Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
AJNR Am J Neuroradiol ; 41(9): 1584-1591, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32819894

RESUMO

BACKGROUND AND PURPOSE: Slowly expanding/evolving lesions measured by conventional T1-weighted/T2-weighted brain MR imaging may contribute to progressive disability accumulation in MS. We evaluated the longitudinal change in myelin and axonal tissue integrity in white matter slowly expanding/evolving lesions by means of the magnetization transfer ratio and DTI radial diffusivity. MATERIALS AND METHODS: Slowly expanding/evolving lesions were detected within the Study to Assess the Efficacy, Safety, Tolerability, and Pharmacokinetics of BIIB033 in Participants With Relapsing Forms of Multiple Sclerosis When Used Concurrently With Avonex (SYNERGY) Phase 2 clinical trial dataset (NCT01864148), comprising patients with relapsing-remitting and secondary-progressive MS (n = 299) with T1-weighted/T2-weighted MR imaging at all trial time points (baseline to week 72). RESULTS: Compared with non-slowly expanding/evolving lesions (areas not classified as slowly expanding/evolving lesion) of baseline nonenhancing T2 lesions, slowly expanding/evolving lesions had a lower normalized magnetization transfer ratio and greater DTI radial diffusivity, both in patients with relapsing-remitting MS (n = 242) and secondary-progressive MS (n = 57, P < .001 for all). Although the changes with time in both the normalized magnetization transfer ratio and DTI radial diffusivity between slowly expanding/evolving lesions and non-slowly expanding/evolving lesions were positively correlated (P < .001), a decrease in the normalized magnetization transfer ratio and a greater increase in DTI radial diffusivity were observed in slowly expanding/evolving lesions versus non-slowly expanding/evolving lesions from baseline to week 72 in relapsing-remitting MS and secondary-progressive MS (P < .001 for all). CONCLUSIONS: Patterns of longitudinal change in the normalized magnetization transfer ratio and DTI radial diffusivity in slowly expanding/evolving lesions were consistent with progressive demyelination and tissue loss, as seen in smoldering white matter MS plaques.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Doenças Desmielinizantes/diagnóstico por imagem , Esclerose Múltipla/diagnóstico por imagem , Esclerose Múltipla/tratamento farmacológico , Substância Branca/diagnóstico por imagem , Adulto , Encéfalo/diagnóstico por imagem , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Doenças Desmielinizantes/tratamento farmacológico , Doenças Desmielinizantes/patologia , Imagem de Difusão por Ressonância Magnética , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla/patologia , Substância Branca/efeitos dos fármacos , Substância Branca/patologia
2.
J Biol Chem ; 276(18): 14602-6, 2001 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-11278268

RESUMO

Homozygosity for the aly point mutation in NF-kappaB-inducing kinase (NIK) results in alymphoplasia in mice, a phenotype similar to that of homozygosity for deletion of the lymphotoxin beta receptor (LTbetaR). We now find that NF-kappaB activation by Epstein-Barr virus latent membrane protein 1 (LMP1) or by an LMP1 transmembrane domain chimera with the LTbetaR signaling domain in human embryonic kidney 293 cells is selectively inhibited by a wild type dominant negative NIK comprised of amino acids 624-947 (DN-NIK) and not by aly DN-NIK. In contrast, LMP1/CD40 is inhibited by both wild type (wt) and aly DN-NIK. LMP1, an LMP1 transmembrane domain chimera with the LTbetaR signaling domain, and LMP1/CD40 activate NF-kappaB in wt or aly murine embryo fibroblasts. Although wt and aly NIK do not differ in their in vitro binding to tumor necrosis factor receptor-associated factor 1, 2, 3, or 6 or in their in vivo association with tumor necrosis factor receptor-associated factor 2 and differ marginally in their very poor binding to IkappaB kinase beta (IKKbeta), only wt NIK is able to bind to IKKalpha. These data are compatible with a model in which activation of NF-kappaB by LMP1 and LTbetaR is mediated by an interaction of NIK or a NIK-like kinase with IKKalpha that is abrogated by the aly mutation. On the other hand, CD40 mediates NF-kappaB activation through a kinase that interacts with a different component of the IKK complex.


Assuntos
Mutação , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/genética , Receptores do Fator de Necrose Tumoral/metabolismo , Proteínas da Matriz Viral/metabolismo , Sequência de Bases , Antígenos CD40/metabolismo , Linhagem Celular , Primers do DNA , Humanos , Quinase I-kappa B , Receptor beta de Linfotoxina , Mutagênese Sítio-Dirigida , Proteínas Serina-Treonina Quinases/metabolismo , Quinase Induzida por NF-kappaB
3.
Proc Natl Acad Sci U S A ; 97(13): 7470-5, 2000 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-10852966

RESUMO

The X-linked lymphoproliferative syndrome (XLP) is a genetic disorder in which affected males have a morbid or fatal response to Epstein-Barr virus infection. The XLP deficiency has been mapped to a gene encoding a 128-residue protein, SH2D1A, which is comprised principally of a Src homology 2 (SH2) domain. We now report that SH2D1A associates with Dok1, a protein that interacts with Ras-GAP, Csk, and Nck. An SH2D1A SH2 domain mutant that has been identified in XLP does not associate with Dok1, in accord with the hypothesis that this interaction is linked to XLP. The association of SH2D1A with Dok1 also depends on phosphorylation of Dok1 Y(449) in the sequence ALYSQVQK. Further, overexpression of SH2D1A is found to activate NF-kappaB in 293T cells. NF-kappaB activation by SH2D1A does not depend on the wild-type SH2 domain and is inhibited by a dominant-negative IkappaB kinase beta. Thus, SH2D1A can affect multiple intracellular signaling pathways that are potentially important in the normal effective host response to Epstein-Barr virus infection.


Assuntos
Proteínas de Transporte/genética , Proteínas de Ligação a DNA , Peptídeos e Proteínas de Sinalização Intracelular , Transtornos Linfoproliferativos/genética , NF-kappa B/genética , Fosfoproteínas/genética , Proteínas de Ligação a RNA , Proteínas de Transporte/metabolismo , Infecções por Vírus Epstein-Barr/genética , Regulação da Expressão Gênica , Predisposição Genética para Doença , Humanos , Transtornos Linfoproliferativos/metabolismo , Masculino , NF-kappa B/metabolismo , Fosfoproteínas/metabolismo , Proteína Associada à Molécula de Sinalização da Ativação Linfocitária , Síndrome , Células Tumorais Cultivadas , Cromossomo X
4.
Proc Natl Acad Sci U S A ; 97(11): 6055-60, 2000 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-10811897

RESUMO

Epstein-Barr virus (EBV) transforms B lymphocytes into lymphoblastoid cell lines usurping the Notch and tumor necrosis factor receptor pathways to effect transcription including NF-kappaB activation. To determine whether NF-kappaB activity is essential in the growth and survival of EBV-transformed lymphoblastoid cell lines, a nondegradable IkappaBalpha mutant was expressed under tetracycline regulation. Despite continued Bcl-2 and Bcl-x/L expression, NF-kappaB inhibition induced apoptosis as evidenced by poly(ADP-ribose) polymerase cleavage, nuclear condensation and fragmentation, and hypodiploid DNA content. Both caspase 3 and 8 activation and loss of mitochondrial membrane potential were observed in apoptotic cells. However, caspase inhibition failed to block apoptosis. These experiments indicate that NF-kappaB inhibitors may be useful in the therapy of EBV-induced cellular proliferation.


Assuntos
Apoptose/fisiologia , Linfócitos B/citologia , Transformação Celular Viral , Proteínas de Ligação a DNA/fisiologia , Herpesvirus Humano 4/fisiologia , Proteínas I-kappa B , NF-kappa B/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2 , Clorometilcetonas de Aminoácidos/farmacologia , Linfócitos B/virologia , Caspase 3 , Caspase 8 , Caspase 9 , Inibidores de Caspase , Caspases/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Transformada/citologia , Inibidores de Cisteína Proteinase/farmacologia , Proteínas de Ligação a DNA/genética , Desenho de Fármacos , Citometria de Fluxo , Regulação da Expressão Gênica , Humanos , Membranas Intracelulares/fisiologia , Transtornos Linfoproliferativos/tratamento farmacológico , Transtornos Linfoproliferativos/virologia , Potenciais da Membrana , Microscopia Confocal , Antígenos de Histocompatibilidade Menor , Mitocôndrias/fisiologia , Inibidor de NF-kappaB alfa , NF-kappa B/antagonistas & inibidores , Biossíntese de Proteínas , Proteínas/genética , Proteínas Recombinantes de Fusão/fisiologia , Transfecção
5.
Oncogene ; 18(49): 6959-64, 1999 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-10602470

RESUMO

Epstein-Barr virus (EBV) transforms resting primary human B lymphocytes into indefinitely proliferating lymphoblastoid cell lines in vitro and is associated with several human malignancies in vivo. Recombinant EBV genetic analyses combined with in vitro B lymphocyte transformation assays demonstrate that latent infection membrane protein 1 (LMP1) is essential for EBV-mediated lymphocyte transformation. LMP1 has no intrinsic enzymatic activity but instead aggregates cellular proteins of the tumor necrosis factor receptor signaling pathway to activate transcription factor NF-kappaB. Mutants rendering LMP1 defective in these protein interactions are impaired in their abilities to activate NF-kappaB in reporter gene assays. Concordantly, EBV recombinants with LMP1 mutations that are compromised for NF-kappaB activation are impaired for growth transformation. Thus, EBV-mediated growth transformation is genetically and biochemically linked to LMP1-mediated activation of NF-kappaB.


Assuntos
Transformação Celular Viral , Herpesvirus Humano 4/genética , Proteínas I-kappa B , NF-kappa B/fisiologia , Proteínas da Matriz Viral/fisiologia , Apoptose , Linfócitos B/patologia , Proteínas de Ligação a DNA/fisiologia , Humanos , Ativação Linfocitária , Inibidor de NF-kappaB alfa , Proteínas/fisiologia , Receptores do Fator de Necrose Tumoral/fisiologia , Fator 1 Associado a Receptor de TNF
6.
J Virol ; 73(12): 9908-16, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10559303

RESUMO

Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1) is essential for EBV-mediated transformation of primary B lymphocytes. LMP1 spontaneously aggregates in the plasma membrane and enables two transformation effector sites (TES1 and TES2) within the 200-amino-acid cytoplasmic carboxyl terminus to constitutively engage the tumor necrosis factor receptor (TNFR)-associated factors TRAF1, TRAF2, TRAF3, and TRAF5 and the TNFR-associated death domain proteins TRADD and RIP, thereby activating NF-kappaB and c-Jun N-terminal kinase (JNK). To investigate the importance of the 60% of the LMP1 carboxyl terminus that lies between the TES1-TRAF and TES2-TRADD and -RIP binding sites, an EBV recombinant was made that contains a specific deletion of LMP1 codons 232 to 351. Surprisingly, the deletion mutant was similar to wild-type (wt) LMP1 EBV recombinants in its efficiency in transforming primary B lymphocytes into lymphoblastoid cell lines (LCLs). Mutant and wt EBV-transformed LCLs were similarly efficient in long-term outgrowth and in regrowth after endpoint dilution. Mutant and wt LMP1 proteins were also similar in their constitutive association with TRAF1, TRAF2, TRAF3, TRADD, and RIP. Mutant and wt EBV-transformed LCLs were similar in steady-state levels of Bcl2, JNK, and activated JNK proteins. The wt phenotype of recombinants with LMP1 codons 232 to 351 deleted further demarcates TES1 and TES2, underscores their central importance in B-lymphocyte growth transformation, and provides a new perspective on LMP1 sequence variation between TES1 and TES2.


Assuntos
Linfócitos B/virologia , Transformação Celular Viral , Proteínas de Ligação a DNA/metabolismo , Herpesvirus Humano 4/genética , Proteínas de Homeodomínio , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral , Proteínas da Matriz Viral/genética , Linfócitos B/patologia , Sítios de Ligação , Divisão Celular , Células Cultivadas , Códon , Proteínas do Citoesqueleto , Antígenos Nucleares do Vírus Epstein-Barr , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Proteína Quinase 9 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Oligopeptídeos , Peptídeos , Fosforilação , Proteínas Quinases/metabolismo , Proteínas/metabolismo , Proteínas de Ligação a RNA , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Deleção de Sequência , Proteína de Domínio de Morte Associada a Receptor de TNF , Fator 1 Associado a Receptor de TNF , Fator 2 Associado a Receptor de TNF , Fator 3 Associado a Receptor de TNF , Fatores de Transcrição , Proteínas da Matriz Viral/metabolismo
7.
Proc Natl Acad Sci U S A ; 96(22): 12536-41, 1999 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-10535957

RESUMO

Human cytomegalovirus (CMV), a herpesvirus that causes congenital disease and opportunistic infections in immunocompromised individuals, encodes functions that facilitate efficient viral propagation by altering host cell behavior. Here we show that CMV blocks apoptosis mediated by death receptors and encodes a mitochondria-localized inhibitor of apoptosis, denoted vMIA, capable of suppressing apoptosis induced by diverse stimuli. vMIA, a product of the viral UL37 gene, inhibits Fas-mediated apoptosis at a point downstream of caspase-8 activation and Bid cleavage but upstream of cytochrome c release, while residing in mitochondria and associating with adenine nucleotide translocator. These functional properties resemble those ascribed to Bcl-2; however, the absence of sequence similarity to Bcl-2 or any other known cell death suppressors suggests that vMIA defines a previously undescribed class of anti-apoptotic proteins.


Assuntos
Apoptose/genética , Citomegalovirus/genética , Proteínas Estruturais Virais/genética , Linhagem Celular , Citomegalovirus/fisiologia , Células HeLa , Humanos , Replicação Viral/genética
8.
Mol Cell Biol ; 19(8): 5759-67, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10409763

RESUMO

A site in the Epstein-Barr virus (EBV) transforming protein LMP1 that constitutively associates with the tumor necrosis factor receptor 1 (TNFR1)-associated death domain protein TRADD to mediate NF-kappaB and c-Jun N-terminal kinase activation is critical for long-term lymphoblastoid cell proliferation. We now find that LMP1 signaling through TRADD differs from TNFR1 signaling through TRADD. LMP1 needs only 11 amino acids to activate NF-kappaB or synergize with TRADD in NF-kappaB activation, while TNFR1 requires approximately 70 residues. Further, LMP1 does not require TRADD residues 294 to 312 for NF-kappaB activation, while TNFR1 requires TRADD residues 296 to 302. LMP1 is partially blocked for NF-kappaB activation by a TRADD mutant consisting of residues 122 to 293. Unlike TNFR1, LMP1 can interact directly with receptor-interacting protein (RIP) and stably associates with RIP in EBV-transformed lymphoblastoid cell lines. Surprisingly, LMP1 does not require RIP for NF-kappaB activation. Despite constitutive association with TRADD or RIP, LMP1 does not induce apoptosis in EBV-negative Burkitt lymphoma or human embryonic kidney 293 cells. These results add a different perspective to the molecular interactions through which LMP1, TRADD, and RIP participate in B-lymphocyte activation and growth.


Assuntos
Apoptose , Transformação Celular Viral , Regulação da Expressão Gênica , Herpesvirus Humano 4/fisiologia , Proteínas Quinases Ativadas por Mitógeno , NF-kappa B/metabolismo , Proteínas/metabolismo , Proteínas da Matriz Viral/fisiologia , Antígenos CD/fisiologia , Linfócitos B/metabolismo , Linfócitos B/patologia , Linfoma de Burkitt/patologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Linhagem Celular Transformada , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Células Jurkat/metabolismo , Células Jurkat/patologia , Rim , Substâncias Macromoleculares , Modelos Moleculares , Proteína Serina-Treonina Quinases de Interação com Receptores , Receptores do Fator de Necrose Tumoral/fisiologia , Receptores Tipo I de Fatores de Necrose Tumoral , Fator 1 Associado a Receptor de TNF , Transcrição Gênica , Células Tumorais Cultivadas
9.
J Virol ; 72(10): 7900-8, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9733827

RESUMO

In this study, we investigated the induction of cellular gene expression by the Epstein-Barr Virus (EBV) latent membrane protein 1 (LMP1). Previously, LMP1 was shown to induce the expression of ICAM-1, LFA-3, CD40, and EBI3 in EBV-negative Burkitt lymphoma (BL) cells and of the epidermal growth factor receptor (EGF-R) in epithelial cells. We now show that LMP1 expression also increased Fas and tumor necrosis factor receptor-associated factor 1 (TRAF1) in BL cells. LMP1 mediates NF-kappaB activation via two independent domains located in its C-terminal cytoplasmic tail, a TRAF-interacting site that associates with TRAF1, -2, -3, and -5 through a PXQXT/S core motif and a TRADD-interacting site. In EBV-transformed B cells or transiently transfected BL cells, significant amounts of TRAF1, -2, -3, and -5 are associated with LMP1. In epithelial cells, very little TRAF1 is expressed, and only TRAF2, -3, and -5, are significantly complexed with LMP1. The importance of TRAF binding to the PXQXT/S motif in LMP1-mediated gene induction was studied by using an LMP1 mutant that contains alanine point mutations in this motif and fails to associate with TRAFs. This mutant, LMP1(P204A/Q206A), induced 60% of wild-type LMP1 NF-kappaB activation and had approximately 60% of wild-type LMP1 effect on Fas, ICAM-1, CD40, and LFA-3 induction. In contrast, LMP1(P204A/Q206A) was substantially more impaired in TRAF1, EBI3, and EGF-R induction. Thus, TRAF binding to the PXQXT/S motif has a nonessential role in up-regulating Fas, ICAM-1, CD40, and LFA-3 expression and a critical role in up-regulating TRAF1, EBI3, and EGF-R expression. Further, D1 LMP1, an LMP1 mutant that does not aggregate failed to induce TRAF1, EBI3, Fas, ICAM-1, CD40, and LFA-3 expression confirming the essential role for aggregation in LMP1 signaling. Overexpression of a dominant form of IkappaBalpha blocked LMP1-mediated TRAF1, EBI3, Fas, ICAM-1, CD40, and LFA-3 up-regulation, indicating that NF-kappaB is an important component of LMP1-mediated gene induction from both the TRAF- and TRADD-interacting sites.


Assuntos
Regulação Viral da Expressão Gênica , Herpesvirus Humano 4/metabolismo , NF-kappa B/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Proteínas da Matriz Viral/metabolismo , Sítios de Ligação , Linhagem Celular Transformada , Receptores ErbB/genética , Humanos , Molécula 1 de Adesão Intercelular/genética , Ligação Proteica , Ativação Transcricional , Regulação para Cima
10.
Biochemistry ; 36(23): 7169-75, 1997 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-9188717

RESUMO

The transmembrane protein tyrosine phosphatase CD45 functions to activate Src-family member kinase activity in T lymphocytes. The inability to activate p56(lck) in CD45-deficient cells results in a higher threshold of signaling through the T cell receptor. The lymphoid-specific CD45-associated protein, CD45AP, interacts with CD45 through transmembrane interactions. Cells lines and mice deficient in CD45 express CD45AP mRNA, yet the protein is poorly expressed, indicating that CD45 is required for efficient expression of CD45AP. Pulse-chase analysis indicates that CD45 associates with CD45AP within minutes of biosynthesis. Cell surface labeling and coimmunoprecipitation demonstrate that CD45AP associates with surface-expressed CD45. Therefore, CD45AP is localized to the plasma membrane. To further characterize this interaction, chimeric proteins containing mutations in CD45 transmembrane regions were expressed, and their ability to associate with CD45AP was determined. Alanine-scan mutations of the CD45 transmembrane region demonstrate that no single amino acid is essential for the interaction with CD45AP. However, the expression of chimeric transmembrane regions indicates that a minimum of three and a maximum of eight amino acids in this region are sufficient to allow interaction with CD45AP.


Assuntos
Aminoácidos/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Proteínas de Membrana , Fosfoproteínas/metabolismo , Sequência de Aminoácidos , Aminoácidos/química , Animais , Linhagem Celular , Retículo Endoplasmático/metabolismo , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Antígenos Comuns de Leucócito/química , Camundongos , Dados de Sequência Molecular , Fosfoproteínas/química , RNA Mensageiro/metabolismo
11.
Curr Biol ; 7(6): 408-17, 1997 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-9197241

RESUMO

BACKGROUND: Adhesion of leukocytes to the extracellular matrix and to other cells is mediated by members of the integrin family of adhesion molecules. Src family kinases are activated upon integrin-mediated adhesion. In lymphocytes, CD45 is a leukocyte-specific transmembrane protein tyrosine phosphatase that activates Src family kinases associated with B-cell and T-cell antigen receptor signaling by constitutive dephosphorylation of the inhibitory carboxy-terminal tyrosine phosphorylation site. Here, we show that CD45 is also important in downregulating the kinase activity of Src family members during integrin-mediated adhesion in macrophages. RESULTS: We found that CD45 colocalized with beta2 integrin and the Src family kinase p53/56(lyn) to adhesion sites in bone marrow-derived macrophages. Macrophages from CD45(-/-) mice were unable to maintain integrin-mediated adhesion. In adherent macrophages, absence of CD45 led to the hyperphosphorylation and hyperactivation of p56/59(hck) and p53/56(lyn), but not of p58(c-fgr). CD45 directly inactivated p59(hck) but not p56(lck) in transient transfection assays. Furthermore, coexpression of CD45 with p59(hck) or p56(lyn) containing a tyrosine to phenylalanine mutation at the carboxy-terminal negative regulatory site resulted in decreased tyrosine phosphorylation of the Src family member kinases due to dephosphorylation of the potentiating tyrosine phosphorylation site within the kinase domain. CONCLUSIONS: Using primary bone marrow macrophages, these studies demonstrate that CD45 regulates Src family kinases and is required to maintain macrophage adhesion. CD45 decreases Src family kinase activity by dephosphorylating the tyrosine residue located within the kinase domain.


Assuntos
Antígenos CD18/metabolismo , Integrinas/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Macrófagos/fisiologia , Quinases da Família src/metabolismo , Animais , Adesão Celular , Regulação para Baixo , Regulação da Expressão Gênica , Camundongos , Camundongos Mutantes , Fosforilação , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-hck
12.
J Biol Chem ; 270(47): 28103-7, 1995 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-7499298

RESUMO

CD45 is a transmembrane protein-tyrosine phosphatase required for antigen receptor signaling in lymphocytes. CD45 activates the Src family protein-tyrosine kinases, p56lck and p59fyn, by dephosphorylating a negative regulatory tyrosine in the carboxyl terminus. Immunoprecipitation of CD45 precipitates p56lck and CD45AP. Although the function of CD45AP is unknown, it has been proposed to be an adapter between p56lck and CD45. To assess the ability of CD45AP to function as an adapter, we determined the regions required for the interaction with CD45 by expressing chimeric proteins in HeLa cells. CD45AP has a region similar to a potential protein-protein interaction domain, the WW domain. Surprisingly, this domain was not necessary for the association with CD45. Rather, a 40-amino acid sequence encompassing the putative transmembrane domain of CD45AP was sufficient to mediate binding to CD45. Similarly, a 39-amino acid sequence encompassing the CD45 transmembrane region was sufficient to direct the interaction with CD45AP. Expression of p56lck with CD45AP resulted in an interaction that could only be detected by in vitro kinase reaction, suggesting that the association of p56lck and CD45AP is weak. These data support a model in which CD45AP links CD45 with other proteins but not necessarily p56lck.


Assuntos
Antígenos Comuns de Leucócito/química , Antígenos Comuns de Leucócito/metabolismo , Proteínas de Membrana , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Sequência de Aminoácidos , Animais , Western Blotting , Membrana Celular/metabolismo , Clonagem Molecular , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Antígenos Comuns de Leucócito/isolamento & purificação , Proteína Tirosina Quinase p56(lck) Linfócito-Específica , Camundongos , Dados de Sequência Molecular , Fosfoproteínas/isolamento & purificação , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-fyn , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Quinases da Família src/metabolismo
13.
Int Immunol ; 6(2): 169-78, 1994 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-7908828

RESUMO

CD45, the leukocyte-common antigen, is a transmembrane protein tyrosine phosphatase uniquely expressed by cells of hematopoietic origin. We have developed CD4+ and CD8+ T cell clones that are deficient in the expression of CD45 and have previously shown that these cells fail to proliferate in response to antigen or cross-linked CD3. These studies have now been extended to show that stimulation with anti-Thy-1, a mitogenic signal for the CD4+CD45+ and CD8+CD45+ T cells, fails to induce proliferation in the CD45- T cells. Examination of the CD8+CD45- T cells correlates anti-Thy-1 unresponsiveness with a failure to increase in tyrosine phosphorylation. Furthermore, stimulation of CD8+CD45+ T cells with anti-Thy-1 results in an increase in p56lck activity but not in CD8+CD45- T cells. In contrast to the results with anti-Thy-1, both the CD4+CD45- and CD8+CD45- T cells respond to treatment with lectin mitogens, concanavalin A or phytohemagglutinin. Lectin-induced proliferation was inhibited by the addition of cyclosporin A. Treatment of CD45- T cells with PMA and ionomycin also results in proliferation indicating that activation of protein kinase C in conjunction with an increase in intracellular calcium rescues the defect caused by CD45 deficiency. The data suggest that CD45 is required for the activation of tyrosine kinase activity immediate or prior to transmembrane signaling.


Assuntos
Antígenos de Superfície/imunologia , Lectinas/farmacologia , Antígenos Comuns de Leucócito/fisiologia , Glicoproteínas de Membrana/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Anticorpos Monoclonais , Linhagem Celular , Células Clonais , Feminino , Immunoblotting , Ionomicina/farmacologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos CBA , Camundongos Endogâmicos DBA , Proteínas Tirosina Quinases/análise , Acetato de Tetradecanoilforbol/farmacologia , Antígenos Thy-1
15.
Proc Natl Acad Sci U S A ; 90(4): 1402-6, 1993 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-8433999

RESUMO

Stimulation of tyrosine phosphorylation is an early and important event in antigen-induced T-cell activation. T-cell clones deficient in expression of CD45, a transmembrane protein-tyrosine-phosphatase (protein-tyrosine-phosphate phosphohydrolase, EC 3.1.3.48), are impaired in their ability to respond to either antigen or T-cell receptor cross-linking. Analysis of the CD45-deficient CD8+ T-cell clone L3M-93 demonstrates that the Src family members p56lck and p59fyn show increased immunoreactivity with anti-phosphotyrosine antibody and exhibit decreased kinase activity. The site of increased tyrosine phosphorylation in Src family members was identified by comparison of cyanogen bromide peptide maps. Phosphorylation of the C-terminal phosphopeptide, containing the negative regulatory site of tyrosine phosphorylation, from the CD45-deficient cells was increased 8-fold for p56lck and 2-fold for p59fyn. These data suggest that CD45 dephosphorylates the negative regulatory site of multiple Src family members in the cytotoxic T-lymphocyte clone L3 and show a correlation between the ability to respond efficiently to antigen and the dephosphorylation of Src family members by CD45.


Assuntos
Genes src , Antígenos Comuns de Leucócito/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores de Antígenos de Linfócitos T/fisiologia , Transdução de Sinais , Linfócitos T/imunologia , Animais , Membrana Celular/fisiologia , Células Clonais , Brometo de Cianogênio , Immunoblotting , Ativação Linfocitária , Proteína Tirosina Quinase p56(lck) Linfócito-Específica , Fragmentos de Peptídeos/isolamento & purificação , Mapeamento de Peptídeos , Fosforilação , Proteínas Proto-Oncogênicas c-fyn
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA