Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
J Pharmacokinet Pharmacodyn ; 46(1): 65-74, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30661177

RESUMO

Danvatirsen is a Generation 2.5 antisense oligonucleotide under clinical development. Population PK modelling was conducted using data from 3 available danvatirsen Phase I/II studies in oncology patients to investigate the impact of flat dosing on exposure compared to ideal body weight-based dosing. A total of 126 patients who received danvatirsen doses ranging from 1 to 4 mg/kg as monotherapy or in combination with durvalumab, most at 3 mg/kg (n = 70), was used in the danvatirsen population PK analysis. A 2-compartment model with linear elimination described the data well. Covariate analysis revealed ideal body weight was not a significant covariate on the PK of danvatirsen; nor was age, sex or race. The model-based simulation suggested that steady state weekly AUC and Cmax were very similar between 3 mg/kg and 200 mg flat dosing (geometric mean of AUC: 62.5 vs. 63.4 mg h/L and Cmax: 26.2 vs. 26.5 mg/L for two dose groups) with slightly less overall between-subject variability in the flat dosing regimen. The switch to flat dosing was approved by multiple regulatory agencies, including FDA, EMA, PMDA and ANSM. Several ongoing studies have been evaluating flat dosing. Interim analysis from an ongoing study (D5660C00016, NCT03421353) has shown the observed steady state concentration from 200 mg flat dose is in agreement with the model predictions. The population PK model could be further utilized in subsequent exposure-response efficacy and safety modelling.


Assuntos
Neoplasias/tratamento farmacológico , Oligonucleotídeos/administração & dosagem , Oligonucleotídeos/farmacocinética , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacocinética , Peso Corporal/fisiologia , Simulação por Computador , Relação Dose-Resposta a Droga , Feminino , Humanos , Masculino , Taxa de Depuração Metabólica/fisiologia , Pessoa de Meia-Idade , Modelos Biológicos , Neoplasias/metabolismo , Oligonucleotídeos Antissenso/farmacocinética
2.
Nature ; 466(7310): 1115-9, 2010 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-20740013

RESUMO

The activation of pro-inflammatory gene programs by nuclear factor-kappaB (NF-kappaB) is primarily regulated through cytoplasmic sequestration of NF-kappaB by the inhibitor of kappaB (IkappaB) family of proteins. IkappaBbeta, a major isoform of IkappaB, can sequester NF-kappaB in the cytoplasm, although its biological role remains unclear. Although cells lacking IkappaBbeta have been reported, in vivo studies have been limited and suggested redundancy between IkappaBalpha and IkappaBbeta. Like IkappaBalpha, IkappaBbeta is also inducibly degraded; however, upon stimulation by lipopolysaccharide (LPS), it is degraded slowly and re-synthesized as a hypophosphorylated form that can be detected in the nucleus. The crystal structure of IkappaBbeta bound to p65 suggested this complex might bind DNA. In vitro, hypophosphorylated IkappaBbeta can bind DNA with p65 and c-Rel, and the DNA-bound NF-kappaB:IkappaBbeta complexes are resistant to IkappaBalpha, suggesting hypophosphorylated, nuclear IkappaBbeta may prolong the expression of certain genes. Here we report that in vivo IkappaBbeta serves both to inhibit and facilitate the inflammatory response. IkappaBbeta degradation releases NF-kappaB dimers which upregulate pro-inflammatory target genes such as tumour necrosis factor-alpha (TNF-alpha). Surprisingly, absence of IkappaBbeta results in a dramatic reduction of TNF-alpha in response to LPS even though activation of NF-kappaB is normal. The inhibition of TNF-alpha messenger RNA (mRNA) expression correlates with the absence of nuclear, hypophosphorylated-IkappaBbeta bound to p65:c-Rel heterodimers at a specific kappaB site on the TNF-alpha promoter. Therefore IkappaBbeta acts through p65:c-Rel dimers to maintain prolonged expression of TNF-alpha. As a result, IkappaBbeta(-/-) mice are resistant to LPS-induced septic shock and collagen-induced arthritis. Blocking IkappaBbeta might be a promising new strategy for selectively inhibiting the chronic phase of TNF-alpha production during the inflammatory response.


Assuntos
Artrite Experimental/metabolismo , Regulação da Expressão Gênica , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Adjuvantes Imunológicos/farmacologia , Animais , Linhagem Celular , Citocinas/sangue , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos DBA , Camundongos Knockout , Fator de Necrose Tumoral alfa/sangue
3.
Nat Genet ; 33(3): 401-6, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12590264

RESUMO

RNA interference (RNAi) has recently emerged as a specific and efficient method to silence gene expression in mammalian cells either by transfection of short interfering RNAs (siRNAs; ref. 1) or, more recently, by transcription of short hairpin RNAs (shRNAs) from expression vectors and retroviruses. But the resistance of important cell types to transduction by these approaches, both in vitro and in vivo, has limited the use of RNAi. Here we describe a lentiviral system for delivery of shRNAs into cycling and non-cycling mammalian cells, stem cells, zygotes and their differentiated progeny. We show that lentivirus-delivered shRNAs are capable of specific, highly stable and functional silencing of gene expression in a variety of cell types and also in transgenic mice. Our lentiviral vectors should permit rapid and efficient analysis of gene function in primary human and animal cells and tissues and generation of animals that show reduced expression of specific genes. They may also provide new approaches for gene therapy.


Assuntos
Lentivirus/genética , Interferência de RNA , Animais , Sequência de Bases , Antígenos CD8/genética , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Células Cultivadas , Vetores Genéticos , Proteínas de Fluorescência Verde , Proteínas Luminescentes/genética , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Conformação de Ácido Nucleico , RNA/química , RNA/genética , Receptores de Interleucina-2/genética , Células-Tronco
4.
Eur J Immunol ; 41(3): 787-97, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21287546

RESUMO

B-cell-activating factor of the TNF family (BAFF)/BLyS contributes to B-cell homeostasis and function in the periphery. BAFF is expressed as a membrane-bound protein or released by proteolytic cleavage, but the functional importance of this processing event is poorly understood. Mice expressing BAFF with a mutated furin consensus cleavage site, i.e. furin-mutant BAFF (fmBAFF), were not different from BAFF-deficient mice with regard to their B-cell populations and responses to immunization. It is however noteworthy that an alternative processing event releases some soluble BAFF in fmBAFF mice. Mild overexpression (∼ 5-fold) of fmBAFF alone generated intermediate levels of B cells without improving humoral responses to immunization. Processed BAFF was however important for B-cell homeostasis, as peripheral B-cell populations and antibody responses were readily restored by administration of soluble BAFF trimers in BAFF-deficient mice. However, the rescue of CD23 expression in B cells of BAFF-deficient mice required both soluble BAFF trimers and fmBAFF, or a polymeric form of soluble BAFF (BAFF 60-mer). These results point to a predominant role of processed BAFF for B-cell homeostasis and function, and indicate possible accessory roles for membrane-bound BAFF.


Assuntos
Fator Ativador de Células B/genética , Fator Ativador de Células B/metabolismo , Linfócitos B/imunologia , Mutação , Sequência de Aminoácidos , Animais , Formação de Anticorpos , Fator Ativador de Células B/química , Fator Ativador de Células B/deficiência , Sequência de Bases , Sítios de Ligação/genética , Primers do DNA/genética , Furina/química , Células HEK293 , Homeostase , Humanos , Imunoglobulinas/sangue , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Processamento de Proteína Pós-Traducional , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Solubilidade
5.
Biochim Biophys Acta ; 1792(11): 1073-9, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19715759

RESUMO

Polycythemia vera (PV) is a myeloproliferative disorder involving hematopoietic stem cells. A recurrent somatic missense mutation in JAK2 (JAK2V617F) is thought to play a causal role in PV. Therefore, targeting Jak2 will likely provide a molecular mechanism-based therapy for PV. To facilitate the development of such new and specific therapeutics, a suitable and well-characterized preclinical animal model is essential. Although several mouse models of PV have been reported, the spatiotemporal kinetics of PV formation and progression has not been studied. To address this, we created a bone marrow transplant mouse model that co-expresses mutant Jak2 and luciferase 2 (Luc2) genes. Bioluminescent imaging (BLI) was used to visualize disease cells and analyze the kinetics of PV development in vivo. To better understand the molecular mechanism of PV, we generated mice carrying a kinase inactive mutant Jak2 (Jak2K882E), demonstrating that the PV disease was dependent on constitutive activation of the Jak2 kinase activity. We further showed that the Jak2V617F mutation caused increased stem cell renewal activity and impaired cell differentiation, which was at least in part due to deregulated transcriptional programming. The Jak2V617F-Luc2 PV mice will be a useful preclinical model to characterize novel JAK2 inhibitors for the treatment of PV.


Assuntos
Janus Quinase 2/metabolismo , Luciferases/biossíntese , Medições Luminescentes , Policitemia Vera/enzimologia , Policitemia Vera/patologia , Animais , Diferenciação Celular/genética , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/uso terapêutico , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/genética , Luciferases/genética , Camundongos , Camundongos Mutantes , Mutação de Sentido Incorreto , Células NIH 3T3 , Policitemia Vera/tratamento farmacológico , Policitemia Vera/genética , Células-Tronco/enzimologia , Células-Tronco/patologia
6.
J Exp Med ; 198(6): 937-45, 2003 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-12975458

RESUMO

The cellular source of B cell activation factor (BAFF) required for peripheral B cell survival/maturation is unknown. To determine the nature of BAFF-producing cells we established and analyzed reciprocal bone marrow (BM) chimeras with wild-type (WT) and BAFF-deficient mice. The results revealed that BAFF production by radiation-resistant stromal cells is completely sufficient to provide a necessary signal for B cell survival/maturation, as BAFF-/- BM cells transferred into lethally irradiated WT mice gave rise to normal numbers of follicular (FO) and marginal zone (MZ) B cell subpopulations. On the other hand, transfer of WT BM into BAFF-/- lethally irradiated mice resulted only in minimal reconstitution of mature FO B cells and no restoration of MZ B cells. Thus, in the absence of BAFF+/+ stromal cells, BAFF production by BM-derived cells, presumably by macrophages, dendritic cells, and/or neutrophils, was not at all sufficient to support normal B cell homeostasis. Interestingly, immunization of both types of chimeras stimulated high levels of antigen-specific antibody secretion, indicating that either stromal cell- or hematopoietic cell-derived BAFF is sufficient for B cell antibody responses.


Assuntos
Linfócitos B/metabolismo , Homeostase , Proteínas de Membrana/metabolismo , Células Estromais/efeitos da radiação , Fator de Necrose Tumoral alfa/metabolismo , Animais , Fator Ativador de Células B , Células da Medula Óssea/metabolismo , Separação Celular , Sobrevivência Celular , Citometria de Fluxo , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Radiação , Quimera por Radiação , Baço/citologia , Baço/metabolismo , Células Estromais/metabolismo , Fator de Necrose Tumoral alfa/genética
7.
Gastroenterology ; 136(3): 912-23, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19109961

RESUMO

BACKGROUND AND AIMS: Tumor necrosis factor (TNF) superfamily members have attracted attention as new therapeutic targets for treating inflammatory disease. TNF-like weak inducer of apoptosis (TWEAK) is a unique, multifunctional TNF family cytokine that signals through its receptor, fibroblast growth factor-inducible molecule 14 (Fn14). The role of this pathway in the intestine has not been previously reported. METHODS: The 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis model was conducted in TWEAK- or Fn14-deficient mice or in normal mice treated with a TWEAK-blocking monoclonal antibody, and clinical severity, histopathology, immunohistochemistry for cell infiltrates, TWEAK and Fn14, gene expression profiling in the colon, and systemic adaptive immunity were assessed. The effect of TWEAK on colon epithelial cell production of inflammatory mediators was analyzed in vitro. The gamma-irradiation injury model was conducted in TWEAK- or Fn14-deficient mice, and crypt epithelial death was assessed. RESULTS: Colitis severity and histologic scores were significantly reduced by TWEAK pathway deficiency or TWEAK-blocking monoclonal antibody. Neutrophil and macrophage infiltrates, chemokines, cytokines, and matrix metalloproteinase expression were reduced in the TWEAK-deficient colon after TNBS administration; however, systemic adaptive immune responses to trinitrophenyl were not altered. Fn14 is expressed on colon epithelial cells in TNBS colitis, and TWEAK induces epithelial production of pathogenic mediators. TWEAK also regulates intestinal epithelial turnover, as evidenced by reduced epithelial cell death after gamma-irradiation injury in TWEAK and Fn14 knockout mice. CONCLUSIONS: Our studies elucidate a nonredundant TWEAK-intestinal epithelial cell axis and suggest that blocking TWEAK may dampen chronic intestinal inflammation and allow normal epithelial repair.


Assuntos
Colite/metabolismo , Colite/patologia , Mucosa Intestinal/patologia , Receptores do Fator de Necrose Tumoral/metabolismo , Fatores de Necrose Tumoral/metabolismo , Animais , Colite/imunologia , Colo/imunologia , Colo/metabolismo , Colo/patologia , Citocina TWEAK , Modelos Animais de Doenças , Raios gama , Sistema Imunitário/fisiologia , Mediadores da Inflamação/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/patologia , Neutrófilos/patologia , RNA Mensageiro/metabolismo , Receptor de TWEAK , Fatores de Necrose Tumoral/genética , Úlcera/imunologia , Úlcera/metabolismo , Úlcera/patologia
8.
Neuron ; 45(3): 353-9, 2005 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-15694322

RESUMO

Myelin-associated inhibitory factors (MAIFs) are inhibitors of CNS axonal regeneration following injury. The Nogo receptor complex, composed of the Nogo-66 receptor 1 (NgR1), neurotrophin p75 receptor (p75), and LINGO-1, represses axon regeneration upon binding to these myelin components. The limited expression of p75 to certain types of neurons and its temporal expression during development prompted speculation that other receptors are involved in the NgR1 complex. Here, we show that an orphan receptor in the TNF family called TAJ, broadly expressed in postnatal and adult neurons, binds to NgR1 and can replace p75 in the p75/NgR1/LINGO-1 complex to activate RhoA in the presence of myelin inhibitors. In vitro exogenously added TAJ reversed neurite outgrowth caused by MAIFs. Neurons from Taj-deficient mice were more resistant to the suppressive action of the myelin inhibitors. Given the limited expression of p75, the discovery of TAJ function is an important step for understanding the regulation of axonal regeneration.


Assuntos
Inibidores do Crescimento/metabolismo , Proteínas da Mielina/metabolismo , Bainha de Mielina/metabolismo , Regeneração Nervosa/fisiologia , Receptores de Superfície Celular/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Animais , Axônios/fisiologia , Sítios de Ligação/fisiologia , Células CHO , Células COS , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Cricetinae , Proteínas Ligadas por GPI , Ligantes , Substâncias Macromoleculares/metabolismo , Proteínas de Membrana , Camundongos , Camundongos Knockout , Glicoproteína Associada a Mielina/metabolismo , Proteínas do Tecido Nervoso , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Receptor Nogo 1 , Receptor de Fator de Crescimento Neural , Receptores de Fator de Crescimento Neural/metabolismo , Receptores do Fator de Necrose Tumoral/genética , Proteína rhoA de Ligação ao GTP/metabolismo
9.
Nat Neurosci ; 8(6): 745-51, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15895088

RESUMO

The control of myelination by oligodendrocytes in the CNS is poorly understood. Here we show that LINGO-1 is an important negative regulator of this critical process. LINGO-1 is expressed in oligodendrocytes. Attenuation of its function by dominant-negative LINGO-1, LINGO-1 RNA-mediated interference (RNAi) or soluble human LINGO-1 (LINGO-1-Fc) leads to differentiation and increased myelination competence. Attenuation of LINGO-1 results in downregulation of RhoA activity, which has been implicated in oligodendrocyte differentiation. Conversely, overexpression of LINGO-1 leads to activation of RhoA and inhibition of oligodendrocyte differentiation and myelination. Treatment of oligodendrocyte and neuron cocultures with LINGO-1-Fc resulted in highly developed myelinated axons that have internodes and well-defined nodes of Ranvier. The contribution of LINGO-1 to myelination was verified in vivo through the analysis of LINGO-1 knockout mice. The ability to recapitulate CNS myelination in vitro using LINGO-1 antagonists and the in vivo effects seen in the LINGO-1 knockout indicate that LINGO-1 signaling may be critical for CNS myelination.


Assuntos
Sistema Nervoso Central/embriologia , Regulação para Baixo/genética , Bainha de Mielina/metabolismo , Glicoproteína Associada a Mielina/genética , Fibras Nervosas Mielinizadas/metabolismo , Oligodendroglia/metabolismo , Receptores de Superfície Celular/genética , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , Sistema Nervoso Central/crescimento & desenvolvimento , Sistema Nervoso Central/metabolismo , Técnicas de Cocultura , Regulação para Baixo/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Proteínas de Membrana , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Bainha de Mielina/genética , Bainha de Mielina/ultraestrutura , Glicoproteína Associada a Mielina/antagonistas & inibidores , Glicoproteína Associada a Mielina/metabolismo , Fibras Nervosas Mielinizadas/efeitos dos fármacos , Fibras Nervosas Mielinizadas/ultraestrutura , Proteínas do Tecido Nervoso , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/ultraestrutura , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-fyn , Interferência de RNA/efeitos dos fármacos , Interferência de RNA/fisiologia , Nós Neurofibrosos/genética , Nós Neurofibrosos/metabolismo , Nós Neurofibrosos/ultraestrutura , Ratos , Ratos Long-Evans , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Superfície Celular/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Proteína rhoA de Ligação ao GTP/metabolismo , Quinases da Família src/genética , Quinases da Família src/metabolismo
10.
J Clin Endocrinol Metab ; 101(11): 4313-4321, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27459523

RESUMO

CONTEXT: Polycystic ovary syndrome (PCOS), the most common endocrinopathy in women, is characterized by high secretion levels of LH and T. Currently, there is no treatment licensed specifically for PCOS. OBJECTIVE: The objective of this study was to investigate whether a targeted therapy would decrease LH pulse frequency in women with PCOS, subsequently reducing serum LH and T concentrations and thereby presenting a novel therapeutic approach to the management of PCOS. DESIGN: This study is a double-blind, double-dummy, placebo-controlled, phase 2 trial. SETTINGS: University hospitals and private clinical research centers were included. PARTICIPANTS: Women with PCOS aged 18-45 years participated. INTERVENTION: Intervention included AZD4901 (a specific neurokinin-3 [NK3] receptor antagonist) at a dose of 20, 40, or 80 mg/day or matching placebo for 28 days. MAIN OUTCOME MEASURE: Change from baseline in the area under the LH serum concentration-time curve over 8 hours (area under the curve) on day 7 relative to placebo was measured. RESULTS: Of a total 67 randomized patients, 65 were evaluable. On day 7, the following baseline-adjusted changes relative to placebo were observed in patients receiving AZD4901 80 mg/day: 1) a reduction of 52.0% (95% confidence interval [CI], 29.6-67.3%) in LH area under the curve; 2) a reduction of 28.7% (95% CI, 13.9-40.9%) in total T concentration; and 3) a reduction of 3.55 LH pulses/8 hours (95% CI, 2.0-5.1) (all nominal P < .05). CONCLUSIONS: The NK3 receptor antagonist AZD4901 specifically reduced LH pulse frequency and subsequently serum LH and T concentrations, thus presenting NK3 receptor antagonism as a potential approach to treating the central neuroendocrine pathophysiology of PCOS.


Assuntos
Aminoquinolinas/farmacologia , Hormônio Luteinizante/sangue , Avaliação de Resultados em Cuidados de Saúde , Síndrome do Ovário Policístico/tratamento farmacológico , Receptores da Neurocinina-3/antagonistas & inibidores , Sulfonamidas/farmacologia , Testosterona/sangue , Adolescente , Adulto , Aminoquinolinas/administração & dosagem , Método Duplo-Cego , Feminino , Humanos , Hormônio Luteinizante/efeitos dos fármacos , Pessoa de Meia-Idade , Sulfonamidas/administração & dosagem , Adulto Jovem
11.
Cancer Res ; 71(8): 3052-65, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21493594

RESUMO

PDK1 activates AKT suggesting that PDK1 inhibition might suppress tumor development. However, while PDK1 has been investigated intensively as an oncology target, selective inhibitors suitable for in vivo studies have remained elusive. In this study we present the results of in vivo PDK1 inhibition through a universally applicable RNAi approach for functional drug target validation in oncogenic pathway contexts. This approach, which relies on doxycycline-inducible shRNA expression from the Rosa26 locus, is ideal for functional studies of genes like PDK1 where constitutive mouse models lead to strong developmental phenotypes or embryonic lethality. We achieved more than 90% PDK1 knockdown in vivo, a level sufficient to impact physiological functions resulting in hyperinsulinemia and hyperglycemia. This phenotype was reversible on PDK1 reexpression. Unexpectedly, long-term PDK1 knockdown revealed a lack of potent antitumor efficacy in 3 different mouse models of PTEN-deficient cancer. Thus, despite efficient PDK1 knockdown, inhibition of the PI3K pathway was marginal suggesting that PDK1 was not a rate limiting factor. Ex vivo analysis of pharmacological inhibitors revealed that AKT and mTOR inhibitors undergoing clinical development are more effective than PDK1 inhibitors at blocking activated PI3K pathway signaling. Taken together our findings weaken the widely held expectation that PDK1 represents an appealing oncology target.


Assuntos
Neoplasias Experimentais/enzimologia , PTEN Fosfo-Hidrolase/deficiência , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Animais , Técnicas de Silenciamento de Genes , Inativação Gênica , Leucemia Experimental/enzimologia , Leucemia Experimental/genética , Masculino , Camundongos , Camundongos Transgênicos , Neoplasias Experimentais/genética , Proteína Oncogênica v-akt/antagonistas & inibidores , Proteína Oncogênica v-akt/metabolismo , PTEN Fosfo-Hidrolase/genética , Fosforilação , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/genética , Proteínas Serina-Treonina Quinases/genética , Piruvato Desidrogenase Quinase de Transferência de Acetil , Interferência de RNA
12.
Biochem Pharmacol ; 78(4): 382-9, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19413997

RESUMO

Polycythemia vera (PV) is a myeloproliferative disorder characterized by increased red cell mass and splenomegaly in the absence of secondary causes [Tefferi A., Spivak J.L., Polycythemia vera: scientific advances and current practice. Semin Hematol 2005;42(4):206-20.]. Recently, several laboratories have discovered that the vast majority of patients with PV carry a single, activating mutation (V617F) in the pseudokinase domain of Janus kinase 2 (Jak2) [Zhao R, Xing S, Li Z, Fu X, Li Q, Krantz SB, et al., Identification of an acquired JAK2 mutation in polycythemia vera. J Biol Chem 2005;280(24):22788-92; James C, Ugo V, Le Couédic JP, Staerk J, Delhommeau F, Lacout C, et al., A unique clonal JAK2 mutation leading to constitutive signalling causes polycythemia vera. Nature 2005;434(7037):1144-8; Kralovics R, Passamonti F, Buser AS, Teo SS, Tiedt R, Passweg JR, et al., A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med 2005;352(17):1779-90; Levine RL, Wadleigh M, Cools J, Ebert BL, Wernig G, Huntly BJ, et al., Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell 2005;7(4):387-97.]. This discovery has spurred interest in developing therapies for PV via inhibition of Jak2. We induced polycythemia in mice by administering high dose recombinant erythropoietin (Epo) and determined that administration recapitulates almost all of the major and minor diagnostic features of human PV. We then tested a selective, small molecule inhibitor of Jak2 (Jak2i) and showed that this treatment prevents polycythemia. This prevention of polycythemia was accompanied by lower hematocrits, reduced spleen sizes and reductions in Stat5 phosphorylation (pStat5). Surprisingly, Epo rapidly (<1h) induces mobilization of activated erythroid precursors into the blood, thus allowing drug-response relationships to guide discovery. We conclude that inhibition of Jak2 prevents polycythemia in mice, and furthermore present this model as an efficient tool for the discovery of drugs that effectively treat human PV.


Assuntos
Inibidores Enzimáticos/uso terapêutico , Janus Quinase 2/antagonistas & inibidores , Policitemia Vera/fisiopatologia , Policitemia/prevenção & controle , Piridonas/uso terapêutico , Animais , Proliferação de Células , Inibidores Enzimáticos/farmacologia , Células Precursoras Eritroides , Humanos , Janus Quinase 2/metabolismo , Camundongos , Fosforilação , Mielofibrose Primária/fisiopatologia , Proteínas Tirosina Quinases/metabolismo , Piridonas/síntese química , Piridonas/química , Transdução de Sinais , Trombocitemia Essencial , Células Tumorais Cultivadas
13.
Blood ; 111(3): 1004-12, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17942754

RESUMO

The cytokine BAFF binds to the receptors TACI, BCMA, and BAFF-R on B cells, whereas APRIL binds to TACI and BCMA only. The signaling properties of soluble trimeric BAFF (BAFF 3-mer) were compared with those of higher-order BAFF oligomers. All forms of BAFF bound BAFF-R and TACI, and elicited BAFF-R-dependent signals in primary B cells. In contrast, signaling through TACI in mature B cells or plasmablasts was only achieved by higher-order BAFF and APRIL oligomers, all of which were also po-tent activators of a multimerization-dependent reporter signaling pathway. These results indicate that, although BAFF-R and TACI can provide B cells with similar signals, only BAFF-R, but not TACI, can respond to soluble BAFF 3-mer, which is the main form of BAFF found in circulation. BAFF 60-mer, an efficient TACI agonist, was also detected in plasma of BAFF transgenic and nontransgenic mice and was more than 100-fold more active than BAFF 3-mer for the activation of multimerization-dependent signals. TACI supported survival of activated B cells and plasmablasts in vitro, providing a rational basis to explain the immunoglobulin deficiency reported in TACI-deficient persons.


Assuntos
Fator Ativador de Células B/metabolismo , Linfócitos B/citologia , Linfócitos B/metabolismo , Ativação Linfocitária , Proteína Transmembrana Ativadora e Interagente do CAML/metabolismo , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos/imunologia , Formação de Anticorpos/imunologia , Fator Ativador de Células B/química , Fator Ativador de Células B/genética , Receptor do Fator Ativador de Células B/imunologia , Receptor do Fator Ativador de Células B/metabolismo , Linfócitos B/imunologia , Linhagem Celular , Proliferação de Células , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Imunoglobulinas/biossíntese , Imunoglobulinas/imunologia , Ligantes , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Dados de Sequência Molecular , Alinhamento de Sequência , Transdução de Sinais , Baço/imunologia , Proteína Transmembrana Ativadora e Interagente do CAML/deficiência , Proteína Transmembrana Ativadora e Interagente do CAML/genética , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética , Regulação para Cima
14.
J Exp Med ; 205(5): 1049-62, 2008 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-18411337

RESUMO

T helper type 17 (Th17) cells play an important pathogenic function in autoimmune diseases; their regulation, however, is not well understood. We show that the expression of a tumor necrosis factor receptor family member, death receptor 3 (DR3; also known as TNFRSF25), is selectively elevated in Th17 cells, and that TL1A, its cognate ligand, can promote the proliferation of effector Th17 cells. To further investigate the role of the TL1A-DR3 pathway in Th17 regulation, we generated a TL1A-deficient mouse and found that TL1A(-/-) dendritic cells exhibited a reduced capacity in supporting Th17 differentiation and proliferation. Consistent with these data, TL1A(-/-) animals displayed decreased clinical severity in experimental autoimmune encephalomyelitis (EAE). Finally, we demonstrated that during EAE disease progression, TL1A was required for the optimal differentiation as well as effector function of Th17 cells. These observations thus establish an important role of the TL1A-DR3 pathway in promoting Th17 cell function and Th17-mediated autoimmune disease.


Assuntos
Doenças Autoimunes/imunologia , Encefalomielite Autoimune Experimental/imunologia , Antígeno HLA-DR3/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/deficiência , Animais , Encéfalo/imunologia , Encéfalo/patologia , Diferenciação Celular , Divisão Celular , Citocinas/metabolismo , Primers do DNA , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Medula Espinal/imunologia , Medula Espinal/patologia , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia
15.
J Immunol ; 178(7): 4342-50, 2007 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-17371991

RESUMO

The T cell, Ig domain, and mucin domain-1 (TIM-1) gene is associated with Th2 T cell responses and human atopic diseases. The mechanism by which TIM-1 influences T cell responses remains unknown. We demonstrate that TIM-1 is recruited to the TCR-signaling complex via association with CD3. TIM-1 up-regulates TCR-associated signaling events, including phosphorylation of Zap70 and IL-2-inducible T cell kinase. This activity requires TIM-1 tyrosine phosphorylation. TIM-1 expression induces formation of a novel complex that includes PI3K and ITK. Finally, the consequences of TIM-1 activation include increased expression of effector cytokines. These results demonstrate that TIM-1 is a critical component of the human T cell response and provide a mechanistic hypothesis for the role of TIM-1 in disease.


Assuntos
Complexo CD3/metabolismo , Ativação Linfocitária , Glicoproteínas de Membrana/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores Virais/metabolismo , Linfócitos T/imunologia , Complexo CD3/análise , Células Cultivadas , Citocinas/metabolismo , Receptor Celular 1 do Vírus da Hepatite A , Humanos , Glicoproteínas de Membrana/análise , Glicoproteínas de Membrana/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Tirosina Quinases/metabolismo , Receptores Virais/análise , Receptores Virais/genética , Linfócitos T/química , Tirosina/genética , Tirosina/metabolismo , Regulação para Cima , Proteína-Tirosina Quinase ZAP-70/metabolismo
16.
J Immunol ; 179(6): 3872-80, 2007 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-17785824

RESUMO

Loss of Bruton's tyrosine kinase (Btk) function results in mouse Xid disease characterized by a reduction in mature B cells and impaired humoral immune responses. These defects have been mainly attributed to impaired BCR signaling including reduced activation of the classical NF-kappaB pathway. In this study we show that Btk also couples the receptor for B cell-activating factor (BAFF) of the TNF family (BAFF-R) to the NF-kappaB pathway. Loss of Btk results in defective BAFF-mediated activation of both classical and alternative NF-kappaB pathways. Btk appears to regulate directly the classical pathway in response to BAFF such that Btk-deficient B cells exhibit reduced kinase activity of IkappaB kinase gamma-containing complexes and defective IkappaBalpha degradation. In addition, Btk-deficient B cells produce reduced levels of NF-kappaB2 (p100) basally and in response to stimulation via the BCR or BAFF-R, resulting in impaired activation of the alternative NF-kappaB pathway by BAFF. These results suggest that Btk regulates B cell survival by directly regulating the classical NF-kappaB pathway under both BCR and BAFF-R, as well as by inducing the expression of the components of alternative pathway for sustained NF-kappaB activation in response BAFF. Thus, impaired BCR- and BAFF-induced signaling to NF-kappaB may contribute to the observed defects in B cell survival and humoral immune responses in Btk-deficient mice.


Assuntos
Linfócitos B/enzimologia , NF-kappa B/metabolismo , Proteínas Tirosina Quinases/fisiologia , Tirosina Quinase da Agamaglobulinemia , Animais , Receptor do Fator Ativador de Células B/deficiência , Receptor do Fator Ativador de Células B/genética , Receptor do Fator Ativador de Células B/fisiologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos B/patologia , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Proteínas Tirosina Quinases/deficiência , Proteínas Tirosina Quinases/genética , Receptores de Antígenos de Linfócitos B/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia
17.
J Immunol ; 178(4): 2249-61, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-17277130

RESUMO

The TAPR locus containing the TIM gene family is implicated in the development of atopic inflammation in mouse, and TIM-1 allelic variation has been associated with the incidence of atopy in human patient populations. In this study, we show that manipulation of the TIM-1 pathway influences airway inflammation and pathology. Anti-TIM-1 mAbs recognizing distinct epitopes differentially modulated OVA-induced lung inflammation in the mouse. The epitopes recognized by these Abs were mapped, revealing that mAbs to both the IgV and stalk domains of TIM-1 have therapeutic activity. Unexpectedly, mAbs recognizing unique epitopes spanning exon 4 of the mucin/stalk domains exacerbated immune responses. Using Ag recall response studies, we demonstrate that the TIM-1 pathway acts primarily by modulating the production of T(H)2 cytokines. Furthermore, ex vivo cellular experiments indicate that TIM-1 activity controls CD4(+) T cell activity. These studies validate the genetic hypothesis that the TIM-1 locus is linked to the development of atopic disease and suggest novel therapeutic strategies for targeting asthma and other atopic disorders.


Assuntos
Anticorpos Monoclonais/farmacologia , