Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Nat Commun ; 10(1): 4779, 2019 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-31636267

RESUMO

Hyperinflammatory syndromes are life-threatening disorders caused by overzealous immune cell activation and cytokine release, often resulting from defects in negative feedback mechanisms. In the quintessential hyperinflammatory syndrome familial hemophagocytic lymphohistiocytosis (HLH), inborn errors of cytotoxicity result in effector cell accumulation, immune dysregulation and, if untreated, tissue damage and death. Here, we describe a human case with a homozygous nonsense R688* RC3H1 mutation suffering from hyperinflammation, presenting as relapsing HLH. RC3H1 encodes Roquin-1, a posttranscriptional repressor of immune-regulatory proteins such as ICOS, OX40 and TNF. Comparing the R688* variant with the murine M199R variant reveals a phenotypic resemblance, both in immune cell activation, hypercytokinemia and disease development. Mechanistically, R688* Roquin-1 fails to localize to P-bodies and interact with the CCR4-NOT deadenylation complex, impeding mRNA decay and dysregulating cytokine production. The results from this unique case suggest that impaired Roquin-1 function provokes hyperinflammation by a failure to quench immune activation.


Assuntos
Linfo-Histiocitose Hemofagocítica/genética , Proteínas de Ligação a RNA/genética , Ubiquitina-Proteína Ligases/genética , Adolescente , Animais , Códon sem Sentido , Consanguinidade , Ciclosporina/uso terapêutico , Eosinofilia/genética , Eosinofilia/imunologia , Homozigoto , Humanos , Imunofenotipagem , Imunossupressores/uso terapêutico , Proteína Coestimuladora de Linfócitos T Induzíveis/genética , Proteína Coestimuladora de Linfócitos T Induzíveis/imunologia , Proteína Coestimuladora de Linfócitos T Induzíveis/metabolismo , Linfo-Histiocitose Hemofagocítica/tratamento farmacológico , Linfo-Histiocitose Hemofagocítica/imunologia , Masculino , Camundongos , Monócitos/imunologia , Receptores OX40/genética , Receptores OX40/imunologia , Receptores OX40/metabolismo , Recidiva , Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Ubiquitina-Proteína Ligases/imunologia
3.
J Virol ; 92(22)2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30158289

RESUMO

Rabies virus is a neurovirulent RNA virus, which causes about 59,000 human deaths each year. Treatment for rabies does not exist due to incomplete understanding of the pathogenesis. MALT1 mediates activation of several immune cell types and is involved in the proliferation and survival of cancer cells. MALT1 acts as a scaffold protein for NF-κB signaling and a cysteine protease that cleaves substrates, leading to the expression of immunoregulatory genes. Here, we examined the impact of genetic or pharmacological MALT1 inhibition in mice on disease development after infection with the virulent rabies virus strain CVS-11. Morbidity and mortality were significantly delayed in Malt1-/- compared to Malt1+/+ mice, and this effect was associated with lower viral load, proinflammatory gene expression, and infiltration and activation of immune cells in the brain. Specific deletion of Malt1 in T cells also delayed disease development, while deletion in myeloid cells, neuronal cells, or NK cells had no effect. Disease development was also delayed in mice treated with the MALT1 protease inhibitor mepazine and in knock-in mice expressing a catalytically inactive MALT1 mutant protein, showing an important role of MALT1 proteolytic activity. The described protective effect of MALT1 inhibition against infection with a virulent rabies virus is the precise opposite of the sensitizing effect of MALT1 inhibition that we previously observed in the case of infection with an attenuated rabies virus strain. Together, these data demonstrate that the role of immunoregulatory responses in rabies pathogenicity is dependent on virus virulence and reveal the potential of MALT1 inhibition for therapeutic intervention.IMPORTANCE Rabies virus is a neurotropic RNA virus that causes encephalitis and still poses an enormous challenge to animal and public health. Efforts to establish reliable therapeutic strategies have been unsuccessful and are hampered by gaps in the understanding of virus pathogenicity. MALT1 is an intracellular protease that mediates the activation of several innate and adaptive immune cells in response to multiple receptors, and therapeutic MALT1 targeting is believed to be a valid approach for autoimmunity and MALT1-addicted cancers. Here, we study the impact of MALT1 deficiency on brain inflammation and disease development in response to infection of mice with the highly virulent CVS-11 rabies virus. We demonstrate that pharmacological or genetic MALT1 inhibition decreases neuroinflammation and extends the survival of CVS-11-infected mice, providing new insights in the biology of MALT1 and rabies virus infection.


Assuntos
Encéfalo/imunologia , Inflamação/prevenção & controle , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/fisiologia , Vírus da Raiva/imunologia , Raiva/prevenção & controle , Linfócitos T/imunologia , Animais , Encéfalo/metabolismo , Encéfalo/virologia , Células Cultivadas , Inflamação/imunologia , Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/antagonistas & inibidores , Raiva/imunologia , Raiva/metabolismo , Linfócitos T/patologia , Linfócitos T/virologia
4.
J Virol ; 92(8)2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29367251

RESUMO

MALT1 is involved in the activation of immune responses, as well as in the proliferation and survival of certain cancer cells. MALT1 acts as a scaffold protein for NF-κB signaling and a cysteine protease that cleaves substrates, further promoting the expression of immunoregulatory genes. Deregulated MALT1 activity has been associated with autoimmunity and cancer, implicating MALT1 as a new therapeutic target. Although MALT1 deficiency has been shown to protect against experimental autoimmune encephalomyelitis, nothing is known about the impact of MALT1 on virus infection in the central nervous system. Here, we studied infection with an attenuated rabies virus, Evelyn-Rotnycki-Abelseth (ERA) virus, and observed increased susceptibility with ERA virus in MALT1-/- mice. Indeed, after intranasal infection with ERA virus, wild-type mice developed mild transient clinical signs with recovery at 35 days postinoculation (dpi). Interestingly, MALT1-/- mice developed severe disease requiring euthanasia at around 17 dpi. A decreased induction of inflammatory gene expression and cell infiltration and activation was observed in MALT1-/- mice at 10 dpi compared to MALT1+/+ infected mice. At 17 dpi, however, the level of inflammatory cell activation was comparable to that observed in MALT1+/+ mice. Moreover, MALT1-/- mice failed to produce virus-neutralizing antibodies. Similar results were obtained with specific inactivation of MALT1 in T cells. Finally, treatment of wild-type mice with mepazine, a MALT1 protease inhibitor, also led to mortality upon ERA virus infection. These data emphasize the importance of early inflammation and activation of T cells through MALT1 for controlling the virulence of an attenuated rabies virus in the brain.IMPORTANCE Rabies virus is a neurotropic virus which can infect any mammal. Annually, 59,000 people die from rabies. Effective therapy is lacking and hampered by gaps in the understanding of virus pathogenicity. MALT1 is an intracellular protein involved in innate and adaptive immunity and is an interesting therapeutic target because MALT1-deregulated activity has been associated with autoimmunity and cancers. The role of MALT1 in viral infection is, however, largely unknown. Here, we study the impact of MALT1 on virus infection in the brain, using the attenuated ERA rabies virus in different models of MALT1-deficient mice. We reveal the importance of MALT1-mediated inflammation and T cell activation to control ERA virus, providing new insights in the biology of MALT1 and rabies virus infection.


Assuntos
Encéfalo/imunologia , Ativação Linfocitária , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/imunologia , Vírus da Raiva/imunologia , Raiva/imunologia , Linfócitos T/imunologia , Animais , Encéfalo/patologia , Encéfalo/virologia , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Inflamação/virologia , Camundongos , Camundongos Knockout , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/genética , Raiva/genética , Raiva/patologia , Vírus da Raiva/genética , Linfócitos T/patologia
5.
Cell Death Discov ; 3: 17012, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28280602

RESUMO

Rabies virus is a highly neurovirulent RNA virus, which causes about 59000 deaths in humans each year. Previously, we described macrophage cytotoxicity upon infection with rabies virus. Here we examined the type of cell death and the role of specific caspases in cell death and disease development upon infection with two laboratory strains of rabies virus: Challenge Virus Standard strain-11 (CVS-11) is highly neurotropic and lethal for mice, while the attenuated Evelyn-Rotnycki-Abelseth (ERA) strain has a broader cell tropism, is non-lethal and has been used as an oral vaccine for animals. Infection of Mf4/4 macrophages with both strains led to caspase-1 activation and IL-1ß and IL-18 production, as well as activation of caspases-3, -7, -8, and -9. Moreover, absence of caspase-3, but not of caspase-1 and -11 or -7, partially inhibited virus-induced cell death of bone marrow-derived macrophages. Intranasal inoculation with CVS-11 of mice deficient for either caspase-1 and -11 or -7 or both IL-1ß and IL-18 led to general brain infection and lethal disease similar to wild-type mice. Deficiency of caspase-3, on the other hand, significantly delayed the onset of disease, but did not prevent final lethal outcome. Interestingly, deficiency of caspase-1/11, the key executioner of pyroptosis, aggravated disease severity caused by ERA virus, whereas wild-type mice or mice deficient for either caspase-3, -7, or both IL-1ß and IL-18 presented the typical mild symptoms associated with ERA virus. In conclusion, rabies virus infection of macrophages induces caspase-1- and caspase-3-dependent cell death. In vivo caspase-1/11 and caspase-3 differently affect disease development in response to infection with the attenuated ERA strain or the virulent CVS-11 strain, respectively. Inflammatory caspases seem to control attenuated rabies virus infection, while caspase-3 aggravates virulent rabies virus infection.

6.
Cell Death Dis ; 7(6): e2250, 2016 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-27253414

RESUMO

An important regulator of inflammatory signalling is the ubiquitin-editing protein A20 that acts as a break on nuclear factor-κB (NF-κB) activation, but also exerts important cytoprotective functions. A20 knockout mice are cachectic and die prematurely due to excessive multi-organ inflammation. To establish the importance of A20 in liver homeostasis and pathology, we developed a novel mouse line lacking A20 specifically in liver parenchymal cells. These mice spontaneously develop chronic liver inflammation but no fibrosis or hepatocellular carcinomas, illustrating an important role for A20 in normal liver tissue homeostasis. Hepatocyte-specific A20 knockout mice show sustained NF-κB-dependent gene expression in the liver upon tumor necrosis factor (TNF) or lipopolysaccharide injection, as well as hepatocyte apoptosis and lethality upon challenge with sublethal doses of TNF, demonstrating an essential role for A20 in the protection of mice against acute liver failure. Finally, chronic liver inflammation and enhanced hepatocyte apoptosis in hepatocyte-specific A20 knockout mice was associated with increased susceptibility to chemically or high fat-diet-induced hepatocellular carcinoma development. Together, these studies establish A20 as a crucial hepatoprotective factor.


Assuntos
Apoptose , Citoproteção , Hepatócitos/metabolismo , Hepatócitos/patologia , Inflamação/patologia , Neoplasias Hepáticas/patologia , Fígado/patologia , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/metabolismo , Animais , Apoptose/efeitos dos fármacos , Carcinogênese/efeitos dos fármacos , Carcinogênese/metabolismo , Carcinogênese/patologia , Doença Crônica , Citocinas/metabolismo , Citoproteção/efeitos dos fármacos , Dieta Hiperlipídica , Proteína de Domínio de Morte Associada a Fas/metabolismo , Deleção de Genes , Hepatite/metabolismo , Hepatite/patologia , Hepatócitos/efeitos dos fármacos , Inflamação/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Fígado/efeitos dos fármacos , Neoplasias Hepáticas/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Fenótipo , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/deficiência , Fator de Necrose Tumoral alfa/farmacologia
8.
Mucosal Immunol ; 7(5): 1079-93, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24424522

RESUMO

Intestinal damage and severe diarrhea are serious side effects of cancer chemotherapy and constrain the usage of most such therapies. Here we show that interleukin-33 (IL-33) mediates the severe intestinal mucositis in mice treated with irinotecan (CPT-11), a commonly used cancer chemotherapeutic agent. Systemic CPT-11 administration led to severe mucosal damage, diarrhea, and body weight loss concomitant with the induction of IL-33 in the small intestine (SI). This mucositis was markedly reduced in mice deficient in the IL-33R (ST2(-/-)). Moreover, recombinant IL-33 exacerbated the CPT-11-induced mucositis, whereas IL-33 blockade with anti-IL-33 antibody or soluble ST2 markedly attenuated the disease. CPT-11 treatment increased neutrophil accumulation in the SI and adhesion to mesenteric veins. Supernatants from SI explants treated with CPT-11 enhanced transmigration of neutrophils in vitro in an IL-33-, CXCL1/2-, and CXCR2-dependent manner. Importantly, IL-33 blockade reduced mucositis and enabled prolonged CPT-11 treatment of ectopic CT26 colon carcinoma, leading to a beneficial outcome of the chemotherapy. These results suggest that inhibition of the IL-33/ST2 pathway may represent a novel approach to limit mucositis and thus improve the effectiveness of chemotherapy.


Assuntos
Camptotecina/análogos & derivados , Neoplasias do Colo/terapia , Interleucina-33/antagonistas & inibidores , Mucosite/induzido quimicamente , Animais , Antineoplásicos Fitogênicos/farmacologia , Antineoplásicos Fitogênicos/uso terapêutico , Western Blotting , Camptotecina/farmacologia , Linhagem Celular Tumoral , Sinergismo Farmacológico , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Proteína 1 Semelhante a Receptor de Interleucina-1 , Interleucina-33/genética , Interleucina-33/farmacologia , Irinotecano , Camundongos , Camundongos Endogâmicos BALB C , Mucosite/prevenção & controle , Receptores de Interleucina/metabolismo , Proteínas Recombinantes/farmacologia
10.
Cell Death Differ ; 18(12): 1845-53, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21566665

RESUMO

The ubiquitin-editing enzyme A20 (tumor necrosis factor-α-induced protein 3) serves as a critical brake on nuclear factor κB (NF-κB) signaling. In humans, polymorphisms in or near the A20 gene are associated with several inflammatory disorders, including psoriasis. We show here that epidermis-specific A20-knockout mice (A20(EKO)) develop keratinocyte hyperproliferation, but no signs of skin inflammation, such as immune cell infiltration. However, A20(EKO) mice clearly developed ectodermal organ abnormalities, including disheveled hair, longer nails and sebocyte hyperplasia. This phenotype resembles that of mice overexpressing ectodysplasin-A1 (EDA-A1) or the ectodysplasin receptor (EDAR), suggesting that A20 negatively controls EDAR signaling. We found that A20 inhibited EDAR-induced NF-κB signaling independent from its de-ubiquitinating activity. In addition, A20 expression was induced by EDA-A1 in embryonic skin explants, in which its expression was confined to the hair placodes, known to be the site of EDAR expression. In summary, our data indicate that EDAR-induced NF-κB levels are controlled by A20, which functions as a negative feedback regulator, to assure proper skin homeostasis and epidermal appendage development.


Assuntos
Cisteína Endopeptidases/genética , Epiderme/fisiologia , Homeostase , Peptídeos e Proteínas de Sinalização Intracelular/genética , Queratinócitos/metabolismo , NF-kappa B/metabolismo , Animais , Cisteína Endopeptidases/metabolismo , Cisteína Endopeptidases/fisiologia , Ectodisplasinas/farmacologia , Ectodisplasinas/fisiologia , Receptor Edar/agonistas , Receptor Edar/antagonistas & inibidores , Receptor Edar/metabolismo , Epiderme/patologia , Retroalimentação Fisiológica , Genes Reporter , Células HEK293 , Cabelo/anormalidades , Cabelo/embriologia , Humanos , Hiperplasia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Queratinócitos/fisiologia , Antígeno Ki-67/metabolismo , Luciferases/biossíntese , Luciferases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Técnicas de Cultura de Tecidos , Proteína 3 Induzida por Fator de Necrose Tumoral alfa , Fator de Necrose Tumoral alfa/farmacologia , Fator de Necrose Tumoral alfa/fisiologia
11.
J Vet Pharmacol Ther ; 33(6): 564-72, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21062309

RESUMO

The pharmacodynamic properties of tepoxalin, Na-salicylate and ketoprofen were determined in an intravenous lipopolysaccharide (LPS) inflammation model in broiler chickens. The drugs were administered orally at a dose of 30, 50 and 3 mg/kg, respectively. LPS administration induces an increase in the intracellular expression of interleukin (IL)-1ß and IL-6 and the secreted IL-6 plasma concentration. Furthermore, an elevation in body temperature is noted. Despite pretreatment with a single dose of the drugs and LPS administration on the T(max) of the drug after a second dose, no decrease was seen in systemic IL-6 levels. The intracellular expression of IL-1ß in the heterophils was slightly decreased if LPS was administered in combination with each of the three drugs. Tepoxalin and Na-salicylate administration had no significant effect on the LPS-induced increase in prostaglandin E(2) plasma concentration, in contrast to ketoprofen. None of the three drugs were able to influence the elevation in body temperature after LPS administration. The pharmacokinetic properties of Na-salicylate and ketoprofen were not altered in combination with LPS administration. However, LPS significantly decreased the AUC(0→6 h) of the active metabolite of tepoxalin, RWJ-20142, indicating a perfusion-limited elimination for this molecule.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Inflamação/veterinária , Cetoprofeno/farmacologia , Doenças das Aves Domésticas/tratamento farmacológico , Pirazóis/farmacologia , Salicilato de Sódio/farmacologia , Animais , Anti-Inflamatórios não Esteroides/farmacocinética , Anti-Inflamatórios não Esteroides/uso terapêutico , Temperatura Corporal/efeitos dos fármacos , Galinhas , Cromatografia Líquida de Alta Pressão/veterinária , Dinoprostona/sangue , Relação Dose-Resposta a Droga , Feminino , Citometria de Fluxo/veterinária , Inflamação/tratamento farmacológico , Injeções Intravenosas/veterinária , Interleucina-1beta/sangue , Interleucina-6/sangue , Cetoprofeno/farmacocinética , Lipopolissacarídeos/farmacologia , Masculino , Pirazóis/farmacocinética , Salicilato de Sódio/farmacocinética
12.
Avian Pathol ; 38(5): 403-11, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19937527

RESUMO

Intravenous administration of lipopolysaccharide (LPS) from Escherichia coli O127:B8 at a dose of 1,500,000 u/kg body weight evoked a hypothermic response followed by a fever phase in 5-week-old broiler chickens. The hypothermic phase coincided with a severe decrease in blood pressure. We assume that this decrease in blood pressure is, at least partly, responsible for the hypothermic phase of the body temperature curve. LPS administration also caused a decrease in circulating white blood cells. The heterophils were predominantly sequestered in the lungs. In LPS-treated chickens, far more apoptotic leukocytes were present in the circulation, compared with control chickens. The molecular players responsible for the LPS-induced inflammatory response could be TL1A, IL-1beta and IL-6, since a slight increase in their mRNA levels in white blood cells was already seen 1 h after LPS administration. In accordance with these observations, the levels of secreted IL-6 were maximal 3 h after LPS administration. These parameters characterize this LPS-induced inflammation model in broiler chickens.


Assuntos
Galinhas , Modelos Animais de Doenças , Inflamação/fisiopatologia , Animais , Apoptose/efeitos dos fármacos , Expressão Gênica , Hipotensão/induzido quimicamente , Hipotermia/induzido quimicamente , Inflamação/induzido quimicamente , Inflamação/patologia , Injeções Intravenosas , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Leucócitos/efeitos dos fármacos , Leucócitos/patologia , Lipopolissacarídeos/administração & dosagem , Pulmão/efeitos dos fármacos , Pulmão/patologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo
13.
Oncogene ; 27(47): 6131-40, 2008 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-18622428

RESUMO

The epidermal growth factor receptor (EGFR) is frequently overexpressed in various tumours of epidermal origin and is held responsible for tumourigenicity and tumour persistence. Increased nuclear factor (NF)-kappaB activity has been suggested to be involved in the malignant behaviour of EGFR-overexpressing cells. However, the mechanisms that regulate EGF-induced NF-kappaB activation are still largely unknown. Here we show that EGF can induce NF-kappaB-dependent gene expression independently from IkappaBalpha degradation or p100 processing in EGFR-overexpressing HEK293T cells. Moreover, EGF-induced NF-kappaB activation could be inhibited by overexpression of ABINs, which were previously identified as intracellular inhibitors of tumour necrosis factor, interleukin-1 and lipopolysaccharide-induced NF-kappaB activation. Knockdown of ABIN-1 by RNA interference boosted the NF-kappaB response upon EGF stimulation. The C-terminal ubiquitin-binding domain containing region of ABINs was crucial and sufficient for NF-kappaB inhibition. Adenoviral gene transfer of ABINs reduced constitutive NF-kappaB activity as well as the proliferation of EGFR-overexpressing A431 and DU145 human carcinoma cells. Altogether, these results demonstrate an important role for an ABIN-sensitive non-classical NF-kappaB signalling pathway in the proliferation of EGFR-overexpressing tumour cells, and indicate a potential use for ABIN gene therapy in the treatment of cancer.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas de Ligação a DNA/fisiologia , Receptores ErbB/antagonistas & inibidores , NF-kappa B/antagonistas & inibidores , Neoplasias/patologia , Proteínas Adaptadoras de Transdução de Sinal/análise , Proteínas Adaptadoras de Transdução de Sinal/genética , Linhagem Celular Tumoral , Proliferação de Células , Ciclina D1/genética , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/genética , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/análise , Receptores ErbB/fisiologia , Terapia Genética , Humanos , Proteínas I-kappa B/metabolismo , Inibidor de NF-kappaB alfa , NF-kappa B/fisiologia , Subunidade p52 de NF-kappa B/metabolismo , Neoplasias/metabolismo , Neoplasias/terapia , Fosforilação , Estrutura Terciária de Proteína , Interferência de RNA , Transdução de Sinais
14.
Cell Mol Life Sci ; 65(19): 2964-78, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18535784

RESUMO

Toll-like receptors (TLRs) as well as the receptors for tumor necrosis factor (TNF-R) and interleukin-1 (IL-1R) play an important role in innate immunity by regulating the activity of distinct transcription factors such as nuclear factor-kappaB (NF-kappaB). TLR, IL-1R and TNF-R signaling to NF-kappaB converge on a common IkappaB kinase complex that phosphorylates the NF-kappaB inhibitory protein IkappaBalpha. However, upstream signaling components are in large part receptor-specific. Nevertheless, the principles of signaling are similar, involving the recruitment of specific adaptor proteins and the activation of kinase cascades in which protein-protein interactions are controlled by poly-ubiquitination. In this review, we will discuss our current knowledge of NF-kappaB signaling in response to TLR-4, TNF-R and IL-1R stimulation, with a special focus on the similarities and dissimilarities among these pathways.


Assuntos
NF-kappa B/metabolismo , Receptores de Interleucina-1/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais/fisiologia , Receptor 4 Toll-Like/metabolismo , Animais , Humanos , Interleucina-1/metabolismo , Modelos Biológicos
15.
Oncogene ; 27(27): 3780-8, 2008 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-18246123

RESUMO

Interleukin-1beta (IL-1beta) is a cytokine that shares with tumor necrosis factor (TNF) the ability to initiate largely similar signaling pathways, leading to proinflammatory gene expression. In contrast to TNF, however, IL-1beta is not believed to induce tumor cell death. Here we demonstrate that prolonged treatment with IL-1beta, in combination with interferon-gamma (IFNgamma), is cytotoxic for L929 tumor cells. IL-1beta/IFNgamma-induced cytotoxicity requires only minimal amounts of IL-1beta and shows morphological features of necrosis. Although TNF induces a similar response, we could exclude a contribution of endogenous TNF production in the effect of IL-1beta/IFNgamma. Cell death in response to IL-1beta/IFNgamma is independent of caspases, but requires the IL-1beta/IFNgamma-induced production of inducible nitric oxide synthase (iNOS) and NO. Moreover, necrosis and iNOS/NO production could be prevented by treatment of the cells with a p38 mitogen activated protein kinase (p38MAPK) or IkappaB kinase beta inhibitor. Altogether, these findings demonstrate that prolonged exposure to IL-1beta plus IFNgamma induces L929 tumor cell necrosis, via a p38MAPK and nuclear factor-kappaB (NF-kappaB)-dependent signaling pathway, leading to the expression of iNOS and the production of toxic NO levels.


Assuntos
Interferon gama/toxicidade , Interleucina-1beta/toxicidade , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Camundongos , Necrose , Fator de Necrose Tumoral alfa/toxicidade
16.
Oncogene ; 27(26): 3739-45, 2008 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-18212736

RESUMO

Deregulated nuclear factor kappaB (NF-kappaB) activation plays an important role in inflammation and tumorigenesis. ABIN proteins have been characterized as negative regulators of NF-kappaB signaling. However, their mechanism of NF-kappaB inhibition remained unclear. With the help of a yeast two-hybrid screen, we identified ABIN proteins as novel ubiquitin-interacting proteins. The minimal ubiquitin-binding domain (UBD) corresponds to the ABIN homology domain 2 (AHD2) and is highly conserved in ABIN-1, ABIN-2 and ABIN-3. Moreover, this region is also present in NF-kappaB essential modulator/IkappaB kinase gamma (NEMO/IKKgamma) and the NEMO-like protein optineurin, and is therefore termed UBD in ABIN proteins and NEMO (UBAN). Nuclear magnetic resonance studies of the UBAN domain identify it as a novel type of UBD, with the binding surface on ubiquitin being significantly different from the binding surface of other UBDs. ABIN-1 specifically binds ubiquitinated NEMO via a bipartite interaction involving its UBAN and NEMO-binding domain. Mutations in the UBAN domain led to a loss of ubiquitin binding and impaired the NF-kappaB inhibitory potential of ABINs. Taken together, these data illustrate an important role for ubiquitin binding in the negative regulation of NF-kappaB signaling by ABINs and identify UBAN as a novel UBD.


Assuntos
Proteínas de Ligação a DNA/fisiologia , NF-kappa B/antagonistas & inibidores , Ubiquitina/metabolismo , Sítios de Ligação , Linhagem Celular , Proteínas de Ligação a DNA/química , Humanos , NF-kappa B/fisiologia , Estrutura Terciária de Proteína , Transdução de Sinais , Técnicas do Sistema de Duplo-Híbrido
17.
Avian Pathol ; 37(1): 39-44, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18202948

RESUMO

Our objective was to create a standardized and reproducible inflammation model in chickens in order to study the pharmacodynamics of several anti-pyretic and anti-inflammatory drugs. We studied the influence of age and repeated lipopolysaccharide (LPS) administration on body temperature and the correlation of this with concentrations of interleukin-6 and IgM antibodies against LPS in plasma of chickens. Three-week-old and 5-week-old broilers were injected intravenously with LPS from Escherichia coli O127: B8 at a dose of 1 mg/kg. LPS administration was repeated after 2 or 7 days. After the first dose of LPS, the body temperature was initially decreased below normal and then later increased above normal. The second dose of LPS reduced the level of hypothermia and the duration of the febrile phase. Three-week-old birds responded to LPS with a higher maximum body temperature and a greater area under the body temperature versus time curve than 5-week-old chickens (P<0.05). Interleukin-6 reached its highest concentration 3 h after LPS administration and returned to baseline levels after 9 h. A second dose of LPS resulted in a significantly lower peak in interleukin-6. Significant higher levels of antibodies against LPS could be detected 7 days after LPS administration. However, there appeared to be no correlation between the reduced response to LPS and the presence of antibodies.


Assuntos
Envelhecimento/fisiologia , Temperatura Corporal/efeitos dos fármacos , Galinhas/fisiologia , Imunoglobulina M/sangue , Interleucina-6/sangue , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/imunologia , Animais , Galinhas/imunologia , Feminino , Lipopolissacarídeos/farmacologia , Masculino , Fatores de Tempo
18.
J Cell Mol Med ; 12(5A): 1767-76, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18081695

RESUMO

Pseudomonas aeruginosa is an opportunistic bacterial pathogen that forms a serious problem for immunocompromised patients and also the leading cause of mortality in cystic fibrosis. The overall importance of a functional Type III secretion system (T3SS) in P. aeruginosa virulence has been well established, but the underlying mechanisms are still unclear. Using in vitro infected macrophages as w as a murine model of acute lung infection, we show that the Caspase-1 mediated maturation and secretion of IL-1beta needs a translocation competent T3SS and Flagellin, but not the Type III effector proteins ExoS, ExoT and ExoY. However, ExoS was found to negative regulate the P. aeruginosa induced IL-1beta maturation by a mechanism that is dependent on its ADP ribosyltransferase activity. Moreov ExoS deficiency also switched the mode of macrophage death from apoptosis to pro-inflammatory pyroptosis. Altogether, these data demonstrate a dual role for the P. aeruginosa T3SS in the regulation of Caspase-1 mediated IL-1beta production and provide new insights into the mechanisms of immune evasion by this pathogen.


Assuntos
Caspase 1/metabolismo , Interleucina-1beta/metabolismo , Pseudomonas aeruginosa/metabolismo , ADP Ribose Transferases/metabolismo , Animais , Apoptose , Toxinas Bacterianas/metabolismo , Células Cultivadas , Feminino , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos DBA , Infecções por Pseudomonas/genética , Infecções por Pseudomonas/metabolismo , Infecções por Pseudomonas/patologia
19.
Cell Prolif ; 40(5): 640-55, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17877607

RESUMO

OBJECTIVE: Benzoylaminoalkanohydroxamic acids, including 5-(4-dimethylaminobenzoyl)aminovaleric acid hydroxamide (4-Me(2)N-BAVAH), are structural analogues of Trichostatin A, a naturally occurring histone deacetylase inhibitor (HDACi). 4-Me(2)N-BAVAH has been shown to induce histone hyperacetylation and to inhibit proliferation in Friend erythroleukaemia cells in vitro. However, the molecular mechanisms have remained unidentified. MATERIALS AND METHODS: In this study, we evaluated the effects of 4-Me(2)N-BAVAH on proliferation in non-malignant cells, namely epidermal growth factor-stimulated primary rat hepatocytes. RESULTS AND CONCLUSION: We have found that 4-Me(2)N-BAVAH inhibits HDAC activity at non-cytotoxic concentrations and prevents cells from responding to the mitogenic stimuli of epidermal growth factor. This results in an early G(1) cell cycle arrest that is independent of p21 activity, but instead can be attributed to inhibition of cyclin D1 transcription through a mechanism involving inhibition of nuclear factor-kappaB activation. In addition, 4-Me(2)N-BAVAH delays the onset of spontaneous apoptosis in primary rat hepatocyte cultures as evidenced by down-regulation of the pro-apoptotic proteins Bid and Bax, and inhibition of caspase-3 activation.


Assuntos
Hepatócitos/efeitos dos fármacos , Inibidores de Histona Desacetilases , Ácidos Hidroxâmicos/farmacologia , NF-kappa B/antagonistas & inibidores , Ácidos Pentanoicos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Ciclina D , Ciclinas/genética , Regulação para Baixo/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Fase G1/efeitos dos fármacos , Genes Reporter , Hepatócitos/citologia , Hepatócitos/metabolismo , Luciferases/genética , Ratos , Transcrição Gênica/efeitos dos fármacos
20.
Oncogene ; 25(12): 1693-5, 2006 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-16186791

RESUMO

Protein kinases are critically involved in signaling pathways that regulate cell growth, differentiation, activation, and survival. Lck, a member of the Src family of protein tyrosine kinases, plays a key role in T-lymphocyte activation and differentiation. However, under certain conditions Lck is also involved in the induction of apoptosis. In this issue of Oncogene, Samraj et al. used the Lck-defective JCaM1.6 cell line to demonstrate the critical role of Lck in the apoptotic response of T-cell leukemia cells to several chemotherapeutic drugs. They further showed that Lck controls the mitochondrial death pathway by regulating proapoptotic Bak expression. This chemosensitizing effect of Lck is independent of T-cell receptor signaling and does not require the kinase activity of Lck. These findings demonstrate that Lck might be part of two independent signaling pathways leading to either cell proliferation or apoptosis, and reveal a hitherto unrecognized link between Lck, Bak, and chemosensitivity of human leukemic cells.


Assuntos
Antineoplásicos/farmacologia , Apoptose/fisiologia , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/biossíntese , Proteína Killer-Antagonista Homóloga a bcl-2/biossíntese , Animais , Apoptose/efeitos dos fármacos , Humanos , Leucemia/tratamento farmacológico , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Proteína Killer-Antagonista Homóloga a bcl-2/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...