51
|
Yan W, George S, Fotadar U, Tyhovych N, Kamer A, Yost MJ, Price RL, Haggart CR, Holmes JW, Terracio L. Tissue engineering of skeletal muscle. ACTA ACUST UNITED AC 2008; 13:2781-90. [PMID: 17880268 DOI: 10.1089/ten.2006.0408] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Loss of skeletal muscle profoundly affects the health and well-being of patients, and there currently is no way to replace lost muscle. We believe that a key step in the development of a prosthesis for reconstruction of dysfunctional muscular tissue is the ability to reconstitute the in vivo-like 3-dimensional (3D) organization of skeletal muscle in vitro with isolated satellite cells. In our present proof of principle studies, we have successfully constructed a multilayered culture of skeletal muscle cells, derived from neonatal satellite cells, that are distributed in a 3D pattern of organization that mimics many of the features of intact tissue. These multilayered cultures are composed of elongated multinucleated myotubes that are MyoD positive. Histological studies indicate that the multiple layers of myotubes can be distinguished. Expression of muscle-specific markers such as myosin heavy chain, dystrophin, integrin alpha-7, alpha-enolase, and beta-enolase was detected using real-time reverse transcriptase polymerase chain reaction at levels near adult values. Physiological measurements of the engineered skeletal muscle showed that they tetanize and display physiologic force length behavior, although developed force per cross-sectional area was below that of native rat skeletal muscle.
Collapse
Affiliation(s)
- Wentao Yan
- Department of Basic Sciences, New York University, New York, New York 10010, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
52
|
Kragtorp KA, Miller JR. Integrin alpha5 is required for somite rotation and boundary formation in Xenopus. Dev Dyn 2007; 236:2713-20. [PMID: 17685483 DOI: 10.1002/dvdy.21280] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The morphogenesis of somites in Xenopus laevis is characterized by a complex process of cell turning that requires coordinated regulation of cell shape, adhesion, and motility. The integrin alpha5 subunit has been implicated in the formation of somite boundaries in organisms utilizing epithelialization to create morphologically distinct somites, but its function has not been examined in Xenopus. We used a splice-blocking morpholino to knock down expression of integrin alpha5 during somite formation. Loss of integrin alpha5 delayed somite turning and accumulation of integrin beta1 at somite boundaries, and disrupted the fibronectin matrix surrounding developing somites. Irregular somite boundaries with a sparse and discontinuous fibronectin matrix formed upon eventual completion of somite turning. Recovery of somite morphology was improved, but still incomplete in far posterior somites. These data demonstrate that the role of integrin alpha5 in somite boundary formation is conserved in a species using a unique mechanism of somitogenesis.
Collapse
Affiliation(s)
- Katherine A Kragtorp
- Department of Genetics, University of Minnesota, Minneapolis, Minnesota 55455, USA.
| | | |
Collapse
|
53
|
Liu J, Burkin DJ, Kaufman SJ. Increasing alpha 7 beta 1-integrin promotes muscle cell proliferation, adhesion, and resistance to apoptosis without changing gene expression. Am J Physiol Cell Physiol 2007; 294:C627-40. [PMID: 18045857 DOI: 10.1152/ajpcell.00329.2007] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The dystrophin-glycoprotein complex maintains the integrity of skeletal muscle by associating laminin in the extracellular matrix with the actin cytoskeleton. Several human muscular dystrophies arise from defects in the components of this complex. The alpha(7)beta(1)-integrin also binds laminin and links the extracellular matrix with the cytoskeleton. Enhancement of alpha(7)-integrin levels alleviates pathology in mdx/utrn(-/-) mice, a model of Duchenne muscular dystrophy, and thus the integrin may functionally compensate for the absence of dystrophin. To test whether increasing alpha(7)-integrin levels affects transcription and cellular functions, we generated alpha(7)-integrin-inducible C2C12 cells and transgenic mice that overexpress the integrin in skeletal muscle. C2C12 myoblasts with elevated levels of integrin exhibited increased adhesion to laminin, faster proliferation when serum was limited, resistance to staurosporine-induced apoptosis, and normal differentiation. Transgenic expression of eightfold more integrin in skeletal muscle did not result in notable toxic effects in vivo. Moreover, high levels of alpha(7)-integrin in both myoblasts and in skeletal muscle did not disrupt global gene expression profiles. Thus increasing integrin levels can compensate for defects in the extracellular matrix and cytoskeleton linkage caused by compromises in the dystrophin-glycoprotein complex without triggering apparent overt negative side effects. These results support the use of integrin enhancement as a therapy for muscular dystrophy.
Collapse
Affiliation(s)
- Jianming Liu
- Department of Cell and Developmental Biology, University of Illinois, 601 S. Goodwin Ave., B107 Chemical and Life Sciences Laboratory, Urbana, IL 61801, USA
| | | | | |
Collapse
|
54
|
Milner DJ, Kaufman SJ. Alpha7beta1 integrin does not alleviate disease in a mouse model of limb girdle muscular dystrophy type 2F. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 170:609-19. [PMID: 17255329 PMCID: PMC1851849 DOI: 10.2353/ajpath.2007.060686] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Transgenic expression of the alpha7beta1 integrin in the dystrophic mdx/utr-/- mouse decreases development of muscular dystrophy and enhances longevity. To explore the possibility that elevating alpha7beta1 integrin expression could also ameliorate different forms of muscular dystrophy, we used transgenic technology to enhance integrin expression in mice lacking delta-sarcoglycan (delta sgc), a mouse model for human limb girdle muscular dystrophy type 2F. Unlike alpha7 transgenic mdx/utr-/- mice, enhanced alpha7beta1 integrin expression in the delta sgc-null mouse did not alleviate muscular dystrophy in these animals. Expression of the transgene in the delta sgc-null did not alleviate dystrophic histopathology, nor did it decrease cardiomyopathy or restore exercise tolerance. One hallmark of integrin-mediated alleviation of muscular dystrophy in the mdx/utr-/- background is the restoration of myotendinous junction integrity. As assessed by atomic force microscopy, myotendinous junctions from normal and delta sgc-null mice were indistinguishable, thus suggesting the important influence of myotendinous junction integrity on the severity of muscular dystrophy and providing a possible explanation for the inability of enhanced integrin expression to alleviate dystrophy in the delta sgc-null mouse. These results suggest that distinct mechanisms underlie the development of the diseases that arise from deficiencies in dystrophin and sarcoglycan.
Collapse
Affiliation(s)
- Derek J Milner
- Department of Cell and Developmental Biology, University of Illinois, B107 Chemical and Life Sciences Laboratory, 601 South Goodwin Ave., Urbana, IL 61801, USA
| | | |
Collapse
|
55
|
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
|
56
|
Tran T, McNeill KD, Gerthoffer WT, Unruh H, Halayko AJ. Endogenous laminin is required for human airway smooth muscle cell maturation. Respir Res 2006; 7:117. [PMID: 16968549 PMCID: PMC1586013 DOI: 10.1186/1465-9921-7-117] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2006] [Accepted: 09/12/2006] [Indexed: 01/08/2023] Open
Abstract
Background Airway smooth muscle (ASM) contraction underlies acute bronchospasm in asthma. ASM cells can switch between a synthetic-proliferative phenotype and a contractile phenotype. While the effects of extracellular matrix (ECM) components on modulation of ASM cells to a synthetic phenotype have been reported, the role of ECM components on maturation of ASM cells to a contractile phenotype in adult lung is unclear. As both changes in ECM components and accumulation of contractile ASM are features of airway wall remodelling in asthma, we examined the role of the ECM protein, laminin, in the maturation of contractile phenotype in human ASM cells. Methods Human ASM cells were made senescence-resistant by stable expression of human telomerase reverse transcriptase. Maturation to a contractile phenotype was induced by 7-day serum deprivation, as assessed by immunoblotting for desmin and calponin. The role of laminin on ASM maturation was investigated by comparing the effects of exogenous laminin coated on culture plates, and of soluble laminin peptide competitors. Endogenous expression of laminin chains during ASM maturation was also measured. Results Myocyte binding to endogenously expressed laminin was required for ASM phenotype maturation, as laminin competing peptides (YIGSR or GRGDSP) significantly reduced desmin and calponin protein accumulation that otherwise occurs with prolonged serum deprivation. Coating of plastic cell culture dishes with different purified laminin preparations was not sufficient to further promote accumulation of desmin or calponin during 7-day serum deprivation. Expression of α2, β1 and γ1 laminin chains by ASM cells was specifically up-regulated during myocyte maturation, suggesting a key role for laminin-2 in the development of the contractile phenotype. Conclusion While earlier reports suggest exogenously applied laminin slows the spontaneous modulation of ASM to a synthetic phenotype, we show for the first time that endogenously expressed laminin is required for ASM maturation to the contractile phenotype. As endogenously expressed laminin chains α2, β1 and γ1 are uniquely increased during myocyte maturation, these laminin chains may be key in this process. Thus, human ASM maturation appears to involve regulated endogenous expression of a select set of laminin chains that are essential for accumulation of contractile phenotype myocytes.
Collapse
Affiliation(s)
- Thai Tran
- Departments of Physiology and Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
- Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, MB, Canada
- CIHR National Training Program in Allergy and Asthma, University of Manitoba, Winnipeg, MB, Canada
| | - Karol D McNeill
- Departments of Physiology and Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
- Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, MB, Canada
- CIHR National Training Program in Allergy and Asthma, University of Manitoba, Winnipeg, MB, Canada
| | - William T Gerthoffer
- Department of Pharmacology, University of Nevada School of Medicine, Reno, NV, USA
| | - Helmut Unruh
- Section of Thoracic Surgery, University of Manitoba, Winnipeg, MB, Canada
| | - Andrew J Halayko
- Departments of Physiology and Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
- Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, MB, Canada
- CIHR National Training Program in Allergy and Asthma, University of Manitoba, Winnipeg, MB, Canada
- Section of Respiratory Diseases, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
57
|
Boppart MD, Burkin DJ, Kaufman SJ. α7β1-Integrin regulates mechanotransduction and prevents skeletal muscle injury. Am J Physiol Cell Physiol 2006; 290:C1660-5. [PMID: 16421207 DOI: 10.1152/ajpcell.00317.2005] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
α7β1-Integrin links laminin in the extracellular matrix with the cell cytoskeleton and therein mediates transduction of mechanical forces into chemical signals. Muscle contraction and stretching ex vivo result in activation of intracellular signaling molecules that are integral to postexercise injury responses. Because α7β1-integrin stabilizes muscle and provides communication between the matrix and cytoskeleton, the role of this integrin in exercise-induced cell signaling and skeletal muscle damage was assessed in wild-type and transgenic mice overexpressing the α7BX2 chain. We report here that increasing α7β1-integrin inhibits phosphorylation of molecules associated with muscle damage, including the mitogen-activated protein kinases (JNK, p38, and ERK), following downhill running. Likewise, activation of molecules associated with hypertrophy (AKT, mTOR, and p70S6k) was diminished in mice overexpressing integrin. While exercise resulted in Evans blue dye-positive fibers, an index of muscle damage, increased integrin protected mice from injury. Moreover, exercise leads to an increase in α7β1protein. These experiments provide the first evidence that α7β1-integrin is a negative regulator of mechanotransduction in vivo and provides resistance to exercise-induced muscle damage.
Collapse
Affiliation(s)
- Marni D Boppart
- Dept. of Cell and Developmental Biology, University of Illinois, Urbana, IL 61801, USA
| | | | | |
Collapse
|
58
|
Chao JT, Martinez-Lemus LA, Kaufman SJ, Meininger GA, Ramos KS, Wilson E. Modulation of α7-integrin-mediated adhesion and expression by platelet-derived growth factor in vascular smooth muscle cells. Am J Physiol Cell Physiol 2006; 290:C972-80. [PMID: 16282198 DOI: 10.1152/ajpcell.00136.2005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
We showed previously that the expression of α7-integrin in aortic vascular smooth muscle cells (VSMC) is enhanced in a rat model of atherosclerosis. In the present study, we investigated the effects of platelet-derived growth factor (PDGF) on α7-integrin expression and VSMC adhesion and migration. Expression of the α7-integrin gene was determined by real-time RT-PCR, whereas protein levels were determined by fluorescence-activated cell sorting analysis. PDGF increased α7cell surface protein expression (12 and 24 h: 3.3 ± 0.8- and 3.6 ± 0.4-fold, P < 0.05 vs. control) and mRNA levels (24 h: 3.1-fold, P < 0.05 vs. control) in a time-dependent manner. Actinomycin D and cycloheximide attenuated PDGF-induced increases in α7-integrin, indicating the involvement of de novo mRNA and protein synthesis. Treatment with the MAPK inhibitors PD-98059, SP-600125, and SB-203580 attenuated PDGF-induced increases in mRNA. In contrast, PD-98059 and SP-600125, but not SB-203580, attenuated PDGF-induced increases in cell surface protein levels. PDGF-treated VSMC adhered to laminin more efficiently (42 ± 6% increase, P < 0.01), and this increase was partially inhibited by anti-α7-integrin function-blocking antibody. However, PDGF did not alter migration on laminin, and there was no effect of the anti-α7-integrin function-blocking antibody on basal or PDGF-stimulated migration. Immunofluorescence imaging revealed an increase in α7-integrin distribution along the stress fibers. Together, these observations indicate that PDGF enhances α7-integrin expression in VSMC and promotes α7-integrin-mediated adhesion to laminin.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Cell Adhesion/physiology
- Cells, Cultured
- Enzyme Inhibitors/metabolism
- Epidermal Growth Factor/metabolism
- Fibroblast Growth Factors/metabolism
- Humans
- Integrin alpha Chains/genetics
- Integrin alpha Chains/metabolism
- Integrins/metabolism
- Laminin/metabolism
- MAP Kinase Signaling System/physiology
- Mitogen-Activated Protein Kinases/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- Platelet-Derived Growth Factor/metabolism
- Protein Subunits/genetics
- Protein Subunits/metabolism
- Rats
- Rats, Sprague-Dawley
- Transcription, Genetic
- Transforming Growth Factor beta/metabolism
Collapse
Affiliation(s)
- Jun-Tzu Chao
- Division of Vascular Biology, Cardiovascular Research Institute, The Texas A&M University System Health Science Center, 336 Joe Reynolds Medical Bldg., College Station, 77843, USA
| | | | | | | | | | | |
Collapse
|
59
|
Muroya S, Nakajima I, Oe M, Chikuni K. Effect of phase limited inhibition of MyoD expression on the terminal differentiation of bovine myoblasts: no alteration of Myf5 or myogenin expression. Dev Growth Differ 2006; 47:483-92. [PMID: 16179075 DOI: 10.1111/j.1440-169x.2005.00822.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
To investigate the roles played by MyoD in the terminal differentiation of satellite cell-derived myoblasts, the effect of antisense inhibition of MyoD expression was examined in bovine adult myoblast culture, in which inhibition treatment was limited to the terminal differentiation phase. MyoD antisense oligonucleotide DNA (AS-mD) suppressed the formation of multinucleated myotubes in the cell culture. Myotube formation was suppressed even when AS-mD treatment was limited to the period preceding the onset of myotube formation. Reverse transcriptase-polymerase chain reaction (RT-PCR) analysis revealed that treatment with AS-mD suppressed the expression of myosin heavy chain embryonic isoform and troponin T isoforms at 4 days after the induction of differentiation. AS-mD also suppressed the expression of MRF4, but did not alter the expression of either Myf5 or myogenin, in contrast to previous results using mouse cells possessing MyoD(-/-) genetic background. These findings suggest that MyoD controls myogenesis but not Myf5 or myogenin mRNA expression during the terminal differentiation phase. Furthermore, among the alpha4, alpha5, alpha6, and alpha7 integrins, alpha4, alpha5, and alpha7 integrin expression was suppressed by AS-mD treatment, in parallel with the suppression of myotube formation, which suggests that MyoD controls myotube formation by regulating the expression of alpha4, alpha5, and alpha7 integrins.
Collapse
Affiliation(s)
- Susumu Muroya
- Department of Animal Products, National Institute of Livestock and Grassland Science, Tsukuba, Ibaraki 305-0901, Japan.
| | | | | | | |
Collapse
|
60
|
Nishiuchi R, Takagi J, Hayashi M, Ido H, Yagi Y, Sanzen N, Tsuji T, Yamada M, Sekiguchi K. Ligand-binding specificities of laminin-binding integrins: a comprehensive survey of laminin-integrin interactions using recombinant alpha3beta1, alpha6beta1, alpha7beta1 and alpha6beta4 integrins. Matrix Biol 2006; 25:189-97. [PMID: 16413178 DOI: 10.1016/j.matbio.2005.12.001] [Citation(s) in RCA: 283] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2005] [Revised: 11/25/2005] [Accepted: 12/02/2005] [Indexed: 11/29/2022]
Abstract
The interactions of cells with basement membranes are primarily mediated via the engagement of laminins by a group of integrin family proteins, including integrins alpha3beta1, alpha6beta1, alpha7beta1 and alpha6beta4. To explore the ligand-binding specificities of these laminin-binding integrins, we produced these integrins, including two alpha7beta1 splice variants (alpha7X1beta1 and alpha7X2beta1), as soluble recombinant proteins and determined their binding specificities and affinities toward a panel of purified laminin isoforms containing distinct alpha chains. Among the five laminin-binding integrins investigated, alpha3beta1 and alpha6beta4 exhibited a clear specificity for laminin-332 (alpha3beta3gamma2) and laminin-511 (alpha5beta1gamma1)/521 (alpha5beta2gamma1), while integrin alpha6beta1 showed a broad specificity, binding to all laminin isoforms with a preference for laminin-111 (alpha1beta1gamma1), laminin-332 and laminin-511/521. The two alpha7beta1 variants were distinct from alpha3beta1, alpha6beta1 and alpha6beta4 in that they did not bind to laminin-332. alpha7X1beta1 bound to all laminins, except laminin-332, with a preference for laminin-211 (alpha2beta1gamma1)/221 (alpha2beta2gamma1) and laminin-511/521, while alpha7X2beta1 bound preferentially to laminin-111 and laminin-211/221. Laminin-511/521 was the most preferred ligand for all the laminin-binding integrins, except for alpha7X2beta1, whereas laminin-411 was the poorest ligand, capable of binding to alpha6beta1 and alpha7X1beta1 with only modest binding affinities. These comprehensive analyses of the interactions between laminin-binding integrins and a panel of laminins clearly demonstrate that the isoforms of both integrins and laminins differ in their binding specificities and affinities, and provide a molecular basis for better understanding of the adhesive interactions of cells with basement membranes of defined laminin compositions.
Collapse
Affiliation(s)
- Ryoko Nishiuchi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Osaka, 565-0871, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Elola MT, Chiesa ME, Alberti AF, Mordoh J, Fink NE. Galectin-1 receptors in different cell types. J Biomed Sci 2005; 12:13-29. [PMID: 15864736 DOI: 10.1007/s11373-004-8169-5] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2004] [Accepted: 09/07/2004] [Indexed: 02/07/2023] Open
Abstract
Galectins are a family of animal lectins defined by two properties: shared amino acid sequences in their carbohydrate-recognizing domain, and beta-galactoside affinity. A wide variety of biological phenomena are related to galectins, i.e., development, differentiation, morphogenesis, tumor metastasis, apoptosis, RNA splicing, and immunoregulatory function. In this review, we will focus on galectin-1 receptors, and some of the mechanisms by which this lectin affects different cell types. Several galectin-1 receptors are discussed such as CD45, CD7, CD43, CD2, CD3, CD4, CD107, CEA, actin, extracellular matrix proteins such as laminin and fibronectin, glycosaminoglycans, integrins, a beta-lactosamine glycolipid, GM1 ganglioside, polypeptide HBGp82, glycoprotein 90 K/MAC-2BP, CA125 cancer antigen, and pre-B cell receptor.
Collapse
Affiliation(s)
- María T Elola
- Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires, Patricias Argentinas, 435 (1405), Buenos Aires, Argentina.
| | | | | | | | | |
Collapse
|
62
|
Zhao Z, Gruszczynska-Biegala J, Zolkiewska A. ADP-ribosylation of integrin alpha7 modulates the binding of integrin alpha7beta1 to laminin. Biochem J 2005; 385:309-17. [PMID: 15361073 PMCID: PMC1134699 DOI: 10.1042/bj20040590] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The extracellular domain of integrin alpha7 is ADP-ribosylated by an arginine-specific ecto-ADP-ribosyltransferase after adding exogenous NAD+ to intact C2C12 skeletal muscle cells. The effect of ADP-ribosylation on the structure or function of integrin alpha7beta1 has not been explored. In the present study, we show that ADP-ribosylation of integrin alpha7 takes place exclusively in differentiated myotubes and that this post-translational modification modulates the affinity of alpha7beta1 dimer for its ligand, laminin. ADP-ribosylation in the 37-kDa 'stalk' region of alpha7 that takes place at micromolar NAD+ concentrations increases the binding of the alpha7beta1 dimer to laminin. Increased in vitro binding of integrin alpha7beta1 to laminin after ADP-ribosylation of the 37-kDa fragment of alpha7 requires the presence of Mn2+ and it is not observed in the presence of Mg2+. In contrast, ADP-ribosylation of the 63-kDa N-terminal region comprising the ligand-binding site of alpha7 that occurs at approx. 100 microM NAD+ inhibits the binding of integrin alpha7beta1 to laminin. Furthermore, incubation of C2C12 myotubes with NAD+ increases the expression of an epitope on integrin beta1 subunit recognized by monoclonal antibody 9EG7. We discuss our results based on the current models of integrin activation. We also hypothesize that ADP-ribosylation may represent a mechanism of regulation of integrin alpha7beta1 function in myofibres in vivo when the continuity of the membrane is compromised and NAD+ is available as a substrate for ecto-ADP-ribosylation.
Collapse
Affiliation(s)
- Zhefeng Zhao
- Department of Biochemistry, Kansas State University, 104 Willard Hall, Manhattan, KS 66506, U.S.A
| | | | - Anna Zolkiewska
- Department of Biochemistry, Kansas State University, 104 Willard Hall, Manhattan, KS 66506, U.S.A
- To whom correspondence should be addressed (email )
| |
Collapse
|
63
|
Plantman S, Novikova L, Novikov L, Hammarberg H, Wallquist W, Kellerth JO, Cullheim S. Integrin messenger RNAs in the red nucleus after axotomy and neurotrophic administration. Neuroreport 2005; 16:709-13. [PMID: 15858411 DOI: 10.1097/00001756-200505120-00011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Integrins are cell surface receptors known to be important for regeneration in the peripheral nervous system. We have investigated the expression of integrin messenger RNAs in red nucleus neurons of adult rats after axotomy and administration of neurotrophic factors. Using radioactive in situ hybridization, messenger RNA for integrin subunits beta1, alpha3, alpha7 and alphaV could be detected. No change of any alpha subunit could be detected after axotomy. In contrast, a small upregulation of beta1 was detected after lesion. Administration of neurotrophin-3 induced a robust further increase in beta1 messenger RNA levels, whereas brain-derived neurotrophic factor did not. By analogy to the peripheral nervous system, we propose that integrins may be important for a regenerative response in central nervous system neurons.
Collapse
Affiliation(s)
- Stefan Plantman
- Department of Neuroscience, Karolinska Institutet, S-17177 Stockholm, Sweden.
| | | | | | | | | | | | | |
Collapse
|
64
|
Abstract
Muscle injuries are one of the most common traumas occurring in sports. Despite their clinical importance, few clinical studies exist on the treatment of these traumas. Thus, the current treatment principles of muscle injuries have either been derived from experimental studies or been tested only empirically. Although nonoperative treatment results in good functional outcomes in the majority of athletes with muscle injuries, the consequences of failed treatment can be very dramatic, possibly postponing an athlete's return to sports for weeks or even months. Moreover, the recognition of some basic principles of skeletal muscle regeneration and healing processes can considerably help in both avoiding the imminent dangers and accelerating the return to competition. Accordingly, in this review, the authors have summarized the prevailing understanding on the biology of muscle regeneration. Furthermore, they have reviewed the existing data on the different treatment modalities (such as medication, therapeutic ultrasound, physical therapy) thought to influence the healing of injured skeletal muscle. In the end, they extend these findings to clinical practice in an attempt to propose an evidence-based approach for the diagnosis and optimal treatment of skeletal muscle injuries.
Collapse
Affiliation(s)
- Tero A H Järvinen
- Department of Orthopaedics, Tampere University Hospital and University of Tampere, PO Box 2000, FIN-33521 Tampere, Finland
| | | | | | | | | |
Collapse
|
65
|
Burkin DJ, Wallace GQ, Milner DJ, Chaney EJ, Mulligan JA, Kaufman SJ. Transgenic expression of {alpha}7{beta}1 integrin maintains muscle integrity, increases regenerative capacity, promotes hypertrophy, and reduces cardiomyopathy in dystrophic mice. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 166:253-63. [PMID: 15632017 PMCID: PMC1602287 DOI: 10.1016/s0002-9440(10)62249-3] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
We previously reported that enhanced expression of the alpha7beta1 integrin ameliorates the development of muscular dystrophy and extends longevity in alpha7BX2-mdx/utr(-/-) transgenic mice (Burkin DJ, Wallace GQ, Nicol KJ, Kaufman DJ, Kaufman SJ: Enhanced expression of the alpha7beta1 integrin reduces muscular dystrophy and restores viability in dystrophic mice. We now report on the mechanism by which these mice were rescued by the integrin. As a result of increased integrin in alpha7BX2-mdx/utr(-/-) mice the structural integrity of the myotendinous and neuromuscular junctions are maintained. A twofold increase in satellite cells in alpha7BX2-mdx/utr(-/-) skeletal muscle was detected by immunofluorescence using the satellite cell marker c-met. These cells enhanced the regenerative capacity of muscle in the transgenic animals as determined by fusion of BrdUrd-labeled cells into muscle fibers. Increased integrin also leads to hypertrophy. Finally, transgenic expression of alpha7BX2 integrin chain in skeletal muscle secondarily reduces the development of cardiomyopathy, the ultimate cause of death in these animals. We believe this multiplicity of responses to increased alpha7beta1 integrin collectively inhibits the development of muscle disease and increases longevity in these mice.
Collapse
Affiliation(s)
- Dean J Burkin
- Department of Cell and Structural Biology, University of Illinois, B107 Chemical and Life Sciences Laboratory, 601 South Goodwin Ave., Urbana, IL 61801, USA
| | | | | | | | | | | |
Collapse
|
66
|
Lopez MA, Mayer U, Hwang W, Taylor T, Hashmi MA, Jannapureddy SR, Boriek AM. Force transmission, compliance, and viscoelasticity are altered in the alpha7-integrin-null mouse diaphragm. Am J Physiol Cell Physiol 2005; 288:C282-9. [PMID: 15643051 DOI: 10.1152/ajpcell.00362.2003] [Citation(s) in RCA: 22] [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
Alpha7beta1 integrin is a transmembrane structural and receptor protein of skeletal muscles, and the absence of alpha7-integrin causes muscular dystrophy. We hypothesized that the absence of alpha7-integrin alters compliance and viscoelasticity and disrupts the mechanical coupling between passive transverse and axial contractile elements in the diaphragm. In vivo the diaphragm is loaded with pressure, and therefore axial and transverse length-tension relationships are important in assessing its function. We determined diaphragm passive length-tension relationships and the viscoelastic properties of its muscle in 1-month-old alpha7-integrin-null mice and age-matched controls. Furthermore, we measured the isometric contractile properties of the diaphragm from mutant and normal mice in the absence and presence of passive force applied in the transverse direction to fibers in 1-month-old and 5-month-old mutant mice. We found that compared with controls, the diaphragm direction of alpha7-integrin-null mutants showed 1) a significant decrease in muscle extensibility in 1-year-old mice, whereas muscle extensibility increased in the 1-month-old mice; 2) altered muscle viscoelasticity in the transverse direction of the muscle fibers of 1-month-old mice; 3) a significant increase in force-generating capacity in the diaphragms of 1-month-old mice, whereas in 5-month-old mice muscle contractility was depressed; and 4) significant reductions in mechanical coupling between longitudinal and transverse properties of the muscle fibers of 1-month-old mice. These findings suggest that alpha7-integrin serves an important mechanical function in the diaphragm by contributing to passive compliance, viscoelasticity, and modulation of its muscle contractile properties.
Collapse
Affiliation(s)
- M A Lopez
- Department of Medicine, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | |
Collapse
|
67
|
Contacts of Basement Membrane Molecules with Cell Membranes. CURRENT TOPICS IN MEMBRANES 2005. [DOI: 10.1016/s1063-5823(05)56010-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
68
|
Flintoff-Dye NL, Welser J, Rooney J, Scowen P, Tamowski S, Hatton W, Burkin DJ. Role for the α7β1 integrin in vascular development and integrity. Dev Dyn 2005; 234:11-21. [PMID: 16003770 DOI: 10.1002/dvdy.20462] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The alpha7beta1 integrin is a laminin receptor that has been implicated in muscle disease and the development of neuromuscular and myotendinous junctions. Studies have shown the alpha7beta1 integrin is also expressed in nonskeletal muscle tissues. To identify the expression pattern of the alpha7 integrin in these tissues during embryonic development, alpha7 integrin chain knockout mice were generated by a LacZ knockin strategy. In these mice, expression from the alpha7 promoter is reported by beta-galactosidase. From embryonic day (ED) 11.5 to ED14.5, beta-galactosidase was detected in the developing central and peripheral nervous systems and vasculature. The loss of the alpha7 integrin gene resulted in partial embryonic lethality. Several alpha7 null embryos were identified with cerebrovascular hemorrhages and showed reduced vascular smooth muscle cells and cerebral vascularization. The alpha7 null mice that survived to birth exhibited vascular smooth muscle defects, including hyperplasia and hypertrophy. In addition, altered expression of alpha5 and alpha6B integrin chains was detected in the cerebral arteries of alpha7 null mice, which may contribute to the vascular phenotype. Our results demonstrate for the first time that the alpha7beta1 integrin is important for the recruitment or survival of cerebral vascular smooth muscle cells and that this integrin plays an important role in vascular development and integrity.
Collapse
Affiliation(s)
- Nichole L Flintoff-Dye
- Department of Pharmacology, University of Nevada, Manville Health Science Building, Reno, 89557, USA
| | | | | | | | | | | | | |
Collapse
|
69
|
Wallquist W, Zelano J, Plantman S, Kaufman SJ, Cullheim S, Hammarberg H. Dorsal root ganglion neurons up-regulate the expression of laminin-associated integrins after peripheral but not central axotomy. J Comp Neurol 2004; 480:162-9. [PMID: 15514929 DOI: 10.1002/cne.20345] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The favorable prognosis of regeneration in the peripheral nervous system after axonal lesions is generally regarded as dependent on the Schwann cell basal lamina. Laminins, a heterotrimeric group of basal lamina molecules, have been suggested to be among the factors playing this supportive role. For neurons to utilize laminin as a substrate for growth, an expression of laminin binding receptors, integrins, is necessary. In this study, we have examined the expression of laminin binding integrin subunits in dorsal root ganglion (DRG) neurons after transection to either their peripherally projecting axons, as in the sciatic nerve, followed by regeneration, or the centrally projecting axons in dorsal roots, followed by no or weak regenerative activity. In uninjured DRG, immunohistochemical staining revealed a few neurons expressing integrin subunit alpha6, whereas integrin subunits alpha7 and foremost beta1 were expressed in a majority of neurons. After an injury to the sciatic nerve, mRNAs encoding all three integrins were up-regulated in DRG neurons. By anterograde tracing, immunoreactivity for all studied integrins was also found in association with growing axons after a sciatic nerve crush lesion in vivo. In contrast, mRNA levels remained constant in DRG neurons after a dorsal root injury. Together with previous findings, this suggests that integrin subunits alpha6, alpha7, and beta1 have an important role in the regenerative response following nerve injury and that the lack of regenerative capacity following dorsal root injury could in part be explained by the absence of response in integrin regulation.
Collapse
Affiliation(s)
- Wilhelm Wallquist
- Department of Neuroscience, Karolinska Institutet, S-171 77 Stockholm, Sweden.
| | | | | | | | | | | |
Collapse
|
70
|
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: 1.6] [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
|
71
|
Bajanca F, Luz M, Duxson MJ, Thorsteinsdóttir S. Integrins in the mouse myotome: Developmental changes and differences between the epaxial and hypaxial lineage. Dev Dyn 2004; 231:402-15. [PMID: 15366018 DOI: 10.1002/dvdy.20136] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Integrins are cellular adhesion receptors that mediate signaling and play key roles in the development of multicellular organisms. However, their role in the cellular events leading to myotome formation is completely unknown. Here, we describe the expression patterns of the alpha1, alpha4, alpha5, alpha6, and alpha7 integrin subunits in the mouse myotome and correlate them with the expression of several differentiation markers. Our results indicate that these integrin subunits may be differentially involved in the various phases of myogenic determination and differentiation. A detailed characterization of the myogenic cell types expressing the alpha4 and alpha6 subunits showed a regionalization of the myotome and dermomyotome based on cell-adhesion properties. We conclude that alpha6beta1 may be an early marker of epaxial myogenic progenitor cells. In contrast, alpha4beta1 is up-regulated in the intercalated myotome after myocyte differentiation. Furthermore, alpha4beta1 is expressed in the hypaxial dermomyotome and is maintained by early hypaxial myogenic progenitor cells colonizing the myotome.
Collapse
Affiliation(s)
- Fernanda Bajanca
- Departamento de Biologia Animal, Centro de Biologia Ambiental, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | | | | | | |
Collapse
|
72
|
Fukada SI, Higuchi S, Segawa M, Koda KI, Yamamoto Y, Tsujikawa K, Kohama Y, Uezumi A, Imamura M, Miyagoe-Suzuki Y, Takeda S, Yamamoto H. Purification and cell-surface marker characterization of quiescent satellite cells from murine skeletal muscle by a novel monoclonal antibody. Exp Cell Res 2004; 296:245-55. [PMID: 15149854 DOI: 10.1016/j.yexcr.2004.02.018] [Citation(s) in RCA: 137] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2003] [Revised: 02/16/2004] [Indexed: 11/27/2022]
Abstract
A novel monoclonal antibody, SM/C-2.6, specific for mouse muscle satellite cells was established. SM/C-2.6 detects mononucleated cells beneath the basal lamina of skeletal muscle, and the cells co-express M-cadherin. Single fiber analyses revealed that M-cadherin+ mononucleated cells attaching to muscle fibers are stained with SM/C-2.6. SM/C-2.6+ cells, which were freshly purified by FACS from mouse skeletal muscle, became MyoD+ in vitro in proliferating medium, and the cells differentiated into desmin+ and nuclear-MyoD+ myofibers in vitro when placed under differentiation conditions. When the sorted cells were injected into mdx mouse muscles, donor cells differentiated into muscle fibers. Flow cytometric analyses of SM/C-2.6+ cells showed that the quiescent satellite cells were c-kit-, Sca-1-, CD34+, and CD45-. More, SM/C-2.6+ cells were barely included in the side population but in the main population of cells in Hoechst dye efflux assay. These results suggest that SM/C-2.6 identifies and enriches quiescent satellite cells from adult mouse muscle, and that the antibody will be useful as a powerful tool for the characterization of cellular and molecular mechanisms of satellite cell activation and proliferation.
Collapse
Affiliation(s)
- So-ichiro Fukada
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
73
|
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
|
74
|
Chao JT, Meininger GA, Patterson JL, Jones SAL, Partridge CR, Neiger JD, Williams ES, Kaufman SJ, Ramos KS, Wilson E. Regulation of alpha7-integrin expression in vascular smooth muscle by injury-induced atherosclerosis. Am J Physiol Heart Circ Physiol 2004; 287:H381-9. [PMID: 14988073 DOI: 10.1152/ajpheart.00939.2003] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Injury of vascular smooth muscle cells (VSMCs) by allylamine (AAM) leads to phenotypic changes associated with atherogenic progression including increased proliferation, migration, and alterations in cell adhesion. In the present study, the relationship between AAM-induced vascular injury and expression of the alpha(7)-integrin subunit was investigated. The alpha(7)-mRNA and protein expression were examined using real-time RT-PCR, fluorescence-activated cell sorting analysis (FACS), immunohistochemistry, and immunoblotting. In cultured VSMCs from aortas of AAM-treated rats (70 mg/kg for 20 days), alpha(7)-mRNA levels were increased more than twofold compared with control cells. No change was seen in beta(1)-integrin expression. FACS analysis revealed increased cell surface expression of alpha(7)-protein (25 +/- 9%; *P < 0.05). AAM treatment of naive VSMCs enhanced alpha(7)-mRNA expression (2.4 +/- 0.7-fold, mean +/- SE; *P < 0.05). The increased alpha(7)-mRNA expression was attenuated by the amine oxidase inhibitor semicarbazide and the antioxidant pyrrolidine dithiocarbamate, which confirms a role for oxidative stress in modulating alpha(7)-expression. In vivo alpha(7)-mRNA and protein expression were enhanced in the aortas of AAM-treated rats. In addition, increased alpha(7)-integrin expression facilitated AAM VSMC adhesion to laminin more efficiently compared with control (51 +/- 2%; *P < 0.05). Chemical injury induced by AAM significantly enhances alpha(7)-integrin expression in VSMCs. These findings implicate for the first time the expression of alpha(7)-integrin during the response of VSMCs to vascular injury.
Collapse
Affiliation(s)
- Jun-Tzu Chao
- Division of Vascular Biology, Cardiovascular Research Institute, Texas A&M Uniersity System Health Science Center, 336 Joe Reynolds Medical Bldg., College Station, TX 77843, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Springer TA, Wang JH. The three-dimensional structure of integrins and their ligands, and conformational regulation of cell adhesion. ADVANCES IN PROTEIN CHEMISTRY 2004; 68:29-63. [PMID: 15500858 DOI: 10.1016/s0065-3233(04)68002-8] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Integrins are a structurally elaborate family of adhesion molecules that transmit signals bidirectionally across the plasma membrane by undergoing large-scale structural rearrangements. By regulating cell-cell and cell-matrix contacts, integrins participate in a wide-range of biological interactions including development, tissue repair, angiogenesis, inflammation and hemostasis. From a therapeutic standpoint, integrins are probably the most important class of cell adhesion receptors. Structural investigations on integrin-ligand interactions reveal remarkable features in molecular detail. These details include the atomic basis for divalent cation-dependent ligand binding and how conformational signals are propagated long distances from one domain to another between the cytoplasm and the extracellular ligand binding site that regulate affinity for ligand, and conversely, cytosolic signaling pathways.
Collapse
Affiliation(s)
- Timothy A Springer
- CBR Institute for Biomedical Research, Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
76
|
McGrath MJ, Mitchell CA, Coghill ID, Robinson PA, Brown S. Skeletal muscle LIM protein 1 (SLIM1/FHL1) induces alpha 5 beta 1-integrin-dependent myocyte elongation. Am J Physiol Cell Physiol 2003; 285:C1513-26. [PMID: 12917103 DOI: 10.1152/ajpcell.00207.2003] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Skeletal muscle LIM protein 1 (SLIM1/FHL1) contains four and a half LIM domains and is highly expressed in skeletal and cardiac muscle. Elevated SLIM1 mRNA expression has been associated with postnatal skeletal muscle growth and stretch-induced muscle hypertrophy in mice. Conversely, SLIM1 mRNA levels decrease during muscle atrophy. Together, these observations suggest a link between skeletal muscle growth and increased SLIM1 expression. However, the precise function of SLIM1 in skeletal muscle, specifically the role of SLIM1 during skeletal muscle differentiation, is not known. This study investigated the effect of increased SLIM1 expression during skeletal muscle differentiation. Western blot analysis showed an initial decrease followed by an increase in SLIM1 expression during differentiation. Overexpression of SLIM1 in Sol8 or C2C12 skeletal muscle cell lines, at levels observed during hypertrophy, induced distinct effects in differentiating myocytes and undifferentiated reserve cells, which were distinguished by differential staining for two markers of differentiation, MyoD and myogenin. In differentiating skeletal myocytes, SLIM1 overexpression induced hyperelongation, which, by either plating cells on poly-l-lysine or using a series of peptide blockade experiments, was shown to be specifically dependent on ligand binding to the alpha5beta1-integrin, whereas in reserve cells, SLIM1 overexpression induced the formation of multiple cytoplasmic protrusions (branching), which was also integrin mediated. These results suggest that SLIM1 may play an important role during the early stages of skeletal muscle differentiation, specifically in alpha5beta1-integrin-mediated signaling pathways.
Collapse
Affiliation(s)
- Meagan J McGrath
- Dept. of Biochemistry and Molecular Biology, Monash Univ., Wellington Rd., Clayton, VIC 3800, Australia
| | | | | | | | | |
Collapse
|
77
|
Li J, Rao H, Burkin D, Kaufman SJ, Wu C. The muscle integrin binding protein (MIBP) interacts with alpha7beta1 integrin and regulates cell adhesion and laminin matrix deposition. Dev Biol 2003; 261:209-19. [PMID: 12941630 DOI: 10.1016/s0012-1606(03)00304-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Integrins are alphabeta transmembrane receptors that function in key cellular processes, including cell adhesion, differentiation, and extracellular matrix deposition through interactions with extracellular, membrane, and cytoplasmic proteins. We previously identified and cloned a muscle beta1 integrin cytoplasmic binding protein termed MIBP and found that the expression level of MIBP is critical in the decision-making process of terminal myogenic differentiation. We report here that MIBP interacts with the alpha7beta1 integrin but not the alpha5beta1 integrin in C2C12 myoblasts, suggesting an important role of integrin alpha chains in the regulation of the beta1-MIBP interaction. Furthermore, consistent with its selective binding activity toward the alpha7beta1 laminin receptor, we have found that overexpression of MIBP in C2C12 myoblasts resulted in a significant reduction of cell adhesion to laminin and inhibition of laminin matrix deposition. By contrast, neither cell adhesion to fibronectin nor fibronectin matrix deposition was significantly altered in cells overexpressing MIBP. Finally, we show that both the protein level and tyrosine phosphorylation of paxillin, a key signaling molecule involved in the cellular control of myogenic differentiation, are increased by MIBP. These results suggest that MIBP functions in the control of myogenic differentiation by regulating alpha7beta1 integrin-mediated cell interactions with laminin matrix and intracellular signaling through paxillin.
Collapse
Affiliation(s)
- Ji Li
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | | | | | | | | |
Collapse
|
78
|
Previtali SC, Dina G, Nodari A, Fasolini M, Wrabetz L, Mayer U, Feltri ML, Quattrini A. Schwann cells synthesize alpha7beta1 integrin which is dispensable for peripheral nerve development and myelination. Mol Cell Neurosci 2003; 23:210-8. [PMID: 12812754 DOI: 10.1016/s1044-7431(03)00014-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Defects in laminins or laminin receptors are responsible for various neuromuscular disorders, including peripheral neuropathies. Interactions between Schwann cells and their basal lamina are fundamental to peripheral nerve development and successful myelination. Selected laminins are expressed in the endoneurium, and their receptors are developmentally regulated during peripheral nerve formation. Loss-of-function mutations have confirmed the importance and the role of some of these molecules. Here we show for the first time that another laminin receptor, alpha7beta1 integrin, previously described only in neurons, is also expressed in Schwann cells. The expression of alpha7 appears postnatally, such that alpha7beta1 is the last laminin receptor expressed by differentiating Schwann cells. Genetic inactivation of the alpha7 subunit in mice does not affect peripheral nerve formation or the expression of other laminin receptors. Of note, alpha7beta1 is not necessary for basal lamina formation and myelination. Nonetheless, these data taken together with the previous demonstration of impaired axonal regrowth in alpha7-null mice suggest a possible Schwann cell-autonomous role for alpha7 in nerve regeneration.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Basement Membrane/metabolism
- Basement Membrane/pathology
- Basement Membrane/ultrastructure
- Cells, Cultured
- Gene Expression Regulation, Developmental/genetics
- Immunohistochemistry
- Integrins/biosynthesis
- Integrins/deficiency
- Integrins/genetics
- Laminin/genetics
- Laminin/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microscopy, Electron
- Myelin Sheath/metabolism
- Myelin Sheath/ultrastructure
- Myopathies, Structural, Congenital/genetics
- Myopathies, Structural, Congenital/metabolism
- Myopathies, Structural, Congenital/pathology
- Nerve Fibers, Myelinated/metabolism
- Nerve Fibers, Myelinated/ultrastructure
- Nerve Regeneration/genetics
- Peripheral Nerves/growth & development
- Peripheral Nerves/metabolism
- Peripheral Nerves/ultrastructure
- Peripheral Nervous System Diseases/genetics
- Peripheral Nervous System Diseases/metabolism
- Peripheral Nervous System Diseases/physiopathology
- Rats
- Rats, Sprague-Dawley
- Schwann Cells/metabolism
- Schwann Cells/ultrastructure
- Spinal Nerve Roots/growth & development
- Spinal Nerve Roots/metabolism
- Spinal Nerve Roots/ultrastructure
Collapse
Affiliation(s)
- S C Previtali
- Neuropathology Unit, San Raffaele Scientific Institute, Milan, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
79
|
Affiliation(s)
- Ulrike Mayer
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, University of Manchester, M13 9PT Manchester, United Kingdom.
| |
Collapse
|
80
|
Eble JA, Berditchevski F. Purification of integrins and characterization of integrin-associated proteins. Methods Cell Biol 2003; 69:223-46. [PMID: 12070995 DOI: 10.1016/s0091-679x(02)69015-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Affiliation(s)
- Johannes A Eble
- Institute of Physiological Chemistry and Pathobiochemistry, Universität Münster, 48149 Münster, Germany
| | | |
Collapse
|
81
|
Hijikata T, Murakami T, Ishikawa H, Yorifuji H. Plectin tethers desmin intermediate filaments onto subsarcolemmal dense plaques containing dystrophin and vinculin. Histochem Cell Biol 2003; 119:109-23. [PMID: 12610730 DOI: 10.1007/s00418-003-0496-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2002] [Indexed: 01/23/2023]
Abstract
Plectin is a versatile cytoskeletal linker protein that preferentially localizes at interfaces between intermediate filaments and the plasma membrane in muscle, epithelial cells, and other tissues. Its deficiency causes muscular dystrophy with epidermolysis bullosa simplex. To better understand the functional roles of plectin beneath the sarcolemma of skeletal muscles and to gain some insights into the underlying mechanism of plectin-deficient muscular dystrophy, we studied in vivo structural and molecular relationships of plectin to subsarcolemmal cytoskeletal components, such as desmin, dystrophin, and vinculin, in rat skeletal muscles. Immunogold electron microscopy revealed that plectin fine threads tethered desmin intermediate filaments onto subsarcolemmal dense plaques overlying Z-lines and I-bands. These dense plaques were found to contain dystrophin and vinculin, and thus may be the structural basis of costameres. The in vivo association of plectin with desmin, (meta-)vinculin, dystrophin, and actin was demonstrated by immunoprecipitation experiments. Treatment of plectin immunoprecipitates with gelsolin reduced actin, dystrophin, and (meta-)vinculin but not desmin, implicating that subsarcolemmal actin could partly mediate the interaction between plectin and dystrophin or (meta-)vinculin. Altogether, our data suggest that plectin, along with desmin intermediate filaments, might serve a vital structural role in the stabilization of the subsarcolemmal cytoskeleton.
Collapse
Affiliation(s)
- Takao Hijikata
- Department of Anatomy, Gunma University School of Medicine, 3-39-22 Showa-machi, 371-8511 Maebashi, Japan.
| | | | | | | |
Collapse
|
82
|
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: 3.7] [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
|
83
|
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: 1.3] [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
|
84
|
Chang W, Webster DR, Salam AA, Gruber D, Prasad A, Eiserich JP, Bulinski JC. Alteration of the C-terminal amino acid of tubulin specifically inhibits myogenic differentiation. J Biol Chem 2002; 277:30690-8. [PMID: 12070174 DOI: 10.1074/jbc.m204930200] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Detyrosination is an evolutionarily conserved post-translational modification of microtubule polymers that is known to be enhanced during early morphological differentiation of cultured myogenic cells (Gundersen, G. G., Khawaja, S., and Bulinski, J. C. (1989) J. Cell Biol. 109, 2275-2288). We proposed that altering the C terminus of alpha-tubulin by detyrosination plays a role in morphological differentiation. To test our hypothesis, we treated L6 myoblasts with 3-nitrotyrosine (Eiserich, J. P., Estevez, A. G., Bamberg, T. V., Ye, Y. Z., Chumley, P. H., Beckman, J. S., and Freeman, B. A. (1999) Proc. Natl. Acad. Sci. U. S. A. 96, 6365-6375), a nontoxic inhibitor that resulted in high level inhibition of microtubule detyrosination and low level incorporation of nitrotyrosine into microtubules. Even though microtubule stabilization or modification by acetylation still occurred normally, morphological differentiation was blocked; myoblasts neither elongated significantly nor fused. Nitrotyrosine treatment prevented synthesis or activation of markers of myogenic differentiation, including muscle-specific myosin, alpha-actin, integrin alpha(7), and myogenin. Consistent with this, myoblast integrin beta(1A) remained highly expressed. In contrast, the increase in beta-catenin level characteristic of early myogenesis was unaffected by treatment. These results show that the identity of the C-terminal residue of alpha-tubulin modulates microtubule activity, possibly because binding to or signaling from modified microtubules is required for the myogenic program.
Collapse
Affiliation(s)
- Winston Chang
- Department of Biological Sciences, College of Arts & Sciences, Columbia University, New York, NY 10027-2450, USA
| | | | | | | | | | | | | |
Collapse
|
85
|
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: 2.3] [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
|
86
|
Blake DJ, Weir A, Newey SE, Davies KE. Function and genetics of dystrophin and dystrophin-related proteins in muscle. Physiol Rev 2002; 82:291-329. [PMID: 11917091 DOI: 10.1152/physrev.00028.2001] [Citation(s) in RCA: 832] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The X-linked muscle-wasting disease Duchenne muscular dystrophy is caused by mutations in the gene encoding dystrophin. There is currently no effective treatment for the disease; however, the complex molecular pathology of this disorder is now being unravelled. Dystrophin is located at the muscle sarcolemma in a membrane-spanning protein complex that connects the cytoskeleton to the basal lamina. Mutations in many components of the dystrophin protein complex cause other forms of autosomally inherited muscular dystrophy, indicating the importance of this complex in normal muscle function. Although the precise function of dystrophin is unknown, the lack of protein causes membrane destabilization and the activation of multiple pathophysiological processes, many of which converge on alterations in intracellular calcium handling. Dystrophin is also the prototype of a family of dystrophin-related proteins, many of which are found in muscle. This family includes utrophin and alpha-dystrobrevin, which are involved in the maintenance of the neuromuscular junction architecture and in muscle homeostasis. New insights into the pathophysiology of dystrophic muscle, the identification of compensating proteins, and the discovery of new binding partners are paving the way for novel therapeutic strategies to treat this fatal muscle disease. This review discusses the role of the dystrophin complex and protein family in muscle and describes the physiological processes that are affected in Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Derek J Blake
- Medical Research Council, Functional Genetics Unit, Department of Human Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | | | | | | |
Collapse
|
87
|
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: 3.3] [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
|
88
|
Tiger CF, Fougerousse F, Grundström G, Velling T, Gullberg D. alpha11beta1 integrin is a receptor for interstitial collagens involved in cell migration and collagen reorganization on mesenchymal nonmuscle cells. Dev Biol 2001; 237:116-29. [PMID: 11518510 DOI: 10.1006/dbio.2001.0363] [Citation(s) in RCA: 156] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
alpha11beta1 integrin constitutes a recent addition to the integrin family. Here, we present the first in vivo analysis of alpha11 protein and mRNA distribution during human embryonic development. alpha11 protein and mRNA were present in various mesenchymal cells around the cartilage anlage in the developing skeleton in a pattern similar to that described for the transcription factor scleraxis. alpha11 was also expressed by mesenchymal cells in intervertebral discs and in keratocytes in cornea, two sites with highly organized collagen networks. Neither alpha11 mRNA nor alpha11 protein could be detected in myogenic cells in human embryos. The described expression pattern is compatible with alpha11beta1 functioning as a receptor for interstitial collagens in vivo. To test this hypothesis in vitro, full-length human alpha11 cDNA was stably transfected into the mouse satellite cell line C2C12, lacking endogenous collagen receptors. alpha11beta1 mediated cell adhesion to collagens I and IV (with a preference for collagen I) and formed focal contacts on collagens. In addition, alpha11beta1 mediated contraction of fibrillar collagen gels in a manner similar to alpha2beta1, and supported migration on collagen I in response to chemotactic stimuli. Our data support a role for alpha11beta1 as a receptor for interstitial collagens on mesenchymally derived cells and suggest a multifunctional role of alpha11beta1 in the recognition and organization of interstitial collagen matrices during development.
Collapse
Affiliation(s)
- C F Tiger
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, S-75124, Sweden
| | | | | | | | | |
Collapse
|
89
|
Lewis MP, Machell JR, Hunt NP, Sinanan AC, Tippett HL. The extracellular matrix of muscle--implications for manipulation of the craniofacial musculature. Eur J Oral Sci 2001; 109:209-21. [PMID: 11531066 DOI: 10.1034/j.1600-0722.2001.00021.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Successful adaptation of craniofacial skeletal muscle is dependent upon the connective tissue component of the muscle. This is exemplified by procedures such as distraction histo/osteogenesis. The mechanisms underlying remodelling of intramuscular connective tissue are complex and multifactorial and involve extracellular matrix (ECM) molecules, receptors for the ECM (integrins) and enzymes that remodel the ECM (MMPs). This review discusses the current state of knowledge and clinical implications of connective tissue biology as applied to craniofacial skeletal muscle.
Collapse
Affiliation(s)
- M P Lewis
- Department of Orthodontics, Eastman Dental Institute, University College London, UK.
| | | | | | | | | |
Collapse
|
90
|
Bezakova G, Helm JP, Francolini M, Lømo T. Effects of purified recombinant neural and muscle agrin on skeletal muscle fibers in vivo. J Cell Biol 2001; 153:1441-52. [PMID: 11425874 PMCID: PMC2150725 DOI: 10.1083/jcb.153.7.1441] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Aggregation of acetylcholine receptors (AChRs) in muscle fibers by nerve-derived agrin plays a key role in the formation of neuromuscular junctions. So far, the effects of agrin on muscle fibers have been studied in culture systems, transgenic animals, and in animals injected with agrin--cDNA constructs. We have applied purified recombinant chick neural and muscle agrin to rat soleus muscle in vivo and obtained the following results. Both neural and muscle agrin bind uniformly to the surface of innervated and denervated muscle fibers along their entire length. Neural agrin causes a dose-dependent appearance of AChR aggregates, which persist > or = 7 wk after a single application. Muscle agrin does not cluster AChRs and at 10 times the concentration of neural agrin does not reduce binding or AChR-aggregating activity of neural agrin. Electrical muscle activity affects the stability of agrin binding and the number, size, and spatial distribution of the neural agrin--induced AChR aggregates. Injected agrin is recovered from the muscles together with laminin and both proteins coimmunoprecipitate, indicating that agrin binds to laminin in vivo. Thus, the present approach provides a novel, simple, and efficient method for studying the effects of agrin on muscle under controlled conditions in vivo.
Collapse
Affiliation(s)
- G Bezakova
- Department of Physiology, University of Oslo, 0317 Oslo, Norway.
| | | | | | | |
Collapse
|
91
|
Palaiologou AA, Yukna RA, Moses R, Lallier TE. Gingival, dermal, and periodontal ligament fibroblasts express different extracellular matrix receptors. J Periodontol 2001; 72:798-807. [PMID: 11453243 DOI: 10.1902/jop.2001.72.6.798] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Fibroblasts are the predominant cells of the periodontal ligament and the gingiva and have important roles in the function and regeneration of the tooth support apparatus. The goal of this study was to investigate the possible differences in the adhesion properties and expression of extracellular matrix (ECM) receptors among different fibroblast populations. METHODS The adhesion of gingival (GF), dermal (DF), and periodontal ligament fibroblast (PDLF) cultures to ECM proteins (fibronectin, laminin, vitronectin, RGD peptide, collagen type I, and collagen type IV) adsorbed to tissue culture plastic was evaluated fluorometrically. Quantitative reverse transcription-polymerase chain reactions (RT-PCR) were performed using primers specific for 19 integrin subunits to quantify ECM receptor transcript expression. RESULTS Our data demonstrated that GF and PDLF adhere to vitronectin and collagen types I and IV more avidly than do DF. PDLF adhered well to laminin, whereas GF and DF did not. Quantitation of integrin expression demonstrated that the different fibroblast types expressed different integrin transcripts, further demonstrating their innate differences. CONCLUSIONS The 3 fibroblast types studied behave differently and expressed different ECM receptors. However, gingival fibroblasts and periodontal ligament fibroblasts are more similar in their attachment and integrin expression than either is to dermal fibroblasts. Therefore, experiments using DF will not necessarily be valid for oral tissues.
Collapse
Affiliation(s)
- A A Palaiologou
- Louisiana State University Health Sciences Center, School of Dentistry, Department of Periodontology, New Orleans 70119, USA
| | | | | | | |
Collapse
|
92
|
Khan J, Wei JS, Ringnér M, Saal LH, Ladanyi M, Westermann F, Berthold F, Schwab M, Antonescu CR, Peterson C, Meltzer PS. Classification and diagnostic prediction of cancers using gene expression profiling and artificial neural networks. Nat Med 2001; 7:673-9. [PMID: 11385503 PMCID: PMC1282521 DOI: 10.1038/89044] [Citation(s) in RCA: 1233] [Impact Index Per Article: 53.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The purpose of this study was to develop a method of classifying cancers to specific diagnostic categories based on their gene expression signatures using artificial neural networks (ANNs). We trained the ANNs using the small, round blue-cell tumors (SRBCTs) as a model. These cancers belong to four distinct diagnostic categories and often present diagnostic dilemmas in clinical practice. The ANNs correctly classified all samples and identified the genes most relevant to the classification. Expression of several of these genes has been reported in SRBCTs, but most have not been associated with these cancers. To test the ability of the trained ANN models to recognize SRBCTs, we analyzed additional blinded samples that were not previously used for the training procedure, and correctly classified them in all cases. This study demonstrates the potential applications of these methods for tumor diagnosis and the identification of candidate targets for therapy.
Collapse
Affiliation(s)
- J Khan
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Kääriäinen M, Liljamo T, Pelto-Huikko M, Heino J, Järvinen M, Kalimo H. Regulation of α7 integrin by mechanical stress during skeletal muscle regeneration. Neuromuscul Disord 2001; 11:360-9. [PMID: 11369187 DOI: 10.1016/s0960-8966(00)00193-0] [Citation(s) in RCA: 18] [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 continuity of the tendon-myofibre-tendon units disrupted by shearing injury must be re-established during regeneration. We have previously demonstrated in freely moving rats that transected myofibres reinforce their lateral integrin-mediated adhesion, with the maximum around days 5-7. After day 14, most integrin molecules are redistributed to the newly formed myotendinous junctions, by which the ends of regenerating myofibres attach to the scar between the stumps. Here, we analyzed the effects of mechanical stress (free and forced mobilization vs. immobilization and denervation separately and in combination) on the expression of alpha7 integrin and merosin in regenerating myofibres using quantitative in situ hybridization and immunohistochemistry. In all groups, alpha7 integrin expression was upregulated at mRNA level, whereas increased protein accumulation in lateral sarcolemma occurred only in the mobilized groups. The accumulation of merosin was not affected by the stress level. The results demonstrate that active mechanical stress reinforces early lateral integrin-mediated adhesion; molecules may at the same time mediate signals from matrix to cells for adaptation to the altered biomechanical status.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Immunohistochemistry
- In Situ Hybridization
- Integrin alpha Chains
- Laminin/metabolism
- Male
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Muscle Fibers, Skeletal/physiology
- Muscle, Skeletal/injuries
- Muscle, Skeletal/pathology
- Muscle, Skeletal/physiopathology
- Protein Isoforms/genetics
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Reference Values
- Regeneration/physiology
- Stress, Mechanical
- Wounds and Injuries/physiopathology
Collapse
Affiliation(s)
- M Kääriäinen
- Medical School and the Institute of Medical Technology, University of Tampere, Tampere, Finland
| | | | | | | | | | | |
Collapse
|
94
|
Blanco-Bose WE, Yao CC, Kramer RH, Blau HM. Purification of mouse primary myoblasts based on alpha 7 integrin expression. Exp Cell Res 2001; 265:212-20. [PMID: 11302686 DOI: 10.1006/excr.2001.5191] [Citation(s) in RCA: 110] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Fundamental insights have come from the study of myogenesis. Primary myoblasts isolated directly from muscle tissue more closely approximate myogenesis than established cell lines. However, contamination of primary muscle cultures with nonmyogenic cells can complicate the results. To overcome this problem, we previously described a method for myoblast purification based on novel culture conditions (T. A. Rando and H. M. Blau, 1994, J. Cell Biol. 125, 1275--1287). Here we report a refinement of this method that leads directly to an enriched population of mouse primary myoblasts, within significantly fewer population doublings. The method described here avoids using adhesion as a criterion for selection. This advance capitalizes on the ability of the antibody CA5.5 to recognize alpha 7 integrin, a muscle-specific cell surface antigen. Enrichment of myoblasts to greater than 95% of the cell population can be achieved by a single round of flow cytometry or magnetic bead separation. This is the first description of a mouse myoblast purification method based on a cell-type-specific antigen. The ease of this procedure for isolating primary myoblasts should expand the opportunities for (1) using these cells in cell transplantation studies in animal models of human disease, (2) isolating and characterizing mutant myoblasts from transgenic animals, and (3) allowing in vitro studies of molecules that regulate muscle cell growth, differentiation, and neoplasia.
Collapse
Affiliation(s)
- W E Blanco-Bose
- Department of Molecular Pharmacology, Stanford University School of Medicine, Stanford, California 94305-5175, USA
| | | | | | | |
Collapse
|
95
|
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
|
96
|
Burkin DJ, Wallace GQ, Nicol KJ, Kaufman DJ, Kaufman SJ. Enhanced expression of the alpha 7 beta 1 integrin reduces muscular dystrophy and restores viability in dystrophic mice. J Cell Biol 2001; 152:1207-18. [PMID: 11257121 PMCID: PMC2199213 DOI: 10.1083/jcb.152.6.1207] [Citation(s) in RCA: 220] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Muscle fibers attach to laminin in the basal lamina using two distinct mechanisms: the dystrophin glycoprotein complex and the alpha 7 beta 1 integrin. Defects in these linkage systems result in Duchenne muscular dystrophy (DMD), alpha 2 laminin congenital muscular dystrophy, sarcoglycan-related muscular dystrophy, and alpha 7 integrin congenital muscular dystrophy. Therefore, the molecular continuity between the extracellular matrix and cell cytoskeleton is essential for the structural and functional integrity of skeletal muscle. To test whether the alpha 7 beta 1 integrin can compensate for the absence of dystrophin, we expressed the rat alpha 7 chain in mdx/utr(-/-) mice that lack both dystrophin and utrophin. These mice develop a severe muscular dystrophy highly akin to that in DMD, and they also die prematurely. Using the muscle creatine kinase promoter, expression of the alpha 7BX2 integrin chain was increased 2.0-2.3-fold in mdx/utr(-/-) mice. Concomitant with the increase in the alpha 7 chain, its heterodimeric partner, beta 1D, was also increased in the transgenic animals. Transgenic expression of the alpha 7BX2 chain in the mdx/utr(-/-) mice extended their longevity by threefold, reduced kyphosis and the development of muscle disease, and maintained mobility and the structure of the neuromuscular junction. Thus, bolstering alpha 7 beta 1 integrin-mediated association of muscle cells with the extracellular matrix alleviates many of the symptoms of disease observed in mdx/utr(-/-) mice and compensates for the absence of the dystrophin- and utrophin-mediated linkage systems. This suggests that enhanced expression of the alpha 7 beta 1 integrin may provide a novel approach to treat DMD and other muscle diseases that arise due to defects in the dystrophin glycoprotein complex. A video that contrasts kyphosis, gait, joint contractures, and mobility in mdx/utr(-/-) and alpha 7BX2-mdx/utr(-/-) mice can be accessed at http://www.jcb.org/cgi/content/full/152/6/1207.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Body Weight
- Contracture/physiopathology
- Creatine Kinase/genetics
- Creatine Kinase, MM Form
- Cytoskeletal Proteins/genetics
- Cytoskeletal Proteins/metabolism
- Dystrophin/genetics
- Dystrophin/metabolism
- Female
- Hindlimb
- Humans
- Integrins/genetics
- Integrins/metabolism
- Isoenzymes/genetics
- Joints
- Kyphosis
- Magnetic Resonance Imaging
- Male
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred mdx
- Mice, Transgenic
- Microscopy, Fluorescence
- Muscle, Skeletal/pathology
- Muscle, Skeletal/physiopathology
- Muscular Dystrophy, Animal/genetics
- Muscular Dystrophy, Animal/pathology
- Muscular Dystrophy, Animal/physiopathology
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/physiopathology
- Neuromuscular Junction/ultrastructure
- Promoter Regions, Genetic
- Rats
- Receptors, Cholinergic/metabolism
- Receptors, Cholinergic/ultrastructure
- Survival Rate
- Transgenes
- Utrophin
Collapse
Affiliation(s)
- Dean J. Burkin
- Department of Cell and Structural Biology, University of Illinois, Urbana, Illinois 61801
| | - Gregory Q. Wallace
- Department of Cell and Structural Biology, University of Illinois, Urbana, Illinois 61801
| | - Kimberly J. Nicol
- Department of Cell and Structural Biology, University of Illinois, Urbana, Illinois 61801
| | | | - Stephen J. Kaufman
- Department of Cell and Structural Biology, University of Illinois, Urbana, Illinois 61801
| |
Collapse
|
97
|
Guan K, Czyz J, Fürst DO, Wobus AM. Expression and cellular distribution of alpha(v)integrins in beta(1)integrin-deficient embryonic stem cell-derived cardiac cells. J Mol Cell Cardiol 2001; 33:521-32. [PMID: 11181020 DOI: 10.1006/jmcc.2000.1326] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
beta(1)integrin-deficient (beta(1)-/-) ES cells showed increased differentiation of cardiac cells characterized by reduced adhesion and high beating frequency. Whereas in whole embryoid body outgrowths of beta(1)-/- cells maximum levels of alpha(v), beta(3)and beta(5)integrin mRNA were delayed and transiently upregulated, in cardiac clusters isolated from beta(1)-/- cells, only beta(3)integrin mRNA levels were enhanced in comparison to wild-type (wt) cells. To answer the question, whether alpha(v)and beta(3)integrins may compensate, at least partially, the loss of beta(1)integrin function during cardiac differentiation, the distribution of alpha(v)and beta(3)integrins in beta(1)-/- and wt pacemaker-like cardiac cells was analyzed. A different distribution of alpha(v)and beta(3)integrins in beta(1)-/- v wt cardiac cells was found. In wt cardiac cells, beta(1)integrin was localized in specialized subsarcolemmal regions, in particular, at focal contacts and costameres, but alpha(v)integrin was diffusely distributed. In contrast, in beta(1)-/- cardiac cells, alpha(v)integrin was preponderantly localized at cell membranes, focal contacts and costameres. beta(3)integrin displayed a diffuse pattern both in wt and in beta(1)-/- pacemaker-like cells at early differentiation stages, whereas at terminal stages, beta(3)was colocalized with sarcomeres in wt, but not in beta(1)-/- pacemaker-like cells. Quantitative immunofluorescence analysis revealed increased alpha(v)and beta(3)integrin levels in beta(1)-/- pacemaker-like cardiac cells. Our results led us to conclude that altered cellular distribution of alpha(v)integrin and upregulation of beta(3)integrin correlate with growth and survival of beta(1)-/- cardiac pacemaker-like cells at an early developmental state. However, alpha(v)and beta(3)integrins cannot functionally compensate the loss of beta(1)integrin during terminal differentiation of cardiac cells implicating that cardiomyocytes require specific beta(1)integrin functions for cardiac specialization.
Collapse
Affiliation(s)
- K Guan
- In Vitro Differentiation Group, IPK Gatersleben, D-06466 Gatersleben, Germany
| | | | | | | |
Collapse
|
98
|
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
|
99
|
Hammarberg H, Wallquist W, Piehl F, Risling M, Cullheim S. Regulation of laminin-associated integrin subunit mRNAs in rat spinal motoneurons during postnatal development and after axonal injury. J Comp Neurol 2000; 428:294-304. [PMID: 11064368 DOI: 10.1002/1096-9861(20001211)428:2<294::aid-cne8>3.0.co;2-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Two important prerequisites for successful axon regeneration are that appropriate extracellular molecules are available for outgrowing axons and that receptors for such molecules are found in the regenerating neuron. Laminins and their receptors in the integrin family are examples of such molecules, and laminin-associated integrin subunits alpha 3, alpha 6, alpha 7, and beta 1 mRNAs have all been detected in adult rat motoneurons. We have here, by use of in situ hybridization histochemistry, examined the normal postnatal development of the expression in motoneurons of these mRNAs and integrin beta 4 mRNA, all of which have been associated with laminin-2. We studied the regulation of these mRNAs, 1-42 days after two types of axotomy in the adult rat (sciatic nerve transection, SNT; ventral root avulsion, VRA) and 1-10 days after SNT in the neonatal animal. During postnatal development, there was a distinct shift in the integrin composition from a stronger expression of the alpha 6 subunit to a very clear dominance of alpha 7 in the adult. All types of axotomy in the adult rat induced initial (1-7 days) large up-regulations of alpha 6, alpha 7 and beta1 subunit mRNAs (250-500%). Only minor changes for alpha 3 mRNA were seen, and beta 4 mRNA could not be detected at all in motoneurons. After adult SNT, the alpha 7 and beta 1 subunits were up-regulated throughout the studied period, and the alpha 6 subunit mRNA was eventually normalized. After VRA, however, the alpha 7 and beta1 levels peaked earlier than after SNT and were normalized at 42 days, whereas alpha 6 mRNA was up-regulated longer than after SNT. Neonatal SNT had much smaller effects on the expression of the studied subunits. The results suggest that an important part of the response to axotomy of motoneurons is to up-regulate receptors for laminin. The developmental shift in integrin subunit composition and the various responses seen in the lesion models indicate that different isoforms of laminin play a role in the regenerative response.
Collapse
Affiliation(s)
- H Hammarberg
- Department of Neuroscience, Karolinska Institute, S-171 77 Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
100
|
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
|