein, it generates an array of glycoforms with the potential to exert variant-specific functions. CSPG4-mediated molecular events are articulated through the interaction with more than 40 putative ligands and the concurrent involvement of the ectodomain and cytoplasmic tail. Alternating inside-out and outside-in signal transductions may thereby be elicited through a tight functional connection of the PG with the cytoskeleton and its regulators. The potential of CSPG4 to influence both types of signaling mechanisms is also asserted by its lateral mobility along the plasma membrane and its intersection with microdomain-restricted internalization and endocytic trafficking. Owing to the multitude of molecular interplays that CSPG4 may engage, and thanks to a differential phosphorylation of its intracellular domain accounted by crosstalking signaling pathways, the PG stands out for its unique capability to affect numerous cellular phenomena, including those purporting pathologic conditions. We discuss here the progresses made in advancing our understanding about the structural-functional bases for the ability of CSPG4 to widely impact on cell behavior, such as to highlight how its multivalency may be exploited to interfere with disease progression.-Tamburini, E., Dallatomasina, A., Quartararo, J., Cortelazzi, B., Mangieri, D., Lazzaretti, M., Perris, R. Structural deciphering of the NG2/CSPG4 proteoglycan multifunctionality.
Collapse
Affiliation(s)
- Elisa Tamburini
- Centre for Molecular and Translational Oncology (COMT), University of Parma, Parma, Italy
| | - Alice Dallatomasina
- Division of Experimental Oncology, Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy; and
| | - Jade Quartararo
- Centre for Molecular and Translational Oncology (COMT), University of Parma, Parma, Italy
| | - Barbara Cortelazzi
- Centre for Molecular and Translational Oncology (COMT), University of Parma, Parma, Italy
| | | | - Mirca Lazzaretti
- Centre for Molecular and Translational Oncology (COMT), University of Parma, Parma, Italy
| | - Roberto Perris
- Centre for Molecular and Translational Oncology (COMT), University of Parma, Parma, Italy
| |
Collapse
|
17
|
Nayak T, Trotter J, Sakry D. The Intracellular Cleavage Product of the NG2 Proteoglycan Modulates Translation and Cell-Cycle Kinetics via Effects on mTORC1/FMRP Signaling. Front Cell Neurosci 2018; 12:231. [PMID: 30131676 PMCID: PMC6090502 DOI: 10.3389/fncel.2018.00231] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 07/16/2018] [Indexed: 12/11/2022] Open
Abstract
The NG2 proteoglycan is expressed by oligodendrocyte precursor cells (OPCs) and is abundantly expressed by tumors such as melanoma and glioblastoma. Functions of NG2 include an influence on proliferation, migration and neuromodulation. Similar to other type-1 membrane proteins, NG2 undergoes proteolysis, generating a large ectodomain, a C-terminal fragment (CTF) and an intracellular domain (ICD) via sequential action of α- and γ-secretases which is enhanced by neuronal activity. Functional roles of NG2 have so far been shown for the full-length protein, the released ectodomain and CTF, but not for the ICD. In this study, we characterized the role of the NG2 ICD in OPC and Human Embryonic Kidney (HEK) cells. Overexpressed ICD is predominantly localized in the cell cytosol, including the distal processes of OPCs. Nuclear localisation of a fraction of the ICD is dependent on Nuclear Localisation Signals. Immunoprecipitation and Mass Spectrometry followed by functional analysis indicated that the NG2 ICD modulates mRNA translation and cell-cycle kinetics. In OPCs and HEK cells, ICD overexpression results in an mTORC1-dependent upregulation of translation, as well as a shift of the cell population toward S-phase. NG2 ICD increases the active (phosphorylated) form of mTOR and modulates downstream signaling cascades, including increased phosphorylation of p70S6K1 and increased expression of eEF2. Strikingly, levels of FMRP, an RNA-binding protein that is regulated by mTOR/p70S6K1/eEF2 were decreased. In neurons, FMRP acts as a translational repressor under activity-dependent control and is mutated in Fragile X Syndrome (FXS). Knock-down of endogenous NG2 in primary OPC reduced translation and mTOR/p70S6K1 phosphorylation in Oli-neu. Here, we identify the NG2 ICD as a regulator of translation in OPCs via modulation of the well-established mTORC1 pathway. We show that FXS-related FMRP signaling is not exclusive to neurons but plays a role in OPCs. This provides a signal cascade in OPC which can be influenced by the neuronal network, since the NG2 ICD has been shown to be generated by constitutive as well as activity-dependent cleavage. Our results also elucidate a possible role of NG2 in tumors exhibiting enhanced rates of translation and rapid cell cycle kinetics.
Collapse
Affiliation(s)
- Tanmoyita Nayak
- Department of Biology, Molecular Cell Biology, Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Jacqueline Trotter
- Department of Biology, Molecular Cell Biology, Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Dominik Sakry
- Department of Biology, Molecular Cell Biology, Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany.,Department of Molecular Neurobiology, Max Planck Institute for Experimental Medicine, Göttingen, Germany
| |
Collapse
|
18
|
NG2/CSPG4 and progranulin in the posttraumatic glial scar. Matrix Biol 2018; 68-69:571-588. [DOI: 10.1016/j.matbio.2017.10.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/05/2017] [Accepted: 10/06/2017] [Indexed: 12/17/2022]
|
19
|
Tang F, Lord MS, Stallcup WB, Whitelock JM. Cell surface chondroitin sulphate proteoglycan 4 (CSPG4) binds to the basement membrane heparan sulphate proteoglycan, perlecan, and is involved in cell adhesion. J Biochem 2018; 163:399-412. [PMID: 29462330 DOI: 10.1093/jb/mvy008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 11/08/2017] [Indexed: 12/25/2022] Open
Abstract
Chondroitin sulphate proteoglycan 4 (CSPG4) is a cell surface proteoglycan highly expressed by tumour, perivascular and oligodendrocyte cells and known to be involved cell adhesion and migration. This study showed that CSPG4 was present as a proteoglycan on the cell surface of two melanoma cell lines, MM200 and Me1007, as well as shed into the conditioned medium. CSPG4 from the two melanoma cell lines differed in the amount of chondroitin sulphate (CS) decoration, as well as the way the protein core was fragmented. In contrast, the CSPG4 expressed by a colon carcinoma cell line, WiDr, was predominantly as a protein core on the cell surface lacking glycosaminoglycan (GAG) chains. This study demonstrated that CSPG4 immunopurified from the melanoma cell lines formed a complex with perlecan synthesized by the same cultured cells. Mechanistic studies showed that CSPG4 bound to perlecan via hydrophobic protein-protein interactions involving multiple sites on perlecan including the C-terminal region. Furthermore, this study revealed that CSPG4 interacted with perlecan to support cell adhesion and actin polymerization. Together these data suggest a novel mechanism by which CSPG4 expressing cells might attach to perlecan-rich matrices so as those found in connective tissues and basement membranes.
Collapse
Affiliation(s)
- Fengying Tang
- Graduate School of Biomedical Engineering, Level 5 Samuels Building, University of New South Wales, Sydney, NSW 2052, Australia
| | - Megan S Lord
- Graduate School of Biomedical Engineering, Level 5 Samuels Building, University of New South Wales, Sydney, NSW 2052, Australia
| | - William B Stallcup
- Tumour Microenvironment and Cancer Immunology Program, Cancer Centre, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - John M Whitelock
- Graduate School of Biomedical Engineering, Level 5 Samuels Building, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
20
|
Schmitt BM, Laschke MW, Rössler OG, Huang W, Scheller A, Menger MD, Ampofo E. Nerve/glial antigen (NG) 2 is a crucial regulator of intercellular adhesion molecule (ICAM)-1 expression. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1865:57-66. [PMID: 28964848 DOI: 10.1016/j.bbamcr.2017.09.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 09/01/2017] [Accepted: 09/26/2017] [Indexed: 12/13/2022]
Abstract
The proteoglycan nerve/glial antigen (NG) 2 is expressed on multiple cell types and mediates cell proliferation and migration. However, little is known about its function in gene regulation. In this study, we demonstrate that in pericytes and glioblastoma cells intercellular adhesion molecule (ICAM)-1, an essential protein for leukocyte adhesion and transmigration, underlies a NG2-dependent expression. As shown by flow cytometry, Western blot analysis and quantitative real-time polymerase chain reaction (qRT-PCR), silencing of NG2 in human placenta-derived pericytes increased the expression of ICAM-1. Pathway analyses revealed that this is mediated by extracellular-regulated-kinases (ERK) 1/2 signaling. Moreover, leukocyte adhesion to NG2 siRNA-treated pericytes was significantly enhanced when compared to scrambled (scr) siRNA-treated control cells. In vivo, we detected increased ICAM-1 protein levels in the retina of mice lacking NG2 expression. To exclude that this novel mechanism is pericyte-specific, we additionally analyzed the expression of ICAM-1 in dependency of NG2 in two glioblastoma cell lines. We found that A1207 and M059K cells exhibit an inverse expression pattern of NG2 and ICAM-1. Finally, downregulation of NG2 in A1207 cells significantly increased ICAM-1 expression. Taken together, these findings indicate that NG2 may represent a promising target for the modulation of ICAM-1-mediated immune responses.
Collapse
Affiliation(s)
- Beate M Schmitt
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| | - Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| | - Oliver G Rössler
- Medical Biochemistry and Molecular Biology, Saarland University, 66421 Homburg/Saar, Germany
| | - Wenhui Huang
- Department of Molecular Physiology, CIPMM (Center for Integrative Physiology and Molecular Medicine), Saarland University, 66421 Homburg/Saar, Germany
| | - Anja Scheller
- Department of Molecular Physiology, CIPMM (Center for Integrative Physiology and Molecular Medicine), Saarland University, 66421 Homburg/Saar, Germany
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| | - Emmanuel Ampofo
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany.
| |
Collapse
|
21
|
Gomez-Zepeda D, Chaves C, Taghi M, Sergent P, Liu WQ, Chhuon C, Vidal M, Picard M, Thioulouse E, Broutin I, Guerrera IC, Scherrmann JM, Parmentier Y, Decleves X, Menet MC. Targeted unlabeled multiple reaction monitoring analysis of cell markers for the study of sample heterogeneity in isolated rat brain cortical microvessels. J Neurochem 2017; 142:597-609. [DOI: 10.1111/jnc.14095] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 05/31/2017] [Accepted: 06/01/2017] [Indexed: 12/30/2022]
Affiliation(s)
- David Gomez-Zepeda
- Inserm; UMR-S 1144; Variabilité de la réponse aux psychotropes; Paris France
- Université Paris Descartes; Paris France
- Université Paris Diderot; Paris France
| | - Catarina Chaves
- Inserm; UMR-S 1144; Variabilité de la réponse aux psychotropes; Paris France
- Université Paris Descartes; Paris France
- Université Paris Diderot; Paris France
| | - Méryam Taghi
- Inserm; UMR-S 1144; Variabilité de la réponse aux psychotropes; Paris France
- Université Paris Descartes; Paris France
- Université Paris Diderot; Paris France
| | - Philippe Sergent
- Technologie Servier; Département de recherche biopharmaceutique; Orléans France
| | - Wang-Qing Liu
- Université Paris Descartes; Paris France
- CNRS; UMR 8638; Chimie Organique; Médicinale et Extractive et Toxicologie Expérimentale; Paris France
| | - Cérina Chhuon
- Plateforme Protéomique 3P5-Necker; SFR Necker; US24; Université Paris Descartes; Paris France
| | - Michel Vidal
- Université Paris Descartes; Paris France
- CNRS; UMR 8638; Chimie Organique; Médicinale et Extractive et Toxicologie Expérimentale; Paris France
- UF Biologie du médicament et toxicologie; Hôpital Cochin, AP HP; Paris France
| | - Martin Picard
- Université Paris Descartes; Paris France
- CNRS, UMR 8015; Laboratoire de cristallographie et RMN biologiques; Paris France
- CNRS UMR 7099; Laboratoire de Biologie Physico-Chimique des Protéines Membranaires; Institut de Biologie Physico-Chimique (IBPC); Paris France
| | | | - Isabelle Broutin
- Université Paris Descartes; Paris France
- CNRS, UMR 8015; Laboratoire de cristallographie et RMN biologiques; Paris France
| | - Ida-Chiara Guerrera
- Plateforme Protéomique 3P5-Necker; SFR Necker; US24; Université Paris Descartes; Paris France
| | - Jean-Michel Scherrmann
- Inserm; UMR-S 1144; Variabilité de la réponse aux psychotropes; Paris France
- Université Paris Descartes; Paris France
- Université Paris Diderot; Paris France
| | - Yannick Parmentier
- Technologie Servier; Département de recherche biopharmaceutique; Orléans France
| | - Xavier Decleves
- Inserm; UMR-S 1144; Variabilité de la réponse aux psychotropes; Paris France
- Université Paris Descartes; Paris France
- Université Paris Diderot; Paris France
- UF Biologie du médicament et toxicologie; Hôpital Cochin, AP HP; Paris France
| | - Marie-Claude Menet
- Inserm; UMR-S 1144; Variabilité de la réponse aux psychotropes; Paris France
- Université Paris Descartes; Paris France
- Université Paris Diderot; Paris France
- Laboratoire d'hormonologie spécialisé et métabolisme; Hôpital Cochin; AP HP; Paris France
| |
Collapse
|
22
|
TLR4 Deficiency Impairs Oligodendrocyte Formation in the Injured Spinal Cord. J Neurosci 2017; 36:6352-64. [PMID: 27277810 DOI: 10.1523/jneurosci.0353-16.2016] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/05/2016] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED Acute oligodendrocyte (OL) death after traumatic spinal cord injury (SCI) is followed by robust neuron-glial antigen 2 (NG2)-positive OL progenitor proliferation and differentiation into new OLs. Inflammatory mediators are prevalent during both phases and can influence the fate of NG2 cells and OLs. Specifically, toll-like receptor (TLR) 4 signaling induces OL genesis in the naive spinal cord, and lack of TLR4 signaling impairs white matter sparing and functional recovery after SCI. Therefore, we hypothesized that TLR4 signaling may regulate oligodendrogenesis after SCI. C3H/HeJ (TLR4-deficient) and control (C3H/HeOuJ) mice received a moderate midthoracic spinal contusion. TLR4-deficient mice showed worse functional recovery and reduced OL numbers compared with controls at 24 h after injury through chronic time points. Acute OL loss was accompanied by reduced ferritin expression, which is regulated by TLR4 and needed for effective iron storage. TLR4-deficient injured spinal cords also displayed features consistent with reduced OL genesis, including reduced NG2 expression, fewer BrdU-positive OLs, altered BMP4 signaling and inhibitor of differentiation 4 (ID4) expression, and delayed myelin phagocytosis. Expression of several factors, including IGF-1, FGF2, IL-1β, and PDGF-A, was altered in TLR4-deficient injured spinal cords compared with wild types. Together, these data show that TLR4 signaling after SCI is important for OL lineage cell sparing and replacement, as well as in regulating cytokine and growth factor expression. These results highlight new roles for TLR4 in endogenous SCI repair and emphasize that altering the function of a single immune-related receptor can dramatically change the reparative responses of multiple cellular constituents in the injured CNS milieu. SIGNIFICANCE STATEMENT Myelinating cells of the CNS [oligodendrocytes (OLs)] are killed for several weeks after traumatic spinal cord injury (SCI), but they are replaced by resident progenitor cells. How the concurrent inflammatory signaling affects this endogenous reparative response is unclear. Here, we provide evidence that immune receptor toll-like receptor 4 (TLR4) supports OL lineage cell sparing, long-term OL and OL progenitor replacement, and chronic functional recovery. We show that TLR4 signaling is essential for acute iron storage, regulating cytokine and growth factor expression, and efficient myelin debris clearance, all of which influence OL replacement. Importantly, the current study reveals that a single immune receptor is essential for repair responses after SCI, and the potential mechanisms of this beneficial effect likely change over time after injury.
Collapse
|
23
|
Rolih V, Barutello G, Iussich S, De Maria R, Quaglino E, Buracco P, Cavallo F, Riccardo F. CSPG4: a prototype oncoantigen for translational immunotherapy studies. J Transl Med 2017; 15:151. [PMID: 28668095 PMCID: PMC5494135 DOI: 10.1186/s12967-017-1250-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 06/21/2017] [Indexed: 12/21/2022] Open
Abstract
Thanks to striking progress in both the understanding of anti-tumor immune response and the characterization of several tumor associated antigens (TAA), a more rational design and more sophisticated strategies for anti-tumor vaccination have been possible. However, the effectiveness of cancer vaccines in clinical trial is still partial, indicating that additional studies are needed to optimize their design and their pre-clinical testing. Indeed, anti-tumor vaccination success relies on the choice of the best TAA to be targeted and on the translational power of the pre-clinical model used to assess its efficacy. The chondroitin sulfate proteoglycan-4 (CSPG4) is a cell surface proteoglycan overexpressed in a huge range of human and canine neoplastic lesions by tumor cells, tumor microenvironment and cancer initiating cells. CSPG4 plays a central role in the oncogenic pathways required for malignant progression and metastatization. Thanks to these features and to its poor expression in adult healthy tissues, CSPG4 represents an ideal oncoantigen and thus an attractive target for anti-tumor immunotherapy. In this review we explore the potential of CSPG4 immune-targeting. Moreover, since it has been clearly demonstrated that spontaneous canine tumors mimic the progression of human malignancies better than any other pre-clinical model available so far, we reported also our results indicating that CSPG4 DNA vaccination is safe and effective in significantly increasing the survival of canine melanoma patients. Therefore, anti-CSPG4 vaccination strategy could have a substantial impact for the treatment of the wider population of spontaneous CSPG4-positive tumor affected dogs with a priceless translational value and a revolutionary implication for human oncological patients.
