Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 13(1): 14556, 2023 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-37666868

RESUMO

Deriving stem cells to regenerate full-thickness human skin is important for treating skin disorders without invasive surgical procedures. Our previous protocol to differentiate human induced pluripotent stem cells (iPSCs) into skin-derived precursor cells (SKPs) as a source of dermal stem cells employs mouse fibroblasts as feeder cells and is therefore unsuitable for clinical use. Herein, we report a feeder-free method for differentiating iPSCs into SKPs by customising culture substrates. We immunohistochemically screened for laminins expressed in dermal papillae (DP) and explored the conditions for inducing the differentiation of iPSCs into SKPs on recombinant laminin E8 (LM-E8) fragments with or without conjugation to domain I of perlecan (PDI), which binds to growth factors through heparan sulphate chains. Several LM-E8 fragments, including those of LM111, 121, 332, 421, 511, and 521, supported iPSC differentiation into SKPs without PDI conjugation. However, the SKP yield was significantly enhanced on PDI-conjugated LM-E8 fragments. SKPs induced on PDI-conjugated LM111-E8 fragments retained the gene expression patterns characteristic of SKPs, as well as the ability to differentiate into adipocytes, osteocytes, and Schwann cells. Thus, PDI-conjugated LM-E8 fragments are promising agents for inducing iPSC differentiation into SKPs in clinical settings.


Assuntos
Diferenciação Celular , Proteoglicanas de Heparan Sulfato , Células-Tronco Pluripotentes Induzidas , Peptídeos e Proteínas de Sinalização Intercelular , Laminina , Fragmentos de Peptídeos , Domínios Proteicos , Pele , Humanos , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proteoglicanas de Heparan Sulfato/química , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Laminina/química , Laminina/farmacologia , Osteócitos/citologia , Osteócitos/efeitos dos fármacos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/farmacologia , Células de Schwann/citologia , Células de Schwann/efeitos dos fármacos , Pele/citologia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/farmacologia
2.
Matrix Biol Plus ; 18: 100133, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37131404

RESUMO

Basement membranes (BMs) are thin, sheet-like extracellular structures that cover the basal side of epithelial and endothelial tissues and provide structural and functional support to adjacent cell layers. The molecular structure of BMs is a fine meshwork that incorporates specialized extracellular matrix proteins. Recently, live visualization of BMs in invertebrates demonstrated that their structure is flexible and dynamically rearranged during cell differentiation and organogenesis. However, the BM dynamics in mammalian tissues remain to be elucidated. We developed a mammalian BM imaging probe based on nidogen-1, a major BM-specific protein. Recombinant human nidogen-1 fused with an enhanced green fluorescent protein (Nid1-EGFP) retains its ability to bind to other BM proteins, such as laminin, type IV collagen, and perlecan, in a solid-phase binding assay. When added to the culture medium of embryoid bodies derived from mouse ES cells, recombinant Nid1-EGFP accumulated in the BM zone of embryoid bodies, and BMs were visualized in vitro. For in vivo BM imaging, a knock-in reporter mouse line expressing human nidogen-1 fused to the red fluorescent protein mCherry (R26-CAG-Nid1-mCherry) was generated. R26-CAG-Nid1-mCherry showed fluorescently labeled BMs in early embryos and adult tissues, such as the epidermis, intestine, and skeletal muscles, whereas BM fluorescence was unclear in several other tissues, such as the lung and heart. In the retina, Nid1-mCherry fluorescence visualized the BMs of vascular endothelium and pericytes. In the developing retina, Nid1-mCherry fluorescence labeled the BM of the major central vessels; however, the BM fluorescence were hardly observed in the peripheral growing tips of the vascular network, despite the presence of endothelial BM. Time-lapse observation of the retinal vascular BM after photobleaching revealed gradual recovery of Nid1-mCherry fluorescence, suggesting the turnover of BM components in developing retinal blood vessels. To the best of our knowledge, this is the first demonstration of in vivo BM imaging using a genetically engineered mammalian model. Although R26-CAG-Nid1-mCherry has some limitations as an in vivo BM imaging model, it has potential applications in the study of BM dynamics during mammalian embryogenesis, tissue regeneration, and pathogenesis.

3.
Microscopy (Oxf) ; 71(6): 357-363, 2022 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-35950724

RESUMO

Most epithelial tissues rapidly become complex during embryonic development while being surrounded by the basement membrane (BM). Thus, the BM shape is thought to change dramatically as the epithelium grows, but the underlying mechanism is not yet clear. Nidogen-1 is ubiquitous in the BM and binds to various other BM components, including laminin and type IV collagen. To elucidate the behavior of the BM during epithelial morphogenesis, we attempted to live-label the developing BM with recombinant human nidogen-1 fused to an enhanced green fluorescent protein (hNid1-EGFP). Submandibular glands of mouse embryos were cultured in glass-bottomed dishes and incubated in media containing hNid1-EGFP. Subsequent confocal microscopy clearly visualized the BMs surrounding the epithelial end buds. On three-dimensional reconstruction from Z-series confocal sections, the epithelial BM was observed as a thin sheet that expanded continuously around the entire epithelial basal surface. Because the explants continued to grow well in the presence of hNid1-EGFP, time-lapse confocal microscopy was performed to follow the dynamics of the BM. We found that the epithelial BM is an adaptive structure that deforms in accordance with the rapid shape changes of the developing epithelium. Furthermore, hNid1-EGFP was found to be incorporated differently into the epithelial BM compared with that reported for fibronectin or type IV collagen, suggesting that individual BM components assemble in different ways to form the BM.


Assuntos
Colágeno Tipo IV , Glândulas Salivares , Humanos , Animais , Camundongos , Membrana Basal
4.
Mol Biol Cell ; 30(1): 56-68, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30379609

RESUMO

Neural stem cells (NSCs) are retained in the adult ventricular-subventricular zone (V-SVZ), a specialized neurogenic niche with a unique cellular architecture. It currently remains unclear whether or how NSCs utilize basement membranes (BMs) in this niche. Here, we examine the molecular compositions and functions of BMs in the adult mouse V-SVZ. Whole-mount V-SVZ immunostaining revealed that fractones, which are fingerlike processes of extravascular BMs, are speckled BMs unconnected to the vasculature, and differ in their molecular composition from vascular BMs. Glial fibrillary acidic protein (GFAP)-positive astrocytes and NSCs produce and adhere to speckled BMs. Furthermore, Gfap-Cre-mediated Lamc1flox(E1605Q) knockin mice, in which integrin-binding activities of laminins are specifically nullified in GFAP-positive cells, exhibit a decreased number and size of speckled BMs and reduced in vitro neurosphere-forming activity. Our results reveal niche activities of fractones/speckled BMs for NSCs and provide molecular insights into how laminin-integrin interactions regulate NSCs in vivo.


Assuntos
Membrana Basal/metabolismo , Ventrículos Laterais/metabolismo , Células-Tronco Neurais/metabolismo , Nicho de Células-Tronco , Animais , Animais Recém-Nascidos , Membrana Basal/irrigação sanguínea , Membrana Basal/citologia , Proliferação de Células , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Epêndima/citologia , Epêndima/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Integrinas/metabolismo , Laminina/metabolismo , Ventrículos Laterais/citologia , Camundongos Endogâmicos C57BL , Mutação/genética , Células-Tronco Neurais/citologia
5.
Circ Res ; 120(8): 1276-1288, 2017 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-28179430

RESUMO

RATIONALE: Lymphatic vasculature constitutes a second vascular system essential for immune surveillance and tissue fluid homeostasis. Maturation of the hierarchical vascular structure, with a highly branched network of capillaries and ducts, is crucial for its function. Environmental cues mediate the remodeling process, but the mechanism that underlies this process is largely unknown. OBJECTIVE: Polydom (also called Svep1) is an extracellular matrix protein identified as a high-affinity ligand for integrin α9ß1. However, its physiological function is unclear. Here, we investigated the role of Polydom in lymphatic development. METHODS AND RESULTS: We generated Polydom-deficient mice. Polydom-/- mice showed severe edema and died immediately after birth because of respiratory failure. We found that although a primitive lymphatic plexus was formed, it failed to undergo remodeling in Polydom-/- embryos, including sprouting of new capillaries and formation of collecting lymphatic vessels. Impaired lymphatic development was also observed after knockdown/knockout of polydom in zebrafish. Polydom was deposited around lymphatic vessels, but secreted from surrounding mesenchymal cells. Expression of Foxc2 (forkhead box protein c2), a transcription factor involved in lymphatic remodeling, was decreased in Polydom-/- mice. Polydom bound to the lymphangiogenic factor Ang-2 (angiopoietin-2), which was found to upregulate Foxc2 expression in cultured lymphatic endothelial cells. Expressions of Tie1/Tie2 receptors for angiopoietins were also decreased in Polydom-/- mice. CONCLUSIONS: Polydom affects remodeling of lymphatic vessels in both mouse and zebrafish. Polydom deposited around lymphatic vessels seems to ensure Foxc2 upregulation in lymphatic endothelial cells, possibly via the Ang-2 and Tie1/Tie2 receptor system.


Assuntos
Células Endoteliais/metabolismo , Linfangiogênese , Vasos Linfáticos/metabolismo , Proteínas/metabolismo , Angiopoietina-2/metabolismo , Animais , Proteínas de Ligação ao Cálcio , Moléculas de Adesão Celular , Comunicação Celular , Células Cultivadas , Edema/genética , Edema/metabolismo , Edema/fisiopatologia , Células Endoteliais/patologia , Endotélio Linfático/anormalidades , Endotélio Linfático/metabolismo , Endotélio Linfático/fisiopatologia , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Humanos , Vasos Linfáticos/anormalidades , Vasos Linfáticos/fisiopatologia , Mesoderma/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Ligação Proteica , Proteínas/genética , Receptor de TIE-1/genética , Receptor de TIE-1/metabolismo , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Transdução de Sinais , Ducto Torácico/anormalidades , Ducto Torácico/metabolismo , Ducto Torácico/fisiopatologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
6.
PLoS One ; 8(7): e68781, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23874761

RESUMO

Loss of function mutations in GPR56, which encodes a G protein-coupled receptor, cause a specific human brain malformation called bilateral frontoparietal polymicrogyria (BFPP). Studies from BFPP postmortem brain tissue and Gpr56 knockout mice have previously showed that GPR56 deletion leads to breaches in the pial basement membrane (BM) and neuronal ectopias during cerebral cortical development. Since α3ß1 integrin also plays a role in pial BM assembly and maintenance, we evaluated whether it functions together with GPR56 in regulating the same developmental process. We reveal that loss of α3 integrin enhances the cortical phenotype associated with Gpr56 deletion, and that neuronal overmigration through a breached pial BM occurs earlier in double knockout than in Gpr56 single knockout mice. These observations provide compelling evidence of the synergism of GPR56 and α3ß1 integrin in regulating the development of cerebral cortex.


Assuntos
Córtex Cerebral/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Membrana Basal/metabolismo , Membrana Basal/patologia , Movimento Celular/genética , Córtex Cerebral/embriologia , Córtex Cerebral/patologia , Colágeno Tipo III/metabolismo , Deleção de Genes , Integrina alfa3beta1/genética , Integrina alfa3beta1/metabolismo , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Fenótipo , Ligação Proteica , Receptores Acoplados a Proteínas G/genética
7.
Matrix Biol ; 32(3-4): 188-95, 2013 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-23357641

RESUMO

Nephronectin is a basement membrane protein comprising five N-terminal epidermal growth factor (EGF)-like repeats, a central linker segment containing an Arg-Gly-Asp (RGD) motif and a C-terminal meprin-A5 protein-receptor protein tyrosine phosphatase µ (MAM) domain. Nephronectin has been shown to interact with α8ß1 integrin through the central linker segment, but its interactions with other molecules remain to be elucidated. Here, we examined the binding of nephronectin to a panel of glycosaminoglycan (GAG) chains. Nephronectin bound strongly to heparin and chondroitin sulfate (CS)-E and moderately to heparan sulfate (HS), but failed to bind to CS-A, CS-C, CS-D, dermatan sulfate and hyaluronic acid. Deletion of the MAM domain severely impaired the binding of nephronectin to heparin but not CS-E, whereas deletion of the EGF-like repeats reduced its binding to CS-E but not heparin, suggesting that nephronectin interacts with CS-E and heparin through the EGF-like repeats and MAM domain, respectively. Consistent with these results, nephronectin bound to agrin and perlecan, which are heparan sulfate proteoglycans (HSPGs) in basement membranes, in HS-dependent manners. Site-directed mutagenesis of the MAM domain revealed that multiple basic amino acid residues in the putative loop regions were involved in the binding of the MAM domain to agrin. The binding of nephronectin to basement membrane HSPGs was further confirmed by in situ nephronectin overlay assays using mouse frozen tissue sections. Taken together, these findings indicate that nephronectin is capable of binding to HSPGs in basement membranes via the MAM domain, and thereby raise the possibility that interactions with basement membrane HSPGs may be involved in the deposition of nephronectin onto basement membranes.


Assuntos
Agrina/química , Proteínas da Matriz Extracelular/química , Proteoglicanas de Heparan Sulfato/química , Agrina/metabolismo , Sequência de Aminoácidos , Animais , Membrana Basal , Embrião de Mamíferos/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Células HEK293 , Proteoglicanas de Heparan Sulfato/metabolismo , Humanos , Rim/metabolismo , Pulmão/metabolismo , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína
8.
Neurosci Res ; 71(4): 421-6, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21920392

RESUMO

Laminins play an important role in neuronal development. However, the effects of each laminin isoform on neurite morphology remain unclear. Here, we examined the effects of particular laminin (LN) isoforms on hippocampal neuron morphology. We found that LN-511 remarkably promoted elongation of both axons and dendrites, but reduced the number of dendrites. LN-511 E8 fragment, which includes the integrin-binding region, promoted axon and dendrite outgrowth and increased the dendrite number, although its effect on axon and dendrite elongation was smaller than that of full-length LN-511. These results suggest that LN-511 regulates axon and dendrite development by integrin-dependent and -independent mechanisms.


Assuntos
Axônios/ultraestrutura , Dendritos/ultraestrutura , Hipocampo/embriologia , Laminina/metabolismo , Neurogênese/fisiologia , Neurônios/ultraestrutura , Animais , Isoformas de Proteínas , Ratos
9.
J Biol Chem ; 285(47): 36645-55, 2010 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-20843788

RESUMO

Activin A, a member of the transforming growth factor-ß family, plays important roles in hormonal homeostasis and embryogenesis. In this study, we produced recombinant human activin A and examined its abilities to bind to extracellular matrix proteins. Recombinant activin A expressed in 293-F cells was purified as complexes of mature dimeric activin A with its pro-region. Among a panel of extracellular matrix proteins tested, recombinant activin A bound to perlecan and agrin, but not to laminins, nidogens, collagens I and IV, fibronectin, and nephronectin. The binding of recombinant activin A to perlecan was inhibited by heparin and high concentrations of NaCl and abolished by heparitinase treatment of perlecan, suggesting that activin A binds to the heparan sulfate chains of perlecan. In support of this possibility, recombinant activin A was capable of directly binding to heparin and heparan sulfate chains. Site-directed mutagenesis of recombinant activin A revealed that clusters of basic amino acid residues, Lys(259)-Lys(263) and Lys(270)-Lys(272), in the pro-region were required for binding to perlecan. Interestingly, deletion of the peptide segment Lys(259)-Gly(277) containing both basic amino acid clusters from the pro-region did not impair the activity of activin A to stimulate Smad-dependent gene expressions, although it completely ablated the perlecan-binding activity. The binding of activin A to basement membrane heparan sulfate proteoglycans through the basic residues in the pro-region was further confirmed by in situ activin A overlay assays using frozen tissue sections. Taken together, the present results indicate that activin A binds to heparan sulfate proteoglycans through its pro-region and thereby regulates its localization within tissues.


Assuntos
Ativinas/metabolismo , Proteoglicanas de Heparan Sulfato/metabolismo , Heparina/metabolismo , Heparitina Sulfato/metabolismo , Proteínas Recombinantes/metabolismo , Ativinas/genética , Sequência de Aminoácidos , Animais , Western Blotting , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Humanos , Técnicas Imunoenzimáticas , Laminina/metabolismo , Luciferases/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ligação Proteica , Homologia de Sequência de Aminoácidos
10.
J Biochem ; 147(4): 565-79, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19996152

RESUMO

The C1q family is characterized by the C-terminally conserved globular C1q (gC1q) domain. Although more than 30 C1q family proteins have been identified in mammals, many of them remain ill-defined with respect to their molecular and biological properties. Here, we report on a novel C1q family protein specifically expressed in the central nervous system (CNS), which we designated neural C1q-like protein (nCLP) 2. nCLP2 was secreted as disulphide-bonded multimers comprising trimeric units. The multimers were stabilized by interchain disulphide bonds involving the cysteine residues in the N-terminal variable region and the C-terminal gC1q domain. The expression of nCLP2 was restricted to several brain regions and retina, including regions associated with memory formation (i.e. hippocampus, entorhinal cortex, anterodorsal thalamic nucleus). Immunoelectron microscopy revealed that nCLP2 was localized in the mossy fibre axons of hippocampal granule cells and their synaptic boutons and clefts, implying that nCLP2 was anterogradely transported in mossy fibres and secreted from the presynaptic termini. These results suggest that nCLP2 plays roles in synaptic function and maintenance in the CNS.


Assuntos
Sistema Nervoso Central/metabolismo , Complemento C1q/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Sequência de Aminoácidos , Animais , Sistema Nervoso Central/citologia , Complemento C1q/química , Complemento C1q/genética , Cisteína/química , Perfilação da Expressão Gênica , Biblioteca Gênica , Camundongos , Camundongos Endogâmicos ICR , Dados de Sequência Molecular , Fibras Musgosas Hipocampais/metabolismo , Fibras Musgosas Hipocampais/ultraestrutura , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/isolamento & purificação , Proteínas Mutantes/metabolismo , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Especificidade de Órgãos , Domínios e Motivos de Interação entre Proteínas , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Multimerização Proteica , Sinais Direcionadores de Proteínas , Transporte Proteico , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Retina/citologia , Retina/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
11.
Proc Natl Acad Sci U S A ; 106(8): 2800-5, 2009 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-19196964

RESUMO

Anti-p200 pemphigoid has been characterized by autoantibodies to an unidentified 200-kDa protein (p200) of the dermal-epidermal junction. The objective of this study was to identify p200. We performed 2D gel electrophoresis of dermal extracts and immunoblotting with patients' sera, followed by MS analysis of a unique protein band. The protein band corresponded to laminin gamma1. Anti-laminin gamma1 mAb reacted with the anti-p200 immunoprecipitates by immunoblotting. Sera from 32 patients with anti-p200 pemphigoid showed 90% reactivity to the recombinant products of laminin gamma1. None of the healthy control sera reacted with laminin gamma1. By immunoblotting, reactivity of a patient's serum with p200 was competitively inhibited by adding anti-laminin gamma1 C-terminus mAb. Purified anti-p200 IgG also inhibited the reactivity of this mAb to dermal laminin gamma1. Most laminin gamma1-positive sera showed reactivity with recombinant laminin gamma1 C-terminal E8 fragment. Reactivity of patients' sera and purified IgG to dermal laminin gamma1 was higher than reactivity to blood vessel laminin gamma1 under reducing conditions. These results suggest that laminin gamma1 is the autoantigen for patients with anti-p200 pemphigoid. The autoantibodies may specifically recognize dermal laminin gamma1 with unique posttranslational modifications. The epitope is localized to the 246 C-terminal amino acids within the coiled-coil domain. The 9 C-terminal residues are known to be critically involved in laminin recognition by integrins.


Assuntos
Anticorpos Monoclonais/imunologia , Autoanticorpos/imunologia , Laminina/imunologia , Penfigoide Bolhoso/imunologia , Sequência de Aminoácidos , Autoanticorpos/química , Western Blotting , Eletroforese em Gel Bidimensional , Epitopos/química , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Imunoprecipitação , Dados de Sequência Molecular , Especificidade de Órgãos , Pele/imunologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
12.
Proc Natl Acad Sci U S A ; 105(35): 12849-54, 2008 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-18757743

RESUMO

Extracellular matrix (ECM), which provides critical scaffolds for all adhesive cells, regulates proliferation, differentiation, and apoptosis. Different cell types employ customized ECMs, which are thought to play important roles in the generation of so-called niches that contribute to cell-specific functions. The molecular entities of these customized ECMs, however, have not been elucidated. Here, we describe a strategy for transcriptome-wide identification of ECM proteins based on computational screening of >60,000 full-length mouse cDNAs for secreted proteins, followed by in vitro functional assays. These assays screened the candidate proteins for ECM-assembling activities, interactions with other ECM molecules, modifications with glycosaminoglycans, and cell-adhesive activities, and were then complemented with immunohistochemical analysis. We identified 16 ECM proteins, of which seven were localized in basement membrane (BM) zones. The identification of these previously unknown BM proteins allowed us to construct a body map of BM proteins, which represents the comprehensive immunohistochemistry-based expression profiles of the tissue-specific customization of BMs.


Assuntos
Proteínas da Matriz Extracelular/análise , Perfilação da Expressão Gênica , Animais , Membrana Basal/citologia , Membrana Basal/metabolismo , Linhagem Celular , Biologia Computacional , Epitélio/metabolismo , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos ICR , Transporte Proteico , Dente/citologia , Dente/embriologia
13.
Biochem Biophys Res Commun ; 299(3): 498-504, 2002 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-12445830

RESUMO

Laminins, the major basement membrane glycoproteins, are composed of three subunits. We identified a splice variant of the human laminin alpha4 subunit transcript containing 21 extra nucleotides. A heptapeptide sequence, MDCPTIS, was inserted close to the two cysteine residues possibly involved in the intersubunit disulfide bonds. Both the authentic alpha4 subunit (alpha4A) and the variant with the heptapeptide insertion (alpha4B) were readily secreted as laminin-8 trimers (alpha4Abeta1gamma1 or alpha4Bbeta1gamma1) upon cotransfection with expression vectors for the beta1 and gamma1 subunits. The purified recombinant laminin-8 containing the alpha4B subunit was more potent in promoting cell spreading than that containing alpha4A, raising the possibility that the alternative splicing of the alpha4 subunit transcript regulates the cell-adhesive activity of laminin-8. Since both alpha4A and alpha4B transcripts were detected by RT-PCR in several human cell lines, these two isoforms of laminin-8 with differing cell-adhesive activities are present in the basement membranes of human tissues.


Assuntos
Processamento Alternativo , Laminina/genética , Proteínas Recombinantes de Fusão/genética , Adesão Celular/fisiologia , Linhagem Celular , Clonagem Molecular , Humanos , Laminina/metabolismo , Subunidades Proteicas , Proteínas Recombinantes de Fusão/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...