701
|
Nallasamy S, Yoshida K, Akins M, Myers K, Iozzo R, Mahendroo M. Steroid Hormones Are Key Modulators of Tissue Mechanical Function via Regulation of Collagen and Elastic Fibers. Endocrinology 2017; 158:950-962. [PMID: 28204185 PMCID: PMC5460796 DOI: 10.1210/en.2016-1930] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 02/01/2017] [Indexed: 01/12/2023]
Abstract
The extracellular matrix (ECM) plays an active and dynamic role that both reflects and facilitates the functional requirements of a tissue. The mature ECM of the nonpregnant cervix is drastically reorganized during pregnancy to drive changes in tissue mechanics that ensure safe birth. In this study, our research on mice deficient in the proteoglycan decorin have led to the finding that progesterone and estrogen play distinct and complementary roles to orchestrate structural reorganization of both collagen and elastic fibers in the cervix during pregnancy. Abnormalities in collagen and elastic fiber structure and tissue mechanical function evident in the cervix of nonpregnant and early pregnant decorin-null mice transiently recover for the remainder of pregnancy only to return 1 month postpartum. Consistent with the hypothesis that pregnancy levels of progesterone and estrogen may regulate ECM organization and turnover, expressions of factors required for assembly and synthesis of collagen and elastic fibers are temporally regulated, and the ultrastructure of collagen fibrils and elastic fibers is markedly altered during pregnancy in wild-type mice. Finally, utilizing ovariectomized nonpregnant decorin-null mice, we demonstrate structural resolution of collagen and elastic fibers by progesterone or estrogen, respectively, and the potential for both ECM proteins to contribute to mechanical function. These investigations advance understanding of regulatory factors that drive specialized ECM organization and contribute to an understanding of the cervical remodeling process, which may provide insight into potential complications associated with preterm birth that impact 9.6% of live births in the United States.
Collapse
Affiliation(s)
- Shanmugasundaram Nallasamy
- Department of Obstetrics and Gynecology and
- Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Kyoko Yoshida
- Department of Mechanical Engineering, Columbia University, New York, New York 10027; and
| | - Meredith Akins
- Department of Obstetrics and Gynecology and
- Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Kristin Myers
- Department of Mechanical Engineering, Columbia University, New York, New York 10027; and
| | - Renato Iozzo
- Department of Pathology, Anatomy, and Cell Biology
- Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| | - Mala Mahendroo
- Department of Obstetrics and Gynecology and
- Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| |
Collapse
|
702
|
Chaffey PK, Guan X, Wang LX, Tan Z. Introduction: General Aspects of the Chemical Biology of Glycoproteins. CHEMICAL BIOLOGY OF GLYCOPROTEINS 2017. [DOI: 10.1039/9781782623823-00001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
This chapter is meant to serve as an introduction to the remainder of the book by providing general background on the chemical biology of glycoproteins as well as a brief review of the chapters that follow. The purpose here is to introduce some basic concepts common to many forms of glycosylation for those readers who may be unfamiliar with the field. We begin with a discussion of the strategies and methods used to study protein glycosylation. During the overview, an effort is made to highlight a few relevant aspects of chemical glycobiology, including glycoprotein biosynthesis and a brief description of the synthesis and function of glycoproteins. Finally, we have a summary of the contributions from chemical biology over the years. It is our hope that, after reading this introductory chapter, the reader will have a broad view of the chemical glycobiology field as it currently stands and a deeper appreciation for some of the unique ideas that chemical biology brings to the field.
Collapse
Affiliation(s)
- Patrick K. Chaffey
- Department of Chemistry and Biochemistry and BioFrontiers Institute, University of Colorado Boulder CO 80303 USA
| | - Xiaoyang Guan
- Department of Chemistry and Biochemistry and BioFrontiers Institute, University of Colorado Boulder CO 80303 USA
| | - Lai-Xi Wang
- Department of Chemistry and Biochemistry, University of Maryland College Park MD 20742 USA
| | - Zhongping Tan
- Department of Chemistry and Biochemistry and BioFrontiers Institute, University of Colorado Boulder CO 80303 USA
| |
Collapse
|
703
|
Lumican effectively regulates the estrogen receptors-associated functional properties of breast cancer cells, expression of matrix effectors and epithelial-to-mesenchymal transition. Sci Rep 2017; 7:45138. [PMID: 28332606 PMCID: PMC5362815 DOI: 10.1038/srep45138] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 02/17/2017] [Indexed: 12/21/2022] Open
Abstract
Lumican is a small leucine-rich proteoglycan that has been shown to contribute in several physiological processes, but also to exert anticancer activity. On the other hand, it has been recently shown that knockdown of the estrogen receptor α (ERα) in low invasive MCF-7 (ERα+) breast cancer cells and the suppression of ERβ in highly aggressive MDA-MB-231 (ERβ+) cells significantly alter the functional properties of breast cancer cells and the gene expression profile of matrix macromolecules related to cancer progression and cell morphology. In this report, we evaluated the effects of lumican in respect to the ERs-associated breast cancer cell behaviour, before and after suppression of ERs, using scanning electron and confocal microscopies, qPCR and functional assays. Our data pinpointed that lumican significantly attenuated cell functional properties, including proliferation, migration and invasion. Furthermore, it modified cell morphology, inducing cell-cell junctions, evoked EMT/MET reprogramming and suppressed the expression of major matrix effectors (matrix metalloproteinases and EGFR) implicated in breast cancer progression. The effects of lumican were found to be related to the type of breast cancer cells and the ERα/β type. These data support the anticancer activity of lumican and open a new area for the pharmacological targeting of the invasive breast cancer.
Collapse
|
704
|
Napavichayanun S, Aramwit P. Effect of animal products and extracts on wound healing promotion in topical applications: a review. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2017; 28:703-729. [DOI: 10.1080/09205063.2017.1301772] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Supamas Napavichayanun
- Bioactive Resources for Innovative Clinical Applications Research Unit, Chulalongkorn University, Bangkok, Thailand
- Faculty of Pharmaceutical Sciences, Department of Pharmacy Practice, Chulalongkorn University, Bangkok, Thailand
| | - Pornanong Aramwit
- Bioactive Resources for Innovative Clinical Applications Research Unit, Chulalongkorn University, Bangkok, Thailand
- Faculty of Pharmaceutical Sciences, Department of Pharmacy Practice, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
705
|
Hsieh LTH, Nastase MV, Roedig H, Zeng-Brouwers J, Poluzzi C, Schwalm S, Fork C, Tredup C, Brandes RP, Wygrecka M, Huwiler A, Pfeilschifter J, Schaefer L. Biglycan- and Sphingosine Kinase-1 Signaling Crosstalk Regulates the Synthesis of Macrophage Chemoattractants. Int J Mol Sci 2017; 18:ijms18030595. [PMID: 28282921 PMCID: PMC5372611 DOI: 10.3390/ijms18030595] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 02/27/2017] [Accepted: 03/06/2017] [Indexed: 12/14/2022] Open
Abstract
In its soluble form, the extracellular matrix proteoglycan biglycan triggers the synthesis of the macrophage chemoattractants, chemokine (C-C motif) ligand CCL2 and CCL5 through selective utilization of Toll-like receptors (TLRs) and their adaptor molecules. However, the respective downstream signaling events resulting in biglycan-induced CCL2 and CCL5 production have not yet been defined. Here, we show that biglycan stimulates the production and activation of sphingosine kinase 1 (SphK1) in a TLR4- and Toll/interleukin (IL)-1R domain-containing adaptor inducing interferon (IFN)-β (TRIF)-dependent manner in murine primary macrophages. We provide genetic and pharmacological proof that SphK1 is a crucial downstream mediator of biglycan-triggered CCL2 and CCL5 mRNA and protein expression. This is selectively driven by biglycan/SphK1-dependent phosphorylation of the nuclear factor NF-κB p65 subunit, extracellular signal-regulated kinase (Erk)1/2 and p38 mitogen-activated protein kinases. Importantly, in vivo overexpression of soluble biglycan causes Sphk1-dependent enhancement of renal CCL2 and CCL5 and macrophage recruitment into the kidney. Our findings describe the crosstalk between biglycan- and SphK1-driven extracellular matrix- and lipid-signaling. Thus, SphK1 may represent a new target for therapeutic intervention in biglycan-evoked inflammatory conditions.
Collapse
Affiliation(s)
- Louise Tzung-Harn Hsieh
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe Universität, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
| | - Madalina-Viviana Nastase
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe Universität, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
- National Institute for Chemical-Pharmaceutical Research and Development, 112 Vitan Avenue, Bucharest 031299, Romania.
| | - Heiko Roedig
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe Universität, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
| | - Jinyang Zeng-Brouwers
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe Universität, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
| | - Chiara Poluzzi
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe Universität, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
| | - Stephanie Schwalm
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe Universität, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
| | - Christian Fork
- Institut für Kardiovaskulare Physiologie, Klinikum der Goethe-Universität, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
| | - Claudia Tredup
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe Universität, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
| | - Ralf P Brandes
- Institut für Kardiovaskulare Physiologie, Klinikum der Goethe-Universität, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
| | - Malgorzata Wygrecka
- Department of Biochemistry, Faculty of Medicine, Universities of Giessen and Marburg Lung Center, Friedrichstrasse 24, Giessen 35392, Germany.
| | - Andrea Huwiler
- Institute of Pharmacology, University of Bern, Inselspital INO-F, Bern CH-3010, Switzerland.
| | - Josef Pfeilschifter
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe Universität, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
| | - Liliana Schaefer
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe Universität, Theodor-Stern-Kai 7, Frankfurt am Main 60590, Germany.
| |
Collapse
|
706
|
|
707
|
Torres A, Gubbiotti MA, Iozzo RV. Decorin-inducible Peg3 Evokes Beclin 1-mediated Autophagy and Thrombospondin 1-mediated Angiostasis. J Biol Chem 2017; 292:5055-5069. [PMID: 28174297 PMCID: PMC5377817 DOI: 10.1074/jbc.m116.753632] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 02/06/2017] [Indexed: 01/31/2023] Open
Abstract
We previously discovered that systemic delivery of decorin for treatment of breast carcinoma xenografts induces paternally expressed gene 3 (Peg3), an imprinted gene encoding a zinc finger transcription factor postulated to function as a tumor suppressor. Here we found that de novo expression of Peg3 increased Beclin 1 promoter activity and protein expression. This process required the full-length Peg3 as truncated mutants lacking either the N-terminal SCAN domain or the zinc fingers failed to translocate to the nucleus and promote Beclin 1 transcription. Importantly, overexpression of Peg3 in endothelial cells stimulated autophagy and concurrently inhibited endothelial cell migration and evasion from a 3D matrix. Mechanistically, we found that Peg3 induced the secretion of the powerful angiostatic glycoprotein Thrombospondin 1 independently of Beclin 1 transcriptional induction. Thus, we provide a new mechanism whereby Peg3 can simultaneously evoke autophagy in endothelial cells and attenuate angiogenesis.
Collapse
Affiliation(s)
- Annabel Torres
- From the Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| | - Maria A Gubbiotti
- From the Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| | - Renato V Iozzo
- From the Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| |
Collapse
|
708
|
Smits NC, Kobayashi T, Srivastava PK, Skopelja S, Ivy JA, Elwood DJ, Stan RV, Tsongalis GJ, Sellke FW, Gross PL, Cole MD, DeVries JT, Kaplan AV, Robb JF, Williams SM, Shworak NW. HS3ST1 genotype regulates antithrombin's inflammomodulatory tone and associates with atherosclerosis. Matrix Biol 2017; 63:69-90. [PMID: 28126521 DOI: 10.1016/j.matbio.2017.01.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 01/19/2017] [Accepted: 01/20/2017] [Indexed: 12/21/2022]
Abstract
The HS3ST1 gene controls endothelial cell production of HSAT+ - a form of heparan sulfate containing a specific pentasaccharide motif that binds the anticoagulant protein antithrombin (AT). HSAT+ has long been thought to act as an endogenous anticoagulant; however, coagulation was normal in Hs3st1-/- mice that have greatly reduced HSAT+ (HajMohammadi et al., 2003). This finding indicates that HSAT+ is not essential for AT's anticoagulant activity. To determine if HSAT+ is involved in AT's poorly understood inflammomodulatory activities, Hs3st1-/- and Hs3st1+/+ mice were subjected to a model of acute septic shock. Compared with Hs3st1+/+ mice, Hs3st1-/- mice were more susceptible to LPS-induced death due to an increased sensitivity to TNF. For Hs3st1+/+ mice, AT treatment reduced LPS-lethality, reduced leukocyte firm adhesion to endothelial cells, and dilated isolated coronary arterioles. Conversely, for Hs3st1-/- mice, AT induced the opposite effects. Thus, in the context of acute inflammation, HSAT+ selectively mediates AT's anti-inflammatory activity; in the absence of HSAT+, AT's pro-inflammatory effects predominate. To explore if the anti-inflammatory action of HSAT+ also protects against a chronic vascular-inflammatory disease, atherosclerosis, we conducted a human candidate-gene association study on >2000 coronary catheterization patients. Bioinformatic analysis of the HS3ST1 gene identified an intronic SNP, rs16881446, in a putative transcriptional regulatory region. The rs16881446G/G genotype independently associated with the severity of coronary artery disease and atherosclerotic cardiovascular events. In primary endothelial cells, the rs16881446G allele associated with reduced HS3ST1 expression. Together with the mouse data, this leads us to conclude that the HS3ST1 gene is required for AT's anti-inflammatory activity that appears to protect against acute and chronic inflammatory disorders.
Collapse
Affiliation(s)
- Nicole C Smits
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Takashi Kobayashi
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Pratyaksh K Srivastava
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Sladjana Skopelja
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Julianne A Ivy
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Dustin J Elwood
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Radu V Stan
- Department of Pathology, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Gregory J Tsongalis
- Department of Pathology, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Frank W Sellke
- Division of Cardiothoracic Surgery, Brown Medical School, Providence, RI, USA
| | - Peter L Gross
- Department of Medicine, Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Michael D Cole
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, NH, USA; Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - James T DeVries
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Aaron V Kaplan
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - John F Robb
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Scott M Williams
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Nicholas W Shworak
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA; Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.
| |
Collapse
|
709
|
Varun D, Srinivasan GR, Tsai YH, Kim HJ, Cutts J, Petty F, Merkley R, Stephanopoulos N, Dolezalova D, Marsala M, Brafman DA. A robust vitronectin-derived peptide for the scalable long-term expansion and neuronal differentiation of human pluripotent stem cell (hPSC)-derived neural progenitor cells (hNPCs). Acta Biomater 2017; 48:120-130. [PMID: 27989923 DOI: 10.1016/j.actbio.2016.10.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 10/03/2016] [Accepted: 10/26/2016] [Indexed: 12/22/2022]
Abstract
Despite therapeutic advances, neurodegenerative diseases and disorders remain some of the leading causes of mortality and morbidity in the United States. Therefore, cell-based therapies to replace lost or damaged neurons and supporting cells of the central nervous system (CNS) are of great therapeutic interest. To that end, human pluripotent stem cell (hPSC) derived neural progenitor cells (hNPCs) and their neuronal derivatives could provide the cellular 'raw material' needed for regenerative medicine therapies for a variety of CNS disorders. In addition, hNPCs derived from patient-specific hPSCs could be used to elucidate the underlying mechanisms of neurodegenerative diseases and identify potential drug candidates. However, the scientific and clinical application of hNPCs requires the development of robust, defined, and scalable substrates for their long-term expansion and neuronal differentiation. In this study, we rationally designed a vitronectin-derived peptide (VDP) that served as an adhesive growth substrate for the long-term expansion of several hNPC lines. Moreover, VDP-coated surfaces allowed for the directed neuronal differentiation of hNPC at levels similar to cells differentiated on traditional extracellular matrix protein-based substrates. Overall, the ability of VDP to support the long-term expansion and directed neuronal differentiation of hNPCs will significantly advance the future translational application of these cells in treating injuries, disorders, and diseases of the CNS.
Collapse
|
710
|
Karamanos NK. Matrix pathobiology—central roles for proteoglycans and heparanase in health and disease. FEBS J 2017; 284:7-9. [DOI: 10.1111/febs.13945] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Nikos K. Karamanos
- Biochemistry LaboratoryBiochemistry, Biochemical Analysis & Matrix Pathobiology Research GroupDepartment of ChemistryUniversity of Patras Greece
| |
Collapse
|
711
|
Ricard-Blum S. Protein–glycosaminoglycan interaction networks: Focus on heparan sulfate. ACTA ACUST UNITED AC 2017. [DOI: 10.1016/j.pisc.2016.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
712
|
Patel VN, Pineda DL, Hoffman MP. The function of heparan sulfate during branching morphogenesis. Matrix Biol 2017; 57-58:311-323. [PMID: 27609403 PMCID: PMC5329135 DOI: 10.1016/j.matbio.2016.09.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 08/18/2016] [Accepted: 09/01/2016] [Indexed: 02/08/2023]
Abstract
Branching morphogenesis is a fundamental process in the development of diverse epithelial organs such as the lung, kidney, liver, pancreas, prostate, salivary, lacrimal and mammary glands. A unifying theme during organogenesis is the importance of epithelial cell interactions with the extracellular matrix (ECM) and growth factors (GFs). The diverse developmental mechanisms giving rise to these epithelial organs involve many organ-specific GFs, but a unifying paradigm during organogenesis is the regulation of GF activity by heparan sulfates (HS) on the cell surface and in the ECM. This primarily involves the interactions of GFs with the sulfated side-chains of HS proteoglycans. HS is one of the most diverse biopolymers and modulates GF binding and signaling at the cell surface and in the ECM of all tissues. Here, we review what is known about how HS regulates branching morphogenesis of epithelial organs with emphasis on the developing salivary gland, which is a classic model to investigate epithelial-ECM interactions. We also address the structure, biosynthesis, turnover and function of HS during organogenesis. Understanding the regulatory mechanisms that control HS dynamics may aid in the development of therapeutic interventions for diseases and novel strategies for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Vaishali N Patel
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, United States
| | - Dallas L Pineda
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, United States
| | - Matthew P Hoffman
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, United States.
| |
Collapse
|
713
|
Sethi MK, Zaia J. Extracellular matrix proteomics in schizophrenia and Alzheimer's disease. Anal Bioanal Chem 2017; 409:379-394. [PMID: 27601046 PMCID: PMC5203946 DOI: 10.1007/s00216-016-9900-6] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/16/2016] [Accepted: 08/23/2016] [Indexed: 12/17/2022]
Abstract
Brain extracellular matrix (ECM) is a highly organized system that consists of collagens, noncollagenous proteins, glycoproteins, hyaluronan, and proteoglycans. Recognized physiological roles of ECM include developmental regulation, tissue homeostasis, cell migration, cell proliferation, cell differentiation, neuronal plasticity, and neurite outgrowth. Aberrant ECM structure is associated with brain neurodegenerative conditions. This review focuses on two neurodegenerative conditions, schizophrenia and Alzheimer's disease, and summarizes recent findings of altered ECM components, including proteoglycans, glycosaminoglycans, proteins, and glycoproteins, and proteins and genes related to other brain components. The scope includes immunohistochemical, genomics, transcriptomics, proteomics, and glycomics studies, and a critical assessment of current state of proteomic studies for neurodegenerative disorders. The intent is to summarize the ECM molecular alterations associated with neurodegenerative pathophysiology. Graphical Abstract Brain extracellular matrix showing HSPGs, CSPGs, HA, collagens, and other glycoproteins.
Collapse
Affiliation(s)
- Manveen K Sethi
- Center for Biomedical Mass Spectrometry, Department of Biochemistry, Cell Biology & Genomics, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Joseph Zaia
- Center for Biomedical Mass Spectrometry, Department of Biochemistry, Cell Biology & Genomics, Boston University School of Medicine, Boston, MA, 02118, USA.
| |
Collapse
|
714
|
Rosenbloom J, Macarak E, Piera-Velazquez S, Jimenez SA. Human Fibrotic Diseases: Current Challenges in Fibrosis Research. Methods Mol Biol 2017; 1627:1-23. [PMID: 28836191 DOI: 10.1007/978-1-4939-7113-8_1] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Human fibrotic diseases constitute a major health problem worldwide owing to the large number of affected individuals, the incomplete knowledge of the fibrotic process pathogenesis, the marked heterogeneity in their etiology and clinical manifestations, the absence of appropriate and fully validated biomarkers, and, most importantly, the current void of effective disease-modifying therapeutic agents. The fibrotic disorders encompass a wide spectrum of clinical entities including systemic fibrotic diseases such as systemic sclerosis (SSc), sclerodermatous graft vs. host disease, and nephrogenic systemic fibrosis, as well as numerous organ-specific disorders including radiation-induced fibrosis and cardiac, pulmonary, liver, and kidney fibrosis. Although their causative mechanisms are quite diverse and in several instances have remained elusive, these diseases share the common feature of an uncontrolled and progressive accumulation of fibrotic tissue in affected organs causing their dysfunction and ultimate failure. Despite the remarkable heterogeneity in the etiologic mechanisms responsible for the development of fibrotic diseases and in their clinical manifestations, numerous studies have identified activated myofibroblasts as the common cellular element ultimately responsible for the replacement of normal tissues with nonfunctional fibrotic tissue. Critical signaling cascades, initiated primarily by transforming growth factor-β (TGF-β), but also involving numerous cytokines and signaling molecules which stimulate profibrotic reactions in myofibroblasts, offer potential therapeutic targets. Here, we briefly review the current knowledge of the molecular mechanisms involved in the development of tissue fibrosis and point out some of the most important challenges to research in the fibrotic diseases and to the development of effective therapeutic approaches for this often fatal group of disorders. Efforts to further clarify the complex pathogenetic mechanisms of the fibrotic process should be encouraged to attain the elusive goal of developing effective therapies for these serious, untreatable, and often fatal disorders.
Collapse
Affiliation(s)
- Joel Rosenbloom
- The Joan and Joel Rosenbloom Center for Fibrotic Diseases and The Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Edward Macarak
- The Joan and Joel Rosenbloom Center for Fibrotic Diseases and The Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sonsoles Piera-Velazquez
- The Joan and Joel Rosenbloom Center for Fibrotic Diseases and The Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sergio A Jimenez
- The Joan and Joel Rosenbloom Center for Fibrotic Diseases and The Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
715
|
Schaefer L, Tredup C, Gubbiotti MA, Iozzo RV. Proteoglycan neofunctions: regulation of inflammation and autophagy in cancer biology. FEBS J 2017; 284:10-26. [PMID: 27860287 PMCID: PMC5226885 DOI: 10.1111/febs.13963] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 10/27/2016] [Accepted: 11/11/2016] [Indexed: 12/18/2022]
Abstract
Inflammation and autophagy have emerged as prominent issues in the context of proteoglycan signaling. In particular, two small, leucine-rich proteoglycans, biglycan and decorin, play pivotal roles in the regulation of these vital cellular pathways and, as such, are intrinsically involved in cancer initiation and progression. In this minireview, we will address novel functions of biglycan and decorin in inflammation and autophagy, and analyze new emerging signaling events triggered by these proteoglycans, which directly or indirectly modulate these processes. We will critically discuss the dual role of proteoglycan-driven inflammation and autophagy in tumor biology, and delineate the potential mechanisms through which soluble extracellular matrix constituents affect the microenvironment associated with inflammatory and neoplastic diseases.
Collapse
Affiliation(s)
- Liliana Schaefer
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Claudia Tredup
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Maria A. Gubbiotti
- Department of Pathology, Anatomy and Cell Biology, and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, USA
| | - Renato V. Iozzo
- Department of Pathology, Anatomy and Cell Biology, and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, USA
| |
Collapse
|
716
|
Rämisch S, Pramhed A, Tillgren V, Aspberg A, Logan DT. Crystal structure of human chondroadherin: solving a difficult molecular-replacement problem usingde novomodels. ACTA CRYSTALLOGRAPHICA SECTION D-STRUCTURAL BIOLOGY 2017; 73:53-63. [DOI: 10.1107/s205979831601980x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 12/12/2016] [Indexed: 02/08/2023]
Abstract
Chondroadherin (CHAD) is a cartilage matrix protein that mediates the adhesion of isolated chondrocytes. Its protein core is composed of 11 leucine-rich repeats (LRR) flanked by cysteine-rich domains. CHAD makes important interactions with collagen as well as with cell-surface heparin sulfate proteoglycans and α2β1integrins. The integrin-binding site is located in a region of hitherto unknown structure at the C-terminal end of CHAD. Peptides based on the C-terminal human CHAD (hCHAD) sequence have shown therapeutic potential for treating osteoporosis. This article describes a still-unconventional structure solution by phasing withde novomodels, the first of a β-rich protein. Structure determination of hCHAD using traditional, though nonsystematic, molecular replacement was unsuccessful in the hands of the authors, possibly owing to a combination of low sequence identity to other LRR proteins, four copies in the asymmetric unit and weak translational pseudosymmetry. However, it was possible to solve the structure by generating a large number ofde novomodels for the central LRR domain usingRosettaand multiple parallel molecular-replacement attempts usingAMPLE. The hCHAD structure reveals an ordered C-terminal domain belonging to the LRRCT fold, with the integrin-binding motif (WLEAK) being part of a regular α-helix, and suggests ways in which experimental therapeutic peptides can be improved. The crystal structure itself and docking simulations further support that hCHAD dimers form in a similar manner to other matrix LRR proteins.
Collapse
|
717
|
Abstract
Basement membranes are delicate, nanoscale and pliable sheets of extracellular matrices that often act as linings or partitions in organisms. Previously considered as passive scaffolds segregating polarized cells, such as epithelial or endothelial cells, from the underlying mesenchyme, basement membranes have now reached the center stage of biology. They play a multitude of roles from blood filtration to muscle homeostasis, from storing growth factors and cytokines to controlling angiogenesis and tumor growth, from maintaining skin integrity and neuromuscular structure to affecting adipogenesis and fibrosis. Here, we will address developmental, structural and biochemical aspects of basement membranes and discuss some of the pathogenetic mechanisms causing diseases linked to abnormal basement membranes.
Collapse
Affiliation(s)
- Ambra Pozzi
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University, Nashville, TN, United States; Veterans Affairs Hospitals, Nashville, TN, United States.
| | - Peter D Yurchenco
- Department of Pathology and Laboratory Medicine, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - Renato V Iozzo
- Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
718
|
Kechagia M, Papassotiriou I, Gourgoulianis KI. Endocan and the respiratory system: a review. Int J Chron Obstruct Pulmon Dis 2016; 11:3179-3187. [PMID: 28003744 PMCID: PMC5161333 DOI: 10.2147/copd.s118692] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Endocan, formerly called endothelial cell-specific molecule 1, is an endothelial cell-associated proteoglycan that is preferentially expressed by renal and pulmonary endothelium. It is upregulated by proangiogenic molecules as well as by pro-inflammatory cytokines, and since it reflects endothelial activation and dysfunction, it is regarded as a novel tissue and blood-based relevant biomarker. As such, it is increasingly being researched and evaluated in a wide spectrum of healthy and disease pathophysiological processes. Here, we review the present scientific knowledge on endocan, with emphasis on the evidence that underlines its possible clinical value as a prognostic marker in several malignant, inflammatory and obstructive disorders of the respiratory system.
Collapse
Affiliation(s)
- Maria Kechagia
- Respiratory Medicine Department, University of Thessaly Medical School, Larissa
- Department of Clinical Biochemistry, Aghia Sophia Children’s Hospital, Athens, Greece
| | - Ioannis Papassotiriou
- Department of Clinical Biochemistry, Aghia Sophia Children’s Hospital, Athens, Greece
| | | |
Collapse
|
719
|
Piperigkou Z, Bouris P, Onisto M, Franchi M, Kletsas D, Theocharis AD, Karamanos NK. Estrogen receptor beta modulates breast cancer cells functional properties, signaling and expression of matrix molecules. Matrix Biol 2016; 56:4-23. [DOI: 10.1016/j.matbio.2016.05.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 05/08/2016] [Accepted: 05/09/2016] [Indexed: 02/07/2023]
|
720
|
Silk biomaterials functionalized with recombinant domain V of human perlecan modulate endothelial cell and platelet interactions for vascular applications. Colloids Surf B Biointerfaces 2016; 148:130-138. [DOI: 10.1016/j.colsurfb.2016.08.039] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/21/2016] [Accepted: 08/22/2016] [Indexed: 11/21/2022]
|
721
|
Voudouri K, Nikitovic D, Berdiaki A, Kletsas D, Karamanos NK, Tzanakakis GN. IGF-I/EGF and E2 signaling crosstalk through IGF-IR conduit point affects breast cancer cell adhesion. Matrix Biol 2016; 56:95-113. [DOI: 10.1016/j.matbio.2016.06.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 06/22/2016] [Accepted: 06/22/2016] [Indexed: 12/17/2022]
|
722
|
Neill T, Buraschi S, Goyal A, Sharpe C, Natkanski E, Schaefer L, Morrione A, Iozzo RV. EphA2 is a functional receptor for the growth factor progranulin. J Cell Biol 2016; 215:687-703. [PMID: 27903606 PMCID: PMC5146997 DOI: 10.1083/jcb.201603079] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 08/12/2016] [Accepted: 10/19/2016] [Indexed: 01/03/2023] Open
Abstract
The receptor for the growth factor progranulin has remained unclear. Neill et al. show that the Ephrin receptor tyrosine kinase EphA2 is a functional signaling receptor for progranulin and mediates its effects in capillary morphogenesis and autoregulation. Although the growth factor progranulin was discovered more than two decades ago, the functional receptor remains elusive. Here, we discovered that EphA2, a member of the large family of Ephrin receptor tyrosine kinases, is a functional signaling receptor for progranulin. Recombinant progranulin bound with high affinity to EphA2 in both solid phase and solution. Interaction of progranulin with EphA2 caused prolonged activation of the receptor, downstream stimulation of mitogen-activated protein kinase and Akt, and promotion of capillary morphogenesis. Furthermore, we found an autoregulatory mechanism of progranulin whereby a feed-forward loop occurred in an EphA2-dependent manner that was independent of the endocytic receptor sortilin. The discovery of a functional signaling receptor for progranulin offers a new avenue for understanding the underlying mode of action of progranulin in cancer progression, tumor angiogenesis, and perhaps neurodegenerative diseases.
Collapse
Affiliation(s)
- Thomas Neill
- Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107.,Cancer Cell Biology and Signaling Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107
| | - Simone Buraschi
- Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107.,Cancer Cell Biology and Signaling Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107
| | - Atul Goyal
- Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107.,Cancer Cell Biology and Signaling Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107
| | - Catherine Sharpe
- Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107.,Cancer Cell Biology and Signaling Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107
| | - Elizabeth Natkanski
- Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107.,Cancer Cell Biology and Signaling Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107
| | - Liliana Schaefer
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt am Main 60323, Germany
| | - Andrea Morrione
- Department of Urology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107.,Biology of Prostate Cancer Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107
| | - Renato V Iozzo
- Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107 .,Cancer Cell Biology and Signaling Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107
| |
Collapse
|
723
|
García B, Merayo-Lloves J, Rodríguez D, Alcalde I, García-Suárez O, Alfonso JF, Baamonde B, Fernández-Vega A, Vazquez F, Quirós LM. Different Use of Cell Surface Glycosaminoglycans As Adherence Receptors to Corneal Cells by Gram Positive and Gram Negative Pathogens. Front Cell Infect Microbiol 2016; 6:173. [PMID: 27965938 PMCID: PMC5127826 DOI: 10.3389/fcimb.2016.00173] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 11/17/2016] [Indexed: 12/13/2022] Open
Abstract
The epithelium of the cornea is continuously exposed to pathogens, and adhesion to epithelial cells is regarded as an essential first step in bacterial pathogenesis. In this article, the involvement of glycosaminoglycans in the adhesion of various pathogenic bacteria to corneal epithelial cells is analyzed. All microorganisms use glycosaminoglycans as receptors, but arranged in different patterns depending on the Gram-type of the bacterium. The heparan sulfate chains of syndecans are the main receptors, though other molecular species also seem to be involved, particularly in Gram-negative bacteria. Adherence is inhibited differentially by peptides, including heparin binding sequences, indicating the participation of various groups of Gram-positive, and -negative adhesins. The length of the saccharides produces a major effect, and low molecular weight chains inhibit the binding of Gram-negative microorganisms but increase the adherence of Gram-positives. Pathogen adhesion appears to occur preferentially through sulfated domains, and is very dependent on N- and 6-O-sulfation of the glucosamine residue and, to a lesser extent, 2-O sulfation of uronic acid. These data show the differential use of corneal receptors, which could facilitate the development of new anti-infective strategies.
Collapse
Affiliation(s)
- Beatriz García
- Ophthalmology, Vision Sciences and Advanced Therapies Research Group, Instituto Universitario Fernández-Vega, Universidad de OviedoOviedo, Spain; Departmento de Biología Funcional, Universidad de OviedoOviedo, Spain
| | - Jesús Merayo-Lloves
- Ophthalmology, Vision Sciences and Advanced Therapies Research Group, Instituto Universitario Fernández-Vega, Universidad de Oviedo Oviedo, Spain
| | - David Rodríguez
- Departamento de Bioquímica y Biología Molecular, Universidad de OviedoOviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias, Universidad de OviedoOviedo, Spain
| | - Ignacio Alcalde
- Ophthalmology, Vision Sciences and Advanced Therapies Research Group, Instituto Universitario Fernández-Vega, Universidad de Oviedo Oviedo, Spain
| | - Olivia García-Suárez
- Ophthalmology, Vision Sciences and Advanced Therapies Research Group, Instituto Universitario Fernández-Vega, Universidad de OviedoOviedo, Spain; Departmento de Morfología y Biología Celular, Universidad de OviedoOviedo, Spain
| | - José F Alfonso
- Fundación de Investigación Oftalmológica, Instituto Oftalmológico Fernández-Vega Oviedo, Spain
| | - Begoña Baamonde
- Ophthalmology, Vision Sciences and Advanced Therapies Research Group, Instituto Universitario Fernández-Vega, Universidad de Oviedo Oviedo, Spain
| | - Andrés Fernández-Vega
- Fundación de Investigación Oftalmológica, Instituto Oftalmológico Fernández-Vega Oviedo, Spain
| | - Fernando Vazquez
- Ophthalmology, Vision Sciences and Advanced Therapies Research Group, Instituto Universitario Fernández-Vega, Universidad de OviedoOviedo, Spain; Departmento de Biología Funcional, Universidad de OviedoOviedo, Spain; Departmento de Microbiología, Hospital Universitario Central de AsturiasOviedo, Spain
| | - Luis M Quirós
- Ophthalmology, Vision Sciences and Advanced Therapies Research Group, Instituto Universitario Fernández-Vega, Universidad de OviedoOviedo, Spain; Departmento de Biología Funcional, Universidad de OviedoOviedo, Spain
| |
Collapse
|
724
|
Ricard-Blum S, Lisacek F. Glycosaminoglycanomics: where we are. Glycoconj J 2016; 34:339-349. [PMID: 27900575 DOI: 10.1007/s10719-016-9747-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 10/28/2016] [Accepted: 11/01/2016] [Indexed: 01/21/2023]
Abstract
Glycosaminoglycans regulate numerous physiopathological processes such as development, angiogenesis, innate immunity, cancer and neurodegenerative diseases. Cell surface GAGs are involved in cell-cell and cell-matrix interactions, cell adhesion and signaling, and host-pathogen interactions. GAGs contribute to the assembly of the extracellular matrix and heparan sulfate chains are able to sequester growth factors in the ECM. Their biological activities are regulated by their interactions with proteins. The structural heterogeneity of GAGs, mostly due to chemical modifications occurring during and after their synthesis, makes the development of analytical techniques for their profiling in cells, tissues, and biological fluids, and of computational tools for mining GAG-protein interaction data very challenging. We give here an overview of the experimental approaches used in glycosaminoglycomics, of the major GAG-protein interactomes characterized so far, and of the computational tools and databases available to analyze and store GAG structures and interactions.
Collapse
Affiliation(s)
- Sylvie Ricard-Blum
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, UMR 5246 CNRS - Université Lyon 1, INSA Lyon, CPE Lyon, 69622, Villeurbanne Cedex, France.
| | - Frédérique Lisacek
- SIB Swiss Institute of Bioinformatics, 1 Rue Michel-Servet, 1211, Geneva, Switzerland.,Computer Science Department, University of Geneva, Geneva, Switzerland
| |
Collapse
|
725
|
Kobayashi T, Kakizaki I, Nozaka H, Nakamura T. Chondroitin sulfate proteoglycans from salmon nasal cartilage inhibit angiogenesis. Biochem Biophys Rep 2016; 9:72-78. [PMID: 28955991 PMCID: PMC5614546 DOI: 10.1016/j.bbrep.2016.11.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 09/23/2016] [Accepted: 11/14/2016] [Indexed: 12/26/2022] Open
Abstract
Because cartilage lacks nerves, blood vessels, and lymphatic vessels, it is thought to contain factors that inhibit the growth and development of those tissues. Chondroitin sulfate proteoglycans (CSPGs) are a major extracellular component in cartilage. CSPGs contribute to joint flexibility and regulate extracellular signaling via their attached glycosaminoglycan, chondroitin sulfate (CS). CS and CSPG inhibit axonal regeneration; however, their role in blood vessel formation is largely unknown. To clarify the function of CSPG in blood vessel formation, we tested salmon nasal cartilage proteoglycan (PG), a member of the aggrecan family of CSPG, for endothelial capillary-like tube formation. Treatment with salmon PG inhibited endothelial cell adhesion and in vitro tube formation. The anti-angiogenic activity was derived from CS in the salmon PG but not the core protein. Salmon PG also reduced matrix metalloproteinase expression and inhibited angiogenesis in the chick chorioallantoic membrane. All of these data support an anti-angiogenic role for CSPG in cartilage. The role of CSPGs in blood vessel formation in cartilage is largely unknown. Treatment of salmon PG inhibited in vitro and in vivo angiogenesis. The CS portion of salmon PG was responsible for the anti-angiogenic activity. Salmon PG also reduced MMP expression and inhibited cell adhesion. Our results support an anti-angiogenic role for CSPG in cartilage.
Collapse
Key Words
- Aggrecan
- Angiogenesis
- BME, basement membrane extract
- BSA, bovine serum albumin
- CAM, chorioallantoic membrane
- CS, chondroitin sulfate
- CSPG, chondroitin sulfate proteoglycan
- Chondroitin sulfate proteoglycan
- ECM, extracellular matrix
- FAK, focal adhesion kinase
- FBS, fetal bovine serum
- GAG, glycosaminoglycan
- GAPDH, glyceraldehyde-3-phosphate dehydrogenase
- GalNAc, N-acetylgalactosamine
- GdnHCl, guanidine hydrochloride
- GlcUA, glucuronic acid
- Glycosaminoglycan
- HSPG, heparan sulfate proteoglycan
- KSPG, keratin sulfate proteoglycan
- MMP, matrix metalloproteinase
- Matrix metalloproteinase
- OA, osteoarthritis
- PBS, phosphate-buffered saline
- PG, proteoglycan
- UA, uronic acid
- Vascular endothelial cell
Collapse
Affiliation(s)
- Takashi Kobayashi
- Department of Glycotechnology, Center for Advanced Medical Research, Graduate School of Medicine, Hirosaki University, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan.,Departments of Bioscience and Laboratory Medicine, Hirosaki University Graduate School of Health Sciences, 66-1 Hon-cho, Hirosaki, Aomori 036-8564, Japan
| | - Ikuko Kakizaki
- Department of Glycotechnology, Center for Advanced Medical Research, Graduate School of Medicine, Hirosaki University, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Hiroyuki Nozaka
- Departments of Bioscience and Laboratory Medicine, Hirosaki University Graduate School of Health Sciences, 66-1 Hon-cho, Hirosaki, Aomori 036-8564, Japan
| | - Toshiya Nakamura
- Departments of Bioscience and Laboratory Medicine, Hirosaki University Graduate School of Health Sciences, 66-1 Hon-cho, Hirosaki, Aomori 036-8564, Japan
| |
Collapse
|
726
|
Pulsipher A, Qin X, Thomas AJ, Prestwich GD, Oottamasathien S, Alt JA. Prevention of sinonasal inflammation by a synthetic glycosaminoglycan. Int Forum Allergy Rhinol 2016; 7:177-184. [PMID: 27863138 DOI: 10.1002/alr.21865] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 09/12/2016] [Accepted: 09/22/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Glycosaminoglycans (GAGs) are polysaccharides that are distributed on respiratory epithelial cells, endothelial cells, and submucosal glands. Uniquely positioned, certain GAGs exhibit anti-inflammatory properties in respiratory diseases and serve important roles in repairing mucosal surfaces and modulating mucociliary clearance. We hypothesized that topical administration of a synthetic GAG (GM-0111) would prevent sinonasal inflammation in a mouse model of rhinosinusitis (RS). METHODS To test our hypothesis, C57BL/6 mice were intranasally administered fluorescent GM-0111, and sinonasal tissues were examined for coating and penetration ability. To test therapeutic feasibility, mice (n = 6) were given GM-0111 or hyaluronic acid (HA; 800 μg dose) prior to inducing RS with inflammatory molecule LL-37 (115 μg dose). After 24 hours, sinonasal tissues were harvested for histological and biochemical analysis of inflammatory markers (inflammatory cell infiltration, lamina propria [LP] thickening, and neutrophil enzyme myeloperoxidase [MPO]) and cell death. RESULTS GM-0111 was observed within sinonasal tissues 1 hour and 24 hours after intranasal administration, indicating rapid and effective coating and penetration. GM-0111 prevented sinonasal tissues from developing inflammatory changes, with significant reductions in mast cell infiltration (p < 0.05), LP thickening (p < 0.001), and MPO levels (p < 0.01) when compared to tissues treated with LL-37 and those pretreated with HA. GM-0111 reduced cell death within sinonasal tissues in contrast to LL-37-treated tissues. CONCLUSION We report a new synthetic GAG (GM-0111) that uniformly coats and penetrates into the sinonasal mucosa to prevent sinonasal inflammation and cell death in a mouse model of RS.
Collapse
Affiliation(s)
- Abigail Pulsipher
- Division of Head and Neck Surgery, Rhinology-Sinus and Skull Base Surgery Program, Department of Surgery; University of Utah School of Medicine, Salt Lake City, UT.,GlycoMira Therapeutics, Inc., Salt Lake City, UT
| | - Xuan Qin
- Division of Head and Neck Surgery, Rhinology-Sinus and Skull Base Surgery Program, Department of Surgery; University of Utah School of Medicine, Salt Lake City, UT
| | - Andrew J Thomas
- Division of Head and Neck Surgery, Rhinology-Sinus and Skull Base Surgery Program, Department of Surgery; University of Utah School of Medicine, Salt Lake City, UT
| | - Glenn D Prestwich
- Department of Medicinal Chemistry and Center for Therapeutic Biomaterials, Salt Lake City, UT.,GlycoMira Therapeutics, Inc., Salt Lake City, UT
| | - Siam Oottamasathien
- Department of Medicinal Chemistry and Center for Therapeutic Biomaterials, Salt Lake City, UT.,Division of Pediatric Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT
| | - Jeremiah A Alt
- Division of Head and Neck Surgery, Rhinology-Sinus and Skull Base Surgery Program, Department of Surgery; University of Utah School of Medicine, Salt Lake City, UT.,Department of Medicinal Chemistry and Center for Therapeutic Biomaterials, Salt Lake City, UT
| |
Collapse
|
727
|
Extracellular Matrix, a Hard Player in Angiogenesis. Int J Mol Sci 2016; 17:ijms17111822. [PMID: 27809279 PMCID: PMC5133823 DOI: 10.3390/ijms17111822] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 09/30/2016] [Accepted: 10/21/2016] [Indexed: 12/11/2022] Open
Abstract
The extracellular matrix (ECM) is a complex network of proteins, glycoproteins, proteoglycans, and polysaccharides. Through multiple interactions with each other and the cell surface receptors, not only the ECM determines the physical and mechanical properties of the tissues, but also profoundly influences cell behavior and many physiological and pathological processes. One of the functions that have been extensively explored is its impingement on angiogenesis. The strong impact of the ECM in this context is both direct and indirect by virtue of its ability to interact and/or store several growth factors and cytokines. The aim of this review is to provide some examples of the complex molecular mechanisms that are elicited by these molecules in promoting or weakening the angiogenic processes. The scenario is intricate, since matrix remodeling often generates fragments displaying opposite effects compared to those exerted by the whole molecules. Thus, the balance will tilt towards angiogenesis or angiostasis depending on the relative expression of pro- or anti-angiogenetic molecules/fragments composing the matrix of a given tissue. One of the vital aspects of this field of research is that, for its endogenous nature, the ECM can be viewed as a reservoir to draw from for the development of new more efficacious therapies to treat angiogenesis-dependent pathologies.
Collapse
|
728
|
Afratis NA, Karamanou K, Piperigkou Z, Vynios DH, Theocharis AD. The role of heparins and nano-heparins as therapeutic tool in breast cancer. Glycoconj J 2016; 34:299-307. [PMID: 27778131 DOI: 10.1007/s10719-016-9742-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 10/05/2016] [Accepted: 10/10/2016] [Indexed: 01/04/2023]
Abstract
Glycosaminoglycans are integral part of the dynamic extracellular matrix (ECM) network that control crucial biochemical and biomechanical signals required for tissue morphogenesis, differentiation, homeostasis and cancer development. Breast cancer cells communicate with stromal ones to modulate ECM mainly through release of soluble effectors during cancer progression. The intracellular cross-talk between cell surface receptors and estrogen receptors is important for the regulation of breast cancer cell properties and production of ECM molecules. In turn, reorganized ECM-cell surface interface modulates signaling cascades, which regulate almost all aspects of breast cell behavior. Heparan sulfate chains present on cell surface and matrix proteoglycans are involved in regulation of breast cancer functions since they are capable of binding numerous matrix molecules, growth factors and inflammatory mediators thus modulating their signaling. In addition to its anticoagulant activity, there is accumulating evidence highlighting various anticancer activities of heparin and nano-heparin derivatives in numerous types of cancer. Importantly, heparin derivatives significantly reduce breast cancer cell proliferation and metastasis in vitro and in vivo models as well as regulates the expression profile of major ECM macromolecules, providing strong evidence for therapeutic targeting. Nano-formulations of the glycosaminoglycan heparin are possibly novel tools for targeting tumor microenvironment. In this review, the role of heparan sulfate/heparin and its nano-formulations in breast cancer biology are presented and discussed in terms of future pharmacological targeting.
Collapse
Affiliation(s)
- Nikos A Afratis
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26110, Patras, Greece
| | - Konstantina Karamanou
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26110, Patras, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26110, Patras, Greece
| | - Demitrios H Vynios
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26110, Patras, Greece
| | - Achilleas D Theocharis
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26110, Patras, Greece.
| |
Collapse
|
729
|
Wu QH, Ma Y, Ruan CC, Yang Y, Liu XH, Ge Q, Kong LR, Zhang JW, Yan C, Gao PJ. Loss of osteoglycin promotes angiogenesis in limb ischaemia mouse models via modulation of vascular endothelial growth factor and vascular endothelial growth factor receptor 2 signalling pathway. Cardiovasc Res 2016; 113:70-80. [DOI: 10.1093/cvr/cvw220] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 08/29/2016] [Accepted: 09/21/2016] [Indexed: 01/22/2023] Open
|
730
|
Viola M, Brüggemann K, Karousou E, Caon I, Caravà E, Vigetti D, Greve B, Stock C, De Luca G, Passi A, Götte M. MDA-MB-231 breast cancer cell viability, motility and matrix adhesion are regulated by a complex interplay of heparan sulfate, chondroitin-/dermatan sulfate and hyaluronan biosynthesis. Glycoconj J 2016; 34:411-420. [PMID: 27744520 DOI: 10.1007/s10719-016-9735-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 09/23/2016] [Accepted: 09/28/2016] [Indexed: 10/20/2022]
Abstract
Proteoglycans and glycosaminoglycans modulate numerous cellular processes relevant to tumour progression, including cell proliferation, cell-matrix interactions, cell motility and invasive growth. Among the glycosaminoglycans with a well-documented role in tumour progression are heparan sulphate, chondroitin/dermatan sulphate and hyaluronic acid/hyaluronan. While the mode of biosynthesis differs for sulphated glycosaminoglycans, which are synthesised in the ER and Golgi compartments, and hyaluronan, which is synthesized at the plasma membrane, these polysaccharides partially compete for common substrates. In this study, we employed a siRNA knockdown approach for heparan sulphate (EXT1) and heparan/chondroitin/dermatan sulphate-biosynthetic enzymes (β4GalT7) in the aggressive human breast cancer cell line MDA-MB-231 to study the impact on cell behaviour and hyaluronan biosynthesis. Knockdown of β4GalT7 expression resulted in a decrease in cell viability, motility and adhesion to fibronectin, while these parameters were unchanged in EXT1-silenced cells. Importantly, these changes were associated with a decreased expression of syndecan-1, decreased signalling response to HGF and an increase in the synthesis of hyaluronan, due to an upregulation of the hyaluronan synthases HAS2 and HAS3. Interestingly, EXT1-depleted cells showed a downregulation of the UDP-sugar transporter SLC35D1, whereas SLC35D2 was downregulated in β4GalT7-depleted cells, indicating an intricate regulatory network that connects all glycosaminoglycans synthesis. The results of our in vitro study suggest that a modulation of breast cancer cell behaviour via interference with heparan sulphate biosynthesis may result in a compensatory upregulation of hyaluronan biosynthesis. These findings have important implications for the development of glycosaminoglycan-targeted therapeutic approaches for malignant diseases.
Collapse
Affiliation(s)
- Manuela Viola
- Department of Surgical and Morphological Sciences, University of Insubria, Varese, Italy.
| | - Kathrin Brüggemann
- Department of Gynaecology and Obstetrics, Muenster University Hospital, Muenster, Germany
| | - Evgenia Karousou
- Department of Surgical and Morphological Sciences, University of Insubria, Varese, Italy
| | - Ilaria Caon
- Department of Surgical and Morphological Sciences, University of Insubria, Varese, Italy
| | - Elena Caravà
- Department of Surgical and Morphological Sciences, University of Insubria, Varese, Italy
| | - Davide Vigetti
- Department of Surgical and Morphological Sciences, University of Insubria, Varese, Italy
| | - Burkhard Greve
- Department of Radiotherapy-Radiooncology, University Hospital Muenster, Muenster, Germany
| | - Christian Stock
- Institute of Physiology II, University of Muenster, Muenster, Germany.,Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Giancarlo De Luca
- Department of Surgical and Morphological Sciences, University of Insubria, Varese, Italy
| | - Alberto Passi
- Department of Surgical and Morphological Sciences, University of Insubria, Varese, Italy.
| | - Martin Götte
- Department of Gynaecology and Obstetrics, Muenster University Hospital, Muenster, Germany
| |
Collapse
|
731
|
Roles and targeting of the HAS/hyaluronan/CD44 molecular system in cancer. Matrix Biol 2016; 59:3-22. [PMID: 27746219 DOI: 10.1016/j.matbio.2016.10.001] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 10/02/2016] [Accepted: 10/03/2016] [Indexed: 02/07/2023]
Abstract
Synthesis, deposition, and interactions of hyaluronan (HA) with its cellular receptor CD44 are crucial events that regulate the onset and progression of tumors. The intracellular signaling pathways initiated by HA interactions with CD44 leading to tumorigenic responses are complex. Moreover, HA molecules may perform dual functions depending on their concentration and size. Overexpression of variant isoforms of CD44 (CD44v) is most commonly linked to cancer progression, whereas their loss is associated with inhibition of tumor growth. In this review, we highlight that the regulation of HA synthases (HASes) by post-translational modifications, such as O-GlcNAcylation and ubiquitination, environmental factors and the action of microRNAs is important for HA synthesis and secretion in the tumor microenvironment. Moreover, we focus on the roles and interactions of CD44 with various proteins that reside extra- and intracellularly, as well as on cellular membranes with particular reference to the CD44-HA axis in cancer stem cell functions, and the importance of CD44/CD44v6 targeting to inhibit tumorigenesis.
Collapse
|
732
|
Heljasvaara R, Aikio M, Ruotsalainen H, Pihlajaniemi T. Collagen XVIII in tissue homeostasis and dysregulation - Lessons learned from model organisms and human patients. Matrix Biol 2016; 57-58:55-75. [PMID: 27746220 DOI: 10.1016/j.matbio.2016.10.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 09/12/2016] [Accepted: 10/10/2016] [Indexed: 12/13/2022]
Abstract
Collagen XVIII is a ubiquitous basement membrane (BM) proteoglycan produced in three tissue-specific isoforms that differ in their N-terminal non-collagenous sequences, but share collagenous and C-terminal non-collagenous domains. The collagenous domain provides flexibility to the large collagen XVIII molecules on account of multiple interruptions in collagenous sequences. Each isoform has a complex multi-domain structure that endows it with an ability to perform various biological functions. The long isoform contains a frizzled-like (Fz) domain with Wnt-inhibiting activity and a unique domain of unknown function (DUF959), which is also present in the medium isoform. All three isoforms share an N-terminal laminin-G-like/thrombospondin-1 sequence whose specific functions still remain unconfirmed. The proteoglycan nature of the isoforms further increases the functional diversity of collagen XVIII. An anti-angiogenic domain termed endostatin resides in the C-terminus of collagen XVIII and is proteolytically cleaved from the parental molecule during the BM breakdown for example in the process of tumour progression. Recombinant endostatin can efficiently reduce tumour angiogenesis and growth in experimental models by inhibiting endothelial cell migration and proliferation or by inducing their death, but its efficacy against human cancers is still a subject of debate. Mutations in the COL18A1 gene result in Knobloch syndrome, a genetic disorder characterised mainly by severe eye defects and encephalocele and, occasionally, other symptoms. Studies with gene-modified mice have elucidated some aspects of this rare disease, highlighting in particular the importance of collagen XVIII in the development of the eye. Research with model organisms have also helped in determining other structural and biological functions of collagen XVIII, such as its requirement in the maintenance of BM integrity and its emerging roles in regulating cell survival, stem or progenitor cell maintenance and differentiation and inflammation. In this review, we summarise current knowledge on the properties and endogenous functions of collagen XVIII in normal situations and tissue dysregulation. When data is available, we discuss the functions of the distinct isoforms and their specific domains.
Collapse
Affiliation(s)
- Ritva Heljasvaara
- Oulu Center for Cell-Matrix Research, Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, University of Oulu, FIN-90014 Oulu, Finland; Centre for Cancer Biomarkers CCBIO, Department of Biomedicine, University of Bergen, N-5009 Bergen, Norway.
| | - Mari Aikio
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Heli Ruotsalainen
- Oulu Center for Cell-Matrix Research, Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, University of Oulu, FIN-90014 Oulu, Finland
| | - Taina Pihlajaniemi
- Oulu Center for Cell-Matrix Research, Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, University of Oulu, FIN-90014 Oulu, Finland
| |
Collapse
|
733
|
Koo H, Yamada KM. Dynamic cell-matrix interactions modulate microbial biofilm and tissue 3D microenvironments. Curr Opin Cell Biol 2016; 42:102-112. [PMID: 27257751 PMCID: PMC5064909 DOI: 10.1016/j.ceb.2016.05.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 05/07/2016] [Accepted: 05/10/2016] [Indexed: 01/22/2023]
Abstract
Microbial biofilms and most eukaryotic tissues consist of cells embedded in a three-dimensional extracellular matrix. This matrix serves as a scaffold for cell adhesion and a dynamic milieu that provides varying chemical and physical signals to the cells. Besides a vast array of specific molecular components, an extracellular matrix can provide locally heterogeneous microenvironments differing in porosity/diffusion, stiffness, pH, oxygen and metabolites or nutrient levels. Mechanisms of matrix formation, mechanosensing, matrix remodeling, and modulation of cell-cell or cell-matrix interactions and dispersal are being revealed. This perspective article aims to identify such concepts from the fields of biofilm or eukaryotic matrix biology relevant to the other field to help stimulate new questions, approaches, and insights.
Collapse
Affiliation(s)
- Hyun Koo
- Biofilm Research Labs, Levy Center for Oral Health, Department of Orthodontics and Divisions of Pediatric Dentistry & Community Oral Health, School of Dental Medicine, University of Pennsylvania, PA 19104, USA.
| | - Kenneth M Yamada
- Laboratory of Cell and Developmental Biology, Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
734
|
Abstract
The vasculature is essential for proper organ function. Many pathologies are directly and indirectly related to vascular dysfunction, which causes significant morbidity and mortality. A common pathophysiological feature of diseased vessels is extracellular matrix (ECM) remodelling. Analysing the protein composition of the ECM by conventional antibody-based techniques is challenging; alternative splicing or post-translational modifications, such as glycosylation, can mask epitopes required for antibody recognition. By contrast, proteomic analysis by mass spectrometry enables the study of proteins without the constraints of antibodies. Recent advances in proteomic techniques make it feasible to characterize the composition of the vascular ECM and its remodelling in disease. These developments may lead to the discovery of novel prognostic and diagnostic markers. Thus, proteomics holds potential for identifying ECM signatures to monitor vascular disease processes. Furthermore, a better understanding of the ECM remodelling processes in the vasculature might make ECM-associated proteins more attractive targets for drug discovery efforts. In this review, we will summarize the role of the ECM in the vasculature. Then, we will describe the challenges associated with studying the intricate network of ECM proteins and the current proteomic strategies to analyse the vascular ECM in metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- M Lynch
- King's British Heart Foundation Centre, King's College London, London, UK
| | | | | | - M Mayr
- King's British Heart Foundation Centre, King's College London, London, UK.
| |
Collapse
|
735
|
Gubbiotti MA, Vallet SD, Ricard-Blum S, Iozzo RV. Decorin interacting network: A comprehensive analysis of decorin-binding partners and their versatile functions. Matrix Biol 2016; 55:7-21. [PMID: 27693454 DOI: 10.1016/j.matbio.2016.09.009] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Decorin, a prototype small leucine-rich proteoglycan, regulates a vast array of cellular processes including collagen fibrillogenesis, wound repair, angiostasis, tumor growth, and autophagy. This functional versatility arises from a wide array of decorin/protein interactions also including interactions with its single glycosaminoglycan side chain. The decorin-binding partners encompass numerous categories ranging from extracellular matrix molecules to cell surface receptors to growth factors and enzymes. Despite the diversity of the decorin interacting network, two main roles emerge as prominent themes in decorin function: maintenance of cellular structure and outside-in signaling, culminating in anti-tumorigenic effects. Here we present contemporary knowledge regarding the decorin interacting network and discuss in detail the biological relevance of these pleiotropic interactions, some of which could be targeted by therapeutic interventions.
Collapse
Affiliation(s)
- Maria A Gubbiotti
- Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Sylvain D Vallet
- Pericellular and Extracellular Supramolecular Assemblies, Institute of Molecular and Supramolecular Chemistry and Biochemistry, University Claude Bernard, Lyon, France
| | - Sylvie Ricard-Blum
- Pericellular and Extracellular Supramolecular Assemblies, Institute of Molecular and Supramolecular Chemistry and Biochemistry, University Claude Bernard, Lyon, France
| | - Renato V Iozzo
- Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States.
| |
Collapse
|
736
|
Jan AT, Lee EJ, Choi I. Fibromodulin: A regulatory molecule maintaining cellular architecture for normal cellular function. Int J Biochem Cell Biol 2016; 80:66-70. [PMID: 27693429 DOI: 10.1016/j.biocel.2016.09.023] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 09/23/2016] [Accepted: 09/28/2016] [Indexed: 01/02/2023]
Abstract
Fibromodulin (FMOD) is a small leucine-rich proteoglycan that plays roles in a series of biological and pathophysiological processes. The interaction between FMOD and lysyl oxidase (LOX; collagen cross-linking enzyme) helps regulate extracellular matrix composition, and thereby, provides a permissive environment for regulating cellular turnover. FMOD has been mostly studied in the context of matrix component assembly, but during recent years its association with muscle development, cell reprogramming, and the angiogenic program have demonstrated its activities well beyond extracellular matrix maintenance. In fact, the involvement of FMOD in these cellular processes places it the centrum of cellular behaviour and ultimately of tissue properties. Thus, a clear view of the impact FMOD has on tissue integrity would aid its exploitation for tissue modelling and in the treatment of different disorders.
Collapse
Affiliation(s)
- Arif Tasleem Jan
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, Republic of Korea.
| |
Collapse
|
737
|
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: 2.6] [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
|
738
|
Tillgren V, Mörgelin M, Önnerfjord P, Kalamajski S, Aspberg A. The Tyrosine Sulfate Domain of Fibromodulin Binds Collagen and Enhances Fibril Formation. J Biol Chem 2016; 291:23744-23755. [PMID: 27634037 DOI: 10.1074/jbc.m116.730325] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Indexed: 11/06/2022] Open
Abstract
Small leucine-rich proteoglycans interact with other extracellular matrix proteins and are important regulators of matrix assembly. Fibromodulin has a key role in connective tissues, binding collagen through two identified binding sites in its leucine-rich repeat domain and regulating collagen fibril formation in vitro and in vivo Some nine tyrosine residues in the fibromodulin N-terminal domain are O-sulfated, a posttranslational modification often involved in protein interactions. The N-terminal domain mimics heparin, binding proteins with clustered basic amino acid residues. Because heparin affects collagen fibril formation, we investigated whether tyrosine sulfate is involved in fibromodulin interactions with collagen. Using full-length fibromodulin and its N-terminal tyrosine-sulfated domain purified from tissue, as well as recombinant fibromodulin fragments, we found that the N-terminal domain binds collagen. The tyrosine-sulfated domain and the leucine-rich repeat domain both bound to three specific sites along the collagen type I molecule, at the N terminus and at 100 and 220 nm from the N terminus. The N-terminal domain shortened the collagen fibril formation lag phase and tyrosine sulfation was required for this effect. The isolated leucine-rich repeat domain inhibited the fibril formation rate, and full-length fibromodulin showed a combination of these effects. The fibrils formed in the presence of fibromodulin or its fragments showed more organized structure. Fibromodulin and its tyrosine sulfate domain remained bound on the formed fiber. Taken together, this suggests a novel, regulatory function for tyrosine sulfation in collagen interaction and control of fibril formation.
Collapse
Affiliation(s)
- Viveka Tillgren
- From the Departments of Clinical Sciences Lund, Rheumatology and Molecular Skeletal Biology BMC-C12 and
| | - Matthias Mörgelin
- Clinical Sciences Lund, Division of Infection Medicine (BMC) BMC-B14, Lund University, SE-22184 Lund, Sweden
| | - Patrik Önnerfjord
- From the Departments of Clinical Sciences Lund, Rheumatology and Molecular Skeletal Biology BMC-C12 and
| | - Sebastian Kalamajski
- From the Departments of Clinical Sciences Lund, Rheumatology and Molecular Skeletal Biology BMC-C12 and
| | - Anders Aspberg
- From the Departments of Clinical Sciences Lund, Rheumatology and Molecular Skeletal Biology BMC-C12 and
| |
Collapse
|
739
|
Gubbiotti MA, Neill T, Iozzo RV. A current view of perlecan in physiology and pathology: A mosaic of functions. Matrix Biol 2016; 57-58:285-298. [PMID: 27613501 DOI: 10.1016/j.matbio.2016.09.003] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 09/01/2016] [Indexed: 01/06/2023]
Abstract
Perlecan, a large basement membrane heparan sulfate proteoglycan, is expressed in a wide array of tissues where it regulates diverse cellular processes including bone formation, inflammation, cardiac development, and angiogenesis. Here we provide a contemporary review germane to the biology of perlecan encompassing its genetic regulation as well as an analysis of its modular protein structure as it pertains to function. As perlecan signaling from the extracellular matrix converges on master regulators of autophagy, including AMPK and mTOR, via a specific interaction with vascular endothelial growth factor receptor 2, we specifically focus on the mechanism of action of perlecan in autophagy and angiogenesis and contrast the role of endorepellin, the C-terminal fragment of perlecan, in these cellular and morphogenic events.
Collapse
Affiliation(s)
- Maria A Gubbiotti
- Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Thomas Neill
- Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Renato V Iozzo
- Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States.
| |
Collapse
|
740
|
Frey H, Moreth K, Hsieh LTH, Zeng-Brouwers J, Rathkolb B, Fuchs H, Gailus-Durner V, Iozzo RV, de Angelis MH, Schaefer L. A novel biological function of soluble biglycan: Induction of erythropoietin production and polycythemia. Glycoconj J 2016; 34:393-404. [DOI: 10.1007/s10719-016-9722-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 07/21/2016] [Accepted: 08/05/2016] [Indexed: 11/29/2022]
|
741
|
Borza DB. Glomerular basement membrane heparan sulfate in health and disease: A regulator of local complement activation. Matrix Biol 2016; 57-58:299-310. [PMID: 27609404 DOI: 10.1016/j.matbio.2016.09.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/30/2016] [Accepted: 09/01/2016] [Indexed: 12/26/2022]
Abstract
The glomerular basement membrane (GBM) is an essential component of the glomerular filtration barrier. Heparan sulfate proteoglycans such as agrin are major components of the GBM, along with α345(IV) collagen, laminin-521 and nidogen. A loss of GBM heparan sulfate chains is associated with proteinuria in several glomerular diseases and may contribute to the underlying pathology. As the major determinants of the anionic charge of the GBM, heparan sulfate chains have been thought to impart charge selectivity to the glomerular filtration, a view challenged by the negligible albuminuria in mice that lack heparan sulfate in the GBM. Recent studies provide increasing evidence that heparan sulfate chains modulate local complement activation by recruiting complement regulatory protein factor H, the major inhibitor of the alternative pathway in plasma. Factor H selectively inactivates C3b bound to surfaces bearing host-specific polyanions such as heparan sulfate, thus limiting complement activation on self surfaces such as the GBM, which are not protected by cell-bound complement regulators. We discuss mechanisms whereby the acquired loss of GBM heparan sulfate can impair the local regulation of the alternative pathway, exacerbating complement activation and glomerular injury in immune-mediated kidney diseases such as membranous nephropathy and lupus nephritis.
Collapse
Affiliation(s)
- Dorin-Bogdan Borza
- Department of Microbiology and Immunology, Meharry Medical College, 1005 Dr. D. B. Todd, Jr., Blvd., Nashville, TN 37208, USA.
| |
Collapse
|
742
|
Selective demineralisation of dentine extrafibrillar minerals—A potential method to eliminate water-wet bonding in the etch-and-rinse technique. J Dent 2016; 52:55-62. [DOI: 10.1016/j.jdent.2016.07.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 07/14/2016] [Accepted: 07/16/2016] [Indexed: 12/12/2022] Open
|
743
|
Li C, Wang LX. Endoglycosidases for the Synthesis of Polysaccharides and Glycoconjugates. Adv Carbohydr Chem Biochem 2016; 73:73-116. [PMID: 27816108 DOI: 10.1016/bs.accb.2016.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Recent advances in glycobiology have implicated essential roles of oligosaccharides and glycoconjugates in many important biological recognition processes, including intracellular signaling, cell adhesion, cell differentiation, cancer progression, host-pathogen interactions, and immune responses. A detailed understanding of the biological functions, as well as the development of carbohydrate-based therapeutics, often requires structurally well-defined oligosaccharides and glycoconjugates, which are usually difficult to isolate in pure form from natural sources. To meet with this urgent need, chemical and chemoenzymatic synthesis has become increasingly important as the major means to provide homogeneous compounds for functional glycocomics studies and for drug/vaccine development. Chemoenzymatic synthesis, an approach that combines chemical synthesis and enzymatic manipulations, is often the method of choice for constructing complex oligosaccharides and glycoconjugates that are otherwise difficult to achieve by purely chemical synthesis. Among these, endoglycosidases, a class of glycosidases that hydrolyze internal glycosidic bonds in glycoconjugates and polysaccharides, are emerging as a very attractive class of enzymes for synthetic purposes, due to their transglycosylation activity and their capability of transferring oligosaccharide units en bloc in a single step, in contrast to the limitation of monosaccharide transfers by common glycosyltransferases. In this chapter, we provide an overview on the application of endoglycosidases for the synthesis of complex carbohydrates, including oligosaccharides, polysaccharides, glycoproteins, glycolipids, proteoglycans, and other biologically relevant polysaccharides. The scope, limitation, and future directions of endoglycosidase-catalyzed synthesis are discussed.
Collapse
Affiliation(s)
- Chao Li
- University of Maryland, College Park, MD, United States
| | - Lai-Xi Wang
- University of Maryland, College Park, MD, United States
| |
Collapse
|
744
|
Maioral GCCC, Gomes RCT, Verna C, Simões MDJ, Nader HB, Simões RS, Baracat EC, Soares JM. Concentration of glycosaminoglycan in ovariectomized mice uterus after treatment with ovarian steroids. Gynecol Endocrinol 2016; 32:617-621. [PMID: 26899437 DOI: 10.3109/09513590.2016.1147027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The aim of this study was to evaluate the amount of non- and sulfated glycosaminoglycans in the ovariectomized mice uterus, after treatment with ovarian steroids. For this purpose, 50 adult female mice were divided into five groups with 10 animals/each: control group: CG (ovary intact), and ovariectomized groups: OG (vehicle), EG (estradiol), PG (progesterone) and EPG (estradiol combined to progesterone). The treatments started 30 days after ovariectomy. All the animals were treated for 50 consecutive days. These hormones were administered in a sterile oily solution via gavage. Twenty-four hours after the last treatment, all animals were euthanized, removing the uterine horn for biochemical analyses. To quantify, the hyaluronic acid (HA) used ELISA-like fluorometric assay, and the sulfated glycosaminoglycans (GAGs) used agarose gel electrophoresis. The amount of HA was significantly higher in the group treated with progesterone (PG) compared to the others groups (p < 0.05), and in the group treated with estradiol (EG), the amount of chondroitin/dermatan sulfate was significantly higher compared to the others groups (p < 0.05), and in the group treated with progesterone (PG), the amount of heparan sulfate was significantly lower compared to the others groups, except to control group (p < 0.05). Our results showed that the estroprogestative therapy after long time (50 days) profoundly affected the amount of glycosaminoglycans in uterine. These changes may be indicative of uterine pathology such as the development of tumor.
Collapse
Affiliation(s)
| | - Regina Célia T Gomes
- a Department of Gynecology , Universidade Federal de São Paulo , Brazil
- b Department of Morphology and Genetics , Universidade Federal de São Paulo , Brazil
| | - Carina Verna
- a Department of Gynecology , Universidade Federal de São Paulo , Brazil
| | - Manuel de J Simões
- a Department of Gynecology , Universidade Federal de São Paulo , Brazil
- b Department of Morphology and Genetics , Universidade Federal de São Paulo , Brazil
| | - Helena B Nader
- d Department of Biochemistry , Molecular Biology Division, Universidade Federal de São Paulo , Brazil
| | - Ricardo S Simões
- c Department of Obstetrics and Gynecology , Gynecology Division, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo , Brazil , and
| | - Edmund C Baracat
- c Department of Obstetrics and Gynecology , Gynecology Division, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo , Brazil , and
| | - José Maria Soares
- c Department of Obstetrics and Gynecology , Gynecology Division, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo , Brazil , and
| |
Collapse
|
745
|
Taylan F, Costantini A, Coles N, Pekkinen M, Héon E, Şıklar Z, Berberoğlu M, Kämpe A, Kıykım E, Grigelioniene G, Tüysüz B, Mäkitie O. Spondyloocular Syndrome: Novel Mutations in XYLT2 Gene and Expansion of the Phenotypic Spectrum. J Bone Miner Res 2016; 31:1577-85. [PMID: 26987875 DOI: 10.1002/jbmr.2834] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Revised: 03/06/2016] [Accepted: 03/10/2016] [Indexed: 01/08/2023]
Abstract
Spondyloocular syndrome is an autosomal-recessive disorder with spinal compression fractures, osteoporosis, and cataract. Mutations in XYLT2, encoding isoform of xylosyltransferase, were recently identified as the cause of the syndrome. We report on 4 patients, 2 unrelated patients and 2 siblings, with spondyloocular syndrome and novel mutations in XYLT2. Exome sequencing revealed a homozygous nonsense mutation, NM_022167.3(XYLT2): c.2188C>T, resulting in a premature stop codon (p.Arg730*) in a female patient. The patient presents visual impairment, generalized osteoporosis, short stature with short trunk, spinal compression fractures, and increased intervertebral disc space and hearing loss. We extended our XYLT2 analysis to a cohort of 22 patients with generalized osteoporosis, mostly from consanguineous families. In this cohort, we found by Sanger sequencing 2 siblings and 1 single patient who were homozygous for missense mutations in the XYLT2 gene (p.Arg563Gly and p.Leu605Pro). The patients had osteoporosis, compression fractures, cataracts, and hearing loss. Bisphosphonate treatment in 1 patient resulted in almost complete normalization of vertebral structures by adolescence, whereas treatment response in the others was variable. This report together with a previous study shows that mutations in the XYLT2 gene result in a variable phenotype dominated by spinal osteoporosis, cataract, and hearing loss. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Fulya Taylan
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Alice Costantini
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Nicole Coles
- Department of Pediatric Endocrinology of Metabolism, The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | | | - Elise Héon
- Department of Pediatric Endocrinology of Metabolism, The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Zeynep Şıklar
- Department of Pediatric Endocrinology, School of Medicine, Ankara University, Ankara, Turkey
| | - Merih Berberoğlu
- Department of Pediatric Endocrinology, School of Medicine, Ankara University, Ankara, Turkey
| | - Anders Kämpe
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ertuğrul Kıykım
- Department of Pediatric Metabolism, Cerrahpasa Medicine School, Istanbul University, Istanbul, Turkey
| | - Giedre Grigelioniene
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Beyhan Tüysüz
- Department of Pediatric Genetics, Cerrahpasa Medicine School, Istanbul University, Istanbul, Turkey
| | - Outi Mäkitie
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Folkhälsan Institute of Genetics, Helsinki, Finland
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
746
|
Mikami S, Oya M, Mizuno R, Kosaka T, Ishida M, Kuroda N, Nagashima Y, Katsube KI, Okada Y. Recent advances in renal cell carcinoma from a pathological point of view. Pathol Int 2016; 66:481-90. [PMID: 27461942 DOI: 10.1111/pin.12433] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 06/09/2016] [Accepted: 06/16/2016] [Indexed: 12/20/2022]
Abstract
The purpose of this article is to review the recent advances in renal cell carcinoma (RCC) from a pathological point of view. Because the genetic features and morphological characteristics have become major criteria for the classification of RCC, special techniques, such as immunohistochemistry, are essential to the differential diagnosis of renal tumors. Metastasis is frequently observed among the RCC patients with curative nephrectomy, and extracellular matrix-degrading enzymes, such as matrix metalloproteinases (MMP) and heparanase, play a key role in invasion and metastasis of RCC. Snail and Slug, transcription factors of epithelial-mesenchymal transition (EMT), accelerate cancer cell invasion through downregulation of E-cadherin and up-regulation of MMP. Therapies targeted at the vascular endothelial growth factor pathway have become the standard treatment of metastatic RCC. Although they lead to tumor shrinkage mainly by inhibiting angiogenesis, they have typically been associated with drug resistance. The mechanism of the resistance remains largely unknown, but complex events including re-activation of angiogenesis, EMT and cancer stem cells, and immune escape are implicated in the refractory response to the therapy. Recent advances of the research on RCC have caused the changes of classification and therapy, and pathologists should take overall view of these as integrated pathology.
Collapse
Affiliation(s)
- Shuji Mikami
- Division of Diagnostic Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Ryuichi Mizuno
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Takeo Kosaka
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Masaru Ishida
- Department of Urology, Keio University School of Medicine, Tokyo, Japan.,Department of Urology, Saiseikai Tobu Hospital, Yokohama, Japan
| | - Naoto Kuroda
- Department of Diagnostic Pathology, Kochi Red Cross Hospital, Kochi, Japan
| | - Yoji Nagashima
- Department of Surgical Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | | | - Yasunori Okada
- Department of Pathophysiology for Locomotive and Neoplastic Diseases, Juntendo University Graduate School of Medicine Tokyo, Japan
| |
Collapse
|
747
|
Goyal A, Gubbiotti MA, Chery DR, Han L, Iozzo RV. Endorepellin-evoked Autophagy Contributes to Angiostasis. J Biol Chem 2016; 291:19245-56. [PMID: 27435676 DOI: 10.1074/jbc.m116.740266] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Indexed: 12/27/2022] Open
Abstract
Endorepellin, the C-terminal domain of perlecan, is an angiostatic molecule that acts as a potent inducer of autophagy via its interaction with VEGFR2. In this study, we examined the effect of endorepellin on endothelial cells using atomic force microscopy. Soluble endorepellin caused morphological and biophysical changes such as an increase in cell surface roughness and cell height. Surprisingly, these changes were not accompanied by alterations in the endothelial cell elastic modulus. We discovered that endorepellin-induced autophagic flux led to co-localization of mammalian target of rapamycin with LC3-positive autophagosomes. Endorepellin functioned upstream of AMP-activated kinase α, as compound C, an inhibitor of AMP-activated kinase α, abrogated endorepellin-mediated activation and co-localization of Beclin 1 and LC3, thereby reducing autophagic progression. Functionally, we discovered that both endorepellin and Torin 1, a canonical autophagic inducer, blunted ex vivo angiogenesis. We conclude that autophagy is a novel mechanism by which endorepellin promotes angiostasis independent of nutrient deprivation.
Collapse
Affiliation(s)
- Atul Goyal
- From the Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107 and
| | - Maria A Gubbiotti
- From the Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107 and
| | - Daphney R Chery
- the School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104
| | - Lin Han
- the School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104
| | - Renato V Iozzo
- From the Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107 and
| |
Collapse
|
748
|
Gronau T, Krüger K, Prein C, Aszodi A, Gronau I, Iozzo RV, Mooren FC, Clausen-Schaumann H, Bertrand J, Pap T, Bruckner P, Dreier R. Forced exercise-induced osteoarthritis is attenuated in mice lacking the small leucine-rich proteoglycan decorin. Ann Rheum Dis 2016; 76:442-449. [PMID: 27377816 DOI: 10.1136/annrheumdis-2016-209319] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 06/05/2016] [Accepted: 06/14/2016] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Interterritorial regions of articular cartilage matrix are rich in decorin, a small leucine-rich proteoglycan and important structural protein, also involved in many signalling events. Decorin sequesters transforming growth factor β (TGFβ), thereby regulating its activity. Here, we analysed whether increased bioavailability of TGFβ in decorin-deficient (Dcn-/-) cartilage leads to changes in biomechanical properties and resistance to osteoarthritis (OA). METHODS Unchallenged knee cartilage was analysed by atomic force microscopy (AFM) and immunohistochemistry. Active transforming growth factor β-1 (TGFβ1) content within cultured chondrocyte supernatants was measured by ELISA. Quantitative real-time (RT)-PCR was used to analyse mRNA expression of glycosaminoglycan (GAG)-modifying enzymes in C28/I2 cells following TGFβ1 treatment. In addition, OA was induced in Dcn-/- and wild-type (WT) mice via forced exercise on a treadmill. RESULTS AFM analysis revealed a strikingly higher compressive stiffness in Dcn-/- than in WT cartilage. This was accompanied by increased negative charge and enhanced sulfation of GAG chains, but not by alterations in the levels of collagens or proteoglycan core proteins. In addition, decorin-deficient chondrocytes were shown to release more active TGFβ1. Increased TGFβ signalling led to enhanced Chst11 sulfotransferase expression inducing an increased negative charge density of cartilage matrix. These negative charges might attract more water resulting in augmented compressive stiffness of the tissue. Therefore, decorin-deficient mice developed significantly less OA after forced exercise than WT mice. CONCLUSIONS Our study demonstrates that the disruption of decorin-restricted TGFβ signalling leads to higher stiffness of articular cartilage matrix, rendering joints more resistant to OA. Therefore, the loss of an important structural component can improve cartilage homeostasis.
Collapse
Affiliation(s)
- Tobias Gronau
- Institute of Experimental Musculoskeletal Medicine, University Hospital Münster, Münster, Germany.,Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | - Karsten Krüger
- Institute of Sports Medicine, Justus-Liebig University Giessen, Giessen, Germany
| | - Carina Prein
- Center for Applied Tissue Engineering and Regenerative Medicine (CANTER), Munich University of Applied Sciences and Center for Nanoscience (CeNS), Munich, Germany
| | - Attila Aszodi
- Laboratory of Experimental Surgery and Regenerative Medicine, Department of General, Trauma and Reconstruction Surgery, Ludwig-Maximilians-University, Munich, Germany
| | - Isabel Gronau
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | - Renato V Iozzo
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, USA
| | - Frank C Mooren
- Institute of Sports Medicine, Justus-Liebig University Giessen, Giessen, Germany
| | - Hauke Clausen-Schaumann
- Center for Applied Tissue Engineering and Regenerative Medicine (CANTER), Munich University of Applied Sciences and Center for Nanoscience (CeNS), Munich, Germany
| | - Jessica Bertrand
- Institute of Experimental Musculoskeletal Medicine, University Hospital Münster, Münster, Germany.,Department of Orthopaedic Surgery, Otto-von-Guericke University, Magdeburg, Germany
| | - Thomas Pap
- Institute of Experimental Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - Peter Bruckner
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | - Rita Dreier
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| |
Collapse
|
749
|
Shed proteoglycans in tumor stroma. Cell Tissue Res 2016; 365:643-55. [DOI: 10.1007/s00441-016-2452-4] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 06/08/2016] [Indexed: 12/12/2022]
|
750
|
Tiku ML, Madhan B. Preserving the longevity of long-lived type II collagen and its implication for cartilage therapeutics. Ageing Res Rev 2016; 28:62-71. [PMID: 27133944 DOI: 10.1016/j.arr.2016.04.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 04/26/2016] [Indexed: 11/30/2022]
Abstract
Human life expectancy has been steadily increasing at a rapid rate, but this increasing life span also brings about increases in diseases, dementia, and disability. A global burden of disease 2010 study revealed that hip and knee osteoarthritis ranked the 11th highest in terms of years lived with disability. Wear and tear can greatly influence the quality of life during ageing. In particular, wear and tear of the articular cartilage have adverse effects on joints and result in osteoarthritis. The articular cartilage uses longevity of type II collagen as the foundation around which turnover of proteoglycans and the homeostatic activity of chondrocytes play central roles thereby maintaining the function of articular cartilage in the ageing. The longevity of type II collagen involves a complex interaction of the scaffolding needs of the cartilage and its biochemical, structural and mechanical characteristics. The covalent cross-linking of heterotypic polymers of collagens type II, type IX and type XI hold together cartilage, allowing it to withstand ageing stresses. Discerning the biological clues in the armamentarium for preserving cartilage appears to be collagen cross-linking. Therapeutic methods to crosslink in in-vivo are non-existent. However intra-articular injections of polyphenols in vivo stabilize the cartilage and make it resistant to degradation, opening a new therapeutic possibility for prevention and intervention of cartilage degradation in osteoarthritis of aging.
Collapse
Affiliation(s)
- Moti L Tiku
- Rutgers, Robert Wood Johnson Medical School, New Brunswick, NJ, USA.
| | - Balaraman Madhan
- Council of Scientific and Industrial Research - Central Leather Research Institute, Adyar, Chennai, Tamil Nadu, India
| |
Collapse
|