Collapse
Affiliation(s)
- Valeria Rolih
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Turin, Italy
| | - Giuseppina Barutello
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Turin, Italy
| | - Selina Iussich
- Department of Veterinary Sciences, University of Torino, 10095 Grugliasco, Italy
| | - Raffaella De Maria
- Department of Veterinary Sciences, University of Torino, 10095 Grugliasco, Italy
| | - Elena Quaglino
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Turin, Italy
| | - Paolo Buracco
- Department of Veterinary Sciences, University of Torino, 10095 Grugliasco, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Turin, Italy
| | - Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Turin, Italy
| |
Collapse
|
24
|
NG2 Proteoglycan Enhances Brain Tumor Progression by Promoting Beta-1 Integrin Activation in both Cis and Trans Orientations. Cancers (Basel) 2017; 9:cancers9040031. [PMID: 28362324 PMCID: PMC5406706 DOI: 10.3390/cancers9040031] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 03/23/2017] [Accepted: 03/29/2017] [Indexed: 12/23/2022] Open
Abstract
By physically interacting with beta-1 integrins, the NG2 proteoglycan enhances activation of the integrin heterodimers. In glioma cells, co-localization of NG2 and 31 integrin in individual cells (cis interaction) can be demonstrated by immunolabeling, and the NG2-integrin interaction can be confirmed by co-immunoprecipitation. NG2-dependent integrin activation is detected via use of conformationally sensitive monoclonal antibodies that reveal the activated state of the beta-1 subunit in NG2-positive versus NG2-negative cells. NG2-dependent activation of beta-1 integrins triggers downstream activation of FAK and PI3K/Akt signaling, resulting in increased glioma cell proliferation, motility, and survival. Similar NG2-dependent cis activation of beta-1 integrins occurs in microvascular pericytes, leading to enhanced proliferation and motility of these vascular cells. Surprisingly, pericyte NG2 is also able to promote beta-1 integrin activation in closely apposed endothelial cells (trans interaction). Enhanced beta-1 signaling in endothelial cells promotes endothelial maturation by inducing the formation of endothelial junctions, resulting in increased barrier function of the endothelium and increased basal lamina assembly. NG2-dependent beta-1 integrin signaling is therefore important for tumor progression by virtue of its affects not only on the tumor cells themselves, but also on the maturation and function of tumor blood vessels.
Collapse
|
25
|
Maus F, Sakry D, Binamé F, Karram K, Rajalingam K, Watts C, Heywood R, Krüger R, Stegmüller J, Werner HB, Nave KA, Krämer-Albers EM, Trotter J. The NG2 Proteoglycan Protects Oligodendrocyte Precursor Cells against Oxidative Stress via Interaction with OMI/HtrA2. PLoS One 2015; 10:e0137311. [PMID: 26340347 PMCID: PMC4560422 DOI: 10.1371/journal.pone.0137311] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 08/14/2015] [Indexed: 02/06/2023] Open
Abstract
The NG2 proteoglycan is characteristically expressed by oligodendrocyte progenitor cells (OPC) and also by aggressive brain tumours highly resistant to chemo- and radiation therapy. Oligodendrocyte-lineage cells are particularly sensitive to stress resulting in cell death in white matter after hypoxic or ischemic insults of premature infants and destruction of OPC in some types of Multiple Sclerosis lesions. Here we show that the NG2 proteoglycan binds OMI/HtrA2, a mitochondrial serine protease which is released from damaged mitochondria into the cytosol in response to stress. In the cytosol, OMI/HtrA2 initiates apoptosis by proteolytic degradation of anti-apoptotic factors. OPC in which NG2 has been downregulated by siRNA, or OPC from the NG2-knockout mouse show an increased sensitivity to oxidative stress evidenced by increased cell death. The proapoptotic protease activity of OMI/HtrA2 in the cytosol can be reduced by the interaction with NG2. Human glioma expressing high levels of NG2 are less sensitive to oxidative stress than those with lower NG2 expression and reducing NG2 expression by siRNA increases cell death in response to oxidative stress. Binding of NG2 to OMI/HtrA2 may thus help protect cells against oxidative stress-induced cell death. This interaction is likely to contribute to the high chemo- and radioresistance of glioma.
Collapse
Affiliation(s)
- Frank Maus
- Department of Biology, Molecular Cell Biology, Johannes Gutenberg University, Mainz, Germany
| | - Dominik Sakry
- Department of Biology, Molecular Cell Biology, Johannes Gutenberg University, Mainz, Germany
| | - Fabien Binamé
- Department of Biology, Molecular Cell Biology, Johannes Gutenberg University, Mainz, Germany
| | - Khalad Karram
- Department of Biology, Molecular Cell Biology, Johannes Gutenberg University, Mainz, Germany
- Institute for Molecular Medicine, University Medical Center of the Johannes-Gutenberg University, Mainz, Germany
| | - Krishnaraj Rajalingam
- Research Center for Immune Therapy, Institute for Immunology, Johannes Gutenberg University of Mainz, Medical Center Mainz, Mainz, Germany
| | - Colin Watts
- Cambridge University, Dept. Clinical Neurosciences, Division of Neurosurgery, Cambridge, United Kingdom
| | - Richard Heywood
- Cambridge University, Dept. Clinical Neurosciences, Division of Neurosurgery, Cambridge, United Kingdom
| | - Rejko Krüger
- Clinical and Experimental Neuroscience, Luxembourg Center for Systems Biomedicine, University of Luxembourg and Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, and German Center for Neurodegenerative Diseases (DZNE), University of Tübingen, Tübingen, Germany
| | - Judith Stegmüller
- Cellular and Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Hauke B. Werner
- Max Planck Institute of Experimental Medicine, Department of Neurogenetics, Göttingen, Germany
| | - Klaus-Armin Nave
- Max Planck Institute of Experimental Medicine, Department of Neurogenetics, Göttingen, Germany
| | - Eva-Maria Krämer-Albers
- Department of Biology, Molecular Cell Biology, Johannes Gutenberg University, Mainz, Germany
| | - Jacqueline Trotter
- Department of Biology, Molecular Cell Biology, Johannes Gutenberg University, Mainz, Germany
- * E-mail:
| |
Collapse
|
26
|
Sakry D, Trotter J. The role of the NG2 proteoglycan in OPC and CNS network function. Brain Res 2015; 1638:161-166. [PMID: 26100334 DOI: 10.1016/j.brainres.2015.06.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 05/27/2015] [Accepted: 06/02/2015] [Indexed: 01/13/2023]
Abstract
In the normal mammalian CNS, the NG2 proteoglycan is expressed by oligodendrocyte precursor cells (OPC) but not by any other neural cell-type. NG2 is a type-1 membrane protein, exerting multiple roles in the CNS including intracellular signaling within the OPC, with effects on migration, cytoskeleton interaction and target gene regulation. It has been recently shown that the extracellular region of NG2, in addition to an adhesive function, acts as a soluble ECM component with the capacity to alter defined neuronal network properties. This region of NG2 is thus endowed with neuromodulatory properties. In order to generate biologically active fragments yielding these properties, the sequential cleavage of the NG2 protein by α- and γ-secretases occurs. The basal level of constitutive cleavage is stimulated by neuronal network activity. This processing leads to 4 major NG2 fragments which all have been associated with distinct biological functions. Here we summarize these functions, focusing on recent discoveries and their implications for the CNS. This article is part of a Special Issue entitled SI:NG2-glia(Invited only).
Collapse
Affiliation(s)
- Dominik Sakry
- Institute of Molecular Cell Biology; Johannes Gutenberg University of Mainz, Mainz, Germany.
| | - Jacqueline Trotter
- Institute of Molecular Cell Biology; Johannes Gutenberg University of Mainz, Mainz, Germany.
| |
Collapse
|
27
|
Sakry D, Yigit H, Dimou L, Trotter J. Oligodendrocyte precursor cells synthesize neuromodulatory factors. PLoS One 2015; 10:e0127222. [PMID: 25966014 PMCID: PMC4429067 DOI: 10.1371/journal.pone.0127222] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 04/13/2015] [Indexed: 12/20/2022] Open
Abstract
NG2 protein-expressing oligodendrocyte progenitor cells (OPC) are a persisting and major glial cell population in the adult mammalian brain. Direct synaptic innervation of OPC by neurons throughout the brain together with their ability to sense neuronal network activity raises the question of additional physiological roles of OPC, supplementary to generating myelinating oligodendrocytes. In this study we investigated whether OPC express neuromodulatory factors, typically synthesized by other CNS cell types. Our results show that OPC express two well-characterized neuromodulatory proteins: Prostaglandin D2 synthase (PTGDS) and neuronal Pentraxin 2 (Nptx2/Narp). Expression levels of the enzyme PTGDS are influenced in cultured OPC by the NG2 intracellular region which can be released by cleavage and localizes to glial nuclei upon transfection. Furthermore PTGDS mRNA levels are reduced in OPC from NG2-KO mouse brain compared to WT cells after isolation by cell sorting and direct analysis. These results show that OPC can contribute to the expression of these proteins within the CNS and suggest PTGDS expression as a downstream target of NG2 signaling.
Collapse
Affiliation(s)
- Dominik Sakry
- Molecular Cell Biology, Department of Biology, Johannes Gutenberg University Mainz, D-55122 Mainz, Germany
- * E-mail: (JT); (DS)
| | - Hatice Yigit
- Molecular Cell Biology, Department of Biology, Johannes Gutenberg University Mainz, D-55122 Mainz, Germany
| | - Leda Dimou
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University Munich, D-80336 Munich, Germany
| | - Jacqueline Trotter
- Molecular Cell Biology, Department of Biology, Johannes Gutenberg University Mainz, D-55122 Mainz, Germany
- * E-mail: (JT); (DS)
| |
Collapse
|
28
|
Nicolosi PA, Dallatomasina A, Perris R. Theranostic impact of NG2/CSPG4 proteoglycan in cancer. Theranostics 2015; 5:530-44. [PMID: 25767619 PMCID: PMC4350014 DOI: 10.7150/thno.10824] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 12/03/2014] [Indexed: 12/27/2022] Open
Abstract
NG2/CSPG4 is an unusual cell-membrane integral proteoglycan widely recognized to be a prognostic factor, a valuable tool for ex vivo and non-invasive molecular diagnostics and, by virtue of its tight association with malignancy, a tantalizing therapeutic target in several tumour types. Although the biology behind its involvement in cancer progression needs to be better understood, implementation of NG2/CSPG4 in the routine clinical practice is attainable and has the potential to contribute to an improved individualized management of cancer patients. In this context, its polymorphic nature seems to be particularly valuable in the effort to standardize informative diagnostic procedures and consolidate forcible immunotherapeutic treatment strategies. We discuss here the underpinnings for this potential and highlight the benefits of taking advantage of the intra-tumour and inter-patient variability in the regulation of NG2/CSPG4 expression. We envision that NG2/CSPG4 may effectively be exploited in therapeutic interventions aimed at averting resistance to target therapy agents and at interfering with secondary lesion formation and/or tumour recurrence.
Collapse
|
29
|
Joo NE, Miao D, Bermúdez M, Stallcup WB, Kapila YL. Shedding of NG2 by MMP-13 attenuates anoikis. DNA Cell Biol 2015; 33:854-62. [PMID: 25166220 DOI: 10.1089/dna.2014.2399] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Disruption of cell-matrix interactions can lead to anoikis-apoptosis due to loss of matrix contacts. We previously showed that Nerve/glial antigen 2 (NG2) is a novel anoikis receptor. Specifically, overexpression of NG2 leads to anoikis propagation, whereas its suppression leads to anoikis attenuation. Interestingly, NG2 expression decreases in late anoikis, suggesting that NG2 reduction is also critical to this process. Thus, we hypothesized that NG2 undergoes cleavage to curtail anoikis propagation. Further, since matrix metalloproteinases (MMPs) cleave cell surface receptors, play a major role in modulating apoptosis, and are associated with death receptor cleavage during apoptosis, we further hypothesized that cleavage of NG2 could be mediated by MMPs to regulate anoikis. Indeed, anoikis conditions triggered release of the NG2 extracellular domain into condition media during late apoptosis, and this coincided with increased MMP-13 expression. Treatment with an MMP-13 inhibitor and MMP-13 siRNA increased anoikis, since these treatments blocked NG2 release. Further, NG2-positive cells exhibited increased anoikis upon MMP-13 inhibition, whereas MMP-13 inhibition did not increase anoikis in NG2-null cells, corroborating that retention of NG2 on the cell membrane is critical for sustaining anoikis, and its cleavage for mediating anoikis attenuation. Similarly, NG2 suppression with siRNA inhibited NG2 release and anoikis. In contrast, MMP-13 overexpression or exogenous MMP-13 reduced anoikis by more effectively shedding NG2. In conclusion, maintenance of NG2 on the cell surface promotes anoikis propagation, whereas its shedding by MMP-13 actions attenuates anoikis. Given that these findings are derived in the context of periodontal ligament fibroblasts, these data have implications for periodontal inflammation and periodontal disease pathogenesis.
Collapse
Affiliation(s)
- Nam E Joo
- 1 Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan , Ann Arbor, Michigan
| | | | | | | | | |
Collapse
|
30
|
Xiang P, Zhu L, Jiang H, He BP. The activation of NG2 expressing cells is downstream to microglial reaction and mediated by the transforming growth factor beta 1. J Neuroimmunol 2015; 279:50-63. [PMID: 25670001 DOI: 10.1016/j.jneuroim.2015.01.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 12/25/2014] [Accepted: 01/14/2015] [Indexed: 11/30/2022]
Abstract
In the present study, we investigated the mechanism of activation of NG2 expressing cells. Application of microglial inhibitors not only attenuated morphological changes but also significantly retarded increase in the number of NG2 expressing cells. Intracerebral injection of TGF-β1 led to a profound activation of NG2 glia as well as an earlier accumulation of NG2(+)-microglia, whilst inhibition of TGF-β1 Smad2/3 signalling pathway eventually attenuated their active responses. We conclude that the activation of NG2 expressing cells is an event downstream to microglial reaction and TGF-β1 secreted from microglia might play an important role in modulation of the function of NG2 expressing cells.
Collapse
Affiliation(s)
- Ping Xiang
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Lie Zhu
- Department of Plastic Surgery, Chang Zheng Hospital, Shanghai, China
| | - Hua Jiang
- Department of Plastic Surgery, Chang Zheng Hospital, Shanghai, China
| | - Bei Ping He
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
31
|
Nishihara T, Remacle AG, Angert M, Shubayev I, Shiryaev SA, Liu H, Dolkas J, Chernov AV, Strongin AY, Shubayev VI. Matrix metalloproteinase-14 both sheds cell surface neuronal glial antigen 2 (NG2) proteoglycan on macrophages and governs the response to peripheral nerve injury. J Biol Chem 2014; 290:3693-707. [PMID: 25488667 DOI: 10.1074/jbc.m114.603431] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Neuronal glial antigen 2 (NG2) is an integral membrane chondroitin sulfate proteoglycan expressed by vascular pericytes, macrophages (NG2-Mφ), and progenitor glia of the nervous system. Herein, we revealed that NG2 shedding and axonal growth, either independently or jointly, depended on the pericellular remodeling events executed by membrane-type 1 matrix metalloproteinase (MT1-MMP/MMP-14). Using purified NG2 ectodomain constructs, individual MMPs, and primary NG2-Mφ cultures, we demonstrated for the first time that MMP-14 performed as an efficient and unconventional NG2 sheddase and that NG2-Mφ infiltrated into the damaged peripheral nervous system. We then characterized the spatiotemporal relationships among MMP-14, MMP-2, and tissue inhibitor of metalloproteinases-2 in sciatic nerve. Tissue inhibitor of metalloproteinases-2-free MMP-14 was observed in the primary Schwann cell cultures using the inhibitory hydroxamate warhead-based MP-3653 fluorescent reporter. In teased nerve fibers, MMP-14 translocated postinjury toward the nodes of Ranvier and its substrates, laminin and NG2. Inhibition of MMP-14 activity using the selective, function-blocking DX2400 human monoclonal antibody increased the levels of regeneration-associated factors, including laminin, growth-associated protein 43, and cAMP-dependent transcription factor 3, thereby promoting sensory axon regeneration after nerve crush. Concomitantly, DX2400 therapy attenuated mechanical hypersensitivity associated with nerve crush in rats. Together, our findings describe a new model in which MMP-14 proteolysis regulates the extracellular milieu and presents a novel therapeutic target in the damaged peripheral nervous system and neuropathic pain.
Collapse
Affiliation(s)
- Tasuku Nishihara
- From the Departments of Anesthesiology, University of California, San Diego, La Jolla, California 92093, Veterans Affairs San Diego Healthcare System, La Jolla, California 92037, Department of Anesthesiology and Resuscitology, Ehime University, Toon, Ehime 791-0295, Japan
| | - Albert G Remacle
- Sanford-Burnham Medical Research Institute, La Jolla, California 92037, and
| | - Mila Angert
- From the Departments of Anesthesiology, University of California, San Diego, La Jolla, California 92093, Veterans Affairs San Diego Healthcare System, La Jolla, California 92037
| | - Igor Shubayev
- Veterans Affairs San Diego Healthcare System, La Jolla, California 92037
| | - Sergey A Shiryaev
- Sanford-Burnham Medical Research Institute, La Jolla, California 92037, and
| | - Huaqing Liu
- From the Departments of Anesthesiology, University of California, San Diego, La Jolla, California 92093, Veterans Affairs San Diego Healthcare System, La Jolla, California 92037
| | - Jennifer Dolkas
- From the Departments of Anesthesiology, University of California, San Diego, La Jolla, California 92093, Veterans Affairs San Diego Healthcare System, La Jolla, California 92037
| | - Andrei V Chernov
- Sanford-Burnham Medical Research Institute, La Jolla, California 92037, and
| | - Alex Y Strongin
- Sanford-Burnham Medical Research Institute, La Jolla, California 92037, and
| | - Veronica I Shubayev
- From the Departments of Anesthesiology, University of California, San Diego, La Jolla, California 92093, Veterans Affairs San Diego Healthcare System, La Jolla, California 92037,
| |
Collapse
|
32
|
Oligodendrocyte precursor cells modulate the neuronal network by activity-dependent ectodomain cleavage of glial NG2. PLoS Biol 2014; 12:e1001993. [PMID: 25387269 PMCID: PMC4227637 DOI: 10.1371/journal.pbio.1001993] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 09/29/2014] [Indexed: 01/09/2023] Open
Abstract
This study shows that the activity of neurons can trigger shedding of a protein, NG2, from the surface of oligodendrocyte precursor cells; this protein in turn modulates synaptic transmission, revealing a two-way conversation between neurons and glia. The role of glia in modulating neuronal network activity is an important question. Oligodendrocyte precursor cells (OPC) characteristically express the transmembrane proteoglycan nerve-glia antigen 2 (NG2) and are unique glial cells receiving synaptic input from neurons. The development of NG2+ OPC into myelinating oligodendrocytes has been well studied, yet the retention of a large population of synapse-bearing OPC in the adult brain poses the question as to additional functional roles of OPC in the neuronal network. Here we report that activity-dependent processing of NG2 by OPC-expressed secretases functionally regulates the neuronal network. NG2 cleavage by the α-secretase ADAM10 yields an ectodomain present in the extracellular matrix and a C-terminal fragment that is subsequently further processed by the γ-secretase to release an intracellular domain. ADAM10-dependent NG2 ectodomain cleavage and release (shedding) in acute brain slices or isolated OPC is increased by distinct activity-increasing stimuli. Lack of NG2 expression in OPC (NG2-knockout mice), or pharmacological inhibition of NG2 ectodomain shedding in wild-type OPC, results in a striking reduction of N-methyl-D-aspartate (NMDA) receptor-dependent long-term potentiation (LTP) in pyramidal neurons of the somatosensory cortex and alterations in the subunit composition of their α-amino-3-hydroxy-5-methyl-4-isoxazolepr opionicacid (AMPA) receptors. In NG2-knockout mice these neurons exhibit diminished AMPA and NMDA receptor-dependent current amplitudes; strikingly AMPA receptor currents can be rescued by application of conserved LNS protein domains of the NG2 ectodomain. Furthermore, NG2-knockout mice exhibit altered behavior in tests measuring sensorimotor function. These results demonstrate for the first time a bidirectional cross-talk between OPC and the surrounding neuronal network and demonstrate a novel physiological role for OPC in regulating information processing at neuronal synapses. Although glial cells substantially outnumber neurons in the mammalian brain, much remains to be discovered regarding their functions. Among glial cells, oligodendrocyte precursors differentiate into oligodendrocytes, whose function is to enwrap nerves with myelin to ensure proper impulse conduction. However, oligodendrocyte precursors also comprise a stable population in all major regions of the adult brain, making up around 5% of the total number of neurons and glia. Synapses are classically formed between neurons. Nonetheless, oligodendrocyte precursors are unique among glial cells in that they receive direct synaptic input from different types of neurons; whether OPC also send signals to neurons is still unknown. Here we show a bidirectional communication between neurons and oligodendrocyte precursors: neuronal activity regulates the cleavage of a glial membrane protein and the release of an extracellular domain that in turn modulates synaptic transmission between neurons. Our data thus show that a particular subtype of glial cells, oligodendrocyte precursors, functionally integrate into the neuronal network and we link this bidirectional signaling to mouse behavior and disease.
Collapse
|
33
|
Chondroitin sulfate proteoglycan CSPG4 as a novel hypoxia-sensitive marker in pancreatic tumors. PLoS One 2014; 9:e100178. [PMID: 24932730 PMCID: PMC4059742 DOI: 10.1371/journal.pone.0100178] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 05/23/2014] [Indexed: 02/06/2023] Open
Abstract
CSPG4 marks pericytes, undifferentiated precursors and tumor cells. We assessed whether the shed ectodomain of CSPG4 (sCSPG4) might circulate and reflect potential changes in CSPG4 tissue expression (pCSPG4) due to desmoplastic and malignant aberrations occurring in pancreatic tumors. Serum sCSPG4 was measured using ELISA in test (n = 83) and validation (n = 221) cohorts comprising donors (n = 11+26) and patients with chronic pancreatitis (n = 11+20) or neoplasms: benign (serous cystadenoma SCA, n = 13+20), premalignant (intraductal dysplastic IPMNs, n = 9+55), and malignant (IPMN-associated invasive carcinomas, n = 4+14; ductal adenocarcinomas, n = 35+86). Pancreatic pCSPG4 expression was evaluated using qRT-PCR (n = 139), western blot analysis and immunohistochemistry. sCSPG4 was found in circulation, but its level was significantly lower in pancreatic patients than in donors. Selective maintenance was observed in advanced IPMNs and PDACs and showed a nodal association while lacking prognostic relevance. Pancreatic pCSPG4 expression was preserved or elevated, whereby neoplastic cells lacked pCSPG4 or tended to overexpress without shedding. Extreme pancreatic overexpression, membranous exposure and tissuehigh/seralow-discordance highlighted stroma-poor benign cystic neoplasm. SCA is known to display hypoxic markers and coincide with von-Hippel-Lindau and Peutz-Jeghers syndromes, in which pVHL and LBK1 mutations affect hypoxic signaling pathways. In vitro testing confined pCSPG4 overexpression to normal mesenchymal but not epithelial cells, and a third of tested carcinoma cell lines; however, only the latter showed pCSPG4-responsiveness to chronic hypoxia. siRNA-based knockdowns failed to reduce the malignant potential of either normoxic or hypoxic cells. Thus, overexpression of the newly established conditional hypoxic indicator, CSPG4, is apparently non-pathogenic in pancreatic malignancies but might mark distinct epithelial lineage and contribute to cell polarity disorders. Surficial retention on tumor cells renders CSPG4 an attractive therapeutic target. Systemic ‘drop and restoration’ alterations accompanying IPMN and PDAC progression indicate that the interference of pancreatic diseases with local and remote shedding/release of sCSPG4 into circulation deserves broad diagnostic exploration.
Collapse
|
34
|
Sardone F, Traina F, Tagliavini F, Pellegrini C, Merlini L, Squarzoni S, Santi S, Neri S, Faldini C, Maraldi N, Sabatelli P. Effect of mechanical strain on the collagen VI pericellular matrix in anterior cruciate ligament fibroblasts. J Cell Physiol 2014; 229:878-86. [PMID: 24356950 DOI: 10.1002/jcp.24518] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 11/19/2013] [Indexed: 12/15/2022]
Abstract
Cell-extracellular matrix interaction plays a major role in maintaining the structural integrity of connective tissues and sensing changes in the biomechanical environment of cells. Collagen VI is a widely expressed non-fibrillar collagen, which regulates tissues homeostasis. The objective of the present investigation was to extend our understanding of the role of collagen VI in human ACL. This study shows that collagen VI is associated both in vivo and in vitro to the cell membrane of knee ACL fibroblasts, contributing to the constitution of a microfibrillar pericellular matrix. In cultured cells the localization of collagen VI at the cell surface correlated with the expression of NG2 proteoglycan, a major collagen VI receptor. The treatment of ACL fibroblasts with anti-NG2 antibody abolished the localization of collagen VI indicating that collagen VI pericellular matrix organization in ACL fibroblasts is mainly mediated by NG2 proteoglycan. In vitro mechanical strain injury dramatically reduced the NG2 proteoglycan protein level, impaired the association of collagen VI to the cell surface, and promoted cell cycle withdrawal. Our data suggest that the injury-induced alteration of specific cell-ECM interactions may lead to a defective fibroblast self-renewal and contribute to the poor regenerative ability of ACL fibroblasts.
Collapse
Affiliation(s)
- Francesca Sardone
- National Research Council of Italy, Institute of Molecular Genetics, Bologna, Italy; IOR-IRCCS, SC Laboratory of Musculoskeletal Cell Biology, Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Liu Y, Lieberwirth C, Jia X, Curtis JT, Meredith M, Wang ZX. Chemosensory cues affect amygdaloid neurogenesis and alter behaviors in the socially monogamous prairie vole. Eur J Neurosci 2014; 39:1632-41. [PMID: 24641515 DOI: 10.1111/ejn.12531] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 01/24/2014] [Accepted: 01/28/2014] [Indexed: 12/11/2022]
Abstract
The current study examined the effects of pheromonal exposure on adult neurogenesis and revealed the role of the olfactory pathways on adult neurogenesis and behavior in the socially monogamous prairie vole (Microtus ochrogaster). Subjects were injected with a cell proliferation marker [5-bromo-2'-deoxyuridine (BrdU)] and then exposed to their own soiled bedding or bedding soiled by a same- or opposite-sex conspecific. Exposure to opposite-sex bedding increased BrdU labeling in the amygdala (AMY), but not the dentate gyrus (DG), of female, but not male, voles, indicating a sex-, stimulus-, and brain region-specific effect. The removal of the main olfactory bulbs or lesioning of the vomeronasal organ (VNOX) in females reduced BrdU labeling in the AMY and DG, and inhibited the male bedding-induced BrdU labeling in the AMY, revealing the importance of an intact olfactory pathway for amygdaloid neurogenesis. VNOX increased anxiety-like behavior and altered social preference, but it did not affect social recognition memory in female voles. VNOX also reduced the percentage of BrdU-labeled cells that co-expressed the neuronal marker TuJ1 in the AMY, but not the DG. Together, our data indicate the importance of the olfactory pathway in mediating brain plasticity in the limbic system as well as its role in behavior.
Collapse
Affiliation(s)
- Y Liu
- Department of Psychology, Florida State University, 1107 W. Call Street, Tallahassee, FL, 32306, USA; Program in Neuroscience, Florida State University, 1107 W. Call Street, Tallahassee, FL, 32306, USA
| | | | | | | | | | | |
Collapse
|
36
|
Binamé F. Transduction of extracellular cues into cell polarity: the role of the transmembrane proteoglycan NG2. Mol Neurobiol 2014; 50:482-93. [PMID: 24390567 DOI: 10.1007/s12035-013-8610-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 12/08/2013] [Indexed: 01/23/2023]
Abstract
Resident progenitor cells expressing nerve/glial antigen 2 (NG2) such as oligodendrocyte precursor cells (OPC) and pericytes persist in the adult brain. The transmembrane proteoglycan NG2 regulates migration of both these cell types in response to growth factors or specific components of the extracellular matrix. This role of NG2 is linked to the control of cell polarity. The polarization of OPC toward an acute lesion in the brain is impaired in NG2-deficient mice, supporting this concept. A review of the signaling pathways impinged on by NG2 reveals key proteins of cell polarity: phosphatidylinositol 3-kinase, focal adhesion kinase, Rho GTPases, and polarity complex proteins. In the scope of cell migration, I discuss here how the interplay of NG2 with signaling transmitted by extracellular cues can control the establishment of cell polarity, and I propose a model to integrate the apparent opposite effects of NG2 on cellular dynamics.
Collapse
Affiliation(s)
- Fabien Binamé
- Molecular Cell Biology, Department of Biology, Johannes Gutenberg University of Mainz, Mainz, Germany,
| |
Collapse
|
37
|
NG2 regulates directional migration of oligodendrocyte precursor cells via Rho GTPases and polarity complex proteins. J Neurosci 2013; 33:10858-74. [PMID: 23804106 DOI: 10.1523/jneurosci.5010-12.2013] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The transmembrane proteoglycan NG2 is expressed by oligodendrocyte precursor cells (OPC), which migrate to axons during developmental myelination and remyelinate in the adult after migration to injured sites. Highly invasive glial tumors also express NG2. Despite the fact that NG2 has been implicated in control of OPC migration, its mode of action remains unknown. Here, we show in vitro and in vivo that NG2 controls migration of OPC through the regulation of cell polarity. In stab wounds in adult mice we show that NG2 controls orientation of OPC toward the wound. NG2 stimulates RhoA activity at the cell periphery via the MUPP1/Syx1 signaling pathway, which favors the bipolar shape of migrating OPC and thus directional migration. Upon phosphorylation of Thr-2256, downstream signaling of NG2 switches from RhoA to Rac stimulation. This triggers process outgrowth through regulators of front-rear polarity and we show using a phospho-mimetic form of NG2 that indeed NG2 recruits proteins of the CRB and the PAR polarity complexes to stimulate Rac activity via the GEF Tiam1. Our findings demonstrate that NG2 is a core organizer of Rho GTPase activity and localization in the cell, which controls OPC polarity and directional migration. This work also reveals CRB and PAR polarity complexes as new effectors of NG2 signaling in the establishment of front-rear polarity.
Collapse
|
38
|
Lord MS, Whitelock JM. Recombinant production of proteoglycans and their bioactive domains. FEBS J 2013; 280:2490-510. [DOI: 10.1111/febs.12197] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 02/04/2013] [Accepted: 02/15/2013] [Indexed: 12/11/2022]
Affiliation(s)
- Megan S. Lord
- Graduate School of Biomedical Engineering; The University of New South Wales; Sydney; NSW; Australia
| | - John M. Whitelock
- Graduate School of Biomedical Engineering; The University of New South Wales; Sydney; NSW; Australia
| |
Collapse
|
39
|
Moransard M, Dann A, Staszewski O, Fontana A, Prinz M, Suter T. NG2 expressed by macrophages and oligodendrocyte precursor cells is dispensable in experimental autoimmune encephalomyelitis. ACTA ACUST UNITED AC 2011; 134:1315-30. [PMID: 21596769 DOI: 10.1093/brain/awr070] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Increased expression of the chondroitin proteoglycan NG2 is a prominent feature in central nervous system injury with unknown cellular source and biological relevance. Here, we describe the first detailed analysis of experimental autoimmune encephalomyelitis in NG2 knockout mice and NG2 knockout bone marrow chimeras. We show that both macrophages and oligodendrocyte progenitor cells express and secrete NG2 in response to transforming growth factor-β. A subpopulation of macrophages expresses NG2 within leucocyte infiltrates in the central nervous system, but only oligodendrocyte progenitor cells contribute to NG2 accumulation. Notably, NG2 plays no role in experimental autoimmune encephalomyelitis initiation, progression or recuperation. In concurrence, the immune response is unaltered in NG2-deficient mice as are the extent of central nervous system damage and degree of remyelination.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Antigens/genetics
- Antigens/metabolism
- Antigens, CD/metabolism
- Cell Proliferation
- Cells, Cultured
- Disease Models, Animal
- Disease Progression
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Flow Cytometry
- Glial Fibrillary Acidic Protein/metabolism
- Macrophages/metabolism
- Macrophages/ultrastructure
- Mice
- Mice, Inbred C57BL
- Microscopy, Electron, Transmission
- Neurons/metabolism
- Neurons/pathology
- Oligodendroglia/metabolism
- Oligodendroglia/ultrastructure
- Proteoglycans/genetics
- Proteoglycans/metabolism
- RNA, Messenger/metabolism
- Spinal Cord/pathology
- Stem Cells/metabolism
- Transforming Growth Factor beta/metabolism
- Transforming Growth Factor beta/pharmacology
- Up-Regulation/drug effects
- Up-Regulation/physiology
Collapse
Affiliation(s)
- Martijn Moransard
- Clinical Immunology, University Hospital Zurich, Häldeliweg 4, CH-8044 Zürich, Switzerland.
| | | | | | | | | | | |
Collapse
|
40
|
Abstract
The lack of effective conventional therapies for the treatment of advanced stage melanoma has stimulated interest in the development of novel strategies for the management of patients with malignant melanoma. Among them, immunotherapy has attracted much attention because of the potential role played by immunological events in the clinical course of melanoma. For many years, T cell-based immunotherapy has been emphasized in part because of the disappointing results of the monoclonal antibody (mAb)-based clinical trials conducted in the early 1980s and in part because of the postulated major role played by T cells in tumor growth control. More recently, mAb-based therapies have gained in popularity given their clinical and commercial success for a variety of malignant diseases. As a result, there has been increased interest in identifying and characterizing antibody-defined melanoma antigens. Among them, the chondroitin sulfate proteoglycan 4 (CSPG4), also known as high molecular weight-melanoma associated antigen (HMW-MAA) or melanoma chondroitin sulfate proteoglycan (MCSP), has attracted much attention in recent years because of the growing experimental evidence that it fulfills two requirements for immunotherapy to be therapeutically effective: (1) targeting of cancer stem cells (CSC) and (2) development of combinatorial therapies to counteract the escape mechanisms driven by the genetic instability of tumor cells. With this in mind, in this chapter, we have reviewed recent information related to the distribution of CSPG4 on various types of tumors, including CSC, its expression on pericytes in the tumor microenvironment, its recognition by T cells, its role in cell biology as well as the potential mechanisms underlying the ability of CSPG4-specific immunity to control malignant cell growth.
Collapse
|
41
|
Hill RA, Natsume R, Sakimura K, Nishiyama A. NG2 cells are uniformly distributed and NG2 is not required for barrel formation in the somatosensory cortex. Mol Cell Neurosci 2011; 46:689-98. [PMID: 21292011 DOI: 10.1016/j.mcn.2011.01.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Revised: 01/14/2011] [Accepted: 01/24/2011] [Indexed: 11/20/2022] Open
Abstract
The somatosensory barrel cortex in the rodent forms during the first postnatal week setting up a periphery related map with each whisker represented as a bundle of thalamocortical axons (TCAs) in layer IV. The centers of each barrel (hollows) contain the densely packed TCAs, while the areas between each barrel (septa) form a boundary between each barrel. NG2 chondroitin sulfate proteoglycan (CSPG) expressing cells (NG2 cells, polydendrocytes) make up a unique population of glial cells that receive synaptic like input and form close contacts with growing axons. In the present study we investigated the developmental distribution of NG2 cells in the barrel cortex to determine if they display preferential septa distribution similar to other extracellular and cell surface CSPGs. Immunohistochemistry for NG2 and platelet-derived growth factor receptor alpha (PDGFRα) in NG2DsRedBAC transgenic mice showed uniform distribution of NG2 cells and processes in barrel hollows and septa at postnatal (P) days 5, 6, 7, 8, 14, and 30. Changes in the barrel pattern formation caused by cauterization of one row of whiskers at P1 resulted in corresponding changes in extracellular and cell surface CSPG distribution at P7 but no detectable changes in NG2 cell bodies and processes. Furthermore, no abnormalities in barrel formation or reorganization were detected in NG2 knockout mice. These observations suggest that NG2 cells are unlikely to play an inhibitory boundary role on TCA growth and that NG2 expression is not necessary for normal barrel formation.
Collapse
Affiliation(s)
- Robert A Hill
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269, USA
| | | | | | | |
Collapse
|
42
|
Horiuchi M, Lindsten T, Pleasure D, Itoh T. Differing in vitro survival dependency of mouse and rat NG2+ oligodendroglial progenitor cells. J Neurosci Res 2010; 88:957-70. [PMID: 19908280 PMCID: PMC2872551 DOI: 10.1002/jnr.22262] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
NG2 chondroitin sulfate proteoglycan is a surface marker of oligodendroglial progenitor cells (OPCs) in various species. In contrast to well-studied rat OPCs, however, we found that purified mouse NG2 surface positive cells (NG2(+) cells) require additional activation of cyclic AMP (cAMP) signaling for survival in a medium containing 30% B104 neuroblastoma conditioned medium supplemented with fibroblast growth factor-2 (B104CM+FGF2), whereas B104CM+FGF2 alone is sufficient for survival and selective proliferation of rat OPCs. After induction of in vitro differentiation, more than 90% of mouse NG2(+) cells became O4-positive, and a majority expressed myelin basic protein by 5 day of differentiation, which confirmed the identity of isolated mouse NG2(+) cells as OPCs. In comparison to rat OPCs, mouse OPCs in B104CM+FGF2 were less motile, and demonstrated lower basal phosphorylation levels of ERK1/2 and cAMP response element-binding protein (CREB) and a higher incidence of apoptosis mediated by the intrinsic pathway. Transient up-regulation of cAMP-CREB signaling partially inhibited apoptosis of mouse OPCs independently of the ERK pathway. This study demonstrates a difference in trophic requirements between mouse and rat OPCs, with an essential role for cAMP signaling to preserve viability of mouse OPCs.
Collapse
Affiliation(s)
- Makoto Horiuchi
- Department of Neurology, University of California, Davis, School of Medicine, Sacramento, California 95817
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California and University California, Davis, School of Medicine, Sacramento, California 95817
| | - Tullia Lindsten
- Department of Pathology and Laboratory Medicine, and Abramson Family Cancer Research Institute, The University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - David Pleasure
- Department of Neurology, University of California, Davis, School of Medicine, Sacramento, California 95817
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California and University California, Davis, School of Medicine, Sacramento, California 95817
| | - Takayuki Itoh
- Department of Neurology, University of California, Davis, School of Medicine, Sacramento, California 95817
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California and University California, Davis, School of Medicine, Sacramento, California 95817
| |
Collapse
|
43
|
Nishiyama A, Komitova M, Suzuki R, Zhu X. Polydendrocytes (NG2 cells): multifunctional cells with lineage plasticity. Nat Rev Neurosci 2009; 10:9-22. [DOI: 10.1038/nrn2495] [Citation(s) in RCA: 647] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
44
|
Karram K, Goebbels S, Schwab M, Jennissen K, Seifert G, Steinhäuser C, Nave KA, Trotter J. NG2-expressing cells in the nervous system revealed by the NG2-EYFP-knockin mouse. Genesis 2008; 46:743-57. [DOI: 10.1002/dvg.20440] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
45
|
Stallcup WB, Huang FJ. A role for the NG2 proteoglycan in glioma progression. Cell Adh Migr 2008; 2:192-201. [PMID: 19262111 PMCID: PMC2634088 DOI: 10.4161/cam.2.3.6279] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2008] [Accepted: 05/13/2008] [Indexed: 01/18/2023] Open
Abstract
Many human gliomas carry markers characteristic of oligodendrocyte progenitor cells (such as Olig-2, PDGF alpha receptor and NG2 proteoglycan), suggesting these progenitors as the cells of origin for glioma initiation. This review considers the potential roles of the NG2 proteoglycan in glioma progression. NG2 is expressed not only by glioma cells and by oligodendrocyte progenitors, but also by pericytes associated with the tumor microvasculature. The proteoglycan may therefore promote tumor vascularization and recruitment of normal progenitors to the tumor mass, in addition to mediating expansion of the transformed cell population. Along with potentiating growth factor signaling and serving as a cell surface receptor for extracellular matrix components, NG2 also has the ability to mediate activation of beta-1 integrins. These molecular interactions allow the proteoglycan to contribute to critical processes such as cell proliferation, cell motility and cell survival.
Collapse
Affiliation(s)
- William B Stallcup
- Burnham Institute for Medical Research, Cancer Research Center, La Jolla, California 92037, USA.
| | | |
Collapse
|
46
|
Xiong J, Wang Y, Zhu Z, Liu J, Wang Y, Zhang C, Hammes HP, Lang F, Feng Y. NG2 proteoglycan increases mesangial cell proliferation and extracellular matrix production. Biochem Biophys Res Commun 2007; 361:960-7. [PMID: 17686464 DOI: 10.1016/j.bbrc.2007.07.113] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2007] [Accepted: 07/20/2007] [Indexed: 12/12/2022]
Abstract
As a membrane-spanning protein, NG2 chondroitin sulfate proteoglycan interacts with molecules on both sides of plasma membrane. The present study explored the role of NG2 in the pathogenesis of diabetic nephropathy. In the normal kidneys, NG2 was observed predominantly in glomerular mesangium, Bowman's capsule and interstitial vessels. Both mRNA and protein expression in kidneys was significantly higher in strepozotocin-induced diabetic rats than that in normal rats. In the cultured rat mesangial cell line HBZY-1, overexpression of NG2 promoted mesangial cell proliferation and extracellular matrix (ECM) production, such as type VI collagen and laminin. Furthermore, target knockdown of NG2 resulted in decreased cell proliferation and ECM formation. The observations suggest that NG2 is up-regulated in diabetic nephropathy. It actively participates in the development and progression of glomerulosclerosis by stimulating proliferation of mesangial cells and deposition of ECM.
Collapse
Affiliation(s)
- Jing Xiong
- Department of Nephrology, Union Hospital, Tongji Medical College, Huangzhong University of Science & Technology, Wuhan, China
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Massey JM, Amps J, Viapiano MS, Matthews RT, Wagoner MR, Whitaker CM, Alilain W, Yonkof AL, Khalyfa A, Cooper NGF, Silver J, Onifer SM. Increased chondroitin sulfate proteoglycan expression in denervated brainstem targets following spinal cord injury creates a barrier to axonal regeneration overcome by chondroitinase ABC and neurotrophin-3. Exp Neurol 2007; 209:426-45. [PMID: 17540369 PMCID: PMC2270474 DOI: 10.1016/j.expneurol.2007.03.029] [Citation(s) in RCA: 138] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2007] [Revised: 03/24/2007] [Accepted: 03/27/2007] [Indexed: 01/11/2023]
Abstract
Increased chondroitin sulfate proteoglycan (CSPG) expression in the vicinity of a spinal cord injury (SCI) is a primary participant in axonal regeneration failure. However, the presence of similar increases of CSPG expression in denervated synaptic targets well away from the primary lesion and the subsequent impact on regenerating axons attempting to approach deafferented neurons have not been studied. Constitutively expressed CSPGs within the extracellular matrix and perineuronal nets of the adult rat dorsal column nuclei (DCN) were characterized using real-time PCR, Western blot analysis and immunohistochemistry. We show for the first time that by 2 days and through 3 weeks following SCI, the levels of NG2, neurocan and brevican associated with reactive glia throughout the DCN were dramatically increased throughout the DCN despite being well beyond areas of trauma-induced blood brain barrier breakdown. Importantly, regenerating axons from adult sensory neurons microtransplanted 2 weeks following SCI between the injury site and the DCN were able to regenerate rapidly within white matter (as shown previously by Davies et al. [Davies, S.J., Goucher, D.R., Doller, C., Silver, J., 1999. Robust regeneration of adult sensory axons in degenerating white matter of the adult rat spinal cord. J. Neurosci. 19, 5810-5822]) but were unable to enter the denervated DCN. Application of chondroitinase ABC or neurotrophin-3-expressing lentivirus in the DCN partially overcame this inhibition. When the treatments were combined, entrance by regenerating axons into the DCN was significantly augmented. These results demonstrate both an additional challenge and potential treatment strategy for successful functional pathway reconstruction after SCI.
Collapse
Affiliation(s)
- James M. Massey
- M.D./Ph.D. Program, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
- Department of Anatomical Sciences & Neurobiology, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
- Department of Neurological Surgery, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
- Department of Kentucky Spinal Cord Injury Research Center, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
| | - Jeremy Amps
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, 44106
| | - Mariano S. Viapiano
- Department of Neurobiology, Yale University School of Medicine, New Haven, Connecticut, 06520
| | - Russell. T. Matthews
- Department of Neurobiology, Yale University School of Medicine, New Haven, Connecticut, 06520
| | - Michelle R. Wagoner
- Department of Neurological Surgery, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
- Department of Kentucky Spinal Cord Injury Research Center, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
| | - Christopher M. Whitaker
- Department of Anatomical Sciences & Neurobiology, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
- Department of Neurological Surgery, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
- Department of Kentucky Spinal Cord Injury Research Center, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
| | - Warren Alilain
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, 44106
| | - Alicia L. Yonkof
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, 44106
| | - Abdelnaby Khalyfa
- Department of Anatomical Sciences & Neurobiology, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
| | - Nigel G. F. Cooper
- Department of Anatomical Sciences & Neurobiology, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
| | - Jerry Silver
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, 44106
- Address for correspondence, proofs, and reprint requests: Stephen M. Onifer, Ph.D., Spinal Cord and Brain Injury Research Center, University of Kentucky, BBSRB B365, 741 South Limestone Street, Lexington, KY, 40536-0509, U.S.A., TELEPHONE: (859) 323-5226, FAX: (859) 257-5737, EMAIL:
| | - Stephen M. Onifer
- Department of Anatomical Sciences & Neurobiology, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
- Department of Neurological Surgery, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
- Department of Kentucky Spinal Cord Injury Research Center, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
| |
Collapse
|
48
|
Virgintino D, Girolamo F, Errede M, Capobianco C, Robertson D, Stallcup WB, Perris R, Roncali L. An intimate interplay between precocious, migrating pericytes and endothelial cells governs human fetal brain angiogenesis. Angiogenesis 2007; 10:35-45. [PMID: 17225955 DOI: 10.1007/s10456-006-9061-x] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2006] [Accepted: 12/11/2006] [Indexed: 10/23/2022]
Abstract
In order to better understand the process of angiogenesis in the developing human brain, we have examined the spatial relationship and relative contributions of endothelial cells and pericytes, the two primary cell types involved in vessel growth, together with their relation with the vascular basement membrane. Pericytes were immunolocalized through use of the specific markers nerve/glial antigen 2 (NG2) proteoglycan, endosialin (CD248) and the platelet-derived growth factor receptor beta (PDGFR-beta), while endothelial cells were identified by the pan-endothelial marker CD31 and the blood brain barrier (BBB)-specific markers claudin-5 and glucose transporter isoform 1 (GLUT-1). The quantitative analysis demonstrates that microvessels of the fetal human telencephalon are characterized by a continuous layer of activated/angiogenic NG2 pericytes, which tightly invest endothelial cells and participate in the earliest stages of vessel growth. Immunolabelling with anti-active matrix metalloproteinase-2 (aMMP-2) and anti-collagen type IV antibodies revealed that aMMP-2 producing endothelial cells and pericytes are both associated with the vascular basement membrane during vessel sprouting. Detailed localization of the two vascular cell types during angiogenesis suggests that growing microvessels of the human telencephalon are formed by a pericyte-driven angiogenic process in which the endothelial cells are preceded and guided by migrating pericytes during organization of the growing vessel wall.
Collapse
Affiliation(s)
- Daniela Virgintino
- Department of Human Anatomy and Histology, University of Bari School of Medicine, Piazza Giulio Cesare, 11, Bari 70124, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
The fibrous scar that develops after central nervous system (CNS) injury is considered a major impediment for axonal regeneration. It consists of a dense collagen IV meshwork, which serves as a binding matrix for numerous other extracellular matrix components and inhibitory molecules like proteoglycans and semaphorins, but also growth-promoting factors. Inhibition of collagen matrix formation in brain and spinal cord lesions leads to axonal regeneration and functional recovery, although collagen IV per se is not inhibitory for axonal outgrowth. This review focuses on the molecular properties of the collagen IV matrix and its interactions with various molecules that are expressed after CNS lesion. Moreover, studies on collagen expression and matrix formation after injury of regenerating versus non-regenerating nervous systems are reviewed. Major differences in collagen deposition in the CNS and the peripheral nervous system (PNS) and differences in specific cell responses to extracellular matrix deposition in the lesion area are discussed. Therapeutic treatments aiming at suppression of fibrous scarring have been shown to promote axon regeneration in various lesion paradigms of the mammalian CNS.
Collapse
Affiliation(s)
- Nicole Klapka
- Molecular Neurobiology Laboratory, Department of Neurology, Heinrich-Heine University, Düsseldorf, Germany
| | | |
Collapse
|
50
|
Abstract
NG2 is a high-molecular-weight chondroitin sulphate proteoglycan found on the surfaces of oligodendrocyte precursor cells (OPCs). Here we review the history and biology of OPCs with an emphasis on their functions after experimentally induced CNS injury. Injury to brain or spinal cord results in the rapid accumulation of NG2-expressing OPCs in the glial scar that forms at the injury site. The glial scar is considered a biochemical and physical barrier to successful axon regeneration and the functional properties of NG2 suggest that it, along with other macromolecules, participates in the creation of this growth-inhibitory environment. NG2 is an important target for therapies designed to promote successful axon regrowth.
Collapse
Affiliation(s)
- Andrew M Tan
- Department of Neurobiology and Behavior, State University of New York, Stony Brook, NY 11794, USA
| | | | | |
Collapse
|