Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Nat Immunol ; 9(12): 1388-98, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18978794

RESUMO

The transcription factors Foxo1, Foxo3 and Foxo4 modulate cell fate 'decisions' in diverse systems. Here we show that Foxo1-dependent gene expression was critical at many stages of B cell differentiation. Early deletion of Foxo1 caused a substantial block at the pro-B cell stage due to a failure to express interleukin 7 receptor-alpha. Foxo1 inactivation in late pro-B cells resulted in an arrest at the pre-B cell stage due to lower expression of the recombination-activating genes Rag1 and Rag2. Deletion of Foxo1 in peripheral B cells led to fewer lymph node B cells due to lower expression of L-selectin and failed class-switch recombination due to impaired upregulation of the gene encoding activation-induced cytidine deaminase. Thus, Foxo1 regulates a transcriptional program that is essential for early B cell development and peripheral B cell function.


Assuntos
Linfócitos B/citologia , Diferenciação Celular/imunologia , Fatores de Transcrição Forkhead/imunologia , Animais , Linfócitos B/imunologia , Southern Blotting , Proteínas de Ligação a DNA/imunologia , Proteínas de Ligação a DNA/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Expressão Gênica/imunologia , Rearranjo Gênico do Linfócito B/genética , Proteínas de Homeodomínio/imunologia , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Mutantes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/imunologia , Células-Tronco/metabolismo , Transcrição Gênica/imunologia
2.
Res Sq ; 2023 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-37461531

RESUMO

Background: Rheumatoid arthritis is a chronic systemic autoimmune disease that involves transformation of the lining of synovial joints into an invasive and destructive tissue. Synovial fibroblasts become transformed, invading and destroying bone and cartilage of the affected joint(s). Due to the significant role these cells play in the progression of the disease process, developing a therapeutic strategy to target and inhibit their invasive destructive nature could help patients who are affiicted with this debilitating disease. Gingival-derived mesenchymal stem cells are known to possess immunomodulatory properties and have been studied extensively as potential cell-based therapeutics for several autoimmune disorders. Methods: A chimeric human/mouse model of synovitis was created by surgically implanting SCID mice with a piece of human articular cartilage surrounded by RASF. Mice were injected once with either GMSC or GMSCExo at 5-7 days post-implantation. Histology and IHC were used to assess RASF invasion of the cartilage. Flow cytometry was used to understand the homing ability of GMSC in vivo and the incidence of apoptosis of RASF in vitro. Results: We demonstrate that both GMSC and GMSCExo are potent inhibitors of the deleterious effects of RASF. Both treatments were effective in inhibiting the invasive destructive properties of RASF as well as the potential of these cells to migrate to secondary locations and attack the cartilage. GMSC home to the site of the implant and induce programmed cell death of the RASF. Conclusions: Our results indicate that both GMSC and GMSCExo can block the pathological effects of RASF in this chimeric model of RA. A single dose of either GMSC or GMSCExo can inhibit the deleterious effects of RASF. These treatments can also block the invasive migration of the RASF, suggesting that they can inhibit the spread of RA to other joints. Because the gingival tissue is harvested with little difficulty, relatively small amounts of tissue are required to expand the cells, the simple in vitro expansion process, and the increasing technological advances in the production of therapeutic exosomes, we believe that GMSCExo are excellent candidates as a potential therapeutic for RA.

3.
Arthritis Res Ther ; 25(1): 211, 2023 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-37885040

RESUMO

BACKGROUND: Rheumatoid arthritis is a chronic systemic autoimmune disease that involves transformation of the lining of synovial joints into an invasive and destructive tissue. Synovial fibroblasts become transformed, invading and destroying the bone and cartilage of the affected joint(s). Due to the significant role these cells play in the progression of the disease process, developing a therapeutic strategy to target and inhibit their invasive destructive nature could help patients who are afflicted with this debilitating disease. Gingival-derived mesenchymal stem cells are known to possess immunomodulatory properties and have been studied extensively as potential cell-based therapeutics for several autoimmune disorders. METHODS: A chimeric human/mouse model of synovitis was created by surgically implanting SCID mice with a piece of human articular cartilage surrounded by RASF. Mice were injected once with either GMSC or GMSCExo at 5-7 days post-implantation. Histology and IHC were used to assess RASF invasion of the cartilage. Flow cytometry was used to understand the homing ability of GMSC in vivo and the incidence of apoptosis of RASF in vitro. RESULTS: We demonstrate that both GMSC and GMSCExo are potent inhibitors of the deleterious effects of RASF. Both treatments were effective in inhibiting the invasive destructive properties of RASF as well as the potential for these cells to migrate to secondary locations and attack the cartilage. GMSC home to the site of the implant and induce programmed cell death of the RASF. CONCLUSIONS: Our results indicate that both GMSC and GMSCExo can block the pathological effects of RASF in this chimeric model of RA. A single dose of either GMSC or GMSCExo can inhibit the deleterious effects of RASF. These treatments can also block the invasive migration of the RASF, suggesting that they can inhibit the spread of RA to other joints. Because the gingival tissue is harvested with little difficulty, relatively small amounts of tissue are required to expand the cells, the simple in vitro expansion process, and the increasing technological advances in the production of therapeutic exosomes, we believe that GMSCExo are excellent candidates as a potential therapeutic for RA.


Assuntos
Artrite Reumatoide , Exossomos , Células-Tronco Mesenquimais , Humanos , Animais , Camundongos , Membrana Sinovial/metabolismo , Exossomos/metabolismo , Células Cultivadas , Camundongos SCID , Artrite Reumatoide/metabolismo , Células-Tronco Mesenquimais/metabolismo , Fibroblastos/metabolismo
4.
Front Immunol ; 14: 1090177, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38939646

RESUMO

Introduction: Distinct, disease-associated intracellular miRNA (miR) expression profiles have been observed in peripheral blood mononuclear cells (PBMCs) of systemic lupus erythematous (SLE) patients. Additionally, we have identified novel estrogenic responses in PBMCs from SLE patients and demonstrated that estrogen upregulates toll-like receptor (TLR)7 and TLR8 expression. TLR7 and TLR8 bind viral-derived single-stranded RNA to stimulate innate inflammatory responses, but recent studies have shown that miR-21, mir-29a, and miR-29b can also bind and activate these receptors when packaged and secreted in extracellular vesicles (EVs). The objective of this study was to evaluate the association of EV-encapsulated small RNA species in SLE and examine the therapeutic approach of miR inhibition in humanized mice. Methods: Plasma-derived EVs were isolated from SLE patients and quantified. RNA was then isolated and bulk RNA-sequencing reads were analyzed. Also, PBMCs from active SLE patients were injected into immunodeficient mice to produce chimeras. Prior to transfer, the PBMCs were incubated with liposomal EVs containing locked nucleic acid (LNA) antagonists to miR-21, mir-29a, and miR-29b. After three weeks, blood was collected for both immunophenotyping and cytokine analysis; tissue was harvested for histopathological examination. Results: EVs were significantly increased in the plasma of SLE patients and differentially expressed EV-derived small RNA profiles were detected compared to healthy controls, including miR-21, mir-29a, and miR-29b. LNA antagonists significantly reduced proinflammatory cytokines and histopathological infiltrates in the small intestine, liver, and kidney, as demonstrated by H&E-stained tissue sections and immunohistochemistry measuring human CD3. Discussion: These data demonstrate distinct EV-derived small RNA signatures representing SLE-associated biomarkers. Moreover, targeting upregulated EV-encapsulated miR signaling by antagonizing miRs that may bind to TLR7 and TLR8 reveals a novel therapeutic opportunity to suppress autoimmune-mediated inflammation and pathogenesis in SLE.


Assuntos
Modelos Animais de Doenças , Vesículas Extracelulares , Leucócitos Mononucleares , Lúpus Eritematoso Sistêmico , MicroRNAs , Receptor 7 Toll-Like , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/metabolismo , Humanos , Animais , MicroRNAs/genética , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/imunologia , Camundongos , Feminino , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/imunologia , Receptor 7 Toll-Like/metabolismo , Receptor 7 Toll-Like/genética , Inflamação/imunologia , Receptor 8 Toll-Like/metabolismo , Receptor 8 Toll-Like/genética , Adulto , Masculino , Pessoa de Meia-Idade , Camundongos SCID
5.
Nat Cell Biol ; 6(9): 831-9, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15322554

RESUMO

The tyrosine phosphatase PTP-MEG2 is targeted by its amino-terminal Sec14p homology domain to the membrane of secretory vesicles. There it regulates vesicle size by promoting homotypic vesicle fusion by a mechanism that requires its catalytic activity. Here, we identify N-ethylmaleimide-sensitive factor (NSF), a key regulator of vesicle fusion, as a substrate for PTP-MEG2. PTP-MEG2 reduced the phosphotyrosine content of NSF and co-localized with NSF and syntaxin 6 in intact cells. Furthermore, endogenous PTP-MEG2 co-immunoprecipitated with endogenous NSF. Phosphorylation of NSF at Tyr 83, as well as an acidic substitution at the same site, increased its ATPase activity and prevented alphaSNAP binding. Conversely, expression of a Y83F mutant of NSF caused spontaneous fusion events. Our results suggest that the molecular mechanism by which PTP-MEG2 promotes secretory vesicle fusion involves the local release of NSF from a tyrosine-phosphorylated, inactive state. This represents a novel mechanism for localized regulation of NSF and the first demonstrated role for a protein tyrosine phosphatase in the regulated secretory pathway.


Assuntos
Fusão de Membrana , Proteínas Tirosina Fosfatases/fisiologia , Vesículas Secretórias/fisiologia , Humanos , Membranas Intracelulares/fisiologia , Células Jurkat , Proteínas de Membrana/metabolismo , Microscopia Eletrônica , Proteínas Sensíveis a N-Etilmaleimida , Fosforilação , Fosfotirosina , Ligação Proteica , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Tirosina Fosfatases não Receptoras , Proteínas Qa-SNARE , Vesículas Secretórias/enzimologia , Proteínas de Transporte Vesicular/metabolismo
6.
Mol Cell Biol ; 26(5): 1806-16, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16479000

RESUMO

Protein kinase C theta (PKC theta) is unique among PKC isozymes in its translocation to the center of the immune synapse in T cells and its unique downstream signaling. Here we show that the hematopoietic protein tyrosine phosphatase (HePTP) also accumulates in the immune synapse in a PKC theta-dependent manner upon antigen recognition by T cells and is phosphorylated by PKC theta at Ser-225, which is required for lipid raft translocation. Immune synapse translocation was completely absent in antigen-specific T cells from PKC theta-/- mice. In intact T cells, HePTP-S225A enhanced T-cell receptor (TCR)-induced NFAT/AP-1 transactivation, while the acidic substitution mutant was as efficient as wild-type HePTP. We conclude that HePTP is phosphorylated in the immune synapse by PKC theta and thereby targeted to lipid rafts to temper TCR signaling. This represents a novel mechanism for the active immune synapse recruitment and activation of a phosphatase in TCR signaling.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Isoenzimas/metabolismo , Microdomínios da Membrana/metabolismo , Proteína Quinase C/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Linfócitos T/metabolismo , Animais , Domínio Catalítico , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Isoenzimas/genética , Células Jurkat/imunologia , Células Jurkat/metabolismo , Camundongos , Fosforilação , Proteína Quinase C/genética , Proteína Quinase C-theta , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases não Receptoras , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Serina/metabolismo , Transdução de Sinais , Linfócitos T/imunologia , Transgenes
7.
Toxicol Lett ; 160(2): 121-6, 2006 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-16125885

RESUMO

Carbon nanotubes are a man-made form of carbon that did not exist in our environment until very recently. Due to their unique chemical, physical, optical, and magnetic properties, carbon nanotubes have found many uses in industrial products and in the field of nanotechnology, including in nanomedicine. However, very little is yet known about the toxicity of carbon nanotubes. Here, we compare the toxicity of pristine and oxidized multi-walled carbon nanotubes on human T cells and find that the latter are more toxic and induce massive loss of cell viability through programmed cell death at doses of 400 microg/ml, which corresponds to approximately 10 million carbon nanotubes per cell. Pristine, hydrophobic, carbon nanotubes were less toxic and a 10-fold lower concentration of either carbon nanotube type were not nearly as toxic. Our results suggest that carbon nanotubes indeed can be very toxic at sufficiently high concentrations and that careful toxicity studies need to be undertaken particularly in conjunction with nanomedical applications of carbon nanotubes.


Assuntos
Apoptose , Nanotubos de Carbono/toxicidade , Linfócitos T/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Humanos , Células Jurkat , Nanotubos de Carbono/química , Nanotubos de Carbono/ultraestrutura , Oxirredução , Receptores de Antígenos de Linfócitos T/química , Receptores de Antígenos de Linfócitos T/metabolismo
8.
Mol Immunol ; 41(6-7): 687-700, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15220004

RESUMO

The molecular mechanisms of signal transduction have been the focus of intense research during the last decade. In T cells, much of the work has centered on protein tyrosine kinase-mediated signaling from the TCR and cytokine receptors, while the study of protein tyrosine phosphatases has lagged behind. Nevertheless, it has now become clear that many protein tyrosine phosphatases play equally important roles in T cell physiology and that no kinase-regulated system would work without the counterbalancing participation of phosphatases. In fact, we have learned that many processes are regulated primarily on the phosphatase side. This minireview summarizes the current state-of-the art in our understanding of the regulation and biology of protein tyrosine phosphatases in T lymphocyte physiology.


Assuntos
Proteínas Tirosina Fosfatases/fisiologia , Linfócitos T/enzimologia , Fosfatase 3 de Especificidade Dupla , Retículo Endoplasmático/fisiologia , Humanos , Antígenos Comuns de Leucócito/imunologia , Antígenos Comuns de Leucócito/fisiologia , Proteínas Quinases Ativadas por Mitógeno/imunologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Fosforilação , Proteínas Tirosina Fosfatases/imunologia , Linfócitos T/imunologia , Quinases da Família src/imunologia , Quinases da Família src/fisiologia
9.
Methods Mol Biol ; 699: 97-118, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21116981

RESUMO

BD FACS™ CAP (CAP = combinatorial antibody profile) is a screening tool for rapid characterization of human cell surface protein expression profiles using semi-automated high-throughput flow cytometry. The current configuration consists of 229 directly conjugated antibodies arrayed in a 96-well plate as three-color cocktails, which enables the characterization of each of the 229 individual surface markers. Each individual cell type of interest is analyzed on the 96-well screening plates and the data are acquired on a flow cytometer equipped with a high-throughput sampler. The expression level of each marker for each cell type is then calculated using semiautomated custom flow cytometry software. The process of characterizing these surface markers in a highly efficient manner using BD FACS™ CAP is enabled by automated liquid handling for staining, automated flow cytometry for data acquisition, and standardized algorithms for automated data analysis.


Assuntos
Anticorpos/imunologia , Citometria de Fluxo , Perfilação da Expressão Gênica/métodos , Antígenos de Superfície/genética , Antígenos de Superfície/imunologia , Células Cultivadas , Análise por Conglomerados , Processamento Eletrônico de Dados , Corantes Fluorescentes/metabolismo , Perfilação da Expressão Gênica/instrumentação , Ensaios de Triagem em Larga Escala , Humanos , Controle de Qualidade , Sensibilidade e Especificidade , Software , Coloração e Rotulagem
10.
J Biol Chem ; 280(10): 9400-8, 2005 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-15615724

RESUMO

To avoid detection and targeting by the immune system, the plague-causing bacterium Yersinia pestis uses a type III secretion system to deliver a set of inhibitory proteins into the cytoplasm of immune cells. One of these proteins is an exceptionally active tyrosine phosphatase termed YopH, which paralyzes lymphocytes and macrophages by dephosphorylating critical tyrosine kinases and signal transduction molecules. Because Y. pestis strains lacking YopH are avirulent, we set out to develop small molecule inhibitors for YopH. We used a novel and cost-effective approach, in which leads from a chemical library screening were analyzed and computationally docked into the crystal structure of YopH. This resulted in the identification of a series of novel YopH inhibitors with nanomolar Ki values, as well as the structural basis for inhibition. Our inhibitors lack the polar phosphate-mimicking moiety of rationally designed tyrosine phosphatase inhibitors, and they readily entered live cells and rescued them from YopH-induced tyrosine dephosphorylation, signaling paralysis, and cell death. These inhibitors may become useful for treating the lethal infection by Y. pestis.


Assuntos
Proteínas da Membrana Bacteriana Externa/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Salicilatos/farmacologia , Yersinia pestis/enzimologia , Proteínas de Bactérias/antagonistas & inibidores , Calorimetria , Desenho de Fármacos , Cinética , Plasmídeos , Probabilidade , Proteínas Recombinantes/antagonistas & inibidores
11.
J Biol Chem ; 280(11): 10388-94, 2005 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-15632192

RESUMO

To evade the immune system, the etiologic agent of plague, Yersinia pestis, injects an exceptionally active tyrosine phosphatase called YopH into host cells using a type III secretion system. We recently reported that YopH acutely inhibits T cell antigen receptor signaling by dephosphorylating the Lck tyrosine kinase. Here, we show that prolonged presence of YopH in primary T cells or Jurkat T leukemia cells causes apoptosis, detected by annexin V binding, mitochondrial breakdown, caspase activation, and internucleosomal fragmentation. YopH also causes cell death when expressed in HeLa cells, and this cell death was inhibited by YopH-specific small molecule inhibitors. Cell death induced by YopH was also prevented by caspase inhibition or co-expression of Bcl-xL. We conclude that YopH not only paralyzes T cells acutely, but also ensures that the cells will not recover to induce a protective immune response but instead undergo mitochondrially regulated programmed cell death.


Assuntos
Apoptose , Proteínas da Membrana Bacteriana Externa/fisiologia , Mitocôndrias/patologia , Proteínas Tirosina Fosfatases/fisiologia , Linfócitos T/patologia , Yersinia pestis/enzimologia , Anexina A5/química , Anexina A5/farmacologia , Proteínas da Membrana Bacteriana Externa/metabolismo , Caspases/metabolismo , Morte Celular , Separação Celular , Fragmentação do DNA , Ativação Enzimática , Citometria de Fluxo , Células HeLa , Humanos , Células Jurkat , Mitocôndrias/metabolismo , Mutagênese Sítio-Dirigida , Nucleossomos/metabolismo , Fosforilação , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais , Linfócitos T/citologia , Transfecção , Proteína bcl-X
12.
J Neurosci Res ; 70(5): 665-70, 2002 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-12424734

RESUMO

4-hydroxynoneal (HNE), an end product of lipid peroxidation, induces apoptosis in many cell types, including neural cells. HNE toxicity is often accompanied by activation of the c-Jun N-terminal kinase/stress-activated protein kinase (JNK/SAPK) pathway. Here we have evaluated the hypothesis that the primary JNK associated with neurons, JNK3, contributes to HNE-induced neuronal apoptosis. First, we demonstrate that HNE induces caspase-dependent apoptosis in sympathetic neurons. Second, we show that HNE-induced c-Jun phosphorylation and c-jun induction are attenuated in JNK3-deficient neurons. Third, we show that HNE neurotoxicity is significantly inhibited by JNK3 deficiency. In summary, these results indicate that JNK3 plays a critical role in HNE-induced c-Jun activation and apoptosis in sympathetic neurons.


Assuntos
Aldeídos/toxicidade , Inibidores de Cisteína Proteinase/toxicidade , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neurônios/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-jun/genética , Gânglio Cervical Superior/citologia , Animais , Apoptose/fisiologia , Células Cultivadas , Imunofluorescência , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Camundongos , Camundongos Mutantes , Proteína Quinase 10 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/genética , Neurônios/citologia , Estresse Oxidativo/fisiologia , Fosforilação , Proteínas Tirosina Quinases/genética , Ratos
13.
J Neurochem ; 85(4): 999-1005, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12716431

RESUMO

Reactive oxygen species are a necessary triggering event for apoptosis of sympathetic neurons after nerve growth factor (NGF) withdrawal. Reactive oxygen species can lead to the generation of 4-hydroxynonenal (HNE), a highly reactive aldehyde that forms adducts with proteins. This covalent modification can activate or inhibit signal transduction pathways involved in the induction of apoptosis. This process may be clinically relevant because HNE-adduct immunoreactivity increases in several disease states. Here we evaluate the role of HNE-adducts in sympathetic neurons undergoing NGF-deprivation-induced apoptosis, a model of developmental programmed cell death. We show that HNE-adduct immunoreactivity is dramatically increased after NGF-withdrawal in an NADPH oxidase-dependent manner. Moreover, HNE-adducts appear to contribute to NGF-deprivation-induced apoptotic signal transduction because microinjected HNE-adduct antiserum protects sympathetic neurons from NGF withdrawal. In conclusion, this report suggests the direct contribution of endogenously generated HNE in the stimulation of apoptotic signal transduction in neurons.


Assuntos
Aldeídos/metabolismo , Apoptose/fisiologia , Fator de Crescimento Neural/farmacologia , Neurônios/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Soros Imunes/farmacologia , Microinjeções , NADPH Oxidases/metabolismo , Fator de Crescimento Neural/deficiência , Neurônios/citologia , Neurônios/metabolismo , Proteínas/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Sistemas do Segundo Mensageiro/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Sistema Nervoso Simpático/citologia
14.
J Neurosci Res ; 72(2): 185-90, 2003 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-12671992

RESUMO

Caveolae are cholesterol-rich, membrane microdomains that appear critical to signaling between extracellular and intracellular macromolecules as well as cholesterol homeostasis. Caveolae formation is modulated by caveolin, a protein family that is the proteinaceous hallmark of caveolae. Very little is known regarding the events that modulate caveolin expression and regulation in neurons. To detect caveolin expression in neurons, primary rat hippocampal neurons were harvested at embryonic day 18, maintained for 7 days in vitro, and then analyzed for caveolin immunofluorescence. Caveolin-1 immunoreactivity was detected in cells that were identified as neurons by morphology and concurrent microtubule-associated protein (MAP2) staining. Changes in caveolin-1 expression were evaluated by reverse transcriptase-polymerase chain reaction (RT-PCR) analyses of RNA isolated from hippocampal neurons treated with glutamate receptor agonists. Glutamate induced a concentration-dependent increase in caveolin-1 mRNA. The largest increases in caveolin-1 mRNA were detected after 6 hours of treatment. Kainate and AMPA both mimicked glutamate effects on caveolin-1 mRNA expression. Western blot analyses revealed that caveolin was induced at the protein level as well. Taken together, these data suggest that glutamate can regulate caveolin expression through kainate and AMPA ionotropic glutamate receptors.


Assuntos
Caveolinas/genética , Regulação da Expressão Gênica/fisiologia , Ácido Glutâmico/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Animais , Caveolina 1 , Caveolinas/metabolismo , Membrana Celular/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Relação Dose-Resposta a Droga , Feto/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/citologia , Neurotoxinas/farmacologia , RNA Mensageiro/genética , Ratos , Receptores de AMPA/metabolismo , Receptores de Ácido Caínico/metabolismo , Transdução de Sinais/fisiologia
15.
J Biol Chem ; 279(6): 4922-8, 2004 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-14623872

RESUMO

A key virulence factor for Yersinia pestis, the etiologic agent of plague, is the tyrosine phosphatase YopH, which the bacterium injects into host cells. We report that treatment of human T lymphocytes with a recombinant membrane-permeable YopH resulted in severe reduction in intracellular tyrosine phosphorylation and inhibition of T cell activation. The primary signal transducer for the T cell antigen receptor, the Lck tyrosine kinase, was specifically precipitated by a substrate-trapping YopH mutant, and Lck was dephosphorylated at its positive regulatory site, Tyr-394, in cells containing active YopH. By turning off Lck, YopH blocks T cell antigen receptor signaling at its very first step, effectively preventing the development of a protective immune response against this lethal bacterium.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/química , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Yersinia pestis/enzimologia , Sequência de Aminoácidos , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/genética , Humanos , Técnicas In Vitro , Células Jurkat , Ativação Linfocitária , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fosforilação , Proteínas Tirosina Fosfatases/química , Proteínas Tirosina Fosfatases/genética , Receptores de Antígenos de Linfócitos T/antagonistas & inibidores , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/microbiologia , Tirosina/química , Virulência , Yersinia pestis/genética , Yersinia pestis/patogenicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA