Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 12(1): 4379, 2021 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-34282144

RESUMO

The transcription factor IRF5 has been implicated as a therapeutic target for the autoimmune disease systemic lupus erythematosus (SLE). However, IRF5 activation status during the disease course and the effects of IRF5 inhibition after disease onset are unclear. Here, we show that SLE patients in both the active and remission phase have aberrant activation of IRF5 and interferon-stimulated genes. Partial inhibition of IRF5 is superior to full inhibition of type I interferon signaling in suppressing disease in a mouse model of SLE, possibly due to the function of IRF5 in oxidative phosphorylation. We further demonstrate that inhibition of IRF5 via conditional Irf5 deletion and a newly developed small-molecule inhibitor of IRF5 after disease onset suppresses disease progression and is effective for maintenance of remission in mice. These results suggest that IRF5 inhibition might overcome the limitations of current SLE therapies, thus promoting drug discovery research on IRF5 inhibitors.


Assuntos
Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/metabolismo , Animais , Autoanticorpos/imunologia , Doenças Autoimunes , Feminino , Regulação da Expressão Gênica , Humanos , Imunidade Inata , Imunoglobulina G , Fatores Reguladores de Interferon/efeitos dos fármacos , Rim/patologia , Lúpus Eritematoso Sistêmico/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Interferon alfa e beta , Transdução de Sinais , Fatores de Transcrição , Quinases da Família src
2.
J Ethnopharmacol ; 269: 113752, 2021 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-33359858

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Traditionally, the roots of Angelica reflexa B.Y.Lee (AR) have been used to treat cough, phlegm, neuralgia, and arthralgia in Northeast Asia. AIM OF THE STUDY: The anti-asthmatic effect of AR root extract (ARE) was determined using a murine airway allergic inflammation model and the primary T cell polarization assay. MATERIALS AND METHODS: To evaluate the anti-asthmatic effect of ARE, inflammatory cell infiltration was determined histologically and inflammatory mediators were measured in bronchoalveolar lavage fluid (BALF). Furthermore, the effects of AREs on Th2 cell differentiation and activation were determined by western blotting and flow cytometry. RESULTS: Asthmatic phenotypes were alleviated by ARE treatment, which reduced mucus production, inflammatory cell infiltration (especially eosinophilia), and type 2 cytokine levels in BALF. ARE administration to mice reduced the number of activated Th2 (CD4+CD25+) cells and level of GATA3 in the lungs. Furthermore, ARE treatment inhibited the differentiation of Th2 cells in primary cell culture systems via interferon regulatory factor 4 (IRF4) signaling. CONCLUSIONS: Our findings indicate that the anti-asthmatic effect of AREs is mediated by the reduction in Th2 cell activation by regulating IRF4.


Assuntos
Angelica/química , Antiasmáticos/farmacologia , Asma/tratamento farmacológico , Hipersensibilidade/tratamento farmacológico , Extratos Vegetais/farmacologia , Pneumonia/tratamento farmacológico , Células Th2/efeitos dos fármacos , Animais , Antiasmáticos/química , Antiasmáticos/uso terapêutico , Asma/induzido quimicamente , Asma/imunologia , Líquido da Lavagem Broncoalveolar/imunologia , Citocinas/metabolismo , Feminino , Fator de Transcrição GATA3/efeitos dos fármacos , Fator de Transcrição GATA3/metabolismo , Hipersensibilidade/imunologia , Fatores Reguladores de Interferon/efeitos dos fármacos , Fatores Reguladores de Interferon/metabolismo , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/toxicidade , Extratos Vegetais/química , Extratos Vegetais/uso terapêutico , Raízes de Plantas/química , Pneumonia/induzido quimicamente , Pneumonia/metabolismo , Pneumonia/patologia , Eosinofilia Pulmonar/induzido quimicamente , Eosinofilia Pulmonar/tratamento farmacológico , Células RAW 264.7 , Células Th2/imunologia
3.
Osteoarthritis Cartilage ; 29(2): 235-247, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33248223

RESUMO

OBJECTIVE: Fibronectin is a matrix protein that is fragmented during cartilage degradation in osteoarthritis (OA). Treatment of chondrocytes with fibronectin fragments (FN-f) has been used to model OA in vitro, but the system has not been fully characterized. This study sought to define the transcriptional response of chondrocytes to FN-f, and directly compare it to responses traditionally observed in OA. DESIGN: Normal human femoral chondrocytes isolated from tissue donors were treated with either FN-f or PBS (control) for 3, 6, or 18 h. RNA-seq libraries were compared between time-matched FN-f and control samples in order to identify changes in gene expression over time. Differentially expressed genes were compared to a published OA gene set and used for pathway, transcription factor motif, and kinome analysis. RESULTS: FN-f treatment resulted in 3,914 differentially expressed genes over the time course. Genes that are up- or downregulated in OA were significantly up- (P < 0.00001) or downregulated (P < 0.0004) in response to FN-f. Early response genes were involved in proinflammatory pathways, whereas many late response genes were involved in ferroptosis. The promoters of upregulated genes were enriched for NF-κB, AP-1, and IRF motifs. Highly upregulated kinases included CAMK1G, IRAK2, and the uncharacterized kinase DYRK3, while growth factor receptors TGFBR2 and FGFR2 were downregulated. CONCLUSIONS: FN-f treatment of normal human articular chondrocytes recapitulated many key aspects of the OA chondrocyte phenotype. This in vitro model is promising for future OA studies, especially considering its compatibility with genomics and genome-editing techniques.


Assuntos
Cartilagem Articular/citologia , Condrócitos/efeitos dos fármacos , Fibronectinas/farmacologia , Expressão Gênica/efeitos dos fármacos , Osteoartrite/genética , Proteína Quinase Tipo 1 Dependente de Cálcio-Calmodulina/efeitos dos fármacos , Proteína Quinase Tipo 1 Dependente de Cálcio-Calmodulina/genética , Condrócitos/metabolismo , Fêmur , Expressão Gênica/genética , Humanos , Técnicas In Vitro , Fatores Reguladores de Interferon/efeitos dos fármacos , Fatores Reguladores de Interferon/genética , Quinases Associadas a Receptores de Interleucina-1/efeitos dos fármacos , Quinases Associadas a Receptores de Interleucina-1/genética , NF-kappa B/efeitos dos fármacos , NF-kappa B/genética , Osteoartrite/metabolismo , Fragmentos de Peptídeos/farmacologia , Fenótipo , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/efeitos dos fármacos , Proteínas Tirosina Quinases/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/efeitos dos fármacos , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Receptor do Fator de Crescimento Transformador beta Tipo II/efeitos dos fármacos , Receptor do Fator de Crescimento Transformador beta Tipo II/genética , Fator de Transcrição AP-1/efeitos dos fármacos , Fator de Transcrição AP-1/genética
4.
PLoS Pathog ; 13(10): e1006647, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28968468

RESUMO

Interferon regulatory factor-8 (IRF-8) is critical for Th1 cell differentiation and negatively regulates myeloid cell development including myeloid-derived suppressor cells (MDSC). MDSC expand during infection with various pathogens including the gastrointestinal (GI) nematode Heligmosomoides polygyrus bakeri (Hpb). We investigated if IRF-8 contributes to Th2 immunity to Hpb infection. Irf8 expression was down-regulated in MDSC from Hpb-infected C57BL/6 (B6) mice. IRF-8 deficient Irf8-/- and BXH-2 mice had significantly higher adult worm burdens than B6 mice after primary or challenge Hpb infection. During primary infection, MDSC expanded to a significantly greater extent in mesenteric lymph nodes (MLN) and spleens of Irf8-/- and BXH-2 than B6 mice. CD4+GATA3+ T cells numbers were comparable in MLN of infected B6 and IRF-8 deficient mice, but MLN cells from infected IRF-8 deficient mice secreted significantly less parasite-specific IL-4 ex vivo. The numbers of alternatively activated macrophages in MLN and serum levels of Hpb-specific IgG1 and IgE were also significantly less in infected Irf8-/- than B6 mice. The frequencies of antigen-experienced CD4+CD11ahiCD49dhi cells that were CD44hiCD62L- were similar in MLN of infected Irf8-/- and B6 mice, but the proportions of CD4+GATA3+ and CD4+IL-4+ T cells were lower in infected Irf8-/- mice. CD11b+Gr1+ cells from naïve or infected Irf8-/- mice suppressed CD4+ T cell proliferation and parasite-specific IL-4 secretion in vitro albeit less efficiently than B6 mice. Surprisingly, there were significantly more CD4+ T cells in infected Irf8-/- mice, with a higher frequency of CD4+CD25+Foxp3+ T (Tregs) cells and significantly higher numbers of Tregs than B6 mice. In vivo depletion of MDSC and/or Tregs in Irf8-/- mice did not affect adult worm burdens, but Treg depletion resulted in higher egg production and enhanced parasite-specific IL-5, IL-13, and IL-6 secretion ex vivo. Our data thus provide a previously unrecognized role for IRF-8 in Th2 immunity to a GI nematode.


Assuntos
Gastroenteropatias/imunologia , Fatores Reguladores de Interferon/imunologia , Células Supressoras Mieloides/imunologia , Infecções por Nematoides/imunologia , Nematospiroides dubius/imunologia , Células Th2/imunologia , Animais , Células Cultivadas , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Fatores Reguladores de Interferon/efeitos dos fármacos , Fatores Reguladores de Interferon/genética , Interleucina-4/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Linfócitos T Reguladores/imunologia
5.
Arthritis Rheumatol ; 69(7): 1495-1501, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28426911

RESUMO

OBJECTIVE: Gain-of-function mutations in TMEM173, encoding the stimulator of interferon (IFN) genes (STING) protein, underlie a novel type I interferonopathy that is minimally responsive to conventional immunosuppressive therapies and associated with high frequency of childhood morbidity and mortality. STING gain-of-function causes constitutive oversecretion of IFN. This study was undertaken to determine the effects of a TANK-binding kinase 1 (TBK-1)/IKKɛ inhibitor (BX795) on secretion and signaling of IFN in primary peripheral blood mononuclear cells (PBMCs) from patients with mutations in STING. METHODS: PBMCs from 4 patients with STING-associated disease were treated with BX795. The effect of BX795 on IFN pathways was assessed by Western blotting and an IFNß reporter assay, as well as by quantification of IFNα in cell lysates, staining for STAT-1 phosphorylation, and measurement of IFN-stimulated gene (ISG) messenger RNA (mRNA) expression. RESULTS: Treatment of PBMCs with BX795 inhibited the phosphorylation of IFN regulatory factor 3 and IFNß promoter activity induced in HEK 293T cells by cyclic GMP-AMP or by genetic activation of STING. In vitro exposure to BX795 inhibited IFNα production in PBMCs of patients with STING-associated disease without affecting cell survival. In addition, BX795 decreased STAT-1 phosphorylation and ISG mRNA expression independent of IFNα blockade. CONCLUSION: These findings demonstrate the effect of BX795 on reducing type I IFN production and IFN signaling in cells from patients with gain-of-function mutations in STING. A combined inhibition of TBK-1 and IKKɛ therefore holds potential for the treatment of patients carrying STING mutations, and may also be relevant in other type I interferonopathies.


Assuntos
Fator Regulador 3 de Interferon/efeitos dos fármacos , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/efeitos dos fármacos , Interferon-alfa/efeitos dos fármacos , Interferon beta/efeitos dos fármacos , Leucócitos Mononucleares/efeitos dos fármacos , Proteínas de Membrana/efeitos dos fármacos , Pirimidinas/farmacologia , Tiofenos/farmacologia , Western Blotting , Criança , Células HEK293 , Humanos , Quinase I-kappa B/antagonistas & inibidores , Técnicas In Vitro , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Fatores Reguladores de Interferon/efeitos dos fármacos , Fatores Reguladores de Interferon/genética , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/genética , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/metabolismo , Interferon-alfa/imunologia , Interferon beta/imunologia , Leucócitos Mononucleares/imunologia , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Mutação , Nucleotídeos Cíclicos/farmacologia , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT1/efeitos dos fármacos , Fator de Transcrição STAT1/metabolismo
6.
Int Immunopharmacol ; 28(1): 257-63, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26093273

RESUMO

Acute rejection is a serious and life-threatening complication of liver transplantation (LTx). Tacrolimus (TAC) is a potent immunosuppressant used in experimental and clinical transplantation. Interferon regulatory factor 4 (IRF4) plays key roles as a transcription factor in the immune response. This study explored the role of IRF4 in acute rejection after LTx using TAC treatment. Here, LTx was performed in DA (RT1(n)) and Lewis (LEW) (RT1(l)) rats. The recipients were immunosuppressed with TAC (1.5mg/kg/day subcutaneously) or saline. Liver grafts were harvested 1, 3, 5, 7, and 10 days after LTx for histology, immunohistochemistry, western blotting and real-time PCR. Splenic mononuclear cells were activated with different doses of TAC. The nuclear factor of activated T cells (NFAT) signal pathway and CD4+ T subset-related transcription factors were assessed. The results showed that TAC treatment prolonged the survival of liver allografts in recipients, significantly attenuated hepatic tissue injury and improved liver function. IRF4 expression in grafts was down-regulated after TAC treatment. TAC inhibited the expression of IRF4, NFAT, Foxp3 and RORγt in splenic mononuclear cells in vitro. In conclusions, our studies showed that TAC attenuated acute rejection responses after LTx. This attenuation might depend on the TAC-NFAT-IRF4 signal pathway, which is crucial for the function of T helper subsets (Treg and Th17 cells) in acute rejection after LTx. These findings contribute to our understanding of the immune pharmacological mechanism of TAC to prevent rejection in LTx rats.


Assuntos
Rejeição de Enxerto/genética , Imunossupressores/farmacologia , Fatores Reguladores de Interferon/biossíntese , Transplante de Fígado , Tacrolimo/farmacologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Regulação para Baixo/efeitos dos fármacos , Rejeição de Enxerto/patologia , Fatores Reguladores de Interferon/efeitos dos fármacos , Fígado/patologia , Ativação de Macrófagos/efeitos dos fármacos , Masculino , Monócitos/efeitos dos fármacos , Ratos , Ratos Endogâmicos Lew , Transdução de Sinais/efeitos dos fármacos , Análise de Sobrevida , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/imunologia
7.
J Antibiot (Tokyo) ; 67(5): 379-86, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24549153

RESUMO

VirF is an AraC-type transcriptional regulator responsible for activating the transcription of virulence genes required for the intracellular invasion and cell-to-cell spread of Shigella flexneri. Gene disruption studies have validated VirF as a potential target for an anti-virulence therapy to treat shigellosis by determining that VirF is necessary for virulence, but not required for bacterial viability. Using a bacteria-based, ß-galactosidase reporter assay we completed a high-throughput screening (HTS) campaign monitoring VirF activity in the presence of over 140,000 small molecules. From our screening campaign, we identified five lead compounds to pursue in tissue culture-based invasion and cell-to-cell spread assays, and toxicity screens. Our observations of activity in these models for infection have validated our approach of targeting virulence regulation and have allowed us to identify a promising chemical scaffold from our HTS for hit-to-lead development. Interestingly, differential effects on invasion versus cell-to-cell spread suggest that the compounds' efficacies may depend, in part, on the specific promoter that VirF is recognizing.


Assuntos
Antibacterianos/farmacologia , Ensaios de Triagem em Larga Escala/métodos , Fatores Reguladores de Interferon/efeitos dos fármacos , Shigella flexneri/efeitos dos fármacos , Proteínas Virais/efeitos dos fármacos , Fatores de Virulência , Algoritmos , Antibacterianos/isolamento & purificação , Interpretação Estatística de Dados , Genes Reporter/genética , Células HeLa , Humanos , Indicadores e Reagentes , Testes de Sensibilidade Microbiana , Plasmídeos/genética , Reprodutibilidade dos Testes , Shigella flexneri/genética , Shigella flexneri/crescimento & desenvolvimento , Virulência/genética , beta-Galactosidase/genética
8.
PLoS One ; 8(12): e82867, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24386121

RESUMO

OM-85 (Broncho-Vaxom®, Broncho-Munal®, Ommunal®, Paxoral®, Vaxoral®), a product made of the water soluble fractions of 21 inactivated bacterial strain patterns responsible for respiratory tract infections, is used for the prevention of recurrent upper respiratory tract infections and acute exacerbations in chronic obstructive pulmonary disease patients. OM-85 is able to potentiate both innate and adaptive immune responses. However, the molecular mechanisms responsible for OM-85 activation are still largely unknown. Purpose of this study was to investigate the impact of OM-85 stimulation on human dendritic cell functions. We show that OM-85 selectively induced NF-kB and MAPK activation in human DC with no detectable action on the interferon regulatory factor (IRF) pathway. As a consequence, chemokines (i.e. CXCL8, CXCL6, CCL3, CCL20, CCL22) and B-cell activating cytokines (i.e. IL-6, BAFF and IL-10) were strongly upregulated. OM-85 also synergized with the action of classical pro-inflammatory stimuli used at suboptimal concentrations. Peripheral blood mononuclear cells from patients with COPD, a pathological condition often associated with altered PRR expression pattern, fully retained the capability to respond to OM-85. These results provide new insights on the molecular mechanisms of OM-85 activation of the immune response and strengthen the rational for its use in clinical settings.


Assuntos
Extratos Celulares/farmacologia , Células Dendríticas/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases , NF-kappa B/fisiologia , Quimiocinas/genética , Quimiocinas/metabolismo , Células Dendríticas/fisiologia , Sinergismo Farmacológico , Humanos , Fatores Reguladores de Interferon/efeitos dos fármacos , Leucócitos Mononucleares/efeitos dos fármacos , NF-kappa B/efeitos dos fármacos , NF-kappa B/metabolismo , Doença Pulmonar Obstrutiva Crônica/sangue , Regulação para Cima
9.
Antivir Ther ; 16(5): 657-66, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21817187

RESUMO

BACKGROUND: Kaposi's sarcoma-associated herpesvirus (KSHV) is associated with several malignant diseases, including Kaposi's sarcoma, primary effusion lymphoma (PEL) and multicentric Castleman's disease. The objectives of this study were to investigate the use of peptide-conjugated phosphorodiamidate morpholino oligomers (PPMOs) against KSHV early lytic genes and to assess their efficacy in severe combined immunodeficiency disease (SCID) mice against PEL engraftment. PPMOs are short, single-stranded DNA analogues that contain a backbone of morpholine rings and phosphorodiamidate linkages and have high delivery efficiency into cells. METHODS: PEL cells were treated with PPMOs against viral interferon regulatory factor 1 (vIRF-1) and expression of vIRF-1 was analysed. PPMOs against vIRF-1 and viral interleukin-6 (vIL-6) were evaluated against PEL cell engraftment in SCID mice. The PPMOs were incubated with BCBL-1 cells and then introduced into the peritoneal cavities of SCID mice, followed by 9 more doses of PPMOs administered at 2-day intervals. At weeks 3 and 9 after BCBL-1 delivery, peritoneal lavage was collected and the ratio of PEL cells among total cells was determined by flow cytometry analysis. RESULTS: Treatment of PEL cells with PPMOs against vIRF-1 led to a reduction of vIRF-1 expression in a dose-dependent manner. Reduction of vIRF-1 expression resulted in higher levels of cellular interferon regulatory factor 3 and of signal transducer and activator of transcription 1. SCID mice receiving a PPMO against vIL-6 had no engraftment of PEL cells and remained healthy throughout the 120-day study. CONCLUSIONS: This study showed that PPMOs can be effective antiviral agents against KSHV. Blocking the expression of early lytic genes might be beneficial for the control of KSHV-associated malignant diseases.


Assuntos
Antivirais/uso terapêutico , Infecções por Herpesviridae/tratamento farmacológico , Herpesvirus Humano 8/genética , Linfoma de Efusão Primária/tratamento farmacológico , Morfolinos/farmacologia , Animais , Antivirais/imunologia , Antivirais/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Desenho de Fármacos , Feminino , Infecções por Herpesviridae/genética , Herpesvirus Humano 8/efeitos dos fármacos , Fatores Reguladores de Interferon/efeitos dos fármacos , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo , Interleucina-6/análise , Interleucina-6/genética , Interleucina-6/metabolismo , Linfoma de Efusão Primária/genética , Linfoma de Efusão Primária/patologia , Linfoma de Efusão Primária/virologia , Camundongos , Camundongos SCID , Terapia de Alvo Molecular , Morfolinos/imunologia , Morfolinos/uso terapêutico , Fator de Transcrição STAT1/efeitos dos fármacos , Fator de Transcrição STAT1/genética , Sarcoma de Kaposi/genética , Sarcoma de Kaposi/patologia , Sarcoma de Kaposi/virologia , Imunodeficiência Combinada Severa/genética , Transfecção , Proteínas Virais/efeitos dos fármacos , Proteínas Virais/genética , Proteínas Virais/metabolismo
10.
J Immunol ; 183(10): 6646-56, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19864597

RESUMO

LPS is an immunostimulatory component of Gram-negative bacteria. Acting on the immune system in a systemic fashion, LPS exposes the body to the hazard of septic shock. In this study we report that cysteine-rich secretory protein LCCL domain containing 2 (CRISPLD2/Crispld2; human and mouse/rat versions, respectively), expressed by multitissues and leukocytes, is a novel LPS-binding protein. As a serum protein, median CRISPLD2 concentrations in health volunteers and umbilical cord blood samples are 607 microg/ml and 290 microg/ml, respectively. Human peripheral blood granulocytes and mononuclear cells including monocytes, NK cells, and T cells spontaneously release CRISPLD2 (range, 0.2-0.9 microg/ml) and enhance CRISPLD2 secretion (range, 1.5-4.2 microg/ml) in response to stimulation of both LPS and humanized anti-human TLR4-IgA Ab in vitro. CRISPLD2 exhibits significant LPS binding affinity similar to that of soluble CD14, prevents LPS binding to target cells, reduces LPS-induced TNF-alpha and IL-6 production, and protects mice against endotoxin shock. In in vivo experiments, serum Crispld2 concentrations increased in response to a nontoxic dose of LPS and correlated negatively with LPS lethality, suggesting that CRISPLD2 serum concentrations not only are indicators of the degree of a body's exposure to LPS but also reflect an individual's LPS sensitivity.


Assuntos
Moléculas de Adesão Celular/imunologia , Fatores Reguladores de Interferon/imunologia , Lipopolissacarídeos/imunologia , Proteínas Recombinantes/imunologia , Choque Séptico/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Moléculas de Adesão Celular/sangue , Moléculas de Adesão Celular/efeitos dos fármacos , Moléculas de Adesão Celular/metabolismo , Feminino , Granulócitos/imunologia , Granulócitos/metabolismo , Humanos , Fatores Imunológicos/farmacologia , Fatores Reguladores de Interferon/sangue , Fatores Reguladores de Interferon/efeitos dos fármacos , Fatores Reguladores de Interferon/metabolismo , Interleucina-6/imunologia , Interleucina-6/metabolismo , Estimativa de Kaplan-Meier , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Monócitos/imunologia , Monócitos/metabolismo , Proteínas Recombinantes/farmacologia , Choque Séptico/prevenção & controle , Linfócitos T/imunologia , Linfócitos T/metabolismo , Receptor 4 Toll-Like/imunologia , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA