1
|
Critcher M, Pang JM, Huang ML. Mapping the FGF2 Interactome Identifies a Functional Proteoglycan Coreceptor. ACS Chem Biol 2025; 20:105-116. [PMID: 39704408 DOI: 10.1021/acschembio.4c00475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Fibroblast growth factor 2 (FGF2) is a multipotent growth factor and signaling protein that exhibits broad functions across multiple cell types. These functions are often initiated by binding to growth factor receptors and fine-tuned by glycosaminoglycan (GAG)-modified proteins called proteoglycans. The various outputs of FGF2 signaling and functions arise from a dynamic and cell type-specific set of binding partners. However, the interactome of FGF2 has yet to be comprehensively determined. Moreover, the identity of the proteoglycan proteins carrying GAG chains is often overlooked and remains unknown in most cell contexts. Here, we perform peroxidase-catalyzed live cell proximity labeling using an engineered APEX2-FGF2 fusion protein to map the interactome of FGF2. Across two cell lines with established and distinct FGF2-driven functions, we greatly expand upon the known FGF2 interactome, identifying >600 new putative FGF2 interactors. Notably, our results demonstrate a key role for the GAG binding capacity of FGF2 in modulating its interactome.
Collapse
Affiliation(s)
- Meg Critcher
- Department of Chemistry, Scripps Research, 10550 N Torrey Pines Rd, La Jolla, California 92037, United States
- Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, 10550 N Torrey Pines Rd, La Jolla California 92037, United States
| | - Jia Meng Pang
- Department of Chemistry, Scripps Research, 10550 N Torrey Pines Rd, La Jolla, California 92037, United States
- Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, 10550 N Torrey Pines Rd, La Jolla California 92037, United States
| | - Mia L Huang
- Department of Chemistry, Scripps Research, 10550 N Torrey Pines Rd, La Jolla, California 92037, United States
- Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, 10550 N Torrey Pines Rd, La Jolla California 92037, United States
| |
Collapse
|
2
|
Li M, Wang Y, Lin X, Yang H, Zhang X, Bai Y, Li X, Zhang L, Cheng F, Cao C, Zhou Q. Evaluation of antitumor potential of an anti-glypican-1 monoclonal antibody in preclinical lung cancer models reveals a distinct mechanism of action. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:600-626. [PMID: 38966167 PMCID: PMC11220310 DOI: 10.37349/etat.2024.00238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/26/2024] [Indexed: 07/06/2024] Open
Abstract
Aim The main objective of this study was to investigate the antitumor effect of a mouse anti-human glypican-1 (GPC1) monoclonal antibody (mAb) on non-small cell lung carcinoma (NSCLC) and associated molecular mechanisms. Methods The anti-proliferative and anti-migratory activities of anti-GPC1 mAb were examined in A549 and H460 NSCLC cells and LL97A lung fibroblasts. The inhibitory effect of anti-GPC1 mAb on tumor growth was evaluated in an orthotopic lung tumor model. Results The in vitro study showed that anti-GPC1 mAb profoundly inhibited the anchorage-independent growth of A549 and H460 NSCLC cells and exhibited relatively high cytotoxic activities towards LL97A lung fibroblasts, A549/LL97A and H460/LL97A coculture spheroids. Moreover, anti-GPC1 mAb significantly decreased the expression of phospho-Src (p-Src; Tyr416), p-Akt (Ser473) and β-catenin in the co-cultured LL97A lung fibroblasts, and the expression of phospho-mitogen-activated protein kinase kinase (p-MEK; Ser217/221) and phospho-90 kDa ribosomal s6 kinase (p-p90RSK; Ser380) in co-cultured A549 cells. When anti-GPC1 mAb was administered to tumor-bearing mice, the inhibitory effect of anti-GPC1 mAb on the orthotopic lung tumor growth was not statistically significant. Nonetheless, results of Western blot analysis showed significant decrease in the phosphorylation of fibroblast growth factor receptor 1 (FGFR1) at Tyr766, Src at Tyr416, extracellular signal-regulated kinase (ERK) at Thr202/Tyr204, 90 kDa ribosomal S6 kinase (RSK) at Ser380, glycogen synthase kinases 3α (GSK3α) at Ser21 and GSK3β at Ser9 in tumor tissues. These data implicate that anti-GPC1 mAb treatment impairs the interaction between tumor cells and tumor associated fibroblasts by attenuating the paracrine FGFR signal transduction. Conclusions The relatively potent cytotoxicity of anti-GPC1 mAb in lung fibroblasts and its potential inhibitory effect on the paracrine FGFR signal transduction warrant further studies on the combined use of this mAb with targeted therapeutics to improve therapeutic outcomes in lung cancer.
Collapse
Affiliation(s)
- Minghua Li
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Yanhong Wang
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Xiaoyang Lin
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Haiqiang Yang
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Xiaolin Zhang
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Yun Bai
- MegaNano Biotech Inc., Tampa, FL 33612, USA
| | - Xiankun Li
- Zhengzhou Molecular Diagnosis Engineering Technology Research Center, Zhengzhou 450001, Henan Province, China
| | - Lulu Zhang
- Zhengzhou Molecular Diagnosis Engineering Technology Research Center, Zhengzhou 450001, Henan Province, China
| | - Feng Cheng
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Chuanhai Cao
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Qingyu Zhou
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
3
|
Loffredo LF, Surpur A, Ringham OR, Li F, de Los Santos-Alexis K, Arpaia N. Heparan sulfate regulates amphiregulin signaling towards reparative lung mesenchymal cells during influenza A infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.25.591175. [PMID: 38712053 PMCID: PMC11071614 DOI: 10.1101/2024.04.25.591175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Amphiregulin (Areg), a growth factor produced by regulatory T (Treg) cells to facilitate tissue repair/regeneration, contains a heparan sulfate (HS) binding domain. How HS, a highly sulfated glycan subtype that alters growth factor signaling, influences Areg repair/regeneration functions is unclear. Here we report that inhibition of HS in various cell lines and primary lung mesenchymal cells (LMC) qualitatively alters downstream signaling and highlights the existence of HS-dependent vs. -independent Areg transcriptional signatures. Utilizing a panel of cell lines with targeted deletions in HS synthesis-related genes, we found that the presence of the glypican family of heparan sulfate proteoglycans is critical for Areg signaling and confirmed this dependency in primary LMC by siRNA-mediated knockdown. Furthermore, in the context of influenza A (IAV) infection in vivo , we found that an Areg-responsive subset of reparative LMC upregulate glypican-4 and HS. Conditional deletion of HS primarily within this LMC subset resulted in reduced blood oxygen saturation following infection with IAV, with no changes in viral load. Finally, we found that co-culture of HS-knockout LMC with IAV-induced Treg cells results in reduced LMC responses. Collectively, this study reveals the essentiality of HS on a specific lung mesenchymal population as a mediator of Treg cell-derived Areg reparative signaling during IAV infection.
Collapse
|
4
|
Cengiz Winter N, Karakaya M, Mosen P, Brusius I, Anlar B, Haliloglu G, Winter D, Wirth B. Proteomic Investigation of Differential Interactomes of Glypican 1 and a Putative Disease-Modifying Variant of Ataxia. J Proteome Res 2023; 22:3081-3095. [PMID: 37585105 PMCID: PMC10476613 DOI: 10.1021/acs.jproteome.3c00402] [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: 07/05/2023] [Indexed: 08/17/2023]
Abstract
In a currently 13-year-old girl of consanguineous Turkish parents, who developed unsteady gait and polyneuropathy at the ages of 3 and 6 years, respectively, we performed whole genome sequencing and identified a biallelic missense variant c.424C>T, p.R142W in glypican 1 (GPC1) as a putative disease-associated variant. Up to date, GPC1 has not been associated with a neuromuscular disorder, and we hypothesized that this variant, predicted as deleterious, may be causative for the disease. Using mass spectrometry-based proteomics, we investigated the interactome of GPC1 WT and the missense variant. We identified 198 proteins interacting with GPC1, of which 16 were altered for the missense variant. This included CANX as well as vacuolar ATPase (V-ATPase) and the mammalian target of rapamycin complex 1 (mTORC1) complex members, whose dysregulation could have a potential impact on disease severity in the patient. Importantly, these proteins are novel interaction partners of GPC1. At 10.5 years, the patient developed dilated cardiomyopathy and kyphoscoliosis, and Friedreich's ataxia (FRDA) was suspected. Given the unusually severe phenotype in a patient with FRDA carrying only 104 biallelic GAA repeat expansions in FXN, we currently speculate that disturbed GPC1 function may have exacerbated the disease phenotype. LC-MS/MS data are accessible in the ProteomeXchange Consortium (PXD040023).
Collapse
Affiliation(s)
- Nur Cengiz Winter
- Institute
of Human Genetics, University Hospital Cologne, 50931 Cologne, Germany
- Center
for Molecular Medicine Cologne, University
of Cologne, 50931 Cologne, Germany
| | - Mert Karakaya
- Institute
of Human Genetics, University Hospital Cologne, 50931 Cologne, Germany
- Center
for Molecular Medicine Cologne, University
of Cologne, 50931 Cologne, Germany
- Center
for Rare Diseases Cologne, University Hospital
of Cologne, 50931 Cologne, Germany
| | - Peter Mosen
- Institute
for Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, 53115 Bonn, Germany
| | - Isabell Brusius
- Institute
of Human Genetics, University Hospital Cologne, 50931 Cologne, Germany
| | - Banu Anlar
- Department
of Pediatrics, Division of Pediatric Neurology, Hacettepe University Faculty of Medicine, 06230 Ankara, Turkey
| | - Goknur Haliloglu
- Department
of Pediatrics, Division of Pediatric Neurology, Hacettepe University Faculty of Medicine, 06230 Ankara, Turkey
| | - Dominic Winter
- Institute
for Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, 53115 Bonn, Germany
| | - Brunhilde Wirth
- Institute
of Human Genetics, University Hospital Cologne, 50931 Cologne, Germany
- Center
for Molecular Medicine Cologne, University
of Cologne, 50931 Cologne, Germany
- Center
for Rare Diseases Cologne, University Hospital
of Cologne, 50931 Cologne, Germany
| |
Collapse
|
5
|
Colin-Pierre C, El Baraka O, Danoux L, Bardey V, André V, Ramont L, Brézillon S. Regulation of stem cell fate by HSPGs: implication in hair follicle cycling. NPJ Regen Med 2022; 7:77. [PMID: 36577752 PMCID: PMC9797564 DOI: 10.1038/s41536-022-00267-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 11/30/2022] [Indexed: 12/29/2022] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) are part of proteoglycan family. They are composed of heparan sulfate (HS)-type glycosaminoglycan (GAG) chains covalently linked to a core protein. By interacting with growth factors and/or receptors, they regulate numerous pathways including Wnt, hedgehog (Hh), bone morphogenic protein (BMP) and fibroblast growth factor (FGF) pathways. They act as inhibitor or activator of these pathways to modulate embryonic and adult stem cell fate during organ morphogenesis, regeneration and homeostasis. This review summarizes the knowledge on HSPG structure and classification and explores several signaling pathways regulated by HSPGs in stem cell fate. A specific focus on hair follicle stem cell fate and the possibility to target HSPGs in order to tackle hair loss are discussed in more dermatological and cosmeceutical perspectives.
Collapse
Affiliation(s)
- Charlie Colin-Pierre
- Université de Reims Champagne-Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France.
- CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire-MEDyC, Reims, France.
- BASF Beauty Care Solutions France SAS, Pulnoy, France.
| | | | - Louis Danoux
- BASF Beauty Care Solutions France SAS, Pulnoy, France
| | | | - Valérie André
- BASF Beauty Care Solutions France SAS, Pulnoy, France
| | - Laurent Ramont
- Université de Reims Champagne-Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France
- CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire-MEDyC, Reims, France
- CHU de Reims, Service Biochimie-Pharmacologie-Toxicologie, Reims, France
| | - Stéphane Brézillon
- Université de Reims Champagne-Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France
- CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire-MEDyC, Reims, France
| |
Collapse
|
6
|
Schüler SC, Liu Y, Dumontier S, Grandbois M, Le Moal E, Cornelison DDW, Bentzinger CF. Extracellular matrix: Brick and mortar in the skeletal muscle stem cell niche. Front Cell Dev Biol 2022; 10:1056523. [PMID: 36523505 PMCID: PMC9745096 DOI: 10.3389/fcell.2022.1056523] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/03/2022] [Indexed: 11/30/2022] Open
Abstract
The extracellular matrix (ECM) is an interconnected macromolecular scaffold occupying the space between cells. Amongst other functions, the ECM provides structural support to tissues and serves as a microenvironmental niche that conveys regulatory signals to cells. Cell-matrix adhesions, which link the ECM to the cytoskeleton, are dynamic multi-protein complexes containing surface receptors and intracellular effectors that control various downstream pathways. In skeletal muscle, the most abundant tissue of the body, each individual muscle fiber and its associated muscle stem cells (MuSCs) are surrounded by a layer of ECM referred to as the basal lamina. The core scaffold of the basal lamina consists of self-assembling polymeric laminins and a network of collagens that tether proteoglycans, which provide lateral crosslinking, establish collateral associations with cell surface receptors, and serve as a sink and reservoir for growth factors. Skeletal muscle also contains the fibrillar collagenous interstitial ECM that plays an important role in determining tissue elasticity, connects the basal laminae to each other, and contains matrix secreting mesenchymal fibroblast-like cell types and blood vessels. During skeletal muscle regeneration fibroblast-like cell populations expand and contribute to the transitional fibronectin-rich regenerative matrix that instructs angiogenesis and MuSC function. Here, we provide a comprehensive overview of the role of the skeletal muscle ECM in health and disease and outline its role in orchestrating tissue regeneration and MuSC function.
Collapse
Affiliation(s)
- Svenja C. Schüler
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Yuguo Liu
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Simon Dumontier
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Michel Grandbois
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Emmeran Le Moal
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - DDW Cornelison
- Division of Biological Sciences Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
| | - C. Florian Bentzinger
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
7
|
Sparn C, Dimou E, Meyer A, Saleppico R, Wegehingel S, Gerstner M, Klaus S, Ewers H, Nickel W. Glypican-1 drives unconventional secretion of Fibroblast Growth Factor 2. eLife 2022; 11:75545. [PMID: 35348113 PMCID: PMC8986318 DOI: 10.7554/elife.75545] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 03/24/2022] [Indexed: 11/13/2022] Open
Abstract
Fibroblast Growth Factor 2 (FGF2) is a tumor cell survival factor that is transported into the extracellular space by an unconventional secretory mechanism. Cell surface heparan sulfate proteoglycans are known to play an essential role in this process. Unexpectedly, we found that among the diverse sub-classes consisting of syndecans, perlecans, glypicans and others, Glypican-1 (GPC1) is the principle and rate-limiting factor that drives unconventional secretion of FGF2. By contrast, we demonstrate GPC1 to be dispensable for FGF2 signaling into cells. We provide first insights into the structural basis for GPC1-dependent FGF2 secretion, identifying disaccharides with N-linked sulfate groups to be enriched in the heparan sulfate chains of GPC1 to which FGF2 binds with high affinity. Our findings have broad implications for the role of GPC1 as a key molecule in tumor progression.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Helge Ewers
- Institut für Chemie und Biochemie, Freie Universität Berlin
| | | |
Collapse
|
8
|
Inhibition of glypican-1 expression induces an activated fibroblast phenotype in a human bone marrow-derived stromal cell-line. Sci Rep 2021; 11:9262. [PMID: 33927256 PMCID: PMC8084937 DOI: 10.1038/s41598-021-88519-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 04/13/2021] [Indexed: 11/25/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) are the most abundant stromal cell type in the tumor microenvironment. CAFs orchestrate tumor-stromal interactions, and contribute to cancer cell growth, metastasis, extracellular matrix (ECM) remodeling, angiogenesis, immunomodulation, and chemoresistance. However, CAFs have not been successfully targeted for the treatment of cancer. The current study elucidates the significance of glypican-1 (GPC-1), a heparan sulfate proteoglycan, in regulating the activation of human bone marrow-derived stromal cells (BSCs) of fibroblast lineage (HS-5). GPC-1 inhibition changed HS-5 cellular and nuclear morphology, and increased cell migration and contractility. GPC-1 inhibition also increased pro-inflammatory signaling and CAF marker expression. GPC-1 induced an activated fibroblast phenotype when HS-5 cells were exposed to prostate cancer cell conditioned media (CCM). Further, treatment of human bone-derived prostate cancer cells (PC-3) with CCM from HS-5 cells exhibiting GPC-1 loss increased prostate cancer cell aggressiveness. Finally, GPC-1 was expressed in mouse tibia bone cells and present during bone loss induced by mouse prostate cancer cells in a murine prostate cancer bone model. These data demonstrate that GPC-1 partially regulates the intrinsic and extrinsic phenotype of human BSCs and transformation into activated fibroblasts, identify novel functions of GPC-1, and suggest that GPC-1 expression in BSCs exerts inhibitory paracrine effects on the prostate cancer cells. This supports the hypothesis that GPC-1 may be a novel pharmacological target for developing anti-CAF therapeutics to control cancer.
Collapse
|
9
|
Salinas-Marín R, Villanueva-Cabello TM, Martínez-Duncker I. Biology of Proteoglycans and Associated Glycosaminoglycans. COMPREHENSIVE GLYCOSCIENCE 2021:63-102. [DOI: 10.1016/b978-0-12-819475-1.00065-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
10
|
Ravikumar M, Smith RAA, Nurcombe V, Cool SM. Heparan Sulfate Proteoglycans: Key Mediators of Stem Cell Function. Front Cell Dev Biol 2020; 8:581213. [PMID: 33330458 PMCID: PMC7710810 DOI: 10.3389/fcell.2020.581213] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/29/2020] [Indexed: 12/11/2022] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) are an evolutionarily ancient subclass of glycoproteins with exquisite structural complexity. They are ubiquitously expressed across tissues and have been found to exert a multitude of effects on cell behavior and the surrounding microenvironment. Evidence has shown that heterogeneity in HSPG composition is crucial to its functions as an essential scaffolding component in the extracellular matrix as well as a vital cell surface signaling co-receptor. Here, we provide an overview of the significance of HSPGs as essential regulators of stem cell function. We discuss the various roles of HSPGs in distinct stem cell types during key physiological events, from development through to tissue homeostasis and regeneration. The contribution of aberrant HSPG production to altered stem cell properties and dysregulated cellular homeostasis characteristic of cancer is also reviewed. Finally, we consider approaches to better understand and exploit the multifaceted functions of HSPGs in influencing stem cell characteristics for cell therapy and associated culture expansion strategies.
Collapse
Affiliation(s)
- Maanasa Ravikumar
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Raymond Alexander Alfred Smith
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Victor Nurcombe
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University-Imperial College London, Singapore, Singapore
| | - Simon M Cool
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
11
|
Benard E, Casey NP, Inderberg EM, Wälchli S. SJI 2020 special issue: A catalogue of Ovarian Cancer targets for CAR therapy. Scand J Immunol 2020; 92:e12917. [PMID: 32557659 DOI: 10.1111/sji.12917] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/09/2020] [Accepted: 06/09/2020] [Indexed: 12/14/2022]
Abstract
Ovarian Cancer (OC) is currently difficult to cure, mainly due to its late detection and the advanced state of the disease at the time of diagnosis. Therefore, conventional treatments such as debulking surgery and combination chemotherapy are rarely able to control progression of the tumour, and relapses are frequent. Alternative therapies are currently being evaluated, including immunotherapy and advanced T cell-based therapy. In the present review, we will focus on a description of those Chimeric Antigen Receptors (CARs) that have been validated in the laboratory or are being tested in the clinic. Numerous target antigens have been defined due to the identification of OC biomarkers, and many are being used as CAR targets. We provide an exhaustive list of these constructs and their current status. Despite being innovative and efficient, the OC-specific CARs face a barrier to their clinical efficacy: the tumour microenvironment (TME). Indeed, effector cells expressing CARs have been shown to be severely inhibited, rendering the CAR T cells useless once at the tumour site. Herein, we give a thorough description of the highly immunosuppressive OC TME and present recent studies and innovations that have enabled CAR T cells to counteract this negative environment and to destroy tumours.
Collapse
Affiliation(s)
- Emmanuelle Benard
- Translational Research Unit, Section for Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Nicholas P Casey
- Translational Research Unit, Section for Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Else Marit Inderberg
- Translational Research Unit, Section for Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Sébastien Wälchli
- Translational Research Unit, Section for Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
12
|
Heparan Sulfate Proteoglycans Biosynthesis and Post Synthesis Mechanisms Combine Few Enzymes and Few Core Proteins to Generate Extensive Structural and Functional Diversity. Molecules 2020; 25:molecules25184215. [PMID: 32937952 PMCID: PMC7570499 DOI: 10.3390/molecules25184215] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 02/06/2023] Open
Abstract
Glycosylation is a common and widespread post-translational modification that affects a large majority of proteins. Of these, a small minority, about 20, are specifically modified by the addition of heparan sulfate, a linear polysaccharide from the glycosaminoglycan family. The resulting molecules, heparan sulfate proteoglycans, nevertheless play a fundamental role in most biological functions by interacting with a myriad of proteins. This large functional repertoire stems from the ubiquitous presence of these molecules within the tissue and a tremendous structural variety of the heparan sulfate chains, generated through both biosynthesis and post synthesis mechanisms. The present review focusses on how proteoglycans are “gagosylated” and acquire structural complexity through the concerted action of Golgi-localized biosynthesis enzymes and extracellular modifying enzymes. It examines, in particular, the possibility that these enzymes form complexes of different modes of organization, leading to the synthesis of various oligosaccharide sequences.
Collapse
|
13
|
EDMD-Causing Emerin Mutant Myogenic Progenitors Exhibit Impaired Differentiation Using Similar Mechanisms. Cells 2020; 9:cells9061463. [PMID: 32549231 PMCID: PMC7349064 DOI: 10.3390/cells9061463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/05/2020] [Accepted: 06/09/2020] [Indexed: 11/17/2022] Open
Abstract
Mutations in the gene encoding emerin (EMD) cause Emery–Dreifuss muscular dystrophy (EDMD1), an inherited disorder characterized by progressive skeletal muscle wasting, irregular heart rhythms and contractures of major tendons. The skeletal muscle defects seen in EDMD are caused by failure of muscle stem cells to differentiate and regenerate the damaged muscle. However, the underlying mechanisms remain poorly understood. Most EDMD1 patients harbor nonsense mutations and have no detectable emerin protein. There are three EDMD-causing emerin mutants (S54F, Q133H, and Δ95–99) that localize correctly to the nuclear envelope and are expressed at wildtype levels. We hypothesized these emerin mutants would share in the disruption of key molecular pathways involved in myogenic differentiation. We generated myogenic progenitors expressing wildtype emerin and each EDMD1-causing emerin mutation (S54F, Q133H, Δ95–99) in an emerin-null (EMD−/y) background. S54F, Q133H, and Δ95–99 failed to rescue EMD−/y myogenic differentiation, while wildtype emerin efficiently rescued differentiation. RNA sequencing was done to identify pathways and networks important for emerin regulation of myogenic differentiation. This analysis significantly reduced the number of pathways implicated in EDMD1 muscle pathogenesis.
Collapse
|
14
|
Li J, Jia L, Hao Z, Xu Y, Shen J, Ma C, Wu J, Zhao T, Zhi Y, Li P, Li J, Zhu B, Sun S. Site-Specific N-Glycoproteomic Analysis Reveals Upregulated Sialylation and Core Fucosylation during Transient Regeneration Loss in Neonatal Mouse Hearts. J Proteome Res 2020; 19:3191-3200. [PMID: 32425043 DOI: 10.1021/acs.jproteome.0c00172] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Myocardial infarction (MI) is one of the leading causes of deaths worldwide. Because of the incapability of regeneration, the cardiomyocyte loss with MI is replaced by fibrotic scar tissue, which eventually leads to heart failure. Reconstructing regeneration of an adult human heart has been recognized as a promising strategy for cardiac therapeutics. A neonatal mouse heart, which possesses transient regenerative capacity at the first week after birth, represents an ideal model to investigate processes associated with cardiac regeneration. In this work, an integrated glycoproteomic and proteomic analysis was performed to investigate the differences in glycoprotein abundances and site-specific glycosylation between postneonatal day 1 (P1) and day 7 (P7) of mouse hearts. By large-scale profiling and quantifying more than 2900 intact N-glycopeptides in neonatal mouse hearts, we identified 227 altered N-glycopeptides between P1 and P7 hearts. By extracting protein changes from the global proteome data, the normalized glycosylation changes for site-specific glycans were obtained, which showed heterogeneity on glycosites and glycoproteins. Systematic analysis of the glycosylation changes demonstrated an overall upregulation of sialylation and core fucosylation in P7 mice. Notably, the upregulated sialylation was a comprehensive result of increased sialylated glycans with Neu5Gc, with both Neu5Gc and core fucose, and decreased sialylated glycans with Neu5Ac. The upregulated core fucosylation resulted from the increase of glycans containing both core fucose and Neu5Gc but not glycans containing sole core fucose. These data provide a valuable resource for future functional and mechanism studies on heart regeneration and discovery of novel therapeutic targets. All mass spectrometry proteomic data have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the data set identifier PXD017139.
Collapse
Affiliation(s)
- Jun Li
- College of Life Science, Northwest University, Xi'an, Shaanxi province 710069, China
| | - Li Jia
- College of Life Science, Northwest University, Xi'an, Shaanxi province 710069, China
| | - Zhifang Hao
- College of Life Science, Northwest University, Xi'an, Shaanxi province 710069, China
| | - Yintai Xu
- College of Life Science, Northwest University, Xi'an, Shaanxi province 710069, China
| | - Jiechen Shen
- College of Life Science, Northwest University, Xi'an, Shaanxi province 710069, China
| | - Chen Ma
- College of Life Science, Northwest University, Xi'an, Shaanxi province 710069, China
| | - Jingyu Wu
- College of Life Science, Northwest University, Xi'an, Shaanxi province 710069, China
| | - Ting Zhao
- College of Life Science, Northwest University, Xi'an, Shaanxi province 710069, China
| | - Yuan Zhi
- College of Life Science, Northwest University, Xi'an, Shaanxi province 710069, China
| | - Pengfei Li
- College of Life Science, Northwest University, Xi'an, Shaanxi province 710069, China
| | - Jing Li
- College of Life Science, Northwest University, Xi'an, Shaanxi province 710069, China
| | - Bojing Zhu
- College of Life Science, Northwest University, Xi'an, Shaanxi province 710069, China
| | - Shisheng Sun
- College of Life Science, Northwest University, Xi'an, Shaanxi province 710069, China
| |
Collapse
|
15
|
Csapo R, Gumpenberger M, Wessner B. Skeletal Muscle Extracellular Matrix - What Do We Know About Its Composition, Regulation, and Physiological Roles? A Narrative Review. Front Physiol 2020; 11:253. [PMID: 32265741 PMCID: PMC7096581 DOI: 10.3389/fphys.2020.00253] [Citation(s) in RCA: 225] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/05/2020] [Indexed: 12/20/2022] Open
Abstract
Skeletal muscle represents the largest body-composition component in humans. In addition to its primary function in the maintenance of upright posture and the production of movement, it also plays important roles in many other physiological processes, including thermogenesis, metabolism and the secretion of peptides for communication with other tissues. Research attempting to unveil these processes has traditionally focused on muscle fibers, i.e., the contractile muscle cells. However, it is a frequently overlooked fact that muscle fibers reside in a three-dimensional scaffolding that consists of various collagens, glycoproteins, proteoglycans, and elastin, and is commonly referred to as extracellular matrix (ECM). While initially believed to be relatively inert, current research reveals the involvement of ECM cells in numerous important physiological processes. In interaction with other cells, such as fibroblasts or cells of the immune system, the ECM regulates muscle development, growth and repair and is essential for effective muscle contraction and force transmission. Since muscle ECM is highly malleable, its texture and, consequently, physiological roles may be affected by physical training and disuse, aging or various diseases, such as diabetes. With the aim to stimulate increased efforts to study this still poorly understood tissue, this narrative review summarizes the current body of knowledge on (i) the composition and structure of the ECM, (ii) molecular pathways involved in ECM remodeling, (iii) the physiological roles of muscle ECM, (iv) dysregulations of ECM with aging and disease as well as (v) the adaptations of muscle ECM to training and disuse.
Collapse
Affiliation(s)
- Robert Csapo
- Research Unit for Orthopaedic Sports Medicine and Injury Prevention, Institute for Sports Medicine, Alpine Medicine & Health Tourism, UMIT - Private University for Health Sciences, Medical Informatics and Technology, Hall, Austria
| | - Matthias Gumpenberger
- Research Unit for Orthopaedic Sports Medicine and Injury Prevention, Institute for Sports Medicine, Alpine Medicine & Health Tourism, UMIT - Private University for Health Sciences, Medical Informatics and Technology, Hall, Austria
| | - Barbara Wessner
- Department of Sports Medicine, Exercise Physiology and Prevention, Centre for Sport Science and University Sports, University of Vienna, Vienna, Austria
| |
Collapse
|
16
|
Listik E, Toma L. Glypican-1 in human glioblastoma: implications in tumorigenesis and chemotherapy. Oncotarget 2020; 11:828-845. [PMID: 32180897 PMCID: PMC7061737 DOI: 10.18632/oncotarget.27492] [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: 10/23/2019] [Accepted: 01/29/2020] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma is one of the most common malignant brain tumors, with which patients have a mean survival of 24 months. Glypican-1 has been previously shown to be overexpressed in human glioblastoma and to be negatively correlated with patient’s survival. This study aimed to investigate how glypican-1 influences the tumoral profile of human glioblastoma using in vitro cell line models. By downregulating the expression of glypican-1 in U-251 MG cells, we observed that the cellular growth and proliferation were highly reduced, in which cells were significantly shifted towards G0 as opposed to G1 phases. Cellular migration was severely affected, and glypican-1 majorly impacted the affinity towards laminin-binding of glioblastoma U-251 MG cells. This proteoglycan was highly prevalent in glioblastoma cells, being primarily localized in the cellular membrane and extracellular vesicles, occasionally with glypican-3. Glypican-1 could also be found in cell-cell junctions with syndecan-4 but was not identified in lipid rafts in this study. Glypican-1-silenced cells were much more susceptible to temozolomide than in U-251 MG itself. Therefore, we present evidence not only to support facts that glypican-1 is an elementary macromolecule in glioblastoma tumoral microenvironment but also to introduce this proteoglycan as a promising therapeutic target for this lethal tumor.
Collapse
Affiliation(s)
- Eduardo Listik
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Leny Toma
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
17
|
Carmody LC, Blau H, Danis D, Zhang XA, Gourdine JP, Vasilevsky N, Krawitz P, Thompson MD, Robinson PN. Significantly different clinical phenotypes associated with mutations in synthesis and transamidase+remodeling glycosylphosphatidylinositol (GPI)-anchor biosynthesis genes. Orphanet J Rare Dis 2020; 15:40. [PMID: 32019583 PMCID: PMC7001271 DOI: 10.1186/s13023-020-1313-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 01/21/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Defects in the glycosylphosphatidylinositol (GPI) biosynthesis pathway can result in a group of congenital disorders of glycosylation known as the inherited GPI deficiencies (IGDs). To date, defects in 22 of the 29 genes in the GPI biosynthesis pathway have been identified in IGDs. The early phase of the biosynthetic pathway assembles the GPI anchor (Synthesis stage) and the late phase transfers the GPI anchor to a nascent peptide in the endoplasmic reticulum (ER) (Transamidase stage), stabilizes the anchor in the ER membrane using fatty acid remodeling and then traffics the GPI-anchored protein to the cell surface (Remodeling stage). RESULTS We addressed the hypothesis that disease-associated variants in either the Synthesis stage or Transamidase+Remodeling-stage GPI pathway genes have distinct phenotypic spectra. We reviewed clinical data from 58 publications describing 152 individual patients and encoded the phenotypic information using the Human Phenotype Ontology (HPO). We showed statistically significant differences between the Synthesis and Transamidase+Remodeling Groups in the frequencies of phenotypes in the musculoskeletal system, cleft palate, nose phenotypes, and cognitive disability. Finally, we hypothesized that phenotypic defects in the IGDs are likely to be at least partially related to defective GPI anchoring of their target proteins. Twenty-two of one hundred forty-two proteins that receive a GPI anchor are associated with one or more Mendelian diseases and 12 show some phenotypic overlap with the IGDs, represented by 34 HPO terms. Interestingly, GPC3 and GPC6, members of the glypican family of heparan sulfate proteoglycans bound to the plasma membrane through a covalent GPI linkage, are associated with 25 of these phenotypic abnormalities. CONCLUSIONS IGDs associated with Synthesis and Transamidase+Remodeling stages of the GPI biosynthesis pathway have significantly different phenotypic spectra. GPC2 and GPC6 genes may represent a GPI target of general disruption to the GPI biosynthesis pathway that contributes to the phenotypes of some IGDs.
Collapse
Affiliation(s)
- Leigh C Carmody
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032, USA
| | - Hannah Blau
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032, USA
| | - Daniel Danis
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032, USA
| | - Xingman A Zhang
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032, USA
| | | | | | - Peter Krawitz
- Institute of Genomic Statistics and Bioinformatics, University of Bonn, Bonn, Germany
| | - Miles D Thompson
- Department of Pediatrics, UCSD School of Medicine, La Jolla, CA, 92093, USA
| | - Peter N Robinson
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032, USA.
- Institute for Systems Genomics, University of Connecticut, Farmington, CT, USA.
| |
Collapse
|
18
|
Velleman SG, Clark DL, Tonniges JR. The effect of nutrient restriction and syndecan-4 or glypican-1 knockdown on the differentiation of turkey pectoralis major satellite cells differing in age and growth selection. Poult Sci 2020; 98:6078-6090. [PMID: 31180126 DOI: 10.3382/ps/pez304] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 05/16/2019] [Indexed: 11/20/2022] Open
Abstract
Skeletal muscle growth is mediated by the proliferation and differentiation of satellite cells, whose activity is affected by both nutrition and the expression of syndecan-4 and glypican-1. Previous research has not addressed if there is an interactive effect of nutrition with the expression of syndecan-4 and glypican-1. Thus, the objective of the current study was to determine if the response of satellite cells to nutrient restriction was altered by syndecan-4 or glypican-1 knockdown and if age and growth selection are factors. Satellite cells were isolated from pectoralis major muscle of 1-day, 7-wk, and 16-wk-old turkeys selected for increased 16-wk body weight (F line) and the randombred control (RBC2) line from which the F line was selected. Syndecan-4 or glypican-1 expression was knocked down in both lines using small interfering RNAs along with nutrient restriction of 0 or 20% of the standard cell culture medium either applied during proliferation with subsequent normal differentiation medium (RN) or during differentiation with preceding normal proliferation medium (NR). For both lines, nutrient restriction and syndecan-4 or glypican-1 knockdown had an independent and additive effect on satellite cell differentiation at 72 h of differentiation except for 1 d satellite cells. The 1 d satellite cell differentiation was increased by RN treatment, but when combined with syndecan-4 or glypican-1 knockdown, the increase in differentiation was negated. At 48 h of differentiation, syndecan-4 knockdown in 7 and 16 wk satellite cells and glypican-1 knockdown in 7 wk cells cancelled the effect of the RN treatment, but enhanced the effect of NR treatment at 24 h of differentiation. Growth selection had little effect on the interaction between nutrient restriction and syndecan-4 or glypican-1 knockdown. Taken together, these data demonstrate that the satellite cell response to nutrition is dependent on the expression of syndecan-4 and glypican-1 in an age-dependent manner with growth selection having little impact.
Collapse
Affiliation(s)
- Sandra G Velleman
- The Ohio State University/Ohio Agricultural Research and Development Center, Wooster, OH 44691, USA
| | - Daniel L Clark
- The Ohio State University/Ohio Agricultural Research and Development Center, Wooster, OH 44691, USA
| | - Jeffrey R Tonniges
- The Ohio State University/Ohio Agricultural Research and Development Center, Wooster, OH 44691, USA
| |
Collapse
|
19
|
Xu B, Zhang M, Perlingeiro RCR, Shen W. Skeletal Muscle Constructs Engineered from Human Embryonic Stem Cell Derived Myogenic Progenitors Exhibit Enhanced Contractile Forces When Differentiated in a Medium Containing EGM‐2 Supplements. ACTA ACUST UNITED AC 2019; 3:e1900005. [DOI: 10.1002/adbi.201900005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 10/08/2019] [Indexed: 12/22/2022]
Affiliation(s)
- Bin Xu
- Department of Biomedical Engineering University of Minnesota Minneapolis MN 55455 USA
| | - Mengen Zhang
- Department of Biomedical Engineering University of Minnesota Minneapolis MN 55455 USA
| | - Rita C. R. Perlingeiro
- Department of Medicine University of Minnesota Minneapolis MN 55455 USA
- Stem Cell Institute and Institute for Engineering in Medicine University of Minnesota Minneapolis Minnesota 55455 USA
| | - Wei Shen
- Department of Biomedical Engineering University of Minnesota Minneapolis MN 55455 USA
- Stem Cell Institute and Institute for Engineering in Medicine University of Minnesota Minneapolis Minnesota 55455 USA
| |
Collapse
|
20
|
Quach ND, Kaur SP, Eggert MW, Ingram L, Ghosh D, Sheth S, Nagy T, Dawson MR, Arnold RD, Cummings BS. Paradoxical Role of Glypican-1 in Prostate Cancer Cell and Tumor Growth. Sci Rep 2019; 9:11478. [PMID: 31391540 PMCID: PMC6685992 DOI: 10.1038/s41598-019-47874-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 07/25/2019] [Indexed: 02/07/2023] Open
Abstract
Recent studies suggest that glypican-1 (GPC-1) is a biomarker for prostate cancer, but there are few studies elucidating the role of GPC-1 in prostate cancer progression. We observed high expression of GPC-1 in more aggressive prostate cancer cell lines such as PC-3 and DU-145. While inhibition of GPC-1 expression in PC-3 cells decreased cell growth and migration in vitro, it surprisingly increased cell proliferation and migration in DU-145 cells, suggesting that the role of GPC-1 is cell type-dependent. Further, GPC-1 inhibition increased PC-3 tumor size in NCr nude mice xenografts. We hypothesized that the discrepancy between the in vitro and in vivo data is mediated by stromal cells in the tumor microenvironment. Thus, we tested the effect of tumor conditioned media (TCM) on gene expression in human mesenchymal stem cells and fibroblasts. Treatment of stromal cells with TCM from PC-3 cells transfected with GPC-1 shRNA increased the expression of migration markers, endocrine/paracrine biomolecules, and extracellular matrix components. Additionally, the decreased cell growth in GPC-1 knockdown PC-3 cells was rescued by coculturing with stromal cells. These data demonstrate the paradoxical role that GPC-1 plays in prostate cancer cell growth by interacting with stromal cells and through ECM remodeling and endocrine/paracrine signaling.
Collapse
Affiliation(s)
- Nhat D Quach
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, USA.,Department of Molecular Pharmacology, Physiology, & Biotechnology, Brown University, Providence, RI, USA
| | - Sukhneeraj Pal Kaur
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, USA
| | - Matthew W Eggert
- Department of Drug Discovery & Development, Auburn University, Auburn, AL, USA
| | - Lishann Ingram
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, USA
| | - Deepraj Ghosh
- Department of Molecular Pharmacology, Physiology, & Biotechnology, Brown University, Providence, RI, USA
| | - Sheela Sheth
- Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Tamas Nagy
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Michelle R Dawson
- Department of Molecular Pharmacology, Physiology, & Biotechnology, Brown University, Providence, RI, USA.,Center for Biomedical Engineering, Brown University, Providence, RI, USA.,School of Engineering, Brown University, Providence, RI, USA
| | - Robert D Arnold
- Department of Drug Discovery & Development, Auburn University, Auburn, AL, USA.,Interdisciplinary Toxicology Program, University of Georgia, Athens, GA, USA
| | - Brian S Cummings
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, USA. .,Interdisciplinary Toxicology Program, University of Georgia, Athens, GA, USA.
| |
Collapse
|
21
|
Role of glypicans in regulation of the tumor microenvironment and cancer progression. Biochem Pharmacol 2019; 168:108-118. [PMID: 31251939 DOI: 10.1016/j.bcp.2019.06.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 06/20/2019] [Indexed: 12/28/2022]
Abstract
Glypicans are evolutionary conserved, cell surface heparan sulfate (HS) proteoglycans that are attached to the cell membrane via a glycosylphosphatidylinositol (GPI) anchor. Glypicans interact with a broad class of soluble and insoluble ligands, such as morphogens, growth factors, chemokines, receptors and components of the extracellular matrix (ECM). Such versatility comes from their ability to interact through both their HS chains and core protein. Glypicans are involved in cellular and tissue development, morphogenesis and cell motility. They exhibit differential expression in several cancers, acting as both tumor promoters and inhibitors in a cancer type-specific manner. They also influence tumor stroma by facilitating angiogenesis, ECM remodeling and alteration of immune cell functions. Glypicans have emerged as a new therapeutic moiety, whose functions can be exploited in the field of targeted therapies and precision medicine in cancer. This is demonstrated by the emergence of several anti-glypican antibody-based immunologics that have been recently developed and are being evaluated in clinical trials. This review will focus on glypican structure and function with an emphasis on their expression in various cancers. Discussion will also center on the potential of glypicans to be therapeutic targets for inhibition of cancer cell growth.
Collapse
|
22
|
Cross-Talk between Fibroblast Growth Factor Receptors and Other Cell Surface Proteins. Cells 2019; 8:cells8050455. [PMID: 31091809 PMCID: PMC6562592 DOI: 10.3390/cells8050455] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 05/08/2019] [Accepted: 05/13/2019] [Indexed: 12/14/2022] Open
Abstract
Fibroblast growth factors (FGFs) and their receptors (FGFRs) constitute signaling circuits that transmit signals across the plasma membrane, regulating pivotal cellular processes like differentiation, migration, proliferation, and apoptosis. The malfunction of FGFs/FGFRs signaling axis is observed in numerous developmental and metabolic disorders, and in various tumors. The large diversity of FGFs/FGFRs functions is attributed to a great complexity in the regulation of FGFs/FGFRs-dependent signaling cascades. The function of FGFRs is modulated at several levels, including gene expression, alternative splicing, posttranslational modifications, and protein trafficking. One of the emerging ways to adjust FGFRs activity is through formation of complexes with other integral proteins of the cell membrane. These proteins may act as coreceptors, modulating binding of FGFs to FGFRs and defining specificity of elicited cellular response. FGFRs may interact with other cell surface receptors, like G-protein-coupled receptors (GPCRs) or receptor tyrosine kinases (RTKs). The cross-talk between various receptors modulates the strength and specificity of intracellular signaling and cell fate. At the cell surface FGFRs can assemble into large complexes involving various cell adhesion molecules (CAMs). The interplay between FGFRs and CAMs affects cell–cell interaction and motility and is especially important for development of the central nervous system. This review summarizes current stage of knowledge about the regulation of FGFRs by the plasma membrane-embedded partner proteins and highlights the importance of FGFRs-containing membrane complexes in pathological conditions, including cancer.
Collapse
|
23
|
Shimizu Y, Suzuki T, Yoshikawa T, Endo I, Nakatsura T. Next-Generation Cancer Immunotherapy Targeting Glypican-3. Front Oncol 2019; 9:248. [PMID: 31024850 PMCID: PMC6469401 DOI: 10.3389/fonc.2019.00248] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 03/18/2019] [Indexed: 12/16/2022] Open
Abstract
Glypican-3 (GPC3), a 65 kD protein consisting of 580 amino acids, is a heparan sulfate proteoglycan bound to the cell membrane by glycosylphosphatidylinositol. This protein is expressed in the liver and the kidney of healthy fetuses but is hardly expressed in adults, except in the placenta. Contrarily, GPC3 is specifically expressed in hepatocellular carcinoma (HCC), ovarian clear cell carcinoma, melanoma, squamous cell carcinoma of the lung, hepatoblastoma, nephroblastoma (Wilms tumor), yolk sac tumor, and some pediatric cancers. Although the precise function of GPC3 remains unclear, it has been strongly suggested that it is related to the malignant transformation of HCC. We identified GPC3 as a promising target for cancer immunotherapy and have been working on the development of cancer immunotherapeutic agents targeting it through clinical trials. In some trials, it was revealed that the GPC3 peptide vaccines we developed using human leukocyte antigen-A24- and A2-restricted GPC3-derived peptides could induce GPC3-specific cytotoxic T cells in most vaccinated patients and thereby improve their prognosis. To further improve the clinical efficacy of cancer immunotherapy targeting GPC3, we are also developing next-generation therapeutic strategies using T cells engineered to express antigen-specific T-cell receptor or chimeric antigen receptor. In addition, we have successfully monitored the levels of serum full-length GPC3 protein, which is somehow secreted in the blood. The utility of GPC3 as a biomarker for predicting tumor recurrence and treatment efficacy is now being considered. In this review article, we summarize the results of clinical trials carried out by our team and describe the novel agent targeting the cancer-specific shared antigen, GPC3.
Collapse
Affiliation(s)
- Yasuhiro Shimizu
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan.,Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Toshihiro Suzuki
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Toshiaki Yoshikawa
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tetsuya Nakatsura
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| |
Collapse
|
24
|
Issop Y, Hathazi D, Khan MM, Rudolf R, Weis J, Spendiff S, Slater CR, Roos A, Lochmüller H. GFPT1 deficiency in muscle leads to myasthenia and myopathy in mice. Hum Mol Genet 2019; 27:3218-3232. [PMID: 29905857 PMCID: PMC6121184 DOI: 10.1093/hmg/ddy225] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 06/05/2018] [Indexed: 11/13/2022] Open
Abstract
Glutamine-fructose-6-phosphate transaminase 1 (GFPT1) is the rate-limiting enzyme in the hexosamine biosynthetic pathway which yields precursors required for protein and lipid glycosylation. Mutations in GFPT1 and other genes downstream of this pathway cause congenital myasthenic syndrome (CMS) characterized by fatigable muscle weakness owing to impaired neurotransmission. The precise pathomechanisms at the neuromuscular junction (NMJ) owing to a deficiency in GFPT1 is yet to be discovered. One of the challenges we face is the viability of Gfpt1−/− knockout mice. In this study, we use Cre/LoxP technology to generate a muscle-specific GFPT1 knockout mouse model, Gfpt1tm1d/tm1d, characteristic of the human CMS phenotype. Our data suggest a critical role for muscle derived GFPT1 in the development of the NMJ, neurotransmission, skeletal muscle integrity and highlight that a deficiency in skeletal muscle alone is sufficient to cause morphological postsynaptic NMJ changes that are accompanied by presynaptic alterations despite the conservation of neuronal GFPT1 expression. In addition to the conventional morphological NMJ changes and fatigable muscle weakness, Gfpt1tm1d/tm1d mice display a progressive myopathic phenotype with the presence of tubular aggregates in muscle, characteristic of the GFPT1-CMS phenotype. We further identify an upregulation of skeletal muscle proteins glypican-1, farnesyltransferase/geranylgeranyltransferase type-1 subunit α and muscle-specific kinase, which are known to be involved in the differentiation and maintenance of the NMJ. The Gfpt1tm1d/tm1d model allows for further investigation of pathophysiological consequences on genes and pathways downstream of GFPT1 likely to involve misglycosylation or hypoglycosylation of NMJs and muscle targets.
Collapse
Affiliation(s)
- Yasmin Issop
- John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle University, Newcastle, UK
| | - Denisa Hathazi
- Leibniz-Institut für Analytische Wissenschaften-ISAS e.V, Dortmund, Germany
| | - Muzamil Majid Khan
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - Rüdiger Rudolf
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany.,Interdisciplinary Center for Neurosciences, University of Heidelberg, Heidelberg, Germany.,Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, Mannheim, Germany
| | - Joachim Weis
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Sally Spendiff
- John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle University, Newcastle, UK
| | - Clarke R Slater
- Institute of Neuroscience, Newcastle University, Newcastle, UK
| | - Andreas Roos
- John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle University, Newcastle, UK.,Leibniz-Institut für Analytische Wissenschaften-ISAS e.V, Dortmund, Germany
| | - Hanns Lochmüller
- John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle University, Newcastle, UK.,Department of Neuropediatrics and Muscle Disorders,Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany.,Centro Nacional de Análisis Genómico (CNAG-CRG), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain
| |
Collapse
|
25
|
Sapoń K, Janas T, Sikorski AF, Janas T. Polysialic acid chains exhibit enhanced affinity for ordered regions of membranes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1861:245-255. [DOI: 10.1016/j.bbamem.2018.07.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 06/25/2018] [Accepted: 07/19/2018] [Indexed: 12/28/2022]
|
26
|
Avin KG, Vallejo JA, Chen NX, Wang K, Touchberry CD, Brotto M, Dallas SL, Moe SM, Wacker MJ. Fibroblast growth factor 23 does not directly influence skeletal muscle cell proliferation and differentiation or ex vivo muscle contractility. Am J Physiol Endocrinol Metab 2018; 315:E594-E604. [PMID: 29558205 PMCID: PMC6230710 DOI: 10.1152/ajpendo.00343.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 01/29/2018] [Accepted: 02/15/2018] [Indexed: 02/06/2023]
Abstract
Skeletal muscle dysfunction accompanies the clinical disorders of chronic kidney disease (CKD) and hereditary hypophosphatemic rickets. In both disorders, fibroblast growth factor 23 (FGF23), a bone-derived hormone regulating phosphate and vitamin D metabolism, becomes chronically elevated. FGF23 has been shown to play a direct role in cardiac muscle dysfunction; however, it is unknown whether FGF23 signaling can also directly induce skeletal muscle dysfunction. We found expression of potential FGF23 receptors ( Fgfr1-4) and α-Klotho in muscles of two animal models (CD-1 and Cy/+ rat, a naturally occurring rat model of chronic kidney disease-mineral bone disorder) as well as C2C12 myoblasts and myotubes. C2C12 proliferation, myogenic gene expression, oxidative stress marker 8-OHdG, intracellular Ca2+ ([Ca2+]i), and ex vivo contractility of extensor digitorum longus (EDL) or soleus muscles were assessed after treatment with various amounts of FGF23. FGF23 (2-100 ng/ml) did not alter C2C12 proliferation, expression of myogenic genes, or oxidative stress after 24- to 72-h treatment. Acute or prolonged FGF23 treatment up to 6 days did not alter C2C12 [Ca2+]i handling, nor did acute treatment with FGF23 (9-100 ng/ml) affect EDL and soleus muscle contractility. In conclusion, although skeletal muscles express the receptors involved in FGF23-mediated signaling, in vitro FGF23 treatments failed to directly alter skeletal muscle development or function under the conditions tested. We hypothesize that other endogenous substances may be required to act in concert with FGF23 or apart from FGF23 to promote muscle dysfunction in hereditary hypophosphatemic rickets and CKD.
Collapse
Affiliation(s)
- Keith G Avin
- Department of Physical Therapy, School of Health and Rehabilitation Sciences, Indiana University , Indianapolis, Indiana
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana
| | - Julian A Vallejo
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City , Kansas City, Missouri
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City , Kansas City, Missouri
| | - Neal X Chen
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana
| | - Kun Wang
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City , Kansas City, Missouri
| | - Chad D Touchberry
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City , Kansas City, Missouri
| | - Marco Brotto
- College of Nursing and Health Innovation, Bone-Muscle Collaborative Sciences, University of Texas-Arlington , Arlington, Texas
| | - Sarah L Dallas
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City , Kansas City, Missouri
| | - Sharon M Moe
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana
- Roudebush Veterans Administration Medical Center , Indianapolis, Indiana
| | - Michael J Wacker
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City , Kansas City, Missouri
| |
Collapse
|
27
|
Signaling network involved in the GPC3-induced inhibition of breast cancer progression: role of canonical Wnt pathway. J Cancer Res Clin Oncol 2018; 144:2399-2418. [PMID: 30267212 DOI: 10.1007/s00432-018-2751-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 09/11/2018] [Indexed: 12/17/2022]
Abstract
PURPOSE We have shown that GPC3 overexpression in breast cancer cells inhibits in vivo tumor progression, by acting as a metastatic suppressor. GPC3-overexpressing cells are less clonogenic, viable and motile, while their homotypic adhesion is increased. We have presented evidences indicating that GPC3 inhibits canonical Wnt and Akt pathways, while non-canonical Wnt and p38MAPK cascades are activated. In this study, we aimed to investigate whether GPC3-induced Wnt signaling inhibition modulates breast cancer cell properties as well as to describe the interactions among pathways modulated by GPC3. METHODS Fluorescence microscopy, qRT-PCR microarray, gene reporter assay and Western blotting were performed to determine gene expression levels, signaling pathway activities and molecule localization. Lithium was employed to activate canonical Wnt pathway and treated LM3-GPC3 cell viability, migration, cytoskeleton organization and homotypic adhesion were assessed using MTS, wound healing, phalloidin staining and suspension growth assays, respectively. RESULTS We provide new data demonstrating that GPC3 blocks-also at a transcriptional level-both autocrine and paracrine canonical Wnt activities, and that this inhibition is required for GPC3 to modulate migration and homotypic adhesion. Our results indicate that GPC3 is secreted into the extracellular media, suggesting that secreted GPC3 competes with Wnt factors or interacts with them and thus prevents Wnt binding to Fz receptors. We also describe the complex network of interactions among GPC3-modulated signaling pathways. CONCLUSION GPC3 is operating through an intricate molecular signaling network. From the balance of these interactions, the inhibition of breast metastatic spread induced by GPC3 emerges.
Collapse
|
28
|
Keller-Pinter A, Szabo K, Kocsis T, Deak F, Ocsovszki I, Zvara A, Puskas L, Szilak L, Dux L. Syndecan-4 influences mammalian myoblast proliferation by modulating myostatin signalling and G1/S transition. FEBS Lett 2018; 592:3139-3151. [PMID: 30129974 PMCID: PMC6221024 DOI: 10.1002/1873-3468.13227] [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: 02/06/2018] [Revised: 07/27/2018] [Accepted: 08/17/2018] [Indexed: 11/07/2022]
Abstract
Myostatin, a TGF‐β superfamily member, is a negative regulator of muscle growth. Here we describe how myostatin activity is regulated by syndecan‐4, a ubiquitous transmembrane heparan sulfate proteoglycan. During muscle regeneration the levels of both syndecan‐4 and promyostatin decline gradually after a sharp increase, concurrently with the release of mature myostatin. Promyostatin and syndecan‐4 co‐immunoprecipitate, and the interaction is heparinase‐sensitive. ShRNA‐mediated silencing of syndecan‐4 reduces C2C12 myoblast proliferation via blocking the progression from G1‐ to S‐phase of the cell cycle, which is accompanied by elevated levels of myostatin and p21(Waf1/Cip1), and decreases in cyclin E and cyclin D1 expression. Our results suggest that syndecan‐4 functions as a reservoir for promyostatin regulating the local bioavailability of mature myostatin.
Collapse
Affiliation(s)
- Aniko Keller-Pinter
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Hungary
| | - Kitti Szabo
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Hungary
| | - Tamas Kocsis
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Hungary
| | | | - Imre Ocsovszki
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Hungary
| | - Agnes Zvara
- Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Laszlo Puskas
- Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Laszlo Szilak
- Szilak Laboratories Bioinformatics & Molecule-Design Ltd., Szeged, Hungary
| | - Laszlo Dux
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Hungary
| |
Collapse
|
29
|
Yang Z, Song Z, Nie X, Yang J, Zhu C, Guo K, Gu Y. Characteristic properties of muscular-derived extracellular matrix and its application in rat abdominal wall defects. Regen Med 2018; 13:503-517. [PMID: 30028644 DOI: 10.2217/rme-2017-0116] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM To investigate the behavior of porcine-derived decellularized muscular matrix (DMM) in vitro and compare the performance of this biological mesh with that of acellular dermal matrix from pig in a full abdominal wall defect model. MATERIALS & METHODS To describe the in vitro properties of the DMM scaffold with extracellular matrix histological analysis, growth factor quantification and scanning electron microscopy analysis. To compare structural and functional remodeling between acellular dermal matrix and DMM implants in a rodent full abdominal wall defect model. RESULTS & CONCLUSION The results demonstrated that cellular components were effectively removed in the DMM scaffold, which also maintained a 3D architecture, biochemical components and strong mechanical properties. In vivo experiments confirm that the DMM mesh could promote remodeling and reconstruction of functional skeletal muscle tissue.
Collapse
Affiliation(s)
- Zhi Yang
- Department of General Surgery, Hernia & Abdominal Wall Surgery Center of Shanghai Jiao Tong University, Shanghai Ninth People's Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai 200011, PR China.,Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, PR China
| | - Zhicheng Song
- Department of General Surgery, Hernia & Abdominal Wall Surgery Center of Shanghai Jiao Tong University, Shanghai Ninth People's Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai 200011, PR China
| | - Xin Nie
- Department of General Surgery, Hernia & Abdominal Wall Surgery Center of Shanghai Jiao Tong University, Shanghai Ninth People's Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai 200011, PR China
| | - Jianjun Yang
- Department of General Surgery, Hernia & Abdominal Wall Surgery Center of Shanghai Jiao Tong University, Shanghai Ninth People's Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai 200011, PR China
| | - Chenfang Zhu
- Department of General Surgery, Hernia & Abdominal Wall Surgery Center of Shanghai Jiao Tong University, Shanghai Ninth People's Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai 200011, PR China
| | - Kaijin Guo
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, PR China
| | - Yan Gu
- Department of General Surgery, Hernia & Abdominal Wall Surgery Center of Shanghai Jiao Tong University, Shanghai Ninth People's Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai 200011, PR China
| |
Collapse
|
30
|
The effect of syndecan-4 and glypican-1 knockdown on the proliferation and differentiation of turkey satellite cells differing in age and growth rates. Comp Biochem Physiol A Mol Integr Physiol 2018; 223:33-41. [PMID: 29775666 DOI: 10.1016/j.cbpa.2018.05.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/19/2018] [Accepted: 05/08/2018] [Indexed: 11/22/2022]
Abstract
Posthatch skeletal muscle growth requires myogenic satellite cells and the dynamic expression of cell membrane-associated proteins. The membrane associated heparan sulfate proteoglycans, syndecan-4 and glypican-1, link the satellite cell niche to the intracellular environment. Sydnecan-4 and glypican-1 are differentially expressed with age in turkey satellite cells and their over-expression impacts both satellite cell proliferation and differentiation, but their effect on satellite cells from lines with different growth potentials is not known. The objective of the current study was to determine if syndecan-4 and glypican-1 regulation of satellite cell proliferation and differentiation is affected by age and growth selection. Pectoralis major satellite cells isolated at 1 d, 7 and 16-wk of age from a Randombred Control 2 (RBC2) line and a 16-wk body weight (F) line selected from the RBC2 line turkeys were studied. Syndecan-4 and glypican-1 expression was knocked down in both lines. The F-line cells proliferated faster than RBC2 line cells regardless of age, while differentiation tended to be greater in RBC2 line cells than F-line cells at each age. Syndecan-4 knockdown decreased proliferation at 7- and 16-wk but not 1 d cells, and increased differentiation at 1 d and 7 wk but not 16 wk cells. Glypican-1 knockdown differentially affected proliferation depending on cell age, whereas differentiation was decreased for 7- and 16-wk but not 1 d cells. These data suggest syndecan-4 and glypican-1 differentially affected satellite cell function in an age-dependent manner, but had little impact on differences in proliferation and differentiation due to growth selection.
Collapse
|
31
|
The extracellular matrix: Focus on oligodendrocyte biology and targeting CSPGs for remyelination therapies. Glia 2018; 66:1809-1825. [DOI: 10.1002/glia.23333] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 03/06/2018] [Accepted: 03/06/2018] [Indexed: 12/31/2022]
|
32
|
Chui MH, Have C, Hoang LN, Shaw P, Lee CH, Clarke BA. Genomic profiling identifies GPC5 amplification in association with sarcomatous transformation in a subset of uterine carcinosarcomas. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2018; 4:69-78. [PMID: 29416878 PMCID: PMC5783974 DOI: 10.1002/cjp2.89] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 12/10/2017] [Accepted: 12/13/2017] [Indexed: 01/24/2023]
Abstract
Uterine carcinosarcoma, also known as Malignant Mixed Müllerian Tumour, is a high‐grade biphasic neoplasm composed of sarcomatous elements thought to originate via transdifferentiation from high‐grade endometrial carcinoma. To identify molecular factors contributing to the histogenesis of this tumour, we analyzed DNA extracted from matched carcinoma and sarcoma components from 12 cases of carcinosarcoma by a molecular inversion probe microarray to assess genomic copy number alterations (CNAs) and allelic imbalances. Widespread CNAs were identified in tumours with serous histology in the carcinoma component (9/12), while the remaining three cases with endometrioid carcinoma were near‐diploid. Quantification of the extent of genomic aberrations revealed a significant increase in sarcoma relative to carcinoma in tumours with well‐delineated histologic components. Focal amplification of 13q31.3 was identified in 6/12 profiled tumours, of which four harboured the aberration exclusively in the sarcoma component. This result was verified by fluorescence in situ hybridization against GPC5, the only gene situated within the minimal region of amplification. In a validation cohort composed of 97 carcinosarcomas and other uterine sarcomas, amplification of GPC5 (GPC5/CEP13 ratio ≥ 2.2) was identified in 11/97 (11.3%) cases (9/64 carcinosarcoma, 1/3 rhabdomyosarcoma, 1/21 leiomyosarcoma, 0/8 adenosarcoma, 0/1 undifferentiated endometrial sarcoma) and an additional 4 (2.8%) cases had low level gains (GPC5/CEP13 ratio ≥1.5 but <2.2). The functional relevance of Glypican‐5, the gene product of GPC5, in regulating differentiation and lineage commitment was demonstrated in an endometrial carcinoma cell line in vitro. In conclusion, we identified GPC5 amplification as a molecular event mediating epithelial‐mesenchymal transdifferentiation in a subset of uterine carcinosarcomas.
Collapse
Affiliation(s)
- M Herman Chui
- Department of PathologyUniversity Health Network, University of TorontoTorontoCanada
| | - Cherry Have
- Department of PathologyUniversity Health Network, University of TorontoTorontoCanada
| | - Lien N Hoang
- Department of Pathology, BC Cancer AgencyUniversity of British ColumbiaVancouverCanada
| | - Patricia Shaw
- Department of PathologyUniversity Health Network, University of TorontoTorontoCanada
| | - Cheng-Han Lee
- Department of Pathology, BC Cancer AgencyUniversity of British ColumbiaVancouverCanada
| | - Blaise A Clarke
- Department of PathologyUniversity Health Network, University of TorontoTorontoCanada
| |
Collapse
|
33
|
Mashinchian O, Pisconti A, Le Moal E, Bentzinger CF. The Muscle Stem Cell Niche in Health and Disease. Curr Top Dev Biol 2017; 126:23-65. [PMID: 29305000 DOI: 10.1016/bs.ctdb.2017.08.003] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The regulation of stem cells that maintain and regenerate postnatal tissues depends on extrinsic signals originating from their microenvironment, commonly referred to as the stem cell niche. Complex higher-order regulatory interrelationships with the tissue and factors in the systemic circulation are integrated and propagated to the stem cells through the niche. The stem cell niche in skeletal muscle tissue is both a paradigm for a structurally and functionally relatively static niche that maintains stem cell quiescence during tissue homeostasis, and a highly dynamic regenerative niche that is subject to extensive structural remodeling and a flux of different support cell populations. Conditions ranging from aging to chronically degenerative skeletal muscle diseases affect the composition of the niche and thereby impair the regenerative potential of muscle stem cells. A holistic and integrative understanding of the extrinsic mechanisms regulating muscle stem cells in health and disease in a broad systemic context will be imperative for the identification of regulatory hubs in the niche interactome that can be targeted to maintain, restore, or enhance the regenerative capacity of muscle tissue. Here, we review the microenvironmental regulation of muscle stem cells, summarize how niche dysfunction can contribute to disease, and discuss emerging therapeutic implications.
Collapse
Affiliation(s)
- Omid Mashinchian
- Nestlé Institute of Health Sciences, Lausanne, Switzerland; École Polytechnique Fédérale de Lausanne, Doctoral Program in Biotechnology and Bioengineering, Lausanne, Switzerland
| | - Addolorata Pisconti
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Emmeran Le Moal
- Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - C Florian Bentzinger
- Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada.
| |
Collapse
|
34
|
Velleman SG, Song Y. Development and Growth of the Avian Pectoralis Major (Breast) Muscle: Function of Syndecan-4 and Glypican-1 in Adult Myoblast Proliferation and Differentiation. Front Physiol 2017; 8:577. [PMID: 28848451 PMCID: PMC5550705 DOI: 10.3389/fphys.2017.00577] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 07/25/2017] [Indexed: 12/19/2022] Open
Abstract
Muscle fiber number is determined around the time hatch with continued posthatch muscle growth being mediated by the adult myoblast, satellite cell, population of cells. Satellite cells are dynamic in their expression of proteins including the cell membrane associated proteoglycans, syndecan-4 and glypican-1. These proteoglycans play roles in organizing the extracellular environment in the satellite cell niche, cytoskeletal structure, cell-to-cell adhesion, satellite cell migration, and signal transduction. This review article focuses on syndecan-4 and glypican-1 as both are capable of regulating satellite cell responsiveness to fibroblast growth factor 2. Fibroblast growth factor 2 is a potent stimulator of muscle cell proliferation and a strong inhibitor of differentiation. Proteoglycans are composed of a central core protein defined functional domains, and covalently attached glycosaminoglycans and N-glycosylation chains. The functional association of these components with satellite cell function is discussed as well as an emerging role for microRNA regulation of syndecan-4 and glypican-1.
Collapse
Affiliation(s)
- Sandra G Velleman
- Department of Animal Sciences, The Ohio State UniversityWooster, OH, United States
| | - Yan Song
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical SchoolBoston, MA, United States
| |
Collapse
|
35
|
Velleman S, Harding R. Regulation of turkey myogenic satellite cell migration by MicroRNAs miR-128 and miR-24. Poult Sci 2017; 96:1910-1917. [DOI: 10.3382/ps/pew434] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 10/26/2016] [Indexed: 12/13/2022] Open
|
36
|
Griffin J, St-Pierre N, Lilburn M, Wick M. Transcriptional comparison of myogenesis in leghorn and low score normal embryos. Poult Sci 2017; 96:1531-1543. [DOI: 10.3382/ps/pew452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 11/16/2016] [Indexed: 12/13/2022] Open
|
37
|
Rao N, Agmon G, Tierney MT, Ungerleider JL, Braden RL, Sacco A, Christman KL. Engineering an Injectable Muscle-Specific Microenvironment for Improved Cell Delivery Using a Nanofibrous Extracellular Matrix Hydrogel. ACS NANO 2017; 11:3851-3859. [PMID: 28323411 PMCID: PMC5576867 DOI: 10.1021/acsnano.7b00093] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Injection of skeletal muscle progenitors has the potential to be a minimally invasive treatment for a number of diseases that negatively affect vasculature and skeletal muscle, including peripheral artery disease. However, success with this approach has been limited because of poor transplant cell survival. This is primarily attributed to cell death due to extensional flow through the needle, the harsh ischemic environment of the host tissue, a deleterious immune cell response, and a lack of biophysical cues supporting exogenous cell viability. We show that engineering a muscle-specific microenvironment, using a nanofibrous decellularized skeletal muscle extracellular matrix hydrogel and skeletal muscle fibroblasts, improves myoblast viability and maturation in vitro. In vivo, this translates to improved cell survival and engraftment and increased perfusion as a result of increased vascularization. Our results indicate that a combinatorial delivery system, which more fully recapitulates the tissue microenvironment, can improve cell delivery to skeletal muscle.
Collapse
Affiliation(s)
- Nikhil Rao
- Department of Bioengineering and Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, USA, 92037
| | - Gillie Agmon
- Department of Bioengineering and Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, USA, 92037
| | - Matthew T. Tierney
- Graduate School of Biomedical Sciences, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA, 92037
| | - Jessica L. Ungerleider
- Department of Bioengineering and Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, USA, 92037
| | - Rebecca L. Braden
- Department of Bioengineering and Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, USA, 92037
| | - Alessandra Sacco
- Graduate School of Biomedical Sciences, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA, 92037
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA, 92037
| | - Karen L. Christman
- Department of Bioengineering and Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, USA, 92037
| |
Collapse
|
38
|
Pawlikowski B, Vogler TO, Gadek K, Olwin BB. Regulation of skeletal muscle stem cells by fibroblast growth factors. Dev Dyn 2017; 246:359-367. [PMID: 28249356 DOI: 10.1002/dvdy.24495] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 02/16/2017] [Accepted: 02/16/2017] [Indexed: 01/04/2023] Open
Abstract
Fibroblast growth factors (FGFs) are essential for self-renewal of skeletal muscle stem cells (satellite cells) and required for maintenance and repair of skeletal muscle. Satellite cells express high levels of FGF receptors 1 and 4, low levels of FGF receptor 3, and little or no detectable FGF receptor 2. Of the multiple FGFs that influence satellite cell function in culture, FGF2 and FGF6 are the only members that regulate satellite cell function in vivo by activating ERK MAPK, p38α/β MAPKs, PI3 kinase, PLCγ and STATs. Regulation of FGF signaling is complex in satellite cells, requiring Syndecan-4, a heparan sulfate proteoglycan, as well as ß1-integrin and fibronectin. During aging, reduced responsiveness to FGF diminishes satellite cell self-renewal, leading to impaired skeletal muscle regeneration and depletion of satellite cells. Mislocalization of ß1-integrin, reductions in fibronectin, and alterations in heparan sulfate content all contribute to reduced FGF responsiveness in satellite cells. How these cell surface proteins regulate satellite cell self-renewal is incompletely understood. Here we summarize the current knowledge, highlighting the role(s) for FGF signaling in skeletal muscle regeneration, satellite cell behavior, and age-induced muscle wasting. Developmental Dynamics 246:359-367, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Bradley Pawlikowski
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado
| | - Thomas Orion Vogler
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado
| | - Katherine Gadek
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado
| | - Bradley B Olwin
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado
| |
Collapse
|
39
|
The heparanase/heparan sulfate proteoglycan axis: A potential new therapeutic target in sarcomas. Cancer Lett 2016; 382:245-254. [PMID: 27666777 DOI: 10.1016/j.canlet.2016.09.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 09/08/2016] [Accepted: 09/08/2016] [Indexed: 12/29/2022]
Abstract
Heparanase, the only known mammalian endoglycosidase degrading heparan sulfate (HS) chains of HS proteoglycans (HSPG), is a highly versatile protein affecting multiple events in tumor cells and their microenvironment. In several malignancies, deregulation of the heparanase/HSPG system has been implicated in tumor progression, hence representing a valuable therapeutic target. Currently, multiple agents interfering with the heparanase/HSPG axis are under clinical investigation. Sarcomas are characterized by a high biomolecular complexity and multiple levels of interconnection with microenvironment sustaining their growth and progression. The clinical management of advanced diseases remains a challenge. In several sarcoma subtypes, high levels of heparanase expression have been correlated with poor prognosis associated factors. On the other hand, expression of cell surface-associated HSPGs (i.e. glypicans and syndecans) has been found altered in specific sarcoma subtypes. Recent studies provided the preclinical proof-of-principle of the role of the heparanase/HSPG axis as therapeutic target in various sarcoma subtypes. Although currently there are no clinical trials evaluating agents targeting heparanase and/or HSPGs in sarcomas, we here provide arguments for this strategy as potentially able to implement the therapeutic options for sarcoma patients.
Collapse
|
40
|
Kawahara R, Granato DC, Yokoo S, Domingues RR, Trindade DM, Paes Leme AF. Mass spectrometry-based proteomics revealed Glypican-1 as a novel ADAM17 substrate. J Proteomics 2016; 151:53-65. [PMID: 27576135 DOI: 10.1016/j.jprot.2016.08.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 08/02/2016] [Accepted: 08/25/2016] [Indexed: 12/16/2022]
Abstract
ADAM17 (a disintegrin and metalloproteinase 17) is a plasma membrane metalloprotease involved in proteolytic release of the extracellular domain of many cell surface molecules, a process known as ectodomain shedding. Through this process, ADAM17 is implicated in several aspects of tumor growth and metastasis in a broad range of tumors, including head and neck squamous cell carcinomas (HNSCC). In this study, mass spectrometry-based proteomics approaches revealed glypican-1 (GPC1) as a new substrate for ADAM17, and its shedding was confirmed to be metalloprotease-dependent, induced by a pleiotropic agent (PMA) and physiologic ligand (EGF), and inhibited by marimastat. In addition, immunoblotting analysis of GPC1 in the extracellular media from control and ADAM17shRNA pointed to a direct involvement of ADAM17 in the cleavage of GPC1. Moreover, mass spectrometry-based interactome analysis of GPC1 revealed biological functions and pathways related mainly to cellular movement, adhesion and proliferation, which were events also modulated by up regulation of full length and cleavage GPC1. Altogether, we showed that GPC1 is a novel ADAM17 substrate, thus the function of GPC1 may be modulated by proteolysis signaling. BIOLOGICAL SIGNIFICANCE Inhibition of metalloproteases as a therapeutic approach has failed because there is limited knowledge of the degradome of individual proteases as well as the cellular function of cleaved substrates. Using different proteomic techniques, this study uncovered novel substrates that can be modulated by ADAM17 in oral squamous cell carcinoma cell line. Glypican-1 was validated as a novel substrate for ADAM17, with important function in adhesion, proliferation and migration of carcinoma cells. Therefore, this study opens new avenues regarding the proteolysis-mediated function of GPC1 by ADAM17.
Collapse
Affiliation(s)
- Rebeca Kawahara
- Laboratório Nacional de Biociências, LNBio, CNPEM, Campinas, Brazil
| | | | - Sami Yokoo
- Laboratório Nacional de Biociências, LNBio, CNPEM, Campinas, Brazil
| | | | | | | |
Collapse
|
41
|
Akl MR, Nagpal P, Ayoub NM, Tai B, Prabhu SA, Capac CM, Gliksman M, Goy A, Suh KS. Molecular and clinical significance of fibroblast growth factor 2 (FGF2 /bFGF) in malignancies of solid and hematological cancers for personalized therapies. Oncotarget 2016; 7:44735-44762. [PMID: 27007053 PMCID: PMC5190132 DOI: 10.18632/oncotarget.8203] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 03/10/2016] [Indexed: 12/30/2022] Open
Abstract
Fibroblast growth factor (FGF) signaling is essential for normal and cancer biology. Mammalian FGF family members participate in multiple signaling pathways by binding to heparan sulfate and FGF receptors (FGFR) with varying affinities. FGF2 is the prototype member of the FGF family and interacts with its receptor to mediate receptor dimerization, phosphorylation, and activation of signaling pathways, such as Ras-MAPK and PI3K pathways. Excessive mitogenic signaling through the FGF/FGFR axis may induce carcinogenic effects by promoting cancer progression and increasing the angiogenic potential, which can lead to metastatic tumor phenotypes. Dysregulated FGF/FGFR signaling is associated with aggressive cancer phenotypes, enhanced chemotherapy resistance and poor clinical outcomes. In vitro experimental settings have indicated that extracellular FGF2 affects proliferation, drug sensitivity, and apoptosis of cancer cells. Therapeutically targeting FGF2 and FGFR has been extensively assessed in multiple preclinical studies and numerous drugs and treatment options have been tested in clinical trials. Diagnostic assays are used to quantify FGF2, FGFRs, and downstream signaling molecules to better select a target patient population for higher efficacy of cancer therapies. This review focuses on the prognostic significance of FGF2 in cancer with emphasis on therapeutic intervention strategies for solid and hematological malignancies.
Collapse
Affiliation(s)
- Mohamed R. Akl
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Poonam Nagpal
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Nehad M. Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Betty Tai
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Sathyen A. Prabhu
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Catherine M. Capac
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Matthew Gliksman
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Andre Goy
- Lymphoma Division, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - K. Stephen Suh
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| |
Collapse
|
42
|
Flowers SA, Zhou X, Wu J, Wang Y, Makambi K, Kallakury BV, Singer MS, Rosen SD, Davidson B, Goldman R. Expression of the extracellular sulfatase SULF2 is associated with squamous cell carcinoma of the head and neck. Oncotarget 2016; 7:43177-43187. [PMID: 27223083 PMCID: PMC5190016 DOI: 10.18632/oncotarget.9506] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 04/16/2016] [Indexed: 12/15/2022] Open
Abstract
Sulfatase 2 (SULF2), an extracellular sulfatase that alters sulfation on heparan sulfate proteoglycans, is involved in the tumorigenesis and progression of several carcinomas. SULF2 expression has not been evaluated in squamous cell carcinoma of the head and neck (HNSCC). Here we report results of IHC of SULF2 expression in HNSCC tissue. SULF2 was detected in 57% of tumors (n = 40) with a significant increase in intensity and number of stained cells compared to adjacent cancer-free tissue (p-value < 0.01), increasing with cancer stage when comparing stages 1 and 2 to stages 3 and 4 (p-value 0.01). SULF2 was not detected in epithelial cells of cancer-free controls, and expression was independent of patient demographics, tumor location and etiological factors, smoking and HPV infection by p16 IHC analysis. Sandwich ELISA was performed on serum of HNSCC patients (n = 28) and controls (n = 35), and although SULF2 was detectable, no change was observed in HNSCC. Saliva, collected by mouthwash, from HNSCC patients (n = 8) and controls (n = 8) was also tested by ELISA in a preliminary investigation and an increase in SULF2 was observed in HNSCC (p-value 0.041). Overall, this study shows that SULF2 is increased in HNSCC independent of tissue location (oral cavity, oropharynx, larynx and hypopharynx), patient demographics and etiology. Although no change in SULF2 was detected in HNSCC serum, its detection in saliva makes it worthy of further investigation as a potential HNSCC biomarker.
Collapse
Affiliation(s)
- Sarah A. Flowers
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Xin Zhou
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Jing Wu
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Yiwen Wang
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Kepher Makambi
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Bhaskar V. Kallakury
- Department of Pathology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Mark S. Singer
- Department of Anatomy, University of California, San Francisco, CA 94143, USA
| | - Steven D. Rosen
- Department of Anatomy, University of California, San Francisco, CA 94143, USA
| | - Bruce Davidson
- Department of Otolaryngology-Head and Neck Surgery, Medstar Georgetown University Hospital, Washington, DC 20057, USA
| | - Radoslav Goldman
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC 20057, USA
| |
Collapse
|
43
|
Powell D, Velleman S, Cowieson A, Singh M, Muir W. Influence of chick hatch time and access to feed on broiler muscle development. Poult Sci 2016; 95:1433-48. [DOI: 10.3382/ps/pew047] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 01/10/2016] [Indexed: 11/20/2022] Open
|
44
|
Monteforte AJ, Lam B, Das S, Mukhopadhyay S, Wright CS, Martin PE, Dunn AK, Baker AB. Glypican-1 nanoliposomes for potentiating growth factor activity in therapeutic angiogenesis. Biomaterials 2016; 94:45-56. [PMID: 27101205 DOI: 10.1016/j.biomaterials.2016.03.048] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 03/29/2016] [Accepted: 03/30/2016] [Indexed: 12/26/2022]
Abstract
Therapeutic angiogenesis is a highly appealing concept for treating tissues that become ischemic due to vascular disease. A major barrier to the clinical translation of angiogenic therapies is that the patients that are in the greatest need of these treatments often have long term disease states and co-morbidities, such as diabetes and obesity, that make them resistant to angiogenic stimuli. In this study, we identified that human patients with type 2 diabetes have reduced levels of glypican-1 in the blood vessels of their skin. The lack of this key co-receptor in the tissue may make the application of exogenous angiogenic growth factors or cell therapies ineffective. We created a novel therapeutic enhancer for growth factor activity consisting of glypican-1 delivered in a nanoliposomal carrier (a "glypisome"). Here, we demonstrate that glypisomes enhance FGF-2 mediated endothelial cell proliferation, migration and tube formation. In addition, glypisomes enhance FGF-2 trafficking by increasing both uptake and endosomal processing. We encapsulated FGF-2 or FGF-2 with glypisomes in alginate beads and used these to deliver localized growth factor therapy in a murine hind limb ischemia model. Co-delivery of glypisomes with FGF-2 markedly increased the recovery of perfusion and vessel formation in ischemic hind limbs of wild type and diabetic mice in comparison to mice treated with FGF-2 alone. Together, our findings support that glypisomes are effective means for enhancing growth factor activity and may improve the response to local angiogenic growth factor therapies for ischemia.
Collapse
Affiliation(s)
- Anthony J Monteforte
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Brian Lam
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Subhamoy Das
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Somshuvra Mukhopadhyay
- Division of Pharmacology & Toxicology, University of Texas at Austin, Austin, TX, USA; Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA; Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, USA
| | - Catherine S Wright
- Diabetes Research Group, Department of Life Sciences and Institute for Applied Health Research, Glasgow Caledonian University, Glasgow G4 0BA, UK
| | - Patricia E Martin
- Diabetes Research Group, Department of Life Sciences and Institute for Applied Health Research, Glasgow Caledonian University, Glasgow G4 0BA, UK
| | - Andrew K Dunn
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Aaron B Baker
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA; Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, USA; The Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, TX, USA; Institute for Biomaterials, Drug Delivery and Regenerative Medicine, University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
45
|
Patel A, Xue Y, Mukundan S, Rohan LC, Sant V, Stolz DB, Sant S. Cell-Instructive Graphene-Containing Nanocomposites Induce Multinucleated Myotube Formation. Ann Biomed Eng 2016; 44:2036-48. [DOI: 10.1007/s10439-016-1586-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 03/02/2016] [Indexed: 01/19/2023]
|
46
|
Bhullar AS, Putman CT, Mazurak VC. Potential Role of Omega-3 Fatty Acids on the Myogenic Program of Satellite Cells. Nutr Metab Insights 2016; 9:1-10. [PMID: 26884682 PMCID: PMC4747635 DOI: 10.4137/nmi.s27481] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 11/23/2015] [Accepted: 11/25/2015] [Indexed: 12/22/2022] Open
Abstract
Skeletal muscle loss is associated with aging as well as pathological conditions. Satellite cells (SCs) play an important role in muscle regeneration. Omega-3 fatty acids are widely studied in a variety of muscle wasting diseases; however, little is known about their impact on skeletal muscle regeneration. The aim of this review is to evaluate studies examining the effect of omega-3 fatty acids, α-linolenic acid, eicosapentaenoic acid, and docosahexaenoic acid on the regulation of SC proliferation and differentiation. This review highlights mechanisms by which omega-3 fatty acids may modulate the myogenic program of the stem cell population within skeletal muscles and identifies considerations for future studies. It is proposed that minimally three myogenic transcriptional regulatory factors, paired box 7 (Pax7), myogenic differentiation 1 protein, and myogenin, should be measured to confirm the stage of SCs within the myogenic program affected by omega-3 fatty acids.
Collapse
Affiliation(s)
- Amritpal S Bhullar
- M.Sc, Faculty of Agricultural, Life, and Environmental Science, Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, Li Ka Shing Centre for Health Research Innovation, University of Alberta, Edmonton, AB, Canada
| | - Charles T Putman
- PhD, Associate Professor, Faculty of Physical Education and Recreation and Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Vera C Mazurak
- PhD, Associate Professor, Faculty of Agricultural, Life, and Environmental Science, Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, Li Ka Shing Centre for Health Research Innovation, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
47
|
Galimov A, Merry TL, Luca E, Rushing EJ, Mizbani A, Turcekova K, Hartung A, Croce CM, Ristow M, Krützfeldt J. MicroRNA-29a in Adult Muscle Stem Cells Controls Skeletal Muscle Regeneration During Injury and Exercise Downstream of Fibroblast Growth Factor-2. Stem Cells 2016; 34:768-80. [PMID: 26731484 DOI: 10.1002/stem.2281] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Revised: 09/23/2015] [Accepted: 10/31/2015] [Indexed: 01/18/2023]
Abstract
The expansion of myogenic progenitors (MPs) in the adult muscle stem cell niche is critical for the regeneration of skeletal muscle. Activation of quiescent MPs depends on the dismantling of the basement membrane and increased access to growth factors such as fibroblast growth factor-2 (FGF2). Here, we demonstrate using microRNA (miRNA) profiling in mouse and human myoblasts that the capacity of FGF2 to stimulate myoblast proliferation is mediated by miR-29a. FGF2 induces miR-29a expression and inhibition of miR-29a using pharmacological or genetic deletion decreases myoblast proliferation. Next generation RNA sequencing from miR-29a knockout myoblasts (Pax7(CE/+) ; miR-29a(flox/flox) ) identified members of the basement membrane as the most abundant miR-29a targets. Using gain- and loss-of-function experiments, we confirm that miR-29a coordinately regulates Fbn1, Lamc1, Nid2, Col4a1, Hspg2 and Sparc in myoblasts in vitro and in MPs in vivo. Induction of FGF2 and miR-29a and downregulation of its target genes precedes muscle regeneration during cardiotoxin (CTX)-induced muscle injury. Importantly, MP-specific tamoxifen-induced deletion of miR-29a in adult skeletal muscle decreased the proliferation and formation of newly formed myofibers during both CTX-induced muscle injury and after a single bout of eccentric exercise. Our results identify a novel miRNA-based checkpoint of the basement membrane in the adult muscle stem cell niche. Strategies targeting miR-29a might provide useful clinical approaches to maintain muscle mass in disease states such as ageing that involve aberrant FGF2 signaling.
Collapse
Affiliation(s)
- Artur Galimov
- Division of Endocrinology, Diabetes, and Clinical Nutrition, University Zurich and University Hospital Zurich, Zurich, Switzerland.,Competence Center Personalized Medicine, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Troy L Merry
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Edlira Luca
- Division of Endocrinology, Diabetes, and Clinical Nutrition, University Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Elisabeth J Rushing
- Institute of Neuropathology, University Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Amir Mizbani
- Division of Endocrinology, Diabetes, and Clinical Nutrition, University Zurich and University Hospital Zurich, Zurich, Switzerland.,Competence Center Personalized Medicine, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Katarina Turcekova
- Division of Endocrinology, Diabetes, and Clinical Nutrition, University Zurich and University Hospital Zurich, Zurich, Switzerland.,Competence Center Personalized Medicine, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Angelika Hartung
- Division of Endocrinology, Diabetes, and Clinical Nutrition, University Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Carlo M Croce
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University, Columbus, Ohio, USA
| | - Michael Ristow
- Competence Center Personalized Medicine, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Jan Krützfeldt
- Division of Endocrinology, Diabetes, and Clinical Nutrition, University Zurich and University Hospital Zurich, Zurich, Switzerland.,Competence Center Personalized Medicine, ETH Zurich and University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
48
|
Function of Membrane-Associated Proteoglycans in the Regulation of Satellite Cell Growth. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 900:61-95. [DOI: 10.1007/978-3-319-27511-6_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
49
|
Harding RL, Velleman SG. MicroRNA regulation of myogenic satellite cell proliferation and differentiation. Mol Cell Biochem 2015; 412:181-95. [PMID: 26715133 DOI: 10.1007/s11010-015-2625-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 12/08/2015] [Indexed: 01/10/2023]
Abstract
Myogenic satellite cells are stem cells responsible for muscle growth and regeneration. MicroRNAs (miRNAs) play significant roles in regulating numerous cellular processes. Two genes essential to satellite cell function are syndecan-4 and glypican-1. To determine if miRNAs influence myogenic satellite cell function, one miRNA predicted to bind syndecan-4 (miR-128) and two predicted to bind glypican-1 (miR-24 and miR-16) were inhibited in vitro by transfection of inhibitors targeting each miRNA. Inhibition of these miRNAs differentially affected the expression of syndecan-4, glypican-1, and myogenic regulatory factors myoD and myogenin. Inhibition of miR-16 reduced proliferation of satellite cells at 72 h. Inhibition of miR-128 and miR-24 did not affect proliferation. Inhibition of miRNAs reduced differentiation of satellite cells into myotubes at 48 and 72 h except for miR-16, which only affected differentiation at 72 h. Inhibition of all three miRNAs decreased myotube width at 24 h of differentiation and increased myotube width at 48 h of differentiation. Inhibiting these miRNAs also increased the number of nuclei per myotube at 72 h of differentiation. These data demonstrate individual miRNAs regulate genes essential for myogenic satellite cell proliferation and differentiation.
Collapse
Affiliation(s)
- Rachel L Harding
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, 1680 Madison Ave., Wooster, OH, 44691, USA
| | - Sandra G Velleman
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, 1680 Madison Ave., Wooster, OH, 44691, USA.
| |
Collapse
|
50
|
Lu Y, Bradley JS, Siegel PB, Yang N, Johnson SE. Selection for divergent body size alters rates of embryonic skeletal muscle formation and muscle gene expression patterns. Dev Growth Differ 2015; 57:614-24. [PMID: 26660844 DOI: 10.1111/dgd.12250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 10/05/2015] [Accepted: 10/13/2015] [Indexed: 11/28/2022]
Abstract
The impact of divergent selection for body size on embryogenesis is poorly understood. The objective of this experiment was to document skeletal muscle development during embryogenesis in two lines of chickens that display divergent growth as adults. Results reveal that after 54 generations of opposing selection from a common founder population, the embryos from the low weight select (LWS) line develop more rapidly during early embryogenesis than those from the high weight select (HWS) line. Muscle formation during the late embryonic period is more rapid and extensive in the HWS embryo than in the LWS contemporary. Isolated muscle progenitors from embryonic day 10 HWS embryos proliferated more rapidly, forming fibers sooner with a larger size than the LWS cells. The limited myogenic capacity of the LWS progenitor cells is not attributed to altered patterns of expression of Pax7, Pax3 or the myogenic regulatory factor genes. Members of the fibroblast growth factor family are potent mitogens and inhibitors of myoblast differentiation. Transcript abundance of FGF2 and FGF4 was measured in cultures of HWS and LWS progenitors as a function of time. The pattern of expression of FGF4 was similar between HWS and LWS with a large increase between days 1 and 3 followed by a reduction at day 5 of culture. Expression of FGF2 in LWS muscle cells did not change while a significant reduction in FGF2 expression was observed by day 5 in the HWS. Our results indicate that divergent selection for postnatal growth has altered embryonic development.
Collapse
Affiliation(s)
- Yue Lu
- National Engineering Laboratory for Animal Breeding and MOA Key Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.,Department of Animal and Poultry Sciences, Virginia Polytechnic and State University, Blacksburg, Virginia, 24061, USA
| | - Jennifer S Bradley
- Department of Animal and Poultry Sciences, Virginia Polytechnic and State University, Blacksburg, Virginia, 24061, USA
| | - Paul B Siegel
- Department of Animal and Poultry Sciences, Virginia Polytechnic and State University, Blacksburg, Virginia, 24061, USA
| | - Ning Yang
- National Engineering Laboratory for Animal Breeding and MOA Key Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Sally E Johnson
- Department of Animal and Poultry Sciences, Virginia Polytechnic and State University, Blacksburg, Virginia, 24061, USA
| |
Collapse
|