1
|
Yan Y, Quan H, Guo C, Qin Z, Quan T. Alterations of Matrisome Gene Expression in Naturally Aged and Photoaged Human Skin In Vivo. Biomolecules 2024; 14:900. [PMID: 39199288 PMCID: PMC11352887 DOI: 10.3390/biom14080900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/26/2024] [Accepted: 07/23/2024] [Indexed: 09/01/2024] Open
Abstract
The main component of human skin is a collagen-rich extracellular matrix (ECM), known as the matrisome. The matrisome is essential for maintaining the structural integrity and mechanical properties of the skin. Recently, we reported notable decreases in matrisome proteins in natural aging and photoaging human skin. This study aims to investigate the mRNA expression of the core matrisome proteins in human skin, comparing young versus aged and sun-protected versus sun-exposed skin by quantitative real-time PCR and immunostaining. Our findings reveal a notable decrease in core matrisome transcription in aged skin. The mRNA expression of the core matrisome, such as collagen 1A1 (COL1A1), decorin, and dermatopontin, is significantly reduced in aged skin compared to its young skin. Yet, the majority of collagen mRNA expression levels of aged sun-exposed skin are similar to those found in young sun-exposed skin. This discrepancy is primarily attributable to a substantial decrease in collagen transcription in young sun-exposed skin, suggesting early molecular changes in matrisome transcription due to sun exposure, which preceded the emergence of clinical signs of photoaging. These findings shed light on the mRNA transcript profile of major matrisome proteins and their alterations in naturally aged and photoaged human skin, offering valuable insights into skin matrisome biology.
Collapse
Affiliation(s)
- Yan Yan
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (Y.Y.); (C.G.); (Z.Q.)
| | - Hehui Quan
- Lenox Hill Hospital, 100 E 77th St., New York, NY 10075, USA;
| | - Chunfang Guo
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (Y.Y.); (C.G.); (Z.Q.)
| | - Zhaoping Qin
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (Y.Y.); (C.G.); (Z.Q.)
| | - Taihao Quan
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (Y.Y.); (C.G.); (Z.Q.)
| |
Collapse
|
2
|
Yurchenco PD, Kulczyk AW. Polymerizing laminins in development, health, and disease. J Biol Chem 2024; 300:107429. [PMID: 38825010 PMCID: PMC11260871 DOI: 10.1016/j.jbc.2024.107429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/12/2024] [Accepted: 05/26/2024] [Indexed: 06/04/2024] Open
Abstract
Polymerizing laminins are multi-domain basement membrane (BM) glycoproteins that self-assemble into cell-anchored planar lattices to establish the initial BM scaffold. Nidogens, collagen-IV and proteoglycans then bind to the scaffold at different domain loci to create a mature BM. The LN domains of adjacent laminins bind to each other to form a polymer node, while the LG domains attach to cytoskeletal-anchoring integrins and dystroglycan, as well as to sulfatides and heparan sulfates. The polymer node, the repeating unit of the polymer scaffold, is organized into a near-symmetrical triskelion. The structure, recently solved by cryo-electron microscopy in combination with AlphaFold2 modeling and biochemical studies, reveals how the LN surface residues interact with each other and how mutations cause failures of self-assembly in an emerging group of diseases, the LN-lamininopathies, that include LAMA2-related dystrophy and Pierson syndrome.
Collapse
Affiliation(s)
- Peter D Yurchenco
- Department of Pathology & Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA.
| | - Arkadiusz W Kulczyk
- Department of Biochemistry and Microbiology, Institute for Quantitative Biomedicine, Rutgers University, Piscataway, New Jersey, USA
| |
Collapse
|
3
|
Diao B, Sun C, Yu P, Zhao Z, Yang P. LAMA5 promotes cell proliferation and migration in ovarian cancer by activating Notch signaling pathway. FASEB J 2023; 37:e23109. [PMID: 37527216 DOI: 10.1096/fj.202300306r] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 06/18/2023] [Accepted: 07/12/2023] [Indexed: 08/03/2023]
Abstract
LAMA5 (laminin α5) is a member of the laminin family. Despite the recent research implicating LAMA5 in cancer, the function of LAMA5 has remained uncertain in the progression of ovarian cancer (OC). Here, we investigated the functional influences of LAMA5 knockdown on OC in vitro and in vivo. In this study, we used immunohistochemistry (IHC) analysis to detect the relative expression of LAMA5 in OC and non-cancer tissues, and we analyzed its connection with the overall survival (OS) of OC patients. To prove the role of LAMA5 in cell proliferation, migration, and invasion, LAMA5 expression in OC cell lines was inhibited by lentivirus. Compared with normal fallopian tube tissue, epithelial ovarian cancer (EOC) tissue showed critically higher LAMA5 expression levels; additionally, high LAMA5 levels were a poor predictor of OS. We found that cell progression was restrained in LAMA5-knockdown OC cell lines in vivo and in vitro. Finally, LAMA5 might be a commanding inducer of the expression of epithelial-mesenchymal transition (EMT) and Notch signaling pathway-related markers. Together, our research indicates that LAMA5 is highly connected to OC progression as it may play a role in the EMT process through the Notch signaling pathway.
Collapse
Affiliation(s)
- Bowen Diao
- Department of Gynecology, First Affiliated Hospital, Shihezi University, Shihezi, China
- The NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University, Shihezi, China
| | - Chongfeng Sun
- Department of Gynecology, First Affiliated Hospital, Shihezi University, Shihezi, China
- The NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University, Shihezi, China
| | - Panpan Yu
- Department of Gynecology, First Affiliated Hospital, Shihezi University, Shihezi, China
- The NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University, Shihezi, China
| | - Zouyu Zhao
- Department of Gynecology, First Affiliated Hospital, Shihezi University, Shihezi, China
- The NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University, Shihezi, China
| | - Ping Yang
- Department of Gynecology, First Affiliated Hospital, Shihezi University, Shihezi, China
- The NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University, Shihezi, China
| |
Collapse
|
4
|
Nirwane A, Yao Y. Cell-specific expression and function of laminin at the neurovascular unit. J Cereb Blood Flow Metab 2022; 42:1979-1999. [PMID: 35796497 PMCID: PMC9580165 DOI: 10.1177/0271678x221113027] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/08/2022] [Accepted: 06/19/2022] [Indexed: 11/17/2022]
Abstract
Laminin, a major component of the basal lamina (BL), is a heterotrimeric protein with many isoforms. In the CNS, laminin is expressed by almost all cell types, yet different cells synthesize distinct laminin isoforms. By binding to its receptors, laminin exerts a wide variety of important functions. However, due to the reciprocal and cell-specific expression of laminin in different cells at the neurovascular unit, its functions in blood-brain barrier (BBB) maintenance and BBB repair after injury are not fully understood. In this review, we focus on the expression and functions of laminin and its receptors in the neurovascular unit under both physiological and pathological conditions. We first briefly introduce the structures of laminin and its receptors. Next, the expression and functions of laminin and its receptors in the CNS are summarized in a cell-specific manner. Finally, we identify the knowledge gap in the field and discuss key questions that need to be answered in the future. Our goal is to provide a comprehensive overview on cell-specific expression of laminin and its receptors in the CNS and their functions on BBB integrity.
Collapse
Affiliation(s)
- Abhijit Nirwane
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Yao Yao
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
5
|
Falcone S, Nicol T, Blease A, Randles MJ, Angus E, Page A, Tam FWK, Pusey CD, Lennon R, Potter PK. A novel model of nephrotic syndrome results from a point mutation in Lama5 and is modified by genetic background. Kidney Int 2022; 101:527-540. [PMID: 34774562 PMCID: PMC8883398 DOI: 10.1016/j.kint.2021.10.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/22/2021] [Accepted: 10/18/2021] [Indexed: 11/22/2022]
Abstract
Nephrotic syndrome is characterized by severe proteinuria, hypoalbuminaemia, edema and hyperlipidaemia. Genetic studies of nephrotic syndrome have led to the identification of proteins playing a crucial role in slit diaphragm signaling, regulation of actin cytoskeleton dynamics and cell-matrix interactions. The laminin α5 chain is essential for embryonic development and, in association with laminin β2 and laminin γ1, is a major component of the glomerular basement membrane, a critical component of the glomerular filtration barrier. Mutations in LAMA5 were recently identified in children with nephrotic syndrome. Here, we have identified a novel missense mutation (E884G) in the uncharacterized L4a domain of LAMA5 where homozygous mice develop nephrotic syndrome with severe proteinuria with histological and ultrastructural changes in the glomerulus mimicking the progression seen in most patients. The levels of LAMA5 are reduced in vivo and the assembly of the laminin 521 heterotrimer significantly reduced in vitro. Proteomic analysis of the glomerular extracellular fraction revealed changes in the matrix composition. Importantly, the genetic background of the mice had a significant effect on aspects of disease progression from proteinuria to changes in podocyte morphology. Thus, our novel model will provide insights into pathologic mechanisms of nephrotic syndrome and pathways that influence the response to a dysfunctional glomerular basement membrane that may be important in a range of kidney diseases.
Collapse
Affiliation(s)
- Sara Falcone
- Mammalian Genetics Unit, Medical Research Council Harwell Institute, Harwell Campus, Oxfordshire, UK; Centre for Cellular and Molecular Physiology, University of Oxford, Oxford, UK
| | - Thomas Nicol
- Mammalian Genetics Unit, Medical Research Council Harwell Institute, Harwell Campus, Oxfordshire, UK; British Heart Foundation, Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Andrew Blease
- Mammalian Genetics Unit, Medical Research Council Harwell Institute, Harwell Campus, Oxfordshire, UK
| | - Michael J Randles
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Elizabeth Angus
- Biomedical Imaging Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Anton Page
- Biomedical Imaging Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Frederick W K Tam
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Charles D Pusey
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Rachel Lennon
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Paul K Potter
- Mammalian Genetics Unit, Medical Research Council Harwell Institute, Harwell Campus, Oxfordshire, UK; Department Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, UK.
| |
Collapse
|
6
|
Lee S, Lee DS, Jang JH. Recombinant laminin α5 LG1-3 domains support the stemness of human mesenchymal stem cells. Exp Ther Med 2020; 21:166. [PMID: 33456533 DOI: 10.3892/etm.2020.9597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 11/24/2020] [Indexed: 11/06/2022] Open
Abstract
The extracellular matrix components laminin and elastin serve key roles in stem cell therapy. Elastin-like polypeptides (ELPs), derived from a soluble form of elastin, affect the proliferation and differentiation of various types of cells. In the present study, a novel protein was designed containing globular domains 1-3 of laminin α5 (Lα5LG1-3) fused to ELPs (Lα5LG1-3/ELP). Lα5LG1-3/ELP was expressed in Escherichia coli and displayed a molecular size of ~70 kDa on 12% SDS-polyacrylamide gels. The cellular activities, such as cellular adhesion (adhesion assay) and proliferation (MTT cytotoxicity assay), of human mesenchymal stem cells (hMSCs) treated with 1 µg/ml of Lα5LG1-3/ELP were enhanced compared with those of untreated cells. Additionally, the number of undifferentiated hMSCs and their degree of stemness were assessed based on the gene expression levels of the stem cell markers cluster differentiation 90 (CD90), endoglin (CD105) and CD73. The expression levels of these markers were upregulated by 2.42-, 2.29- and 1.92-fold, respectively, in the hMSCs treated with Lα5LG1-3/ELP compared with the levels in untreated controls. Thus, Lα5LG1-3/ELP may be used to enhance the viability of hMSCs and preserve their undifferentiated state, whereby the clinical applications of hMSCs may be improved.
Collapse
Affiliation(s)
- Sujin Lee
- Department of Biochemistry, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| | - Dong-Sung Lee
- College of Pharmacy, Chosun University, Gwangju 61452, Republic of Korea
| | - Jun-Hyeog Jang
- Department of Biochemistry, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| |
Collapse
|
7
|
Suzuki M. Structural and functional analyses of organic molecules regulating biomineralization. Biosci Biotechnol Biochem 2020; 84:1529-1540. [DOI: 10.1080/09168451.2020.1762068] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Abstract
Biomineralization by living organisms are common phenomena observed everywhere. Molluskan shells are representative biominerals that have fine microstructures with controlled morphology, polymorph, and orientation of CaCO3 crystals. A few organic molecules involved in the biominerals play important roles in the formation of such microstructures. Analyses of structure–function relationships for matrix proteins in biominerals revealed that almost all matrix proteins have an acidic region for the binding of calcium ion in CaCO3 crystals and interaction domains for other organic molecules. On the other hand, biomineralization of metal nanoparticles by microorganisms were also investigated. Gold nanoparticles and quantum dots containing cadmium were successfully synthesized by bacteria or a fungus. The analyses of components revealed that glycolipids, oligosaccharides, and lactic acids have key roles to synthesize the gold nanoparticle in Lactobacillus casei as reductants and dispersants. These researches about biomineralization will give new insights for material and environmental sciences in the human society.
Collapse
Affiliation(s)
- Michio Suzuki
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, the University of Tokyo, Tokyo, Japan
| |
Collapse
|
8
|
Arreguin AJ, Colognato H. Brain Dysfunction in LAMA2-Related Congenital Muscular Dystrophy: Lessons From Human Case Reports and Mouse Models. Front Mol Neurosci 2020; 13:118. [PMID: 32792907 PMCID: PMC7390928 DOI: 10.3389/fnmol.2020.00118] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/09/2020] [Indexed: 12/26/2022] Open
Abstract
Laminin α2 gene (LAMA2)-related Congenital Muscular Dystrophy (CMD) was distinguished by a defining central nervous system (CNS) abnormality—aberrant white matter signals by MRI—when first described in the 1990s. In the past 25 years, researchers and clinicians have expanded our knowledge of brain involvement in LAMA2-related CMD, also known as Congenital Muscular Dystrophy Type 1A (MDC1A). Neurological changes in MDC1A can be structural, including lissencephaly and agyria, as well as functional, including epilepsy and intellectual disability. Mouse models of MDC1A include both spontaneous and targeted LAMA2 mutations and range from a partial loss of LAMA2 function (e.g., dy2J/dy2J), to a complete loss of LAMA2 expression (dy3K/dy3K). Diverse cellular and molecular changes have been reported in the brains of MDC1A mouse models, including blood-brain barrier dysfunction, altered neuro- and gliogenesis, changes in synaptic plasticity, and decreased myelination, providing mechanistic insight into potential neurological dysfunction in MDC1A. In this review article, we discuss selected studies that illustrate the potential scope and complexity of disturbances in brain development in MDC1A, and as well as highlight mechanistic insights that are emerging from mouse models.
Collapse
Affiliation(s)
- Andrea J Arreguin
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, United States.,Medical Scientist Training Program (MSTP), Stony Brook University, Stony Brook, NY, United States
| | - Holly Colognato
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
9
|
Simon T, Li L, Wagner C, Zhang T, Saxena V, Brinkman CC, Tostanoski LH, Ostrand-Rosenberg S, Jewell C, Shea-Donohue T, Hippen K, Blazar B, Abdi R, Bromberg JS. Differential Regulation of T-cell Immunity and Tolerance by Stromal Laminin Expressed in the Lymph Node. Transplantation 2019; 103:2075-2089. [PMID: 31343575 PMCID: PMC6768765 DOI: 10.1097/tp.0000000000002774] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Stromal laminins α4 and α5 are differentially regulated in transplant tolerance and immunity, respectively, resulting in altered T-cell trafficking. We hypothesized that laminins directly regulated T-cell activation and polarization. METHODS Human and mouse CD4 T cells were activated in Th1, Th2, Th17, or regulatory T cell (Treg) environments with/without laminin α4 and/or α5. Laminin α5 receptors were blocked with anti-α6 integrin or anti-α-dystroglycan (αDG) monoclonal antibodies, and T-cell polarization was determined. T-cell receptor transgenic TEa CD4 cells that recognized donor alloantigen were transferred into C57BL/6 mice that received alloantigen or cardiac allografts. Laminin receptors were blocked, and TEa T-cell migration and differentiation were assessed. Laminin expression was measured in several models of immunity and tolerance. RESULTS In diverse models, laminins α4 and α5 were differentially regulated. Immunity was associated with decreased laminin α4:α5 ratio, while tolerance was associated with an increased ratio. Laminin α4 inhibited CD4+ T-cell proliferation and Th1, Th2, and Th17 polarization but favored Treg induction. Laminin α5 favored T-cell activation and Th1, Th2, and Th17 polarization and inhibited Treg. Laminin α5 was recognized by T cell integrin α6 and is important for activation and inhibition of Treg. Laminin α5 was also recognized by T cell α-DG and required for Th17 differentiation. Anti-α6 integrin or anti-DG prolonged allograft survival. CONCLUSIONS Laminins α4 and α5 are coinhibitory and costimulatory ligands for human and mouse CD4 T cells, respectively. Laminins and their receptors modulate immune responses by acting as one of the molecular switches for immunity or suppression.
Collapse
Affiliation(s)
- Thomas Simon
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Lushen Li
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Chelsea Wagner
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Tianshu Zhang
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Vikas Saxena
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - C. Colin Brinkman
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Lisa H. Tostanoski
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
| | - Suzanne Ostrand-Rosenberg
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland, USA
| | - Chris Jewell
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
| | - Terez Shea-Donohue
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Keli Hippen
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- The Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Bruce Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- The Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Reza Abdi
- Transplantation Research Center and Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan S. Bromberg
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
10
|
Characterization of dystroglycan binding in adhesion of human induced pluripotent stem cells to laminin-511 E8 fragment. Sci Rep 2019; 9:13037. [PMID: 31506597 PMCID: PMC6737067 DOI: 10.1038/s41598-019-49669-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 08/29/2019] [Indexed: 12/16/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) grow indefinitely in culture and have the potential to regenerate various tissues. In the development of cell culture systems, a fragment of laminin-511 (LM511-E8) was found to improve the proliferation of stem cells. The adhesion of undifferentiated cells to LM511-E8 is mainly mediated through integrin α6β1. However, the involvement of non-integrin receptors remains unknown in stem cell culture using LM511-E8. Here, we show that dystroglycan (DG) is strongly expressed in hiPSCs. The fully glycosylated DG is functionally active for laminin binding, and although it has been suggested that LM511-E8 lacks DG binding sites, the fragment does weakly bind to DG. We further identified the DG binding sequence in LM511-E8, using synthetic peptides, of which, hE8A5-20 (human laminin α5 2688–2699: KTLPQLLAKLSI) derived from the laminin coiled-coil domain, exhibited DG binding affinity and cell adhesion activity. Deletion and mutation studies show that LLAKLSI is the active core sequence of hE8A5-20, and that, K2696 is a critical amino acid for DG binding. We further demonstrated that hiPSCs adhere to hE8A5-20-conjugated chitosan matrices. The amino acid sequence of DG binding peptides would be useful to design substrata for culture system of undifferentiated and differentiated stem cells.
Collapse
|
11
|
Dempsey CE, Bigotti MG, Adams JC, Brancaccio A. Analysis of α-Dystroglycan/LG Domain Binding Modes: Investigating Protein Motifs That Regulate the Affinity of Isolated LG Domains. Front Mol Biosci 2019; 6:18. [PMID: 30984766 PMCID: PMC6450144 DOI: 10.3389/fmolb.2019.00018] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 03/07/2019] [Indexed: 12/25/2022] Open
Abstract
Dystroglycan (DG) is an adhesion complex that links the cytoskeleton to the surrounding extracellular matrix in skeletal muscle and a wide variety of other tissues. It is composed of a highly glycosylated extracellular α-DG associated noncovalently with a transmembrane β-DG whose cytodomain interacts with dystrophin and its isoforms. Alpha-dystroglycan (α-DG) binds tightly and in a calcium-dependent fashion to multiple extracellular proteins and proteoglycans, each of which harbors at least one, or, more frequently, tandem arrays of laminin-globular (LG) domains. Considerable biochemical and structural work has accumulated on the α-DG-binding LG domains, highlighting a significant heterogeneity in ligand-binding properties of domains from different proteins as well as between single and multiple LG domains within the same protein. Here we review biochemical, structural, and functional information on the LG domains reported to bind α-dystroglycan. In addition, we have incorporated bioinformatics and modeling to explore whether specific motifs responsible for α-dystroglycan recognition can be identified within isolated LG domains. In particular, we analyzed the LG domains of slits and agrin as well as those of paradigmatic α-DG non-binders such as laminin-α3. While some stretches of basic residues may be important, no universally conserved motifs could be identified. However, the data confirm that the coordinated calcium atom within the LG domain is needed to establish an interaction with the sugars of α-DG, although it appears that this alone is insufficient to mediate significant α-DG binding. We develop a scenario involving different binding modes of a single LG domain unit, or tandemly repeated units, with α-DG. A variability of binding modes might be important to generate a range of affinities to allow physiological regulation of this interaction, reflecting its crucial biological importance.
Collapse
Affiliation(s)
| | | | - Josephine C Adams
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Andrea Brancaccio
- School of Biochemistry, University of Bristol, Bristol, United Kingdom.,Istituto di Chimica del Riconoscimento Molecolare - CNR, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
12
|
Nirwane A, Yao Y. Laminins and their receptors in the CNS. Biol Rev Camb Philos Soc 2019; 94:283-306. [PMID: 30073746 DOI: 10.1111/brv.12454] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 07/05/2018] [Accepted: 07/09/2018] [Indexed: 01/24/2023]
Abstract
Laminin, an extracellular matrix protein, is widely expressed in the central nervous system (CNS). By interacting with integrin and non-integrin receptors, laminin exerts a large variety of important functions in the CNS in both physiological and pathological conditions. Due to the existence of many laminin isoforms and their differential expression in various cell types in the CNS, the exact functions of each individual laminin molecule in CNS development and homeostasis remain largely unclear. In this review, we first briefly introduce the structure and biochemistry of laminins and their receptors. Next, the dynamic expression of laminins and their receptors in the CNS during both development and in adulthood is summarized in a cell-type-specific manner, which allows appreciation of their functional redundancy/compensation. Furthermore, we discuss the biological functions of laminins and their receptors in CNS development, blood-brain barrier (BBB) maintenance, neurodegeneration, stroke, and neuroinflammation. Last, key challenges and potential future research directions are summarized and discussed. Our goals are to provide a synthetic review to stimulate future studies and promote the formation of new ideas/hypotheses and new lines of research in this field.
Collapse
Affiliation(s)
- Abhijit Nirwane
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 240 W Green Street, Athens, GA 30602, U.S.A
| | - Yao Yao
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 240 W Green Street, Athens, GA 30602, U.S.A
| |
Collapse
|
13
|
Sigmundsson K, Ojala JR, Öhman MK, Österholm AM, Moreno-Moral A, Domogatskaya A, Chong LY, Sun Y, Chai X, Steele JA, George B, Patarroyo M, Nilsson AS, Rodin S, Ghosh S, Stevens MM, Petretto E, Tryggvason K. Culturing functional pancreatic islets on α5-laminins and curative transplantation to diabetic mice. Matrix Biol 2018; 70:5-19. [DOI: 10.1016/j.matbio.2018.03.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/22/2018] [Accepted: 03/22/2018] [Indexed: 12/15/2022]
|
14
|
Yurchenco PD, McKee KK, Reinhard JR, Rüegg MA. Laminin-deficient muscular dystrophy: Molecular pathogenesis and structural repair strategies. Matrix Biol 2017; 71-72:174-187. [PMID: 29191403 DOI: 10.1016/j.matbio.2017.11.009] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/15/2017] [Accepted: 11/16/2017] [Indexed: 12/19/2022]
Abstract
Laminins are large heterotrimers composed of the α, β and γ subunits with distinct tissue-specific and developmentally regulated expression patterns. The laminin-α2 subunit, encoded by the LAMA2 gene, is expressed in skeletal muscle, Schwann cells of the peripheral nerve and astrocytes and pericytes of the capillaries in the brain. Mutations in LAMA2 cause the most common type of congenital muscular dystrophies, called LAMA2 MD or MDC1A. The disorder manifests mostly as a muscular dystrophy but slowing of nerve conduction contributes to the disease. There are severe, non-ambulatory or milder, ambulatory variants, the latter resulting from reduced laminin-α2 expression and/or deficient laminin-α2 function. Lm-211 (α2β1γ1) is responsible for initiating basement membrane assembly. This is primarily accomplished by anchorage of Lm-211 to dystroglycan and α7β1 integrin receptors, polymerization, and binding to nidogen and other structural components. In LAMA2 MD, Lm-411 replaces Lm-211; however, Lm-411 lacks the ability to polymerize and bind to receptors. This results in a weakened basement membrane leading to the disease. The possibility of introducing structural repair proteins that correct the underlying abnormality is an attractive therapeutic goal. Recent studies in mouse models for LAMA2 MD reveal that introduction of laminin-binding linker proteins that restore lost functional activities can substantially ameliorate the disease. This review discusses the underlying mechanism of this repair and compares this approach to other developing therapies employing pharmacological treatments.
Collapse
Affiliation(s)
- Peter D Yurchenco
- Dept. Pathology & Laboratory Medicine, Rutgers University, Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA.
| | - Karen K McKee
- Dept. Pathology & Laboratory Medicine, Rutgers University, Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | | | - Markus A Rüegg
- Biozentrum, University of Basel, 4056 Basel, Switzerland.
| |
Collapse
|
15
|
Simon T, Bromberg JS. Regulation of the Immune System by Laminins. Trends Immunol 2017; 38:858-871. [PMID: 28684207 DOI: 10.1016/j.it.2017.06.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 06/05/2017] [Accepted: 06/06/2017] [Indexed: 01/11/2023]
Abstract
Laminins are trimeric proteins that are major components of the basement membranes that separate endothelia and epithelia from the underlying tissue. Sixteen laminin isoforms have been described, each with distinct tissue expression patterns and functions. While laminins have a critical structural role, recent evidence also indicates that they also impact the migration and functions of immune cells. Laminins are differentially expressed upon immunity or tolerance and orientate the immune response. This review will summarize the structure of laminins, the modulation of their expression, and their interactions with the immune system. Finally, the role of the laminins in autoimmune diseases and transplantation will be discussed.
Collapse
Affiliation(s)
- Thomas Simon
- Departments of Surgery and Microbiology and Immunology, Center for Vascular and Inflammatory Disease, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jonathan S Bromberg
- Departments of Surgery and Microbiology and Immunology, Center for Vascular and Inflammatory Disease, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
16
|
McKee KK, Crosson SC, Meinen S, Reinhard JR, Rüegg MA, Yurchenco PD. Chimeric protein repair of laminin polymerization ameliorates muscular dystrophy phenotype. J Clin Invest 2017; 127:1075-1089. [PMID: 28218617 DOI: 10.1172/jci90854] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 12/30/2016] [Indexed: 12/28/2022] Open
Abstract
Mutations in laminin α2-subunit (Lmα2, encoded by LAMA2) are linked to approximately 30% of congenital muscular dystrophy cases. Mice with a homozygous mutation in Lama2 (dy2J mice) express a nonpolymerizing form of laminin-211 (Lm211) and are a model for ambulatory-type Lmα2-deficient muscular dystrophy. Here, we developed transgenic dy2J mice with muscle-specific expression of αLNNd, a laminin/nidogen chimeric protein that provides a missing polymerization domain. Muscle-specific expression of αLNNd in dy2J mice resulted in strong amelioration of the dystrophic phenotype, manifested by the prevention of fibrosis and restoration of forelimb grip strength. αLNNd also restored myofiber shape, size, and numbers to control levels in dy2J mice. Laminin immunostaining and quantitation of tissue extractions revealed increased Lm211 expression in αLNNd-transgenic dy2J mice. In cultured myotubes, we determined that αLNNd expression increased myotube surface accumulation of polymerization-deficient recombinant laminins, with retention of collagen IV, reiterating the basement membrane (BM) changes observed in vivo. Laminin LN domain mutations linked to several of the Lmα2-deficient muscular dystrophies are predicted to compromise polymerization. The data herein support the hypothesis that engineered expression of αLNNd can overcome polymerization deficits to increase laminin, stabilize BM structure, and substantially ameliorate muscular dystrophy.
Collapse
|
17
|
Aiyelabegan HT, Sadroddiny E. Fundamentals of protein and cell interactions in biomaterials. Biomed Pharmacother 2017; 88:956-970. [PMID: 28178627 DOI: 10.1016/j.biopha.2017.01.136] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/11/2017] [Accepted: 01/23/2017] [Indexed: 12/31/2022] Open
Abstract
The extracellular matrix (ECM) is an active and complex microenvironment with outstanding biomechanical, biophysical, and biochemical characteristics, which can indirectly or directly controls cell adhesion, migration, proliferation, and differentiation, as well as partaking in regeneration and homeostasis of organs and tissues. The ECM has captivated a great deal of attention with the rapid progress of tissue engineering (TE) in the field of regenerative medicine (RM). Approaches to TE, RM and cancer therapy center on the necessity to deliver cell signals to direct cell proliferation and differentiation. These "external signals" are induced from cell-cell, and cell-ECM, interactions, as well as from physico-chemical, mechanical stimuli and growth factors. With the advent of new biomaterials such as casein, we gave a general insight into cell-ECM protein interactions in biomaterials and their applications in TE, RM and cancer therapy. An account of the main ECM molecules and cellular receptors with emphasis on integrins and its ligands was given, their effect on the induction of particular signal transduction pathways is also elucidated.
Collapse
Affiliation(s)
- Hammed Tanimowo Aiyelabegan
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Department of Medical Biotechnology, School of Advanced Technologies in Medicine, International Campus-Tehran University of Medical Sciences (IC-TUMS), Tehran, Iran
| | - Esmaeil Sadroddiny
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Di Russo J, Hannocks MJ, Luik AL, Song J, Zhang X, Yousif L, Aspite G, Hallmann R, Sorokin L. Vascular laminins in physiology and pathology. Matrix Biol 2017; 57-58:140-148. [DOI: 10.1016/j.matbio.2016.06.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 06/28/2016] [Indexed: 12/11/2022]
|
19
|
Penton CM, Badarinarayana V, Prisco J, Powers E, Pincus M, Allen RE, August PR. Laminin 521 maintains differentiation potential of mouse and human satellite cell-derived myoblasts during long-term culture expansion. Skelet Muscle 2016; 6:44. [PMID: 27964750 PMCID: PMC5154152 DOI: 10.1186/s13395-016-0116-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 12/01/2016] [Indexed: 12/03/2022] Open
Abstract
Background Large-scale expansion of myogenic progenitors is necessary to support the development of high-throughput cellular assays in vitro and to advance genetic engineering approaches necessary to develop cellular therapies for rare muscle diseases. However, optimization has not been performed in order to maintain the differentiation capacity of myogenic cells undergoing long-term cell culture. Multiple extracellular matrices have been utilized for myogenic cell studies, but it remains unclear how different matrices influence long-term myogenic activity in culture. To address this challenge, we have evaluated multiple extracellular matrices in myogenic studies over long-term expansion. Methods We evaluated the consequence of propagating mouse and human myogenic stem cell progenitors on various extracellular matrices to determine if they could enhance long-term myogenic potential. For the first time reported, we comprehensively examine the effect of physiologically relevant laminins, laminin 211 and laminin 521, compared to traditionally utilized ECMs (e.g., laminin 111, gelatin, and Matrigel) to assess their capacity to preserve myogenic differentiation potential. Results Laminin 521 supported increased proliferation in early phases of expansion and was the only substrate facilitating high-level fusion following eight passages in mouse myoblast cell cultures. In human myoblast cell cultures, laminin 521 supported increased proliferation during expansion and superior differentiation with myotube hypertrophy. Counterintuitively however, laminin 211, the native laminin isoform in resting skeletal muscle, resulted in low proliferation and poor differentiation in mouse and human cultures. Matrigel performed excellent in short-term mouse studies but showed high amounts of variability following long-term expansion. Conclusions These results demonstrate laminin 521 is a superior substrate for both short-term and long-term myogenic cell culture applications compared to other commonly utilized substrates. Since Matrigel cannot be used for clinical applications, we propose that laminin 521 could possibly be employed in the future to provide myoblasts for cellular therapy directed clinical studies. Electronic supplementary material The online version of this article (doi:10.1186/s13395-016-0116-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christopher M Penton
- Discovery Biology, Tucson Innovation Center, Icagen, Oro Valley, AZ, 85755, USA.
| | | | - Joy Prisco
- Discovery Biology, Tucson Innovation Center, Sanofi, Oro Valley, AZ, 85755, USA
| | - Elaine Powers
- Discovery Biology, Tucson Innovation Center, Sanofi, Oro Valley, AZ, 85755, USA
| | - Mark Pincus
- Discovery Biology, Tucson Innovation Center, Icagen, Oro Valley, AZ, 85755, USA
| | - Ronald E Allen
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, 85721, USA.
| | - Paul R August
- Discovery Biology, Tucson Innovation Center, Icagen, Oro Valley, AZ, 85755, USA.
| |
Collapse
|
20
|
Fleger-Weckmann A, Üstün Y, Kloepper J, Paus R, Bloch W, Chen ZL, Wegner J, Sorokin L, Langbein L, Eckes B, Zigrino P, Krieg T, Nischt R. Deletion of the epidermis derived laminin γ1 chain leads to defects in the regulation of late hair morphogenesis. Matrix Biol 2016; 56:42-56. [DOI: 10.1016/j.matbio.2016.05.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 05/04/2016] [Accepted: 05/08/2016] [Indexed: 12/16/2022]
|
21
|
Rogers RS, Nishimune H. The role of laminins in the organization and function of neuromuscular junctions. Matrix Biol 2016; 57-58:86-105. [PMID: 27614294 DOI: 10.1016/j.matbio.2016.08.008] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 08/10/2016] [Accepted: 08/17/2016] [Indexed: 01/11/2023]
Abstract
The synapse between motor neurons and skeletal muscle is known as the neuromuscular junction (NMJ). Proper alignment of presynaptic and post-synaptic structures of motor neurons and muscle fibers, respectively, is essential for efficient motor control of skeletal muscles. The synaptic cleft between these two cells is filled with basal lamina. Laminins are heterotrimer extracellular matrix molecules that are key members of the basal lamina. Laminin α4, α5, and β2 chains specifically localize to NMJs, and these laminin isoforms play a critical role in maintenance of NMJs and organization of synaptic vesicle release sites known as active zones. These individual laminin chains exert their role in organizing NMJs by binding to their receptors including integrins, dystroglycan, and voltage-gated calcium channels (VGCCs). Disruption of these laminins or the laminin-receptor interaction occurs in neuromuscular diseases including Pierson syndrome and Lambert-Eaton myasthenic syndrome (LEMS). Interventions to maintain proper level of laminins and their receptor interactions may be insightful in treating neuromuscular diseases and aging related degeneration of NMJs.
Collapse
Affiliation(s)
- Robert S Rogers
- Department of Anatomy and Cell Biology, University of Kansas School of Medicine, Kansas City, Kansas, USA.
| | - Hiroshi Nishimune
- Department of Anatomy and Cell Biology, University of Kansas School of Medicine, Kansas City, Kansas, USA.
| |
Collapse
|
22
|
Wegner J, Loser K, Apsite G, Nischt R, Eckes B, Krieg T, Werner S, Sorokin L. Laminin α5 in the keratinocyte basement membrane is required for epidermal-dermal intercommunication. Matrix Biol 2016; 56:24-41. [PMID: 27234307 DOI: 10.1016/j.matbio.2016.05.001] [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: 02/14/2016] [Revised: 05/04/2016] [Accepted: 05/08/2016] [Indexed: 12/22/2022]
Abstract
Laminin α5 is broadly expressed in the epidermal basement membrane (BM) of mature mice and its elimination at this site (Lama5Ker5 mouse) results in hyperproliferation of basal keratinocytes and a delay in hair follicle development, which correlated with upregulation of the dermally-derived laminin α2 and laminin α4 chains in the epidermal BM and of tenascin-C subjacent to the BM. In vitro studies revealed laminin 511 to be strongly adhesive for primary keratinocytes and that loss of laminin α5 does not result in cell autonomous defects in proliferation. Flow cytometry reveals that the loss of laminin α5 resulted in increased numbers of CD45+, CD4+ and CD11b+ immune cells in the skin, which temporo-spatial analyses revealed were detectable only subsequent to the loss of laminin α5 and the appearance of the hyperproliferative keratinocyte phenotype. These findings indicate that immune cell changes are the consequence and not the cause of keratinocyte hyperproliferation. Loss of laminin α5 in the epidermal BM was also associated with changes in the expression of several dermally-derived growth factors involved in keratinocyte proliferation and hair follicle development in adult but not new born Lama5Ker5 skin, including KGF, EGF and KGF-2. In situ binding of FGF-receptor-2α (IIIb)-Fc chimera (FGFR2IIIb) to mouse skin sections revealed decoration of several BMs, including the epidermal BM, which was absent in Lama5Ker5 skin. This indicates reduced levels of FGFR2IIIb ligands, which include KGF and KGF-2, in the epidermal BM of adult Lama5Ker5 skin. Our data suggest an initial inhibitory effect of laminin α5 on basal keratinocyte proliferation and migration, which is exacerbated by subsequent changes in growth factor expression by epidermal and dermal cells, implicating laminin α5 in epidermal-dermal intercommunication.
Collapse
Affiliation(s)
- Jeannine Wegner
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, Germany; Cells-in-Motion Cluster of Excellence, University of Muenster, Germany
| | - Karin Loser
- Cells-in-Motion Cluster of Excellence, University of Muenster, Germany; Department of Dermatology, University of Muenster, Germany
| | - Gunita Apsite
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, Germany; Cells-in-Motion Cluster of Excellence, University of Muenster, Germany
| | | | - Beate Eckes
- Department of Dermatology, University of Cologne, Germany
| | - Thomas Krieg
- Department of Dermatology, University of Cologne, Germany
| | - Sabine Werner
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Switzerland
| | - Lydia Sorokin
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, Germany; Cells-in-Motion Cluster of Excellence, University of Muenster, Germany.
| |
Collapse
|
23
|
HOSHIBA T, TANAKA M. Integrin-independent Cell Adhesion Substrates: Possibility of Applications for Mechanobiology Research. ANAL SCI 2016; 32:1151-1158. [DOI: 10.2116/analsci.32.1151] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Takashi HOSHIBA
- Frontier Center for Organic Materials, Yamagata University
- International Center for Materials Nanoarchitectonics, National Institute for Materials Science
| | - Masaru TANAKA
- Frontier Center for Organic Materials, Yamagata University
- Institute for Materials Chemistry and Engineering, Kyushu University
| |
Collapse
|
24
|
Yamada M, Sekiguchi K. Molecular Basis of Laminin-Integrin Interactions. CURRENT TOPICS IN MEMBRANES 2015; 76:197-229. [PMID: 26610915 DOI: 10.1016/bs.ctm.2015.07.002] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Laminins are composed of three polypeptide chains, designated as α, β, and γ. The C-terminal region of laminin heterotrimers, containing coiled-coil regions, short tails, and laminin globular (LG) domains, is necessary and sufficient for binding to integrins, which are the major laminin receptor class. Laminin recognition by integrins critically requires the α chain LG domains and a glutamic acid residue of the γ chain at the third position from the C-terminus. Furthermore, the C-terminal region of the β chain contains a short amino acid sequence that modulates laminin affinity for integrins. Thus, all three of the laminin chains act cooperatively to facilitate integrin binding. Mammals possess 5 α (α1-5), 3 β (β1-3), and 3 γ (γ1-3) chains, combinations of which give rise to 16 distinct laminin isoforms. Each isoform is expressed in a tissue-specific and developmental stage-specific manner, exerting its functions through binding of integrins. In this review, we detail the current knowledge surrounding the molecular basis and physiological relevance of specific interactions between laminins and integrins, and describe the mechanisms underlying laminin action through integrins.
Collapse
Affiliation(s)
- Masashi Yamada
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| | - Kiyotoshi Sekiguchi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
25
|
Yoshida-Moriguchi T, Campbell KP. Matriglycan: a novel polysaccharide that links dystroglycan to the basement membrane. Glycobiology 2015; 25:702-13. [PMID: 25882296 PMCID: PMC4453867 DOI: 10.1093/glycob/cwv021] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 04/08/2015] [Indexed: 01/01/2023] Open
Abstract
Associations between cells and the basement membrane are critical for a variety of biological events including cell proliferation, cell migration, cell differentiation and the maintenance of tissue integrity. Dystroglycan is a highly glycosylated basement membrane receptor, and is involved in physiological processes that maintain integrity of the skeletal muscle, as well as development and function of the central nervous system. Aberrant O-glycosylation of the α subunit of this protein, and a concomitant loss of dystroglycan's ability to function as a receptor for extracellular matrix (ECM) ligands that bear laminin globular (LG) domains, occurs in several congenital/limb-girdle muscular dystrophies (also referred to as dystroglycanopathies). Recent genetic studies revealed that mutations in DAG1 (which encodes dystroglycan) and at least 17 other genes disrupt the ECM receptor function of dystroglycan and cause disease. Here, we summarize recent advances in our understanding of the enzymatic functions of two of these disease genes: the like-glycosyltransferase (LARGE) and protein O-mannose kinase (POMK, previously referred to as SGK196). In addition, we discuss the structure of the glycan that directly binds the ECM ligands and the mechanisms by which this functional motif is linked to dystroglycan. In light of the fact that dystroglycan functions as a matrix receptor and the polysaccharide synthesized by LARGE is the binding motif for matrix proteins, we propose to name this novel polysaccharide structure matriglycan.
Collapse
Affiliation(s)
- Takako Yoshida-Moriguchi
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology, Department of Internal Medicine, University of Iowa Roy J. and Lucille A. Carver College of Medicine, 4283 Carver Biomedical Research Building, 285 Newton Road, Iowa City, IA 52242-1101, USA
| | - Kevin P Campbell
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology, Department of Internal Medicine, University of Iowa Roy J. and Lucille A. Carver College of Medicine, 4283 Carver Biomedical Research Building, 285 Newton Road, Iowa City, IA 52242-1101, USA
| |
Collapse
|
26
|
VP08R from infectious spleen and kidney necrosis virus is a novel component of the virus-mock basement membrane. J Virol 2014; 88:5491-501. [PMID: 24599992 DOI: 10.1128/jvi.03776-13] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Infectious spleen and kidney necrosis virus (ISKNV), the type species of the genus Megalocytivirus, family Iridoviridae, brings great harm to fish farming. In infected tissues, ISKNV infection is characterized by a unique phenomenon, in that the infected cells are attached by lymphatic endothelial cells (LECs), which are speculated to wall off the infected cells from host immune attack. A viral membrane protein, VP23R, binds and recruits the host nidogen-1 protein to construct a basement membrane (BM)-like structure, termed virus-mock basement membrane (VMBM), on the surface of infected cells to provide attaching sites for LECs. VMBMs do not contain collagen IV protein, which is essential for maintenance of BM integrity and functions. In this study, we identified the VP08R protein encoded by ISKNV. VP08R was predicted to be a secreted protein with a signal peptide but without a transmembrane domain. However, immunofluorescence assays demonstrated that VP08R is located on the plasma membrane of infected cells and shows an expression profile similar to that of VP23R. Coimmunoprecipitation showed that VP08R interacts with both VP23R and nidogen-1, indicating that VP08R is a component of VMBM and is present on the cell membrane by binding to VP23R. Through formation of intermolecular disulfide bonds, VP08R molecules self-organized into a multimer, which may play a role in the maintenance of VMBM integrity and stability. Moreover, the VP08R multimer was easily degraded when the ISKNV-infected cells were lysed, which may be a mechanism for VMBM disassembly when necessary to free LECs and release the mature virions. IMPORTANCE Infectious spleen and kidney necrosis virus (ISKNV; genus Megalocytivirus, family Iridovirus) is most harmful to cultured fishes. In tissues, the ISKNV-infected cells are attached by lymphatic endothelial cells (LECs), which are speculated to segregate the host immune system. A viral membrane protein, VP23R, binds and recruits the host nidogen-1 protein to construct virus-mock basement membranes (VMBMs) on the surface of infected cells to provide attaching sites for LECs. Although VMBMs lack the collagen IV network, which is an essential structural part of true BMs, VMBMs still show an intact structure. An ISKNV-encoded VP08R protein can self-assemble into a multimer and bind both VP23R and nidogen-1 to maintain the integrity and stability of VMBMs. On the basis of these facts, we redrew the putative schematic illustration of the VMBM structure. Our study suggests that the virus adopts a strategy to remodel the cellular matrix and may provide an important reference to elucidate BM functions and the mechanisms of lymphangiogenesis.
Collapse
|
27
|
Nakaya Y, Sukowati EW, Sheng G. Epiblast integrity requires CLASP and Dystroglycan-mediated microtubule anchoring to the basal cortex. ACTA ACUST UNITED AC 2013; 202:637-51. [PMID: 23940118 PMCID: PMC3747297 DOI: 10.1083/jcb.201302075] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Amniote epiblast cells differentiate into mesoderm and endoderm lineages during gastrulation through a process called epithelial-to-mesenchymal transition (EMT). Molecular regulation of gastrulation EMT is poorly understood. Here we show that epiblast epithelial status was maintained by anchoring microtubules to the basal cortex via CLIP-associated protein (CLASP), a microtubule plus-end tracking protein, and Dystroglycan, a transmembrane protein that bridges the cytoskeleton and basement membrane (BM). Mesoderm formation required down-regulation of CLASP and Dystroglycan, and reducing CLASP activity in pregastrulation epiblast cells caused ectopic BM breakdown and disrupted epiblast integrity. These effects were mediated through the CLASP-binding partner LL5. Live-imaging using EB1-enhanced GFP (eGFP) revealed that reducing CLASP and LL5 levels in the epiblast destabilized basal microtubules. We further show that Dystroglycan is localized to basolateral membrane in epiblast cells. Basal but not lateral localization of Dystroglycan was regulated by CLASP. We propose that epiblast-BM interaction requires CLASP- and Dystroglycan-mediated cortical microtubule anchoring, the disruption of which initiates gastrulation EMT.
Collapse
Affiliation(s)
- Yukiko Nakaya
- Laboratory for Early Embryogenesis, Institute of Physical and Chemical Research RIKEN Center for Developmental Biology, Chuo-Ku Kobe, Hyogo 650-0047, Japan
| | | | | |
Collapse
|
28
|
Suzuki M, Iwashima A, Kimura M, Kogure T, Nagasawa H. The molecular evolution of the pif family proteins in various species of mollusks. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2013; 15:145-58. [PMID: 22847736 DOI: 10.1007/s10126-012-9471-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2012] [Accepted: 06/30/2012] [Indexed: 05/04/2023]
Abstract
Various novel proteins have been identified from many kinds of mollusk shells. Although such matrix proteins are believed to play important roles in the calcium carbonate crystal formation of shells, no common proteins that interact with calcium carbonate or that are involved in the molecular mechanisms behind shell formation have been identified. Pif consists of two proteins, Pif 80 and Pif 97, which are encoded by a single mRNA. Pif 80 was identified as a key acidic protein that regulates the formation of the nacreous layer in Pinctada fucata, while Pif 97 has von Willebrand factor type A (VWA) and chitin-binding domains. In this study, we identified Pif homologues from Pinctada margaritifera, Pinctada maxima, Pteria penguin, Mytilus galloprovincialis, and in the genome database of Lottia gigantea in order to compare their primary protein sequences. The VWA and chitin-binding domains are conserved in all Pif 97 homologues, whereas the amino acid sequences of the Pif 80 regions differ markedly among the species. Sequence alignment revealed the presence of a novel significantly conserved sequence between the chitin-binding domain and the C-terminus of Pif 97. Further examination of the Pif 80 regions suggested that they share a sequence that is similar to the laminin G domain. These results indicate that all Pif molecules in bivalves and gastropods may be derived from a common ancestral gene. These comparisons may shed light on the correlation between molecular evolution and morphology in mollusk shell microstructure.
Collapse
Affiliation(s)
- Michio Suzuki
- Department of Earth and Planetary Science, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | | | |
Collapse
|
29
|
Sachs N, Sonnenberg A. Cell-matrix adhesion of podocytes in physiology and disease. Nat Rev Nephrol 2013; 9:200-10. [PMID: 23338211 DOI: 10.1038/nrneph.2012.291] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Cell-matrix adhesion is crucial for maintaining the mechanical integrity of epithelial tissues. Podocytes--a key component of the glomerular filtration barrier--are exposed to permanent transcapillary filtration pressure and must therefore adhere tightly to the underlying glomerular basement membrane (GBM). The major cell-matrix adhesion receptor in podocytes is the integrin α3β1, which connects laminin 521 in the GBM through various adaptor proteins to the intracellular actin cytoskeleton. Other cell-matrix adhesion receptors expressed by podocytes include the integrins α2β1 and αvβ3, α-dystroglycan, syndecan-4 and type XVII collagen. Mutations in genes encoding any of the components critical for podocyte adhesion cause glomerular disease. This Review highlights recent advances in our understanding of the cell biology and genetics of podocyte adhesion with special emphasis on glomerular disease.
Collapse
Affiliation(s)
- Norman Sachs
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | |
Collapse
|
30
|
Yousif LF, Di Russo J, Sorokin L. Laminin isoforms in endothelial and perivascular basement membranes. Cell Adh Migr 2012; 7:101-10. [PMID: 23263631 DOI: 10.4161/cam.22680] [Citation(s) in RCA: 172] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Laminins, one of the major functional components of basement membranes, are found underlying endothelium, and encasing pericytes and smooth muscle cells in the vessel wall. Depending on the type of blood vessel (capillary, venule, postcapillary venule, vein or artery) and their maturation state, both the endothelial and mural cell phenotype vary, with associated changes in laminin isoform expression. Laminins containing the α4 and α5 chains are the major isoforms found in the vessel wall, with the added contribution of laminin α2 in larger vessels. We here summarize current data on the precise localization of these laminin isoforms and their receptors in the different layers of the vessel wall, and their potential contribution to vascular homeostasis.
Collapse
Affiliation(s)
- Lema F Yousif
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | | | | |
Collapse
|
31
|
Affiliation(s)
- Anna Domogatskaya
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77 Stockholm, Sweden; , ,
| | - Sergey Rodin
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77 Stockholm, Sweden; , ,
| | - Karl Tryggvason
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77 Stockholm, Sweden; , ,
| |
Collapse
|
32
|
Spenlé C, Simon-Assmann P, Orend G, Miner JH. Laminin α5 guides tissue patterning and organogenesis. Cell Adh Migr 2012; 7:90-100. [PMID: 23076210 PMCID: PMC3544791 DOI: 10.4161/cam.22236] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Laminins (LM) are extracellular matrix molecules that contribute to and are required for the formation of basement membranes. They participate in the modulation of epithelial/mesenchymal interactions and are implicated in organogenesis and maintenance of organ homeostasis. Among the LM molecules, the LM α5 chain (LMα5) is one of the most widely distributed LM in the developing and mature organism. Its presence in some basement membranes during embryogenesis is absolutely required for maintenance of basement membrane integrity and thus for proper organogenesis. LMα5 also regulates the expression of genes important for major biological processes, in part by repressing or activating signaling pathways, depending upon the physiological context.
Collapse
|
33
|
Katagiri F, Sudo M, Hamakubo T, Hozumi K, Nomizu M, Kikkawa Y. Identification of active sequences in the L4a domain of laminin α5 promoting neurite elongation. Biochemistry 2012; 51:4950-8. [PMID: 22621685 DOI: 10.1021/bi300214g] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Laminin α5 is an extracellular matrix protein containing multiple domains implicated in various biological processes, such as embryogenesis and renal function. In this study, we used recombinant proteins and synthetic peptides to identify amino acid residues within the short arm region of α5 that were critical for neurite outgrowth activity. The short arm of α5 contains three globular domains (LN, L4a, and L4b) and three rodlike elements (LEa, LEb, and LEc). Recombinant proteins comprised of the α5 short arm fused with a Fc tag produced in 293 cells were assayed for PC12 (pheochromocytoma) cell adhesion and neurite outgrowth activities. Although it did not have cell attachment activity, neurite outgrowth was promoted by the recombinant protein. To narrow the region involved in neurite outgrowth activity, two truncated recombinant proteins were produced in 293 cells. A recombinant protein lacking L4a and LEb lost activity. Furthermore, we synthesized 78 partially overlapping peptides representing most of the amino acid sequences of L4a and LEb. Of the peptides, A5-76 [mouse laminin α5 928-939 (TSPDLFRLVFRY) in L4a] exhibited neurite outgrowth activity. Mutagenesis studies showed that Phe(933) and Arg(934) were involved in neurite outgrowth activity. Moreover, inhibition assays using anti-integrin monoclonal antibodies showed that neurite outgrowth on the α5 short arm was partially mediated by integrin α1β1. However, the antibodies to integrin α1 and β1 did not inhibit neurite elongation on the A5-76 peptide. These results suggest that in addition to cellular interactions with the active site in the L4a domain, the binding of integrin α1β1 seems to modulate neurite elongation on the short arm of α5.
Collapse
Affiliation(s)
- Fumihiko Katagiri
- Laboratory of Clinical Biochemistry, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | | | | | | | | | | |
Collapse
|
34
|
Katagiri F, Ishikawa M, Yamada Y, Hozumi K, Kikkawa Y, Nomizu M. Screening of integrin-binding peptides from the laminin α4 and α5 chain G domain peptide library. Arch Biochem Biophys 2012; 521:32-42. [PMID: 22391228 DOI: 10.1016/j.abb.2012.02.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 02/22/2012] [Accepted: 02/23/2012] [Indexed: 02/09/2023]
Abstract
Laminins, a multifunctional protein family of extracellular matrix, interact with various types of integrin. Here, integrin-mediated cell adhesive peptides have been systematically screened in the laminin α4 and α5 chain G domain peptide library consisting of 211 peptides by both the peptide-coated plastic plates and peptide-conjugated Sepharose bead assays using human dermal fibroblasts. Thirteen peptides promoted cell spreading and the activity was specifically inhibited by EDTA. Cell attachment to 11 peptides was inhibited by anti-integrin β1 antibody. Additionally, cell attachment to the A5G81 (AGQWHRVSVRWG) and A5G84 (TWSQKALHHRVP) peptides was specifically inhibited by anti-integrin α3 and α6 antibodies. These results suggest that the A5G81 and A5G84 peptides promote integrin α3β1- and α6β1-mediated cell attachment. Further, most of the integrin-mediated cell adhesive peptides are located in the loop regions in the G domains, suggesting that structure is important for the integrin specific recognition. Integrin binding peptides are useful for understanding laminin functions and have a potential to use for biomaterials and drug development.
Collapse
Affiliation(s)
- Fumihiko Katagiri
- Laboratory of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | | | | | | | | | | |
Collapse
|
35
|
Kusuma N, Anderson RL, Pouliot N. Laminin α5-derived peptides modulate the properties of metastatic breast tumour cells. Clin Exp Metastasis 2011; 28:909-21. [DOI: 10.1007/s10585-011-9422-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Accepted: 09/05/2011] [Indexed: 01/06/2023]
|
36
|
An antibody to the lutheran glycoprotein (Lu) recognizing the LU4 blood type variant inhibits cell adhesion to laminin α5. PLoS One 2011; 6:e23329. [PMID: 21858073 PMCID: PMC3155534 DOI: 10.1371/journal.pone.0023329] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Accepted: 07/13/2011] [Indexed: 11/23/2022] Open
Abstract
Background The Lutheran blood group glycoprotein (Lu), an Ig superfamily (IgSF) transmembrane receptor, is also known as basal cell adhesion molecule (B-CAM). Lu/B-CAM is a specific receptor for laminin α5, a major component of basement membranes in various tissues. Previous reports have shown that Lu/B-CAM binding to laminin α5 contributes to sickle cell vaso-occlusion. However, as there are no useful tools such as function-blocking antibodies or drugs, it is unclear how epithelial and sickled red blood cells adhere to laminin α5 via Lu/B-CAM. Methodology/Principal Findings In this study, we discovered a function-blocking antibody that inhibits Lu binding to laminin α5 using a unique binding assay on tissue sections. To characterize the function-blocking antibody, we identified the site on Lu/B-CAM recognized by this antibody. The extracellular domain of Lu/B-CAM contains five IgSF domains, D1-D2-D3-D4-D5. The antibody epitope was localized to D2, but not to the D3 domain containing the major part of the laminin α5 binding site. Furthermore, mutagenesis studies showed that Arg175, the LU4 blood group antigenic site, was crucial for forming the epitope and the antibody bound sufficiently close to sterically hinder the interaction with α5. Cell adhesion assay using the antibody also showed that Lu/B-CAM serves as a secondary receptor for the adhesion of carcinoma cells to laminin α5. Conclusion/Significance This function-blocking antibody against Lu/B-CAM should be useful for not only investigating cell adhesion to laminin α5 but also for developing drugs to inhibit sickle cell vaso-occlusion.
Collapse
|
37
|
VP23R of infectious spleen and kidney necrosis virus mediates formation of virus-mock basement membrane to provide attaching sites for lymphatic endothelial cells. J Virol 2010; 84:11866-75. [PMID: 20810728 DOI: 10.1128/jvi.00990-10] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Putative open reading frames (ORFs) encoding laminin-like proteins are found in all members of the genus Megalocytivirus, family Iridoviridae. This is the first study that identified the VP23R protein encoded by ORF23R of the infectious spleen and kidney necrosis virus (ISKNV), a member of these genes of megalocytiviruses. The VP23R mRNA covering the ISKNV genomic coordinates 19547 to 22273 was transcribed ahead of the major capsid protein. Immunofluorescence analysis demonstrated that VP23R was expressed on the plasma membrane of the ISKNV-infected cells and could not be a viral envelope protein. Residues 292 to 576 of VP23R are homologous to the laminin γ1III2-6 fragment, which covers the nidogen-binding site. An immunoprecipitation assay showed that VP23R could interact with nidogen-1, and immunohistochemistry showed that nidogen-1 was localized on the outer membrane of the infected cells. Electron microscopy showed that a virus-mock basement membrane (VMBM) was formed on the surface of the infected cells and a layer of endothelial cells (ECs) was attached to the VMBM. The VMBM contained VP23R and nidogen-1 but not collagen IV. The attached ECs were identified as lymphatic endothelial cells (LECs), which have unique feature of overlapping intercellular junctions and can be stained by immunohistochemistry using an antibody against a specific lymphatic marker, Prox-1. Such infection signs have never been described in viruses. Elucidating the functions of LECs attached to the surface of the infected cells may be useful for studies on the pathogenic mechanisms of megalocytiviruses and may also be important for studies on lymphangiogenesis and basement membrane functions.
Collapse
|
38
|
Chen F, Zhao Y, Liu M, Li D, Wu H, Chen H, Zhu Y, Luo F, Zhong J, Zhou Y, Qi Z, Zhang XL. Functional selection of hepatitis C virus envelope E2-binding Peptide ligands by using ribosome display. Antimicrob Agents Chemother 2010; 54:3355-64. [PMID: 20479194 PMCID: PMC2916351 DOI: 10.1128/aac.01357-09] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2009] [Revised: 12/07/2009] [Accepted: 04/29/2010] [Indexed: 02/07/2023] Open
Abstract
Small peptides that inhibit the hepatitis C virus (HCV) at the stage of viral entry have the potential to serve as attractive antiviral drugs. Ribosome display is a cell-free system for in vitro selection of peptides from large random peptide libraries. Thus, we utilized a ribosome display library technique for affinity selection of HCV envelope protein E2-binding peptide ligands. Through 13 rounds of selection, the ribosome display system generated high-affinity 12-mer peptides, and the selected peptide PE2D (MARHRNWPLVMV) demonstrated the highest specificity and affinity to the HCV E2 protein. Furthermore, amino acids 489 to 508 (YPPRPCGIVPAKSVCGPVYC) of E2 were identified as crucial for binding to PE2D. The selected peptides, especially PE2D, not only dramatically blocked E2 protein binding to hepatocytes but also dramatically inhibited HCV cell culture (HCVcc) entry into hepatocytes. HCVcc and HCV particles from HCV patient serum samples could also be specifically captured using PE2D. Our study demonstrates that the newly selected peptide ligand PE2D holds great promise for developing a new molecular probe, a therapeutic drug specifically for HCV, or an early-diagnostic reagent for HCV surface envelope antigen E2.
Collapse
Affiliation(s)
- Fang Chen
- State Key Laboratory of Virology and Department of Immunology and Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, Wuhan University School of Medicine, Wuhan 430071, People's Republic of China, Department of Microbiology, Second Military Medical University, Shanghai 200433, People's Republic of China, The Unit of Viral Hepatitis, Institut Pasteur of Shanghai, Shanghai 200025, People's Republic of China, Jianghan University, College of Life Science, Department of Biotechnology, Wuhan 430056, People's Republic of China
| | - Yinglan Zhao
- State Key Laboratory of Virology and Department of Immunology and Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, Wuhan University School of Medicine, Wuhan 430071, People's Republic of China, Department of Microbiology, Second Military Medical University, Shanghai 200433, People's Republic of China, The Unit of Viral Hepatitis, Institut Pasteur of Shanghai, Shanghai 200025, People's Republic of China, Jianghan University, College of Life Science, Department of Biotechnology, Wuhan 430056, People's Republic of China
| | - Min Liu
- State Key Laboratory of Virology and Department of Immunology and Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, Wuhan University School of Medicine, Wuhan 430071, People's Republic of China, Department of Microbiology, Second Military Medical University, Shanghai 200433, People's Republic of China, The Unit of Viral Hepatitis, Institut Pasteur of Shanghai, Shanghai 200025, People's Republic of China, Jianghan University, College of Life Science, Department of Biotechnology, Wuhan 430056, People's Republic of China
| | - Dongqing Li
- State Key Laboratory of Virology and Department of Immunology and Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, Wuhan University School of Medicine, Wuhan 430071, People's Republic of China, Department of Microbiology, Second Military Medical University, Shanghai 200433, People's Republic of China, The Unit of Viral Hepatitis, Institut Pasteur of Shanghai, Shanghai 200025, People's Republic of China, Jianghan University, College of Life Science, Department of Biotechnology, Wuhan 430056, People's Republic of China
| | - Hongyan Wu
- State Key Laboratory of Virology and Department of Immunology and Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, Wuhan University School of Medicine, Wuhan 430071, People's Republic of China, Department of Microbiology, Second Military Medical University, Shanghai 200433, People's Republic of China, The Unit of Viral Hepatitis, Institut Pasteur of Shanghai, Shanghai 200025, People's Republic of China, Jianghan University, College of Life Science, Department of Biotechnology, Wuhan 430056, People's Republic of China
| | - Haidan Chen
- State Key Laboratory of Virology and Department of Immunology and Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, Wuhan University School of Medicine, Wuhan 430071, People's Republic of China, Department of Microbiology, Second Military Medical University, Shanghai 200433, People's Republic of China, The Unit of Viral Hepatitis, Institut Pasteur of Shanghai, Shanghai 200025, People's Republic of China, Jianghan University, College of Life Science, Department of Biotechnology, Wuhan 430056, People's Republic of China
| | - Yongzhe Zhu
- State Key Laboratory of Virology and Department of Immunology and Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, Wuhan University School of Medicine, Wuhan 430071, People's Republic of China, Department of Microbiology, Second Military Medical University, Shanghai 200433, People's Republic of China, The Unit of Viral Hepatitis, Institut Pasteur of Shanghai, Shanghai 200025, People's Republic of China, Jianghan University, College of Life Science, Department of Biotechnology, Wuhan 430056, People's Republic of China
| | - Fengling Luo
- State Key Laboratory of Virology and Department of Immunology and Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, Wuhan University School of Medicine, Wuhan 430071, People's Republic of China, Department of Microbiology, Second Military Medical University, Shanghai 200433, People's Republic of China, The Unit of Viral Hepatitis, Institut Pasteur of Shanghai, Shanghai 200025, People's Republic of China, Jianghan University, College of Life Science, Department of Biotechnology, Wuhan 430056, People's Republic of China
| | - Jin Zhong
- State Key Laboratory of Virology and Department of Immunology and Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, Wuhan University School of Medicine, Wuhan 430071, People's Republic of China, Department of Microbiology, Second Military Medical University, Shanghai 200433, People's Republic of China, The Unit of Viral Hepatitis, Institut Pasteur of Shanghai, Shanghai 200025, People's Republic of China, Jianghan University, College of Life Science, Department of Biotechnology, Wuhan 430056, People's Republic of China
| | - Yidan Zhou
- State Key Laboratory of Virology and Department of Immunology and Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, Wuhan University School of Medicine, Wuhan 430071, People's Republic of China, Department of Microbiology, Second Military Medical University, Shanghai 200433, People's Republic of China, The Unit of Viral Hepatitis, Institut Pasteur of Shanghai, Shanghai 200025, People's Republic of China, Jianghan University, College of Life Science, Department of Biotechnology, Wuhan 430056, People's Republic of China
| | - Zhongtian Qi
- State Key Laboratory of Virology and Department of Immunology and Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, Wuhan University School of Medicine, Wuhan 430071, People's Republic of China, Department of Microbiology, Second Military Medical University, Shanghai 200433, People's Republic of China, The Unit of Viral Hepatitis, Institut Pasteur of Shanghai, Shanghai 200025, People's Republic of China, Jianghan University, College of Life Science, Department of Biotechnology, Wuhan 430056, People's Republic of China
| | - Xiao-Lian Zhang
- State Key Laboratory of Virology and Department of Immunology and Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, Wuhan University School of Medicine, Wuhan 430071, People's Republic of China, Department of Microbiology, Second Military Medical University, Shanghai 200433, People's Republic of China, The Unit of Viral Hepatitis, Institut Pasteur of Shanghai, Shanghai 200025, People's Republic of China, Jianghan University, College of Life Science, Department of Biotechnology, Wuhan 430056, People's Republic of China
| |
Collapse
|
39
|
Laminin-121--recombinant expression and interactions with integrins. Matrix Biol 2010; 29:484-93. [PMID: 20566382 DOI: 10.1016/j.matbio.2010.05.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2010] [Revised: 05/17/2010] [Accepted: 05/18/2010] [Indexed: 11/22/2022]
Abstract
Laminin-121, previously referred as to laminin-3, was expressed recombinantly in human embryonic kidney (HEK) 293 cells by triple transfection of full-length cDNAs encoding mouse laminin α1, β2 and γ1 chains. The recombinant laminin-121 was purified using Heparin-Sepharose followed by molecular sieve chromatography and shown to be correctly folded by electron microscopy and circular dichroism (CD). The CD spectra of recombinant laminin-121 were very similar to those of laminin-111 isolated from Engelbreth-Holm-Swarm tumor (EHS-laminin) but its T(m) value was smaller than EHS-laminin and recombinant lamnin-111 suggesting that the replacement of the β chain reduced the stability of the coiled-coil structure of laminin-121. Its binding to integrins was compared with EHS-laminin, laminin-3A32 purified from murine epidermal cell line and recombinantly expressed laminins-111, -211 and -221. Laminin-121 showed the highest affinity to α6β1 and α7β1 integrins and furthermore, laminin-121 most effectively supported neurite outgrowth. Together, this suggests that the β2 laminins have higher affinity for integrins than the β1 laminins.
Collapse
|
40
|
Ko MS, Marinkovich MP. Role of dermal-epidermal basement membrane zone in skin, cancer, and developmental disorders. Dermatol Clin 2010; 28:1-16. [PMID: 19945611 DOI: 10.1016/j.det.2009.10.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The dermal-epidermal basement membrane zone is an important epithelial and stromal interface, consisting of an intricately organized collection of intracellular, transmembrane, and extracellular matrix proteins. The basement membrane zone has several main functions including acting as a permeability barrier, forming an adhesive interface between epithelial cells and the underlying matrix, and controlling cellular organization and differentiation. This article identifies key molecular players of the dermal-epidermal membrane zone, and highlights recent research studies that have identified structural and functional roles of these components in the context of various blistering, neoplastic, and developmental syndromes.
Collapse
Affiliation(s)
- Myung S Ko
- Program in Epithelial Biology, Stanford University School of Medicine, 269 Campus Drive, Stanford, CA 94305, USA
| | | |
Collapse
|
41
|
Anderson C, Thorsteinsdóttir S, Borycki AG. Sonic hedgehog-dependent synthesis of laminin alpha1 controls basement membrane assembly in the myotome. Development 2009; 136:3495-504. [PMID: 19783738 DOI: 10.1242/dev.036087] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Basement membranes have essential structural and signalling roles in tissue morphogenesis during embryonic development, but the mechanisms that control their formation are still poorly understood. Laminins are key components of basement membranes and are thought to be essential for initiation of basement membrane assembly. Here, we report that muscle progenitor cells populating the myotome migrate aberrantly in the ventral somite in the absence of sonic hedgehog (Shh) signalling, and we show that this defect is due to the failure to form a myotomal basement membrane. We reveal that expression of Lama1, which encodes laminin alpha1, a subunit of laminin-111, is not activated in Shh(-/-) embryos. Recovery of Lama1 expression or addition of exogenous laminin-111 to Shh(-/-);Gli3(-/-) embryos restores the myotomal basement membrane, demonstrating that laminin-111 is necessary and sufficient to initiate assembly of the myotomal basement membrane. This study uncovers an essential role for Shh signalling in the control of laminin-111 synthesis and in the initiation of basement membrane assembly in the myotome. Furthermore, our data indicate that laminin-111 function cannot be compensated by laminin-511.
Collapse
Affiliation(s)
- Claire Anderson
- Department of Biomedical Science, University of Sheffield, Sheffield, UK
| | | | | |
Collapse
|
42
|
Hidalgo M, Sirour C, Bello V, Moreau N, Beaudry M, Darribère T. In vivo analyzes of dystroglycan function during somitogenesis in Xenopus laevis. Dev Dyn 2009; 238:1332-45. [PMID: 19086027 DOI: 10.1002/dvdy.21814] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Dystroglycan (Dg) is a cell adhesion receptor for laminin that has been reported to play a role in skeletal muscle cell stability, cytoskeletal organization, cell polarity, and signaling. Here we show that Dg is expressed at both the notochord/somite and the intersomitic boundaries, where laminin and fibronectin are accumulated during somitogenesis. Inhibition of Dg function with morpholino antisense oligonucleotides or a dominant negative mutant results in the normal segmentation of the presomitic mesoderm but affects the number, the size, and the integrity of somites. Depletion of Dg disrupts proliferation and alignment of myoblasts without affecting XMyoD and XMRF4 expression. It also leads to defects in laminin deposition at the intersomitic junctions, whereas expression of integrin beta1 subunits and fibronectin assembly occur normally. Our results show that Dg is critical for both proliferation and elongation of somitic cells and that the Dg-cytoplasmic domain is required for the laminin assembly at the intersomitic boundaries. Developmental Dynamics 238:1332-1345, 2009. (c) 2008 Wiley-Liss, Inc.
Collapse
Affiliation(s)
- Magdalena Hidalgo
- Université Pierre et Marie Curie Paris 6 UMR CNRS 7622, Laboratoire de Biologie du Développement, équipe Matrice Extracellulaire et Développement, Paris, France
| | | | | | | | | | | |
Collapse
|
43
|
Carafoli F, Clout NJ, Hohenester E. Crystal structure of the LG1-3 region of the laminin alpha2 chain. J Biol Chem 2009; 284:22786-92. [PMID: 19553699 PMCID: PMC2755686 DOI: 10.1074/jbc.m109.026658] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2009] [Revised: 05/28/2009] [Indexed: 11/17/2022] Open
Abstract
Laminins are large heterotrimeric glycoproteins with many essential functions in basement membrane assembly and function. Cell adhesion to laminins is mediated by a tandem of five laminin G-like (LG) domains at the C terminus of the alpha chain. Integrin binding requires an intact LG1-3 region, as well as contributions from the coiled coil formed by the alpha, beta, and gamma chains. We have determined the crystal structure at 2.8-A resolution of the LG1-3 region of the laminin alpha2 chain (alpha 2LG1-3). The three LG domains adopt typical beta-sandwich folds, with canonical calcium binding sites in LG1 and LG2. LG2 and LG3 interact through a substantial interface, but LG1 is completely dissociated from the LG2-3 pair. We suggest that the missing gamma chain tail may be required to stabilize the interaction between LG1 and LG2-3 in the biologically active conformation. A global analysis of N-linked glycosylation sites shows that the beta-sandwich faces of LG1 are free of carbohydrate modifications in all five laminin alpha chains, suggesting that these surfaces may harbor the integrin binding site. The alpha 2LG1-3 structure provides the first atomic view of the integrin binding region of laminins.
Collapse
Affiliation(s)
- Federico Carafoli
- From the Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Naomi J. Clout
- From the Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Erhard Hohenester
- From the Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| |
Collapse
|
44
|
McKee KK, Capizzi S, Yurchenco PD. Scaffold-forming and Adhesive Contributions of Synthetic Laminin-binding Proteins to Basement Membrane Assembly. J Biol Chem 2009; 284:8984-94. [PMID: 19189961 PMCID: PMC2659255 DOI: 10.1074/jbc.m809719200] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2008] [Indexed: 01/21/2023] Open
Abstract
Laminins that possess three short arms contribute to basement membrane assembly by anchoring to cell surfaces, polymerizing, and binding to nidogen and collagen IV. Although laminins containing the alpha4 and alpha5 subunits are expressed in alpha2-deficient congenital muscular dystrophy, they may be ineffective substitutes because they bind weakly to cell surfaces and/or because they lack the third arm needed for polymerization. We asked whether linker proteins engineered to bind to deficient laminins that provide such missing activities would promote basement membrane assembly in a Schwann cell model. A chimeric fusion protein (alphaLNNd) that adds a short arm terminus to laminin through the nidogen binding locus was generated and compared with the dystrophy-ameliorating protein miniagrin (mAgrin) that binds to the laminin coiled-coil dystroglycan and sulfatides. alphaLNNd was found to mediate laminin binding to collagen IV, to bind to galactosyl sulfatide, and to selectively convert alpha-short arm deletion-mutant laminins LmDeltaalphaLN and LmDeltaalphaLN-L4b into polymerizing laminins. This protein enabled polymerization-deficient laminin but not an adhesion-deficient laminin lacking LG domains (LmDeltaLG) to assemble an extracellular matrix on Schwann cell surfaces. mAgrin, on the other hand, enabled LmDeltaLG to form an extracellular matrix on cell surfaces without increasing accumulation of non-polymerizing laminins. These gain-of-function studies reveal distinct polymerization and anchorage contributions to basement membrane assembly in which the three different LN domains mediate the former, and the LG domains provide primary anchorage with secondary contributions from the alphaLN domain. These findings may be relevant for an understanding of the pathogenesis and treatment of laminin deficiency states.
Collapse
Affiliation(s)
- Karen K McKee
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Piscataway, New Jersey 08854, USA
| | | | | |
Collapse
|
45
|
Gao J, DeRouen MC, Chen CH, Nguyen M, Nguyen NT, Ido H, Harada K, Sekiguchi K, Morgan BA, Miner JH, Oro AE, Marinkovich MP. Laminin-511 is an epithelial message promoting dermal papilla development and function during early hair morphogenesis. Genes Dev 2008; 22:2111-24. [PMID: 18676816 DOI: 10.1101/gad.1689908] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Hair morphogenesis takes place through reciprocal epithelial and mesenchymal signaling; however, the mechanisms controlling signal exchange are poorly understood. Laminins are extracellular proteins that play critical roles in adhesion and signaling. Here we demonstrate the mechanism of how laminin-511 controls hair morphogenesis. Dermal papilla (DP) from laminin-511 mutants showed developmental defects by E16.5, including a failure to maintain expression of the key morphogen noggin. This maintenance was critical as exogenous introduction of noggin or sonic hedgehog (Shh) produced downstream from noggin was sufficient to restore hair follicle development in lama5(-/-) (laminin-511-null) skin. Hair development required the beta1 integrin binding but not the heparin binding domain of laminin-511. Previous studies demonstrated that Shh signaling requires primary cilia, microtubule-based signaling organelles. Laminin-511 mutant DP showed decreased length and structure of primary cilia in vitro and in vivo. Laminin-511, but not laminin-111, restored primary cilia formation in lama5(-/-) mesenchyme and triggered noggin expression in an Shh- and PDGF-dependent manner. Inhibition of laminin-511 receptor beta1 integrin disrupted DP primary cilia formation as well as hair development. These studies show that epithelial-derived laminin-511 is a critical early signal that directs ciliary function and DP maintenance as a requirement for hair follicle downgrowth.
Collapse
Affiliation(s)
- Jing Gao
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Ido H, Ito S, Taniguchi Y, Hayashi M, Sato-Nishiuchi R, Sanzen N, Hayashi Y, Futaki S, Sekiguchi K. Laminin isoforms containing the gamma3 chain are unable to bind to integrins due to the absence of the glutamic acid residue conserved in the C-terminal regions of the gamma1 and gamma2 chains. J Biol Chem 2008; 283:28149-57. [PMID: 18697739 DOI: 10.1074/jbc.m803553200] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Laminins are the major cell adhesive proteins in basement membranes, and consist of three subunits termed alpha, beta, and gamma. Recently, we found that the Glu residue at the third position from the C termini of the gamma1 and gamma2 chains is critically involved in integrin binding by laminins. However, the gamma3 chain lacks this Glu residue, suggesting that laminin isoforms containing the gamma3 chain may be unable to bind to integrins. To address this possibility, we expressed the E8 fragment of laminin-213 and found that it was incapable of binding to integrins. Similarly, the E8 fragment of laminin-113 was expressed and also found to be inactive in binding to integrins, confirming the distinction between the integrin binding activities of gamma3 chain-containing isoforms and those containing the gamma1 or gamma2 chain. To further address the importance of the Glu residue, we swapped the C-terminal four amino acids of the gamma3 chain with the C-terminal nine amino acids of the gamma1 chain, which contain the Glu residue. The resulting chimeric E8 fragment of laminin-213 became fully active in integrin binding, whereas replacement with the nine amino acids of the gamma1 chain after substitution of Gln for the conserved Glu residue failed to restore the integrin binding activity. These results provide both loss-of-function and gain-of-function evidence that laminin isoforms containing the gamma3 chain are unable to bind to integrins due to the absence of the conserved Glu residue, which should play a critical role in integrin binding by laminins.
Collapse
Affiliation(s)
- Hiroyuki Ido
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Kvist AJ, Nyström A, Hultenby K, Sasaki T, Talts JF, Aspberg A. The major basement membrane components localize to the chondrocyte pericellular matrix — A cartilage basement membrane equivalent? Matrix Biol 2008; 27:22-33. [PMID: 17825545 DOI: 10.1016/j.matbio.2007.07.007] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Revised: 07/30/2007] [Accepted: 07/30/2007] [Indexed: 11/23/2022]
Abstract
In this study, we demonstrate that articular cartilage chondrocytes are surrounded by the defining basement membrane proteins laminin, collagen type IV, nidogen and perlecan, and suggest that these form the functional equivalent of a basement membrane. We found by real-time PCR that mouse chondrocytes express these four cardinal components of basement membranes and demonstrated by immunohistochemistry that the proteins are present in bovine and mouse cartilage tissues and are deposited in a thin pericellular structure. Immunoelectron microscopy confirmed high laminin concentration in the pericellular matrix. In cartilage from newborn mice, basement membrane components are widespread in the territorial and interterritorial matrix, while in mature cartilage of adult mice the basement membrane components are localized mainly to a narrow pericellular zone. With progression into old age, this layer becomes less distinct, especially in areas of obvious mechanical attrition. Interestingly, individual laminin subunits were located in different zones of the cartilage, with laminin alpha1 showing preferential localization around a select population of superficial layer chondrocytes. We propose that the chondrocyte, like several other cell types of mesenchymal origin, is surrounded by the functional equivalent of a basement membrane. This structure is presumably involved in maintaining chondrocyte phenotype and viability and may well allow a new understanding of cartilage development and provide clues to the progression of degenerative joint disorders.
Collapse
Affiliation(s)
- Alexander J Kvist
- Department of Experimental Medical Sciences, Lund University, SE-22184 Lund, Sweden
| | | | | | | | | | | |
Collapse
|
48
|
Zhou Y, Jiang D, Thomason DB, Jarrett HW. Laminin-induced activation of Rac1 and JNKp46 is initiated by Src family kinases and mimics the effects of skeletal muscle contraction. Biochemistry 2007; 46:14907-16. [PMID: 18044967 DOI: 10.1021/bi701384k] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Binding of laminin to dystroglycan in the dystrophin glycoprotein complex causes signaling through dystroglycan-syntrophin-grb2-SOS1-Rac1-PAK1-JNK. Laminin binding also causes syntrophin tyrosine phosphorylation to initiate signaling. The kinase responsible was investigated here. PP2 and SU6656, specific inhibitors of Src family kinases, decreased the amount of phosphotyrosine syntrophin and decreased the level of active Rac1 in laminin-treated myoblasts, myotubes, or skeletal muscle microsomes. c-Src and c-Fyn both phosphorylate syntrophin, and inhibition of either with specific siRNAs diminishes the level of syntrophin phosphorylation. When the rat gastrocnemius was contracted, the level of Rac1 activation increased compared to that of the relaxed control muscle and Rac1 colocalized with beta-dystroglycan. Similar results were obtained when the muscle was stretched. Contracted muscle also contained more activated c-Jun N-terminal kinase, JNKp46. E3, an expressed protein containing only laminin domains LG4 and LG5, increased the rate of proliferation of myoblasts, and PP2 prevented cell proliferation. In addition, Src family kinases colocalized with activated Rac1 and with laminin-Sepharose in solid-phase binding assays. Thus, contraction, stretching, or laminin binding causes recruitment of Src family kinase to the dystrophin glycoprotein complex, activating Rac1 and inducing downstream signaling. The DGC likely represents a mechanoreceptor in skeletal muscle-regulating muscle growth in response to muscle activity. Src family kinases play an initiating and critical role.
Collapse
Affiliation(s)
- YanWen Zhou
- Department of Chemistry, University of Texas, San Antonio, Texas 78249, USA
| | | | | | | |
Collapse
|
49
|
von der Mark H, Pöschl E, Lanig H, Sasaki T, Deutzman R, von der Mark K. Distinct Acidic Clusters and Hydrophobic Residues in the Alternative Splice Domains X1 and X2 of α7 Integrins Define Specificity for Laminin Isoforms. J Mol Biol 2007; 371:1188-203. [PMID: 17618648 DOI: 10.1016/j.jmb.2007.05.074] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2007] [Revised: 04/28/2007] [Accepted: 05/24/2007] [Indexed: 11/28/2022]
Abstract
The binding specificity of alpha7beta1 integrins for different laminin isoforms is defined by the X1 and X2 splice domains located in the beta-propeller domain of the alpha7 subunit. In order to gain insight into the mechanism of specific laminin-integrin interactions, we defined laminin-binding epitopes of the alpha7X1 and -X2 domains by single amino acid substitutions and domain swapping between X1 and X2. The interaction of mutated, recombinantly prepared alpha7X1beta1 and alpha7X2beta1 heterodimers with various laminin isoforms was studied by surface plasmon resonance and solid phase binding assays. The data show that distinct clusters of surface-exposed acidic residues located in different positions of the X1 and the X2 loops are responsible for the specific recognition of laminins. These residues are conserved between the respective X1 or X2 splice domains of the alpha7 chains of different species, some also in the corresponding X1/X2 splice domains of alpha6 integrin. Interestingly, ligand binding was also modulated by mutating surface-exposed hydrophobic residues (alpha7X1L205, alpha7X2Y208) at positions corresponding to the fibronectin binding synergy site in alpha5beta1 integrin. Mutations in X1 that affected binding to laminin-1 also affected binding to laminin-8 and -10, but not to the same extent, thus allowing conclusions on the specific role of individual surface epitopes in the selective recognition of laminin-1 versus laminins -8 and -10. The role of the identified epitopes was confirmed by molecular dynamics simulations of wild-type integrins and several inactivating mutations. The analysis of laminin isoform interactions with various X1/X2 chimaera lend further support to the key role of negative surface charges and pointed to an essential contribution of the N-terminal TARVEL sequence of the X1 domain for recognition of laminin-8 and -10. In conclusion, specific surface epitopes containing charged and hydrophobic residues are essential for ligand binding and define specific interactions with laminin isoforms.
Collapse
Affiliation(s)
- Helga von der Mark
- Department of Experimental Medicine I, Nikolaus - Fiebiger Center of Molecular Medicine, University of Erlangen - Nuernberg, 91054 Erlangen, Germany
| | | | | | | | | | | |
Collapse
|
50
|
Mankelow TJ, Burton N, Stefansdottir FO, Spring FA, Parsons SF, Pedersen JS, Oliveira CLP, Lammie D, Wess T, Mohandas N, Chasis JA, Brady RL, Anstee DJ. The Laminin 511/521-binding site on the Lutheran blood group glycoprotein is located at the flexible junction of Ig domains 2 and 3. Blood 2007; 110:3398-406. [PMID: 17638854 PMCID: PMC2200917 DOI: 10.1182/blood-2007-06-094748] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Lutheran blood group glycoprotein, first discovered on erythrocytes, is widely expressed in human tissues. It is a ligand for the alpha5 subunit of Laminin 511/521, an extracellular matrix protein. This interaction may contribute to vaso-occlusive events that are an important cause of morbidity in sickle cell disease. Using x-ray crystallography, small-angle x-ray scattering, and site-directed mutagenesis, we show that the extracellular region of Lutheran forms an extended structure with a distinctive bend between the second and third immunoglobulin-like domains. The linker between domains 2 and 3 appears to be flexible and is a critical determinant in maintaining an overall conformation for Lutheran that is capable of binding to Laminin. Mutagenesis studies indicate that Asp312 of Lutheran and the surrounding cluster of negatively charged residues in this linker region form the Laminin-binding site. Unusually, receptor binding is therefore not a function of the domains expected to be furthermost from the plasma membrane. These studies imply that structural flexibility of Lutheran may be essential for its interaction with Laminin and present a novel opportunity for the development of therapeutics for sickle cell disease.
Collapse
Affiliation(s)
- Tosti J Mankelow
- Bristol Institute for Transfusion Sciences, National Blood Service, Southmead Road, Bristol, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|