1
|
Cloutier G, Seltana A, Fallah S, Beaulieu JF. Integrin α7β1 represses intestinal absorptive cell differentiation. Exp Cell Res 2023; 430:113723. [PMID: 37499931 DOI: 10.1016/j.yexcr.2023.113723] [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: 03/28/2023] [Revised: 07/14/2023] [Accepted: 07/22/2023] [Indexed: 07/29/2023]
Abstract
Intestinal epithelial cell differentiation is a highly controlled and orderly process occurring in the crypt so that cells migrating out to cover the villi are already fully functional. Absorptive cell precursors, which originate from the stem cell population located in the lower third of the crypt, are subject to several cycles of amplification in the transit amplifying (TA) zone, before reaching the terminal differentiation compartment located in the upper third. There is a large body of evidence that absorptive cell differentiation is halted in the TA zone through various epigenetic, transcriptional and intracellular signalling events or mechanisms allowing the transient expansion of this cell population but how these mechanisms are themself regulated remains obscure. One clue can be found in the epithelial cell-matrix microenvironment located all along the crypt-villus axis. Indeed, a previous study from our group revealed that α5-subunit containing laminins such as lamimin-511 and 512 inhibit early stages of differentiation in Caco-2/15 cells. Among potential receptors for laminin 511/512 is the integrin α7β1, which has previously been reported to be expressed in the human intestinal crypts and in early stages of Caco-2/15 cell differentiation. In this study, the effects of knocking down ITGA7 in Caco-2/15 cells were studied using shRNA and CRISPR/Cas9 strategies. Abolition of the α7 integrin subunit resulted in a significant increase in the level of differentiation and polarization markers as well as the morphological features of intestinal cells. Activities of focal adhesion kinase and Src kinase were both reduced in α7-knockdown cells while the three major intestinal pro-differentiation factors CDX2, HNFα1 and HNF4α were overexpressed. Two epigenetic events associated with intestinal differentiation, the reduction of tri-methylated lysine 27 on histone H3 and the increase of acetylation of histone H4 were also observed in α7-knockdown cells. On the other hand, the ablation of α7 had no effect on cell proliferation. In conclusion, these data indicate that integrin α7β1 acts as a major repressor of absorptive cell terminal differentiation in the Caco-2/15 cell model and suggest that the laminin-α7β1 integrin interaction occurring in the transit amplifying zone of the adult intestine is involved in the transient halting of absorptive cell terminal differentiation.
Collapse
Affiliation(s)
- Gabriel Cloutier
- Laboratory of Intestinal Physiopathology, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, J1H 5N4, Canada
| | - Amira Seltana
- Laboratory of Intestinal Physiopathology, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, J1H 5N4, Canada
| | - Sepideh Fallah
- Laboratory of Intestinal Physiopathology, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, J1H 5N4, Canada
| | - Jean-François Beaulieu
- Laboratory of Intestinal Physiopathology, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, J1H 5N4, Canada.
| |
Collapse
|
2
|
Bugiardini E, Nunes AM, Oliveira‐Santos A, Dagda M, Fontelonga TM, Barraza‐Flores P, Pittman AM, Morrow JM, Parton M, Houlden H, Elliott PM, Syrris P, Maas RP, Akhtar MM, Küsters B, Raaphorst J, Schouten M, Kamsteeg E, van Engelen B, Hanna MG, Phadke R, Lopes LR, Matthews E, Burkin DJ. Integrin α7 Mutations Are Associated With Adult-Onset Cardiac Dysfunction in Humans and Mice. J Am Heart Assoc 2022; 11:e026494. [PMID: 36444867 PMCID: PMC9851448 DOI: 10.1161/jaha.122.026494] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 11/02/2022] [Indexed: 11/30/2022]
Abstract
Background Integrin α7β1 is a major laminin receptor in skeletal and cardiac muscle. In skeletal muscle, integrin α7β1 plays an important role during muscle development and has been described as an important modifier of skeletal muscle diseases. The integrin α7β1 is also highly expressed in the heart, but its precise role in cardiac function is unknown. Mutations in the integrin α7 gene (ITGA7) have been reported in children with congenital myopathy. Methods and Results In this study, we described skeletal and cardiac muscle pathology in Itga7-/- mice and 5 patients from 2 unrelated families with ITGA7 mutations. Proband in family 1 presented a homozygous c.806_818del [p.S269fs] variant, and proband in family 2 was identified with 2 intron variants in the ITGA7 gene. The complete absence of the integrin α7 protein in muscle supports the ITGA7 mutations are pathogenic. We performed electrocardiography, echocardiography, or cardiac magnetic resonance imaging, and histological biopsy analyses in patients with ITGA7 deficiency and Itga7-/- mice. The patients exhibited cardiac dysrhythmia and dysfunction from the third decade of life and late-onset respiratory insufficiency, but with relatively mild limb muscle involvement. Mice demonstrated corresponding abnormalities in cardiac conduction and contraction as well as diaphragm muscle fibrosis. Conclusions Our data suggest that loss of integrin α7 causes a novel form of adult-onset cardiac dysfunction indicating a critical role for the integrin α7β1 in normal cardiac function and highlights the need for long-term cardiac monitoring in patients with ITGA7-related congenital myopathy.
Collapse
Affiliation(s)
- Enrico Bugiardini
- Queen Square Centre for Neuromuscular DiseasesQueen Square Institute of Neurology, UCL and National Hospital for Neurology and NeurosurgeryLondonUnited Kingdom
| | - Andreia M. Nunes
- Department of PharmacologyUniversity of Nevada Reno, School of Medicine, Center for Molecular MedicineRenoNV
| | - Ariany Oliveira‐Santos
- Department of PharmacologyUniversity of Nevada Reno, School of Medicine, Center for Molecular MedicineRenoNV
| | - Marisela Dagda
- Department of PharmacologyUniversity of Nevada Reno, School of Medicine, Center for Molecular MedicineRenoNV
| | - Tatiana M. Fontelonga
- Department of PharmacologyUniversity of Nevada Reno, School of Medicine, Center for Molecular MedicineRenoNV
| | - Pamela Barraza‐Flores
- Department of PharmacologyUniversity of Nevada Reno, School of Medicine, Center for Molecular MedicineRenoNV
| | - Alan M. Pittman
- Department of Neuromuscular DiseasesUCL Queen Square Institute of NeurologyLondonUnited Kingdom
- St George’sUniversity of LondonLondonUnited Kingdom
| | - Jasper M. Morrow
- Queen Square Centre for Neuromuscular DiseasesQueen Square Institute of Neurology, UCL and National Hospital for Neurology and NeurosurgeryLondonUnited Kingdom
| | - Matthew Parton
- Queen Square Centre for Neuromuscular DiseasesQueen Square Institute of Neurology, UCL and National Hospital for Neurology and NeurosurgeryLondonUnited Kingdom
| | - Henry Houlden
- Department of Neuromuscular DiseasesUCL Queen Square Institute of NeurologyLondonUnited Kingdom
| | - Perry M. Elliott
- Barts Heart Centre, Barts Health NHS TrustLondonUnited Kingdom
- Centre for Heart Muscle DiseaseInstitute of Cardiovascular Science, University College LondonLondonUnited Kingdom
| | - Petros Syrris
- Centre for Heart Muscle DiseaseInstitute of Cardiovascular Science, University College LondonLondonUnited Kingdom
| | - Roderick P. Maas
- Department of Neurology, Donders Institute for Brain, Cognition and BehaviourRadboud University Medical CenterNijmegenThe Netherlands
| | - Mohammed M. Akhtar
- Barts Heart Centre, Barts Health NHS TrustLondonUnited Kingdom
- Centre for Heart Muscle DiseaseInstitute of Cardiovascular Science, University College LondonLondonUnited Kingdom
| | - Benno Küsters
- Department of PathologyRadboud University Medical CenterNijmegenThe Netherlands
| | - Joost Raaphorst
- Department of Neurology, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam NeuroscienceAmsterdamThe Netherlands
| | - Meyke Schouten
- Department of Human GeneticsRadboud University Medical CenterNijmegenThe Netherlands
| | - Erik‐Jan Kamsteeg
- Department of Human GeneticsRadboud University Medical CenterNijmegenThe Netherlands
| | - Baziel van Engelen
- Department of Neurology, Donders Institute for Brain, Cognition and BehaviourRadboud University Medical CenterNijmegenThe Netherlands
| | - Michael G. Hanna
- Queen Square Centre for Neuromuscular DiseasesQueen Square Institute of Neurology, UCL and National Hospital for Neurology and NeurosurgeryLondonUnited Kingdom
| | - Rahul Phadke
- Division of NeuropathologyUCL Institute of NeurologyLondonUnited Kingdom
- Dubowitz Neuromuscular Centre, MRC Centre for Neuromuscular DiseasesUCL Great Ormond Street Institute of Child HealthLondonUnited Kingdom
| | - Luis R. Lopes
- Barts Heart Centre, Barts Health NHS TrustLondonUnited Kingdom
- Centre for Heart Muscle DiseaseInstitute of Cardiovascular Science, University College LondonLondonUnited Kingdom
| | - Emma Matthews
- The Atkinson Morley Neuromuscular Centre and Regional Neurosciences CentreSt George’s University Hospitals NHS Foundation TrustLondonUnited Kingdom
- Molecular and Clinical Sciences Research Institute, St George’s University of LondonLondonUnited Kingdom
| | - Dean J. Burkin
- Department of PharmacologyUniversity of Nevada Reno, School of Medicine, Center for Molecular MedicineRenoNV
| |
Collapse
|
3
|
Gonzalez Porras MA, Stojkova K, Vaicik MK, Pelowe A, Goddi A, Carmona A, Long B, Qutub AA, Gonzalez A, Cohen RN, Brey EM. Integrins and extracellular matrix proteins modulate adipocyte thermogenic capacity. Sci Rep 2021; 11:5442. [PMID: 33686208 PMCID: PMC7940610 DOI: 10.1038/s41598-021-84828-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 02/08/2021] [Indexed: 12/13/2022] Open
Abstract
Obesity and the metabolic disease epidemic has led to an increase in morbidity and mortality. A rise in adipose thermogenic capacity via activation of brown or beige fat is a potential treatment for metabolic diseases. However, an understanding of how local factors control adipocyte fate is limited. Mice with a null mutation in the laminin α4 (LAMA4) gene (KO) exhibit resistance to obesity and enhanced expression of thermogenic fat markers in white adipose tissue (WAT). In this study, changes in WAT extracellular matrix composition in the absence of LAMA4 were evaluated using liquid chromatography/tandem mass spectrometry. KO-mice showed lower levels of collagen 1A1 and 3A1, and integrins α7 (ITA7) and β1 (ITB1). ITA7-ITB1 and collagen 1A1-3A1 protein levels were lower in brown adipose tissue compared to WAT in wild-type mice. Immunohistochemical staining confirmed lower levels and different spatial distribution of ITA7 in KO-WAT. In culture studies, ITA7 and LAMA4 levels decreased following a 12-day differentiation of adipose-derived stem cells into beige fat, and knock-down of ITA7 during differentiation increased beiging. These results demonstrate that extracellular matrix interactions regulate adipocyte thermogenic capacity and that ITA7 plays a role in beige adipose formation. A better understanding of the mechanisms underlying these interactions can be used to improve systemic energy metabolism and glucose homeostasis.
Collapse
Affiliation(s)
- Maria A Gonzalez Porras
- Department of Biomedical Engineering and Chemical Engineering, AET 1.102, The University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX, 78249, USA
| | - Katerina Stojkova
- Department of Biomedical Engineering and Chemical Engineering, AET 1.102, The University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX, 78249, USA
| | - Marcella K Vaicik
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA
| | - Amanda Pelowe
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Anna Goddi
- Section of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Alanis Carmona
- Section of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Byron Long
- Department of Biomedical Engineering and Chemical Engineering, AET 1.102, The University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX, 78249, USA
| | - Amina A Qutub
- Department of Biomedical Engineering and Chemical Engineering, AET 1.102, The University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX, 78249, USA
| | - Anjelica Gonzalez
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Ronald N Cohen
- Section of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Eric M Brey
- Department of Biomedical Engineering and Chemical Engineering, AET 1.102, The University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX, 78249, USA.
| |
Collapse
|
4
|
Kon S, Atakilit A, Sheppard D. Short form of α9 promotes α9β1 integrin-dependent cell adhesion by modulating the function of the full-length α9 subunit. Exp Cell Res 2011; 317:1774-84. [PMID: 21515257 DOI: 10.1016/j.yexcr.2011.04.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Revised: 04/07/2011] [Accepted: 04/11/2011] [Indexed: 11/25/2022]
Abstract
The α9β1 integrin is a multifunctional receptor that interacts with a variety of ligands including vascular cell adhesion molecule 1, tenascin-C, and osteopontin. A 2.3-kb truncated form of α9 integrin subunit cDNA was identified by searching the Medline database. This splice variant, which we called the short form of α9 integrin (SFα9), encodes a 632-aa isoform lacking transmembrane and cytoplasmic domains, and its authentic expression was verified by PCR and Western blotting. SFα9 is expressed on the cell surface but cannot bind ligand in the absence of the full-length α9 subunit. Over-expression of SFα9 in cells expressing full-length α9 promotes α9-dependent cell adhesion. This promoting effect of SFα9 requires the authentic cytoplasmic domain of the co-expressed full-length α9 subunit. Thus, SFα9 is a novel functional modulator of α9β1 integrin by inside-out signaling.
Collapse
Affiliation(s)
- Shigeyuki Kon
- Lung Biology Center, Department of Medicine, University of California, San Francisco, CA 94143, USA
| | | | | |
Collapse
|
5
|
Van Hoof D, Dormeyer W, Braam SR, Passier R, Monshouwer-Kloots J, Ward-van Oostwaard D, Heck AJR, Krijgsveld J, Mummery CL. Identification of cell surface proteins for antibody-based selection of human embryonic stem cell-derived cardiomyocytes. J Proteome Res 2010; 9:1610-8. [PMID: 20088484 DOI: 10.1021/pr901138a] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The absence of identified cell surface proteins and corresponding antibodies to most differentiated derivatives of human embryonic stem cells (hESCs) has largely limited selection of specific cell types from mixed cell populations to genetic approaches. Here, we describe the use of mass spectrometry (MS)-based proteomics on cell membrane proteins isolated from hESCs that were differentiated into cardiomyocytes to identify candidate proteins for this particular lineage. Quantitative MS distinguished cardiomyocyte-specific plasma membrane proteins that were highly enriched or detected only in cardiomyocytes derived from hESCs and human fetal hearts compared with a heterogeneous pool of hESC-derived differentiated cells. For several candidates, cardiomyocyte-specific expression and cell surface localization were verified by conventional antibody-based methodologies. Using an antibody against elastin microfibril interfacer 2 (EMILIN2), we demonstrate that cardiomyocytes can be sorted from live cell populations. Besides showing that MS-based membrane proteomics is a powerful tool to identify candidate proteins that allow purification of specific cell lineages from heterogeneous populations, this approach generated a plasma membrane proteome profile suggesting signaling pathways that control cell behavior.
Collapse
Affiliation(s)
- Dennis Van Hoof
- Hubrecht Institute, Developmental Biology and Stem Cell Research, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Liu J, Gurpur PB, Kaufman SJ. Genetically determined proteolytic cleavage modulates alpha7beta1 integrin function. J Biol Chem 2008; 283:35668-78. [PMID: 18940796 PMCID: PMC2602887 DOI: 10.1074/jbc.m804661200] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2008] [Revised: 10/14/2008] [Indexed: 01/07/2023] Open
Abstract
The dystrophin-glycoprotein complex and the alpha7beta1 integrin are trans-sarcolemmal linkage systems that connect and transduce contractile forces between muscle fibers and the extracellular matrix. alpha7beta1 is the major laminin binding integrin in skeletal muscle. Different functional variants of this integrin are generated by alternative splicing and post-translational modifications such as glycosylation and ADP-ribosylation. Here we report a species-specific difference in alpha7 chains that results from an intra-peptide proteolytic cleavage, by a serine protease, at the 603RRQ605 site. Site-directed mutagenesis of RRQ to GRQ prevents this cleavage. This RRQ sequence in the alpha7 integrin chain is highly conserved among vertebrates but it is absent in mice. Protein structure modeling indicates this cleavage site is located in an open region between the beta-propeller and thigh domains of the alpha7 chain. Compared with the non-cleavable alpha7 chain, the cleaved form enhances cell adhesion and spreading on laminin. Cleavage of the alpha7 chain is elevated upon myogenic differentiation, and this cleavage may be mediated by urokinase-type plasminogen activator. These results suggest proteolytic cleavage is a novel mechanism that regulates alpha7 integrin functions in skeletal muscle, and that the generation of such cleavage sites is another evolutionary mechanism for expanding and modifying protein functions.
Collapse
Affiliation(s)
- Jianming Liu
- Department of Cell and Developmental Biology, University of Illinois, Urbana, Illinois 61801, USA
| | | | | |
Collapse
|
7
|
Boppart MD, Volker SE, Alexander N, Burkin DJ, Kaufman SJ. Exercise promotes alpha7 integrin gene transcription and protection of skeletal muscle. Am J Physiol Regul Integr Comp Physiol 2008; 295:R1623-30. [PMID: 18784336 DOI: 10.1152/ajpregu.00089.2008] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The alpha7beta1 integrin is increased in skeletal muscle in response to injury-producing exercise, and transgenic overexpression of this integrin in mice protects against exercise-induced muscle damage. The present study investigates whether the increase in the alpha7beta1 integrin observed in wild-type mice in response to exercise is due to transcriptional regulation and examines whether mobilization of the integrin at the myotendinous junction (MTJ) is a key determinant in its protection against damage. A single bout of downhill running exercise selectively increased transcription of the alpha7 integrin gene in 5-wk-old wild-type mice 3 h postexercise, and an increased alpha7 chain was detected in muscle sarcolemma adjacent to tendinous tissue immediately following exercise. The alpha7B, but not alpha7A isoform, was found concentrated and colocalized with tenascin-C in muscle fibers lining the MTJ. To further validate the importance of the integrin in the protection against muscle damage following exercise, muscle injury was quantified in alpha7(-/-) mice. Muscle damage was extensive in alpha7(-/-) mice in response to both a single and repeated bouts of exercise and was largely restricted to areas of high MTJ concentration and high mechanical force near the Achilles tendon. These results suggest that exercise-induced muscle injury selectively increases transcription of the alpha7 integrin gene and promotes a rapid change in the alpha7beta integrin at the MTJ. These combined molecular and cellular alterations are likely responsible for integrin-mediated attenuation of exercise-induced muscle damage.
Collapse
Affiliation(s)
- Marni D Boppart
- Department of Kinesiology and Community Health, University of Illinois, Urbana, Illinois, USA
| | | | | | | | | |
Collapse
|
8
|
Bunch TA, Kendall TL, Shakalya K, Mahadevan D, Brower DL. Modulation of ligand binding by alternative splicing of the alphaPS2 integrin subunit. J Cell Biochem 2007; 102:211-23. [PMID: 17372926 DOI: 10.1002/jcb.21288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The Drosophila alphaPS2 integrin subunit is found in two isoforms. alphaPS2C contains 25 residues not found in alphaPS2m8, encoded by the alternative eighth exon. Previously, it was shown that cells expressing alphaPS2C spread more effectively than alphaPS2m8 cells on fragments of the ECM protein Tiggrin, and that alphaPS2C-containing integrins are relatively insensitive to depletion of Ca(2+). Using a ligand mimetic probe for Tiggrin affinity (TWOW-1), we show that the affinity of alphaPS2CbetaPS for this ligand is much higher than that of alphaPS2m8betaPS. However, the two isoforms become more similar in the presence of activating levels of Mn(2+). Modeling indicates that the exon 8-encoded residues replace the third beta strand of the third blade of the alpha subunit beta-propeller structure, and generate an exaggerated loop between this and the fourth strand. alphaPS2 subunits with the extra loop structure but with an m8-like third strand, or subunits with a C-like strand but an m8-like short loop, both fail to show alphaPS2C-like affinity for TWOW-1. Surprisingly, a single C > m8-like change at the third strand-loop transition point is sufficient to make alphaPS2C require Ca(2+) for function, despite the absence of any known cation binding site in this region. These data indicate that alternative splicing in integrin alpha subunit extracellular domains may affect ligand affinity via relatively subtle alterations in integrin conformation. These results may have relevance for vertebrate alpha6 and alpha7, which are alternatively spliced at the same site.
Collapse
Affiliation(s)
- Thomas A Bunch
- Department of Molecular and Cellular Biology, Arizona Cancer Center, 1515 N. Campbell Ave., Tucson, Arizona 85724, USA.
| | | | | | | | | |
Collapse
|
9
|
Lai SY, Ziober AF, Lee MN, Cohen NA, Falls EM, Ziober BL. Activated Vav2 modulates cellular invasion through Rac1 and Cdc42 in oral squamous cell carcinoma. Oral Oncol 2007; 44:683-8. [PMID: 17996485 DOI: 10.1016/j.oraloncology.2007.08.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2007] [Revised: 08/29/2007] [Accepted: 08/31/2007] [Indexed: 11/15/2022]
Abstract
The Rho family of GTPases regulates cellular adhesion and motility. Guanine nucleotide exchange factors (GEFs) in turn regulate GTPases by promoting nucleotide exchange from GDP to GTP. We have determined that GTP-bound Rac1 is elevated in invasive oral squamous cell carcinoma (OSCC) cell lines. Because of the critical role of invasion in the progression and metastasis of OSCC, we investigated if the GEF Vav2 modulated Rac1 and Cdc42 activation and thus influenced OSCC invasion. Expression levels of Vav2 did not correlate with invasion but phosphorylated or activated Vav2 was associated with the more invasive cell lines. Transfection of activated Vav2 into the immortalized keratinocyte cell line HaCat and a low-level expressing Vav2 invasive OSCC cell line resulted in increased GTP-bound Rac1 and Cdc42 and increased invasion. Thus, activation of Vav2 appears to modulate cellular invasion through specific regulation of Rac1 and Cdc42 activity in OSCC.
Collapse
Affiliation(s)
- Stephen Y Lai
- Molecular Tumor Biology Laboratory, Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania Health System, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | | | | | | | | | | |
Collapse
|
10
|
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
|
11
|
Samson T, Will C, Knoblauch A, Sharek L, von der Mark K, Burridge K, Wixler V. Def-6, a guanine nucleotide exchange factor for Rac1, interacts with the skeletal muscle integrin chain alpha7A and influences myoblast differentiation. J Biol Chem 2007; 282:15730-42. [PMID: 17403664 DOI: 10.1074/jbc.m611197200] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Integrin alpha7beta1 is the major laminin binding integrin receptor of muscle cells. The alpha7 chain occurs in several splice isoforms, of which alpha7A and alpha7B differ in their intracellular domains only. The fact that the expression of alpha7A and alpha7B is tightly regulated during skeletal muscle development suggests different and distinct roles for both isoforms. However, so far, functional properties and interacting proteins were described for the alpha7B chain only. Using a yeast two-hybrid screen, we have found that Def-6, a guanine nucleotide exchange factor for Rac1, binds to the intracellular domain of the alpha7A subunit. The specificity of the Def-6-alpha7A interaction has been shown by direct yeast two-hybrid binding assays and coprecipitation experiments. This is the first description of an alpha7A-specific and -exclusive interaction, because Def-6 did not bind to any other tested integrin cytoplasmic domain. Interestingly, the binding of Def-6 to alpha7A was abolished, when cells were cotransfected with an Src-related kinase, which is known to phosphorylate Def-6 and stimulate its exchange activity. We found expression of Def-6 was not only restricted to T-lymphocytes as described thus far but in a more widespread manner, including different muscle tissues. In cells, Def-6 is seen in newly forming cell protrusions and focal adhesions, and its localization partially overlaps with the alpha7A integrin receptor. C2C12 myoblasts overexpressing Def-6 show a delay of Rac1 inactivation during myogenic differentiation and abnormal myotube formation. Thus, our data suggest a role for Def-6 in the fine regulation of Rac1 during myogenesis with the integrin alpha7A chain guiding this regulation in a spatio-temporal manner.
Collapse
Affiliation(s)
- Thomas Samson
- Department of Cell and Developmental Biology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599, USA.
| | | | | | | | | | | | | |
Collapse
|
12
|
Chernousov MA, Kaufman SJ, Stahl RC, Rothblum K, Carey DJ. α7β1 integrin is a receptor for laminin-2 on Schwann cells. Glia 2007; 55:1134-44. [PMID: 17598176 DOI: 10.1002/glia.20536] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The Schwann cell basal lamina acts as an organizer of peripheral nerve tissue and influences many aspects of cell behavior during development and regeneration. A principal component of the Schwann cell basal lamina is laminin-2. This study was undertaken to identify Schwann cell receptors for laminin-2. We found that among several Schwann cell integrins that can potentially interact with laminin-2, only alpha7beta1 bound to laminin-2-Sepharose. Dystroglycan, a non-integrin Schwann cell receptor for laminin-2 identified previously, was also found to bind to laminin-2-Sepharose. Antibody to the alpha7 integrin subunit partially inhibited Schwann cell adhesion to laminin-2. Small interfering RNA-mediated suppression of either alpha7 integrin or dystroglycan expression decreased adhesion and spreading of Schwann cells on laminin-2, whereas knocking down both proteins together inhibited adhesion and spreading on laminin-2 almost completely. alpha7 integrin and dystroglycan both colocalized with laminin-2 containing basal lamina tubes in differentiating neuron-Schwann cell cocultures. The alpha7beta1 integrin also coprecipitates with focal adhesion kinase in differentiating cocultures. These findings strongly suggest that alpha7beta1 integrin is a Schwann cell receptor for laminin-2 that provides transmembrane linkage between the Schwann cell basal lamina and cytoskeleton.
Collapse
|
13
|
Ziober AF, Falls EM, Ziober BL. The extracellular matrix in oral squamous cell carcinoma: friend or foe? Head Neck 2006; 28:740-9. [PMID: 16649214 DOI: 10.1002/hed.20382] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Oral squamous cell carcinoma is a disfiguring, highly invasive and metastatic cancer. Despite advances in detection and therapy, many patients will continue to face a poor prognosis. It is well established that the predominate factor determining overall survival in patients with oral squamous cell carcinoma is lymph node involvement. Tumor growth and progression to invasive cancer requires tumor cell interactions with the extracellular matrix. An understanding of how the extracellular matrix influences tumor development and invasion is fundamental in the development of new prognostic indicators and treatment strategies for oral squamous cell carcinoma. In this review, we summarize how changes in the extracellular matrix contribute to oral cancer development.
Collapse
Affiliation(s)
- Amy F Ziober
- Department of Otorhinolaryngology-Head and Neck Surgery, Hospital of the University of Pennsylvania, 3400 Spruce Street, 5 Ravdin Building, Philadelphia PA 19104, USA.
| | | | | |
Collapse
|
14
|
Zhao Z, Gruszczynska-Biegala J, Cheuvront T, Yi H, von der Mark H, von der Mark K, Kaufman SJ, Zolkiewska A. Interaction of the disintegrin and cysteine-rich domains of ADAM12 with integrin alpha7beta1. Exp Cell Res 2004; 298:28-37. [PMID: 15242759 DOI: 10.1016/j.yexcr.2004.04.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2003] [Revised: 04/02/2004] [Indexed: 11/17/2022]
Abstract
We describe a novel interaction between the disintegrin and cysteine-rich (DC) domains of ADAM12 and the integrin alpha7beta1. Integrin alpha7beta1 extracted from human embryonic kidney 293 cells transfected with alpha7 cDNA was retained on an affinity column containing immobilized DC domain of ADAM12. 293 cells stably transfected with alpha7 cDNA adhered to DC-coated wells, and this adhesion was partially inhibited by 6A11 integrin alpha7 function-blocking antibody. The X1 and the X2 extracellular splice variants of integrin alpha7 supported equally well adhesion to the DC protein. Integrin alpha7beta1-mediated cell adhesion to DC had different requirements for Mn2+ than adhesion to laminin. Furthermore, integrin alpha7beta1-mediated cell adhesion to laminin, but not to DC, resulted in efficient cell spreading and phosphorylation of focal adhesion kinase (FAK) at Tyr397. We also show that adhesion of L6 myoblasts to DC is mediated in part by the endogenous integrin alpha7beta1 expressed in these cells. Since integrin alpha7 plays an important role in muscle cell growth, stability, and survival, and since ADAM12 has been implicated in muscle development and regeneration, we postulate that the interaction between ADAM12 and integrin alpha7beta1 may be relevant to muscle development, function, and disease. We also conclude that laminin and the DC domain of ADAM12 represent two functional ligands for integrin alpha7beta1, and adhesion to each of these two ligands via integrin alpha7beta1 triggers different cellular responses.
Collapse
Affiliation(s)
- Zhefeng Zhao
- Department of Biochemistry, Kansas State University, Manhattan 66506, USA
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Zagris N, Christopoulos M, Giakoumaki A. Developmentally regulated expression and functional role of alpha7 integrin in the chick embryo. Dev Growth Differ 2004; 46:299-307. [PMID: 15206960 DOI: 10.1111/j.1440-169x.2004.00747.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Integrin alpha 7 beta 1 is a specific cellular receptor for laminin. In the present work, we studied the distribution pattern of the alpha 7 subunit by immunofluorescence and immunoprecipitation and the role of the integrin by blocking antibodies in early chick embryos. alpha 7 immunoreactivity was first detectable in the neural plate during neural furrow formation (stage HH5, early neurula, Hamburger & Hamilton 1951) and its expression was upregulated in the neural folds during primary neurulation. The alpha 7 expression domain spanned the entire neural tube by stage HH8 (4 somites), and was then downregulated and confined to the neuroepithelial cells in the germinal region near the lumen and the ventrolateral margins of the neural tube in embryos by the onset of stage HH17 (29 somites). Expression of alpha 7 in the neural tube was transient suggesting that alpha 7 functions during neural tube closure and axon guidance and may not be required for neuronal differentiation or for the maintenance of the differentiated cell types. alpha 7 immunoreactivity was strong in the newly formed epithelial somites, although this expression was restricted only to the myotome in the mature somites. The most intense alpha 7 immunoreactivity was detectable in the paired heart primordia and the endoderm apposing the heart primordia in embryos at stage HH8. In the developing heart, alpha 7 immunoreactivity was: (i) intense in the myocardium; (ii) milder in the endocardial cushions of the ventricle; (iii) intense in the sinus venosus; (iv) distinct in the associated blood vessels; and (v) undetectable in the dorsal mesocardium of embryos at stage HH17. Inhibition of function of alpha 7 by blocking antibodies showed that alpha 7 integrin-laminin signaling may play a critical role in tissue organization of the neural plate and neural tube closure, in tissue morphogenesis of the heart tube but not in the directional migration of pre-cardiac cells, and in somite epithelialization but not in segment formation in presomitic mesoderm. In embryos treated with alpha 7 antibody, the formation of median somites in place of a notochord was intriguing and suggested that alpha 7 integrin-laminin signaling may have played a role in segment re-specification in the mesoderm.
Collapse
Affiliation(s)
- Nikolas Zagris
- Division of Genetics and Cell and Developmental Biology, Department of Biology, University of Patras, Patras, Greece.
| | | | | |
Collapse
|
16
|
Yeh MG, Ziober BL, Liu B, Lipkina G, Vizirianakis IS, Kramer RH. The beta1 cytoplasmic domain regulates the laminin-binding specificity of the alpha7X1 integrin. Mol Biol Cell 2003; 14:3507-18. [PMID: 12972542 PMCID: PMC196545 DOI: 10.1091/mbc.e02-12-0824] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
During muscle development, the laminin-specific alpha7 integrin is alternatively spliced in the putative ligand-binding domain to yield either the alpha7X1 or the alpha7X2 variant. The relative level of alpha7X1 and alpha7X2 is developmentally regulated. Similarly, the partner beta1 integrin cytoplasmic domain is converted from the beta1A to the beta1D splice variant. To determine whether beta1D modulates the activity of the alpha7 receptor, cells were transfected with alpha7X1 and beta1D cDNA. alpha7X1 coupled with beta1A failed to adhere to laminin-1, whereas cotransfectants expressing alpha7X1 and beta1D showed strong adhesion. Interestingly, alpha7X1 complexed with beta1A and beta1D displayed the same level of poor adhesion to laminin-2/4 or strong adhesion to laminin-10/11. These findings indicate that alpha7 function is regulated not only by X1/X2 in its extracellular domain but also by beta1 cytoplasmic splice variants. It is likely that expression of beta1D alters alpha7X1 binding to laminin isoforms by a process related to ligand affinity modulation. Functional regulation of alpha7beta1 by developmentally regulated splicing events may be important during myogenic differentiation and repair because the integrin mediates adhesion, motility, and cell survival.
Collapse
Affiliation(s)
- Ming-Guang Yeh
- Department of Stomatology, University of California at San Francisco, San Francisco, CA 94143-0512, USA
| | | | | | | | | | | |
Collapse
|
17
|
Affiliation(s)
- Ulrike Mayer
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, University of Manchester, M13 9PT Manchester, United Kingdom.
| |
Collapse
|
18
|
Sensory neuron subtypes have unique substratum preference and receptor expression before target innervation. J Neurosci 2003. [PMID: 12629182 DOI: 10.1523/jneurosci.23-05-01781.2003] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The factors controlling the specification and subsequent differentiation of sensory neurons are poorly understood. Data from embryological manipulations suggest that either sensory neuron fates are specified by the targets they encounter or sensory neurons are considerably more "plastic" with respect to specification than are neurons of the CNS. The prevailing view that sensory neurons are specified late in development is not consistent, however, with the directed outgrowth of sensory neurons to their targets and the characteristic spatial distribution of sensory neuron fates within the peripheral ganglia. To address when in development different classes of sensory neurons can first be distinguished, we investigated the interactions of early dorsal root ganglia neurons with the extracellular matrix before neurite outgrowth to targets. We found that subclasses of sensory neurons in early dorsal root ganglia show different patterns of neurite outgrowth and integrin expression that are predictive of their fates. In the absence of neurotrophins, presumptive proprioceptive neurons extend neurites robustly on both laminin and fibronectin, whereas presumptive cutaneous neurons show a strong preference for laminin. Cutaneous afferents that have innervated targets show a similar strong preference for laminin and show higher levels of integrin alpha7beta1 than do proprioceptive neurons. Finally, presumptive proprioceptive neurons express fibronectin receptors, integrin alpha3beta1, alpha4beta1, and alpha5beta1, at higher levels than do presumptive cutaneous neurons. Our results indicate that subtypes of sensory neurons have unique patterns of neurite outgrowth and receptor expression before target innervation.
Collapse
|
19
|
Guan W, Puthenveedu MA, Condic ML. Sensory neuron subtypes have unique substratum preference and receptor expression before target innervation. J Neurosci 2003; 23:1781-91. [PMID: 12629182 PMCID: PMC6741987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023] Open
Abstract
The factors controlling the specification and subsequent differentiation of sensory neurons are poorly understood. Data from embryological manipulations suggest that either sensory neuron fates are specified by the targets they encounter or sensory neurons are considerably more "plastic" with respect to specification than are neurons of the CNS. The prevailing view that sensory neurons are specified late in development is not consistent, however, with the directed outgrowth of sensory neurons to their targets and the characteristic spatial distribution of sensory neuron fates within the peripheral ganglia. To address when in development different classes of sensory neurons can first be distinguished, we investigated the interactions of early dorsal root ganglia neurons with the extracellular matrix before neurite outgrowth to targets. We found that subclasses of sensory neurons in early dorsal root ganglia show different patterns of neurite outgrowth and integrin expression that are predictive of their fates. In the absence of neurotrophins, presumptive proprioceptive neurons extend neurites robustly on both laminin and fibronectin, whereas presumptive cutaneous neurons show a strong preference for laminin. Cutaneous afferents that have innervated targets show a similar strong preference for laminin and show higher levels of integrin alpha7beta1 than do proprioceptive neurons. Finally, presumptive proprioceptive neurons express fibronectin receptors, integrin alpha3beta1, alpha4beta1, and alpha5beta1, at higher levels than do presumptive cutaneous neurons. Our results indicate that subtypes of sensory neurons have unique patterns of neurite outgrowth and receptor expression before target innervation.
Collapse
MESH Headings
- Animals
- Cell Differentiation/drug effects
- Cell Differentiation/physiology
- Chick Embryo
- Extracellular Matrix/metabolism
- Fibronectins/metabolism
- Fibronectins/pharmacology
- Ganglia, Spinal/cytology
- Ganglia, Spinal/embryology
- Ganglia, Spinal/metabolism
- Integrins/biosynthesis
- Integrins/genetics
- Laminin/metabolism
- Laminin/pharmacology
- Nerve Growth Factor/pharmacology
- Neurites/drug effects
- Neurites/physiology
- Neurons, Afferent/classification
- Neurons, Afferent/cytology
- Neurons, Afferent/drug effects
- Neurons, Afferent/metabolism
- Neurotrophin 3/pharmacology
- RNA, Messenger/biosynthesis
- Receptor, trkA/biosynthesis
- Receptor, trkC/biosynthesis
- Receptors, Cell Surface/biosynthesis
- Receptors, Fibronectin/biosynthesis
Collapse
Affiliation(s)
- Wei Guan
- Department of Neurobiology and Anatomy, University of Utah, School of Medicine, Salt Lake City, Utah 84132-3401, USA
| | | | | |
Collapse
|
20
|
Rosbottom A, Scudamore CL, von der Mark H, Thornton EM, Wright SH, Miller HRP. TGF-beta 1 regulates adhesion of mucosal mast cell homologues to laminin-1 through expression of integrin alpha 7. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:5689-95. [PMID: 12421948 DOI: 10.4049/jimmunol.169.10.5689] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Mucosal mast cells (MMC) or their precursors migrate through the intestinal lamina propria to reside intraepithelially, where expression of mouse mast cell protease-1 indicates the mature phenotype. Alterations in expression of integrins that govern cell adhesion to the extracellular matrix may regulate this process. As the key cytokine mediating differentiation of mouse mast cell protease-1-expressing MMC homologues in vitro, TGF-beta1 was considered a likely candidate for regulation of the integrins that facilitate intraepithelial migration of MMC. Therefore, we examined adhesion of bone marrow-derived mast cells cultured with and without TGF-beta1 to laminin-1, fibronectin, and vitronectin along with expression of integrins likely to regulate this adhesion. Adhesion of PMA-stimulated cultured mast cells to laminin-1 increased from 5.3 +/- 3.6% (mean +/- SEM) in the absence of TGF-beta1 to 58.7 +/- 4.0% (p < 0.05) when cultured mast cells had differentiated into MMC homologues in the presence of TGF-beta1. Increased adhesion of MMC homologues to laminin-1 was also stimulated by FcepsilonRI cross-linking and the calcium ionophore A23187. Expression of the laminin-binding integrin alpha(7) by MMC homologues grown in the presence of TGF-beta1 was demonstrated by RT-PCR and flow cytometry, and preincubation of MMC homologues with the alpha(7)-neutralizing Ab 6A11 inhibited adhesion to laminin-1 by 98% (p < 0.05), demonstrating a novel role for this molecule in adhesion of a hemopoietic cell to laminin-1.
Collapse
Affiliation(s)
- Anne Rosbottom
- Department of Veterinary Pathology, University of Edinburgh, Roslin, Midlothian, UK EH25 9RG
| | | | | | | | | | | |
Collapse
|
21
|
Gullberg DE, Lundgren-Akerlund E. Collagen-binding I domain integrins--what do they do? PROGRESS IN HISTOCHEMISTRY AND CYTOCHEMISTRY 2002; 37:3-54. [PMID: 11876085 DOI: 10.1016/s0079-6336(02)80008-0] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Collagens are the most abundant proteins in the mammalian body and it is well recognized that collagens fulfill an important structural role in the extracellular matrix in a number of tissues. Inactivation of the collagen alpha 1(I) gene in mice results in embryonic lethality and collagen mutations in humans cause defects leading to disease. Integrins constitute a major group of receptors for extracellular matrix components, including collagens. Currently four collagen-binding I domain-containing integrins are known, namely alpha 1 beta 1, alpha 2 beta 1, alpha 10 beta 1 and alpha 11 beta 1. Unlike the undisputed role of collagens as structural elements, the biological importance of integrin mediated cell-collagen interactions is far from clear. This is in part due to the limited information available on the most recent additions of the integrin family, alpha 10 beta 1 and alpha 11 beta 1. Future studies using gene inactivation of individual and multiple integrin genes will allow testing of the hypothesis that collagen-binding integrins have redundant functions but will also shed light on their importance in pathological conditions. In this review we will describe what is currently known about the collagen-binding integrins and discuss their biological functions.
Collapse
Affiliation(s)
- Donald E Gullberg
- Department of Medical Biochemistry and Microbiology, Biomedical Center, Box 582, Uppsala University, S-75123 Uppsala, Sweden.
| | | |
Collapse
|
22
|
Kääriäinen M, Nissinen L, Kaufman S, Sonnenberg A, Järvinen M, Heino J, Kalimo H. Expression of alpha7beta1 integrin splicing variants during skeletal muscle regeneration. THE AMERICAN JOURNAL OF PATHOLOGY 2002; 161:1023-31. [PMID: 12213731 PMCID: PMC1867267 DOI: 10.1016/s0002-9440(10)64263-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Integrin alpha7beta1 is a laminin receptor, both subunits of which have alternatively spliced, developmentally regulated variants. In skeletal muscle beta1 has two major splice variants of the intracellular domain (beta1A and beta1D). alpha7X1 and alpha7X2 represent variants of the alpha7 ectodomain, whereas alpha7A and alpha7B are variants of the intracellular domain. Previously we showed that during early regeneration after transection injury of muscle alpha7 integrin mediates dynamic adhesion of myofibers along their lateral aspects to the extracellular matrix. Stable attachment of myofibers to the extracellular matrix occurs during the third week after injury, when new myotendinous junctions develop at the ends of the regenerating myofibers. Now we have analyzed the relative expression of beta1A/beta1D and alpha7A/alpha7B and alpha7X1/alpha7X2 isoforms during regeneration for 2 to 56 days after transection of rat soleus muscle using reverse transcriptase-polymerase chain reaction and immunohistochemistry. During early regeneration beta1A was the predominant isoform in both the muscle and scar tissue. Expression of muscle-specific beta1D was detected in regenerating myofibers from day 4 onwards, ie, when myogenic mitotic activity began to decrease, and it became more abundant with the progression of regeneration. alpha7B isoform predominated on day 2. Thereafter, the relative expression of alpha7A transcripts increased until day 7 with the concomitant appearance of alpha7A immunoreactivity on regenerating myofibers. Finally, alpha7B again became the predominant variant in highly regenerated myofibers. Similarly as in the controls, alpha7X1 and alpha7X2 isoforms were both expressed throughout the regeneration with a peak in alpha7X1 expression on day 4 coinciding with the dynamic adhesion stage. The results suggest that during regeneration of skeletal muscle the splicing of beta1 and alpha7 integrin subunits is regulated according to functional requirements. alpha7A and alpha7X1 appear to have a specific role during the dynamic phase of adhesion, whereas alpha7B, alpha7X2, and beta1D predominate during stable adhesion.
Collapse
Affiliation(s)
- Minna Kääriäinen
- Medical School and the Institute of Medical Technology, University of Tampere, Finland
| | | | | | | | | | | | | |
Collapse
|
23
|
Integrin alpha 7 beta 1 in muscular dystrophy/myopathy of unknown etiology. THE AMERICAN JOURNAL OF PATHOLOGY 2002; 160:2135-43. [PMID: 12057917 PMCID: PMC1850814 DOI: 10.1016/s0002-9440(10)61162-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
To investigate the role of integrin alpha 7 in muscle pathology, we used a "candidate gene" approach in a large cohort of muscular dystrophy/myopathy patients. Antibodies against the intracellular domain of the integrin alpha 7A and alpha 7B were used to stain muscle biopsies from 210 patients with muscular dystrophy/myopathy of unknown etiology. Levels of alpha 7A and alpha 7B integrin were found to be decreased in 35 of 210 patients (approximately 17%). In six of these patients no integrin alpha 7B was detected. Screening for alpha 7B mutation in 30 of 35 patients detected only one integrin alpha 7 missense mutation (the mutation on the second allele was not found) in a patient presenting with a congenital muscular dystrophy-like phenotype. No integrin alpha 7 gene mutations were identified in all of the other patients showing integrin alpha 7 deficiency. In the process of mutation analysis, we identified a novel integrin alpha 7 isoform presenting 72-bp deletion. This isoform results from a partial deletion of exon 21 due to the use of a cryptic splice site generated by a G to A missense mutation at nucleotide position 2644 in integrin alpha 7 cDNA. This spliced isoform is present in about 12% of the chromosomes studied. We conclude that secondary integrin alpha 7 deficiency is rather common in muscular dystrophy/myopathy of unknown etiology, emphasizing the multiple mechanisms that may modulate integrin function and stability.
Collapse
|
24
|
von der Mark H, Williams I, Wendler O, Sorokin L, von der Mark K, Pöschl E. Alternative splice variants of alpha 7 beta 1 integrin selectively recognize different laminin isoforms. J Biol Chem 2002; 277:6012-6. [PMID: 11744715 DOI: 10.1074/jbc.m102188200] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The integrin alpha(7)beta(1) occurs in several cytoplasmic (alpha(7A), alpha(7B)) and extracellular splice variants (alpha(7X1), alpha(7X2)), which are differentially expressed during development of skeletal and heart muscle. The extracellular variants result from the alternative splicing of exons X1 and X2, corresponding to a segment within the putative ligand binding domain. To study the specificity and affinity of the X1/X2 variants to different laminin isoforms, soluble alpha(7)beta(1) complexes were prepared by recombinant coexpression of the extracellular domains of the alpha- and beta-subunits. The binding of these complexes to purified ligands was measured by solid phase binding assays. Surprisingly, the alternative splice variants revealed different and specific affinities to different laminin isoforms. While the alpha(7X2) variant bound much more strongly to laminin-1 than the alpha(7X1) variant, the latter showed a high affinity binding to laminins-8 and -10/11. Laminin-2, the major laminin isoform in skeletal muscle, was recognized by both variants, whereas none of the two variants were able to interact with laminin-5. A specific blocking antibody inhibited the binding of both variants to all laminins tested, indicating the involvement of common epitopes in alpha(7X1)beta(1) and alpha(7X2)beta(1). Because laminin-8 and -10/11 as well as alpha(7X1) are expressed in developing skeletal and cardiac muscle, these findings suggest that alpha(7X1)beta(1) may represent a physiological receptor with novel specificities for laminin-8 and -10.
Collapse
Affiliation(s)
- Helga von der Mark
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Zentrum für Molekulare Medizin, Department of Experimental Medicine I, 91054 Erlangen, Germany
| | | | | | | | | | | |
Collapse
|
25
|
Klaffky E, Williams R, Yao CC, Ziober B, Kramer R, Sutherland A. Trophoblast-specific expression and function of the integrin alpha 7 subunit in the peri-implantation mouse embryo. Dev Biol 2001; 239:161-75. [PMID: 11784026 DOI: 10.1006/dbio.2001.0404] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
For implantation and placentation to occur, mouse embryo trophoblast cells must penetrate the uterine stroma to make contact with maternal blood vessels. A major component of the uterine epithelial basement membrane and underlying stromal matrix with which they interact is the extracellular matrix protein laminin. We have identified integrin alpha 7 beta 1 as a major receptor for trophoblast-laminin interactions during implantation and yolk sac placenta formation. It is first expressed by trophectoderm cells of the late blastocyst and by all trophectoderm descendants in the early postimplantation embryo through E8.5, then disappears except in cells at the interface between the allantois and the ectoplacental plate. Integrin alpha 7 expression is a general characteristic of the early differentiation stages of rodent trophoblast, given that two different cultured trophoblast cell lines also express this integrin. Trophoblast cells interact with at least three different laminin isoforms (laminins 1, 2/4, and 10/11) in the blastocyst and in the uterus at the time of implantation. Outgrowth assays using function-blocking antibodies show that alpha 7 beta 1 is the major trophoblast receptor for laminin 1 and a functional receptor for laminins 2/4 and 10/11. When trophoblast cells are cultured on substrates of these three laminins, they attach and spread on all three, but show decreased proliferation on laminin 1. These results show that the alpha 7 beta 1 integrin is expressed by trophoblast cells and acts as receptor for several isoforms of laminin during implantation. These interactions are not only important for trophoblast adhesion and spreading but may also play a role in regulating trophectoderm proliferation and differentiation.
Collapse
Affiliation(s)
- E Klaffky
- Department of Cell Biology, University of Virginia Health System, Charlottesville, Virginia 22908-0732, USA
| | | | | | | | | | | |
Collapse
|
26
|
Abstract
The extracellular matrix interacts with cells and promotes and regulates cellular functions such as adhesion, migration, proliferation, differentiation, and morphogenesis. Extracellular molecules are linked to one another by multiple binding domains and form a stable, multifunctional matrix. Cells respond to the extracellular matrix through plasma membrane receptors, which include integrin and non-integrin receptors. The regulation of these interactions requires the coordination of a multiplicity of signals both spatially and temporally.
Collapse
Affiliation(s)
- N Zagris
- Division of Genetics and Cell and Developmental Biology, Department of Biology, University of Patras, Patras, Greece.
| |
Collapse
|
27
|
Blanco-Bose WE, Blau HM. Laminin-induced change in conformation of preexisting alpha7beta1 integrin signals secondary myofiber formation. Dev Biol 2001; 233:148-60. [PMID: 11319864 DOI: 10.1006/dbio.2001.0177] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Two distinct populations of myoblasts, distinguishable by alpha7 integrin expression have been hypothesized to give rise to two phases of myofiber formation in embryonic limb development. We show here that alpha7 integrin is detectable far earlier than previously reported on both "primary" and "secondary" lineage myoblasts and myofibers. An antibody (1211) that recognizes an intracellular epitope allowed detection of alpha7 integrin previously missed using an antibody (H36) that recognizes an extracellular epitope. We found that when myoblasts were isolated and cultured from different developmental stages, H36 only detected alpha7 integrin that was in direct contact with its ligand, laminin. Moreover, alpha7 integrin detection by H36 was reversible and highly localized to subcellular points of contact between myoblasts and laminin-coated 2.8-microm microspheres. Prior to secondary myofiber formation in limb embryogenesis, laminin was present but not in close proximity to clusters of primary myofibers that expressed alpha7 integrin detected by antibody 1211 using deconvolution microscopy. These results suggest that the timing of the interaction of preexisting alpha7 integrin with its ligand, laminin, is a major determinant of allosteric changes that result in an activated form of alpha7 integrin capable of transducing signals from the extracellular matrix commensurate with secondary myofiber formation.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Antibody Specificity
- Antigens, CD/genetics
- Antigens, CD/immunology
- Cell Compartmentation
- Cell Differentiation
- Cells, Cultured
- Collagen/metabolism
- Culture Techniques
- Hindlimb/cytology
- Integrin alpha Chains
- Integrins/chemistry
- Integrins/metabolism
- Laminin/metabolism
- Muscle Fibers, Skeletal/cytology
- Muscle, Skeletal/cytology
- Protein Conformation
- RNA, Messenger
- Rats
- Rats, Sprague-Dawley
- Receptors, Laminin/chemistry
- Receptors, Laminin/metabolism
- Signal Transduction
- Stem Cells/cytology
Collapse
Affiliation(s)
- W E Blanco-Bose
- Department of Molecular Pharmacology, Stanford University School of Medicine, Stanford, California 94305-5175, USA
| | | |
Collapse
|
28
|
Mielenz D, Hapke S, Pöschl E, von Der Mark H, von Der Mark K. The integrin alpha 7 cytoplasmic domain regulates cell migration, lamellipodia formation, and p130CAS/Crk coupling. J Biol Chem 2001; 276:13417-26. [PMID: 11278916 DOI: 10.1074/jbc.m011481200] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The integrin alpha(7)beta(1) is the major laminin-binding integrin in skeletal, heart, and smooth muscle and is a receptor for laminin-1 and -2. It mediates myoblast migration on laminin-1 and -2 and thus might be involved in muscle development and repair. Previously we have shown that alpha(7)B as well as the alpha(7)A and -C splice variants induce cell motility on laminin when transfected into nonmotile HEK293 cells. In this study we have investigated the role of the cytoplasmic domain of alpha(7) in the laminin-induced signal transduction of alpha(7)beta(1) integrin regulating cell adhesion and migration. Deletion of the cytoplasmic domain did not affect assembly of the mutated alpha(7)Deltacyt/beta(1) heterodimer on the cell surface or adhesion of alpha(7)Deltacyt-transfected cells to laminin. The motility of these cells on the laminin-1/E8 fragment, however, was significantly reduced to the level of mock-transfected cells; lamellipodia formation and polarization of the cells were also impaired. Adhesion to the laminin-1/E8 fragment induced tyrosine phosphorylation of the focal adhesion kinase, paxillin, and p130(CAS) as well as the formation of a p130(CAS)-Crk complex in wild-type alpha(7)B-transfected cells. In alpha(7)BDeltacyt cells, however, the extent of p130(CAS) tyrosine formation was reduced and formation of the p130(CAS)-Crk complex was impaired, with unaltered levels of p130(CAS) and Crk protein levels. These findings indicate adhesion-dependent regulation of p130(CAS)/Crk complex formation by the cytoplasmic domain of alpha(7)B integrin after cell adhesion to laminin-1/E8 and imply alpha(7)B-controlled lamellipodia formation and cell migration through the p130(CAS)/Crk protein complex.
Collapse
Affiliation(s)
- D Mielenz
- Klinisch-molekular biologisches Forschungszentrum, Department for Experimental Medicine I, University of Erlangen-Nürnberg, Glückstrasse 6, 91054 Erlangen, Germany
| | | | | | | | | |
Collapse
|
29
|
Vizirianakis IS, Yao CC, Chen Y, Ziober BL, Tsiftsoglou AS, Kramer RH. Transfection of MCF-7 Carcinoma Cells with Human Integrin α7 cDNA Promotes Adhesion to Laminin. Arch Biochem Biophys 2001; 385:108-16. [PMID: 11361006 DOI: 10.1006/abbi.2000.2134] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The laminin-binding alpha7beta1 integrin receptor is highly expressed by skeletal and cardiac muscles, and has been suggested to be a crucial molecule during myogenic cell migration and differentiation. Absence of integrin alpha7 subunit contributes to a form of muscular dystrophy in integrin alpha7 null mice, whereas specific mutations in the alpha7 gene are associated in humans with congenital myopathy. To examine in more detail the potential role of integrin alpha7 in human-related muscular disorders, we cloned alpha7 cDNA by RT-PCR from human skeletal muscle mRNA and then expressed the full-length human integrin alpha7 cDNA by transfection in several cell lines including MCF-7, COS-7, and NIH3T3 cells. The isolated cDNA corresponds to the human alpha7X2B alternative splice form. Expression of human alpha7 was further confirmed by transfection of chimeric human/mouse alpha7 cDNA constructs. To demonstrate the functionality of expressed human alpha7, adhesion experiments with transfected MCF-7 cells have confirmed the specific binding of human alpha7 to laminin. In addition, mouse polyclonal and monoclonal antibodies were generated against the extracellular domain of human alpha7 and used to analyze by flow cytometry MCF-7 and NIH3T3 cells transfected with the full-length of human alpha7 cDNA. These results show for the first time the exogenous expression of functional full-length human alpha7 cDNA, as well as the development of monoclonal antibodies against the human alpha7 extracellular domain. Antibodies developed will be useful for further analysis of human disorders involving alpha7 dysfunction and facilitate isolation of muscle stem cells (satellite cells) and thereby expand the opportunities for genetically modified transplantation treatment of human disease.
Collapse
MESH Headings
- 3T3 Cells
- Alternative Splicing
- Animals
- Antibodies, Monoclonal/metabolism
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Biotin/metabolism
- Blotting, Western
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- COS Cells
- Cell Adhesion
- Cell Line
- Cell Separation
- Cloning, Molecular
- DNA, Complementary/metabolism
- Flow Cytometry
- Humans
- Immunohistochemistry
- Integrin alpha Chains
- Laminin/metabolism
- Mice
- Molecular Sequence Data
- Muscle, Skeletal/metabolism
- Precipitin Tests
- Protein Structure, Tertiary
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Transfection
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- I S Vizirianakis
- Department of Stomatology, University of California at San Francisco, 94143-0512, USA
| | | | | | | | | | | |
Collapse
|
30
|
Abstract
Laminins are a family of trimeric glycoproteins present in the extracellular matrix and the major constituents of basement membranes. Integrins are alpha beta transmembrane receptors that play critical roles in both cell-matrix and cell-cell adhesion. Several members of the integrin family, including alpha 1 beta 1, alpha 2 beta 1, alpha 3 beta 1, alpha 6 beta 1, alpha 7 beta 1 and alpha 6 beta 4 heterodimers serve as laminin receptors on a variety of cell types. This review summarizes recent advances in understanding the involvement of individual integrins in cell interactions with laminins and the roles of laminin-binding integrins in adhesion-mediated events in vertebrates, including embryonic development, cell migration and tumor cell invasiveness, cell proliferation and differentiation, as well as basement membrane assembly. We discuss the regulation of integrin function via alternative splicing of cytoplasmic domains of alpha and beta subunits of the integrin receptors for laminins and present examples of functional collaboration between laminin-binding integrins and non-integrin laminin receptors. Advances in our understanding of the laminin-binding integrins continue to demonstrate the essential roles these receptors play in maintaining cell polarity and tissue architecture.
Collapse
Affiliation(s)
- A M Belkin
- Department of Biochemistry, The Holland Laboratory, American Red Cross, Rockville, Maryland 20855, USA
| | | |
Collapse
|
31
|
Schöber S, Mielenz D, Echtermeyer F, Hapke S, Pöschl E, von der Mark H, Moch H, von der Mark K. The role of extracellular and cytoplasmic splice domains of alpha7-integrin in cell adhesion and migration on laminins. Exp Cell Res 2000; 255:303-13. [PMID: 10694445 DOI: 10.1006/excr.2000.4806] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The major laminin-binding integrin of skeletal, smooth, and heart muscle is alpha7beta1-integrin, which is structurally related to alpha6beta1. It occurs in three cytoplasmic splice variants (alpha7A, -B, and -C) and two extracellular forms (X1 and X2) which are developmentally regulated and differentially expressed in skeletal muscle. Previously, we have shown that ectopic expression of the alpha7beta-integrin splice variant in nonmotile HEK293 cells specifically induced cell locomotion on laminin-1 but not on fibronectin. To investigate the specificity and the mechanism of the alpha7-mediated cell motility, we expressed the three alpha7-chain cytoplasmic splice variants, as well as alpha6A- and alpha6B-integrin subunits in HEK293 cells. Here we show that all three alpha7 splice variants (containing the X2 domain), as well as alpha6A and alpha6B, promote cell attachment and stimulate cell motility on laminin-1 and its E8 fragment. Deletion of the cytoplasmic domain (excluding the GFFKR consensus sequence) from alpha7B resulted in a loss of the motility-enhancing effect. On laminin-2/4 (merosin), the predominant isoform in mature skeletal muscle, only alpha7-expressing cells showed enhanced motility, whereas cells transfected with alpha6A and alpha6B neither attached nor migrated on laminin-2. Adhesion of alpha7-expressing cells to both laminin-1 and laminin-2 was specifically inhibited by a new monoclonal antibody (6A11) specific for alpha7. Expression of the two extracellular splice variants alpha7X1 and alpha7X2 in HEK293 cells conferred different motilities on laminin isoforms: Whereas alpha7X2B promoted cell migration on both laminin-1 and laminin-2, alpha7X1B supported motility only on laminin-2 and not on laminin-1, although both X1 and X2 splice variants revealed similar adhesion rates to laminin-1 and -2. Fluorescence-activated cell sorter analysis revealed a dramatic reduction of surface expression of alpha6-integrin subunits after alpha7A or -B transfection; also, surface expression of alpha1-, alpha3-, and alpha5-integrins was significantly reduced. These results demonstrate selective responses of alpha6- and alpha7-integrins and of the alpha7 splice variants to laminin-1 and -2 and indicate differential roles in laminin-controlled cell adhesion and migration.
Collapse
Affiliation(s)
- S Schöber
- Institute of Experimental Medicine, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, 91054, Germany
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Velling T, Kusche-Gullberg M, Sejersen T, Gullberg D. cDNA cloning and chromosomal localization of human alpha(11) integrin. A collagen-binding, I domain-containing, beta(1)-associated integrin alpha-chain present in muscle tissues. J Biol Chem 1999; 274:25735-42. [PMID: 10464311 DOI: 10.1074/jbc.274.36.25735] [Citation(s) in RCA: 132] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We previously identified a novel integrin alpha-chain in human fetal muscle cells (Gullberg, D., Velling, T., Sjöberg, G., and Sejersen, T. (1995) Dev. Dyn. 204, 57-65). We have now isolated the full-length cDNA for this integrin subunit, alpha(11). The open reading frame of the cDNA encodes a precursor of 1188 amino acids. The predicted mature protein of 1166 amino acids contains seven conserved FG-GAP repeats, an I domain with a metal ion-dependent adhesion site motif, a short transmembrane region, and a unique cytoplasmic domain of 24 amino acids containing the sequence GFFRS. alpha(11), like other I domain integrins, lacks a dibasic cleavage site for generation of a heavy chain and a light chain, and it contains three potential divalent cation binding sites in repeats 5-7. The presence of 22 inserted amino acids in the extracellular stalk portion (amino acids 804-826) distinguishes the alpha(11) integrin sequence from other integrin alpha-chains. Amino acid sequence comparisons reveal the highest identity of 42% with the alpha(10) integrin chain. Immunoprecipitation with antibodies to alpha(11) integrin captures a 145-kDa protein distinctly larger than the 140-kDa alpha(2) integrin chain when analyzed by SDS-polyacrylamide gel electrophoresis under nonreducing conditions. Fluorescence in situ hybridization maps the integrin alpha(11) gene to chromosome 15q23, in the vicinity of an identified locus for Bardet-Biedl syndrome. Based on Northern blotting, integrin alpha(11) mRNA levels are high in the adult human uterus and in the heart and intermediate in skeletal muscle and some other tissues tested. During in vitro myogenic differentiation, alpha(11) mRNA and protein are up-regulated. Studies of ligand binding properties show that alpha(11)beta(1) binds collagen type I-Sepharose, and cultured muscle cells localize alpha(11)beta(1) into focal contacts on collagen type I. Future studies will reveal the importance of alpha(11)beta(1) for muscle development and integrity in adult muscle and other tissues.
Collapse
Affiliation(s)
- T Velling
- Department of Cell and Molecular Biology, BMC, Box 596, Uppsala University, S-751 24 Uppsala, Sweden
| | | | | | | |
Collapse
|
33
|
Flynn KM, Schreibman MP, Yablonsky-Alter E, Banerjee SP. Sexually dimorphic development and binding characteristics of NMDA receptors in the brain of the platyfish. Gen Comp Endocrinol 1999; 115:282-91. [PMID: 10417241 DOI: 10.1006/gcen.1999.7317] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study investigated age- and gender-specific variations in properties of the glutamate N-methyl-d-aspartate receptor (NMDAR) in a freshwater teleost, the platyfish (Xiphophorus maculatus). Prior localization of the immunoreactive (ir)-R1 subunit of the NMDAR protein (R1) in cells of the nucleus olfactoretinalis (NOR), a primary gonadotropin-releasing hormone (GnRH)-containing brain nucleus in the platyfish, suggests that NMDAR, as in mammals, is involved in modulation of the platyfish brain-pituitary-gonad (BPG) axis. The current study shows that the number of cells in the NOR displaying ir-R1 is significantly increased in pubescent and mature female platyfish when compared to immature and senescent animals. In males, there is no significant change in ir-R1 expression in the NOR at any time in their lifespan. The affinity of the noncompetitive antagonist ((3)H)MK-801 for the NMDAR is significantly increased in pubescent females while maximum binding of ((3)H)MK-801 to the receptor reaches a significant maximum in mature females. In males, both MK-801 affinity and maximum binding remain unchanged throughout development. This is the first report of gender differences in the association of NMDA receptors with neuroendocrine brain areas during development. It is also the first report to suggest NMDA receptor involvement in the development of the BPG axis in a nonmammalian vertebrate.
Collapse
Affiliation(s)
- K M Flynn
- City University of New York, Brooklyn, New York 11210, USA
| | | | | | | |
Collapse
|
34
|
Abstract
Integrins are a family of transmembrane proteins composed of heterodimers of alpha and beta subunits. With their extracellular domain they bind extracellular matrix proteins or other cell surface molecules, and their cytoplasmic domain binds to cytoskeletal and signaling proteins. Thus, they are in an ideal position to transfer information from the extracellular environment to the interior of the cell and vice versa. For several integrin subunits, alternative splicing of mRNA leads to variations in the sequence of both extracellular and cytoplasmic domains. Many integrin splice variants have specific expression patterns, but for some time, functional differences between these variants were not evident. Recent experiments using transfected cell lines and gene targeting of specific splice variants have contributed significantly to our understanding of the function of these splice variants. The results indicate that alternative splicing is a mechanism to subtly regulate the ligand binding and signaling activity of integrins.
Collapse
Affiliation(s)
- A A de Melker
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | |
Collapse
|
35
|
Chen MS, Almeida EA, Huovila AP, Takahashi Y, Shaw LM, Mercurio AM, White JM. Evidence that distinct states of the integrin alpha6beta1 interact with laminin and an ADAM. J Cell Biol 1999; 144:549-61. [PMID: 9971748 PMCID: PMC2132920 DOI: 10.1083/jcb.144.3.549] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/1998] [Revised: 11/24/1998] [Indexed: 11/22/2022] Open
Abstract
Integrins can exist in different functional states with low or high binding capacity for particular ligands. We previously provided evidence that the integrin alpha6beta1, on mouse eggs and on alpha6-transfected cells, interacted with the disintegrin domain of the sperm surface protein ADAM 2 (fertilin beta). In the present study we tested the hypothesis that different states of alpha6beta1 interact with fertilin and laminin, an extracellular matrix ligand for alpha6beta1. Using alpha6-transfected cells we found that treatments (e.g., with phorbol myristate acetate or MnCl2) that increased adhesion to laminin inhibited sperm binding. Conversely, treatments that inhibited laminin adhesion increased sperm binding. Next, we compared the ability of fluorescent beads coated with either fertilin beta or with the laminin E8 fragment to bind to eggs. In Ca2+-containing media, fertilin beta beads bound to eggs via an interaction mediated by the disintegrin loop of fertilin beta and by the alpha6 integrin subunit. In Ca2+-containing media, laminin E8 beads did not bind to eggs. Treatment of eggs with phorbol myristate acetate or with the actin disrupting agent, latrunculin A, inhibited fertilin bead binding, but did not induce laminin E8 bead binding. Treatment of eggs with Mn2+ dramatically increased laminin E8 bead binding, and inhibited fertilin bead binding. Our results provide the first evidence that different states of an integrin (alpha6beta1) can interact with an extracellular matrix ligand (laminin) or a membrane-anchored cell surface ligand (ADAM 2).
Collapse
Affiliation(s)
- M S Chen
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia 22908, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Chen H, Sampson NS. Mediation of sperm-egg fusion: evidence that mouse egg alpha6beta1 integrin is the receptor for sperm fertilinbeta. CHEMISTRY & BIOLOGY 1999; 6:1-10. [PMID: 9889149 DOI: 10.1016/s1074-5521(99)80015-5] [Citation(s) in RCA: 92] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
BACKGROUND INTRODUCTION A key step leading to fertilization is the binding of sperm to the egg plasma membrane. When a mammalian sperm reaches the egg plasma membrane, fertilinbeta, an extracellular sperm membrane protein, is believed to bind to an egg plasma membrane receptor triggering fusion. We set out to identify the fertilinbeta binding partner on the egg plasma membrane. RESULTS We synthesized an 125-labeled peptide with the consensus Asp-Glu-Cys-Asp (DECD) sequence of fertilinbeta's disintegrin domain. This peptide contains a benzophenone photoaffinity probe and inhibits sperm-egg fusion. Upon photoactivation in the presence of whole mouse eggs, a single polypeptide was covalently labeled. This polypeptide has been identified by immunoprecipitation as an alpha6 integrin complexed with beta1 integrin. CONCLUSIONS Our experiments establish that small peptides containing the consensus DECD sequence of sperm fertilinbeta bind specifically to an alpha6beta1 integrin receptor on the egg membrane. We conclude that fertilinbeta binds directly to the alpha6beta1 integrin on the egg surface and this partnership mediates sperm-egg fusion.
Collapse
Affiliation(s)
- H Chen
- Department of Chemistry, State University of New York, Stony Brook 11794 3400, USA
| | | |
Collapse
|
37
|
Graner MW, Bunch TA, Baumgartner S, Kerschen A, Brower DL. Splice variants of the Drosophila PS2 integrins differentially interact with RGD-containing fragments of the extracellular proteins tiggrin, ten-m, and D-laminin 2. J Biol Chem 1998; 273:18235-41. [PMID: 9660786 DOI: 10.1074/jbc.273.29.18235] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Two new potential ligands of the Drosophila PS2 integrins have been characterized by functional interaction in cell culture. These potential ligands are a new Drosophila laminin alpha2 chain encoded by the wing blister locus and Ten-m, an extracellular protein known to be involved in embryonic pattern formation. As with previously identified PS2 ligands, both contain RGD sequences, and RGD-containing fragments of these two proteins (DLAM-RGD and TENM-RGD) can support PS2 integrin-mediated cell spreading. In all cases, this spreading is inhibited specifically by short RGD-containing peptides. As previously found for the PS2 ligand tiggrin (and the tiggrin fragment TIG-RGD), TENM-RGD induces maximal spreading of cells expressing integrin containing the alphaPS2C splice variant. This is in contrast to DLAM-RGD, which is the first Drosophila polypeptide shown to interact preferentially with cells expressing the alphaPS2 m8 splice variant. The betaPS integrin subunit also varies in the presumed ligand binding region as a result of alternative splicing. For TIG-RGD and TENM-RGD, the beta splice variant has little effect, but for DLAM-RGD, maximal cell spreading is supported only by the betaPS4A form of the protein. Thus, the diversity in PS2 integrins due to splicing variations, in combination with diversity of matrix ligands, can greatly enhance the functional complexity of PS2-ligand interactions in the developing animal. The data also suggest that the splice variants may alter regions of the subunits that are directly involved in ligand interactions, and this is discussed with respect to models of integrin structure.
Collapse
Affiliation(s)
- M W Graner
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona 85721, USA
| | | | | | | | | |
Collapse
|
38
|
Zolkiewska A, Thompson WC, Moss J. Interaction of integrin alpha 7 beta 1 in C2C12 myotubes and in solution with laminin. Exp Cell Res 1998; 240:86-94. [PMID: 9570924 DOI: 10.1006/excr.1998.4002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The dimer of integrin alpha 7 and beta 1 is a major laminin-binding receptor in skeletal muscle. We studied interactions of integrin alpha 7 beta 1 with the extracellular matrix protein laminin in solution and in intact cells. Integrin alpha 7 beta 1 bound to EHS laminin (laminin-1, composed of alpha 1, beta 1, and gamma 1 chains), but not to endogenous laminin expressed in C2C12 myotubes. Northern blot analysis demonstrated that C2C12 myotubes synthesized laminin-1 alpha, beta, and gamma subunits mRNAs. C2C12 laminin was, however, immunologically distinct from EHS laminin; it was not recognized by 5D3 anti-laminin-1 monoclonal antibody, whereas 5A2 and LT3 antibodies reacted equally well with C2C12 and EHS laminins. Following deglycosylation of EHS laminin, separation of the subunits by SDS-PAGE, Western blotting, and partial amino acid sequencing of the protein bands, the epitope recognized by 5D3 antibody was localized to the gamma 1 laminin chain. Following binding in vitro, the complex of EHS laminin and integrin alpha 7 beta 1 was subject to chemical cross-linking. The two proteins did not undergo cross-linking at the cell surface, consistent with the fact that in intact, resting myotubes integrin alpha 7 beta 1 interacted poorly with EHS laminin, which may reflect a limited accessibility of integrin alpha 7 beta 1 in the membrane to laminin or an inactive state of the integrin.
Collapse
Affiliation(s)
- A Zolkiewska
- Pulmonary-Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland 20892-1590, USA
| | | | | |
Collapse
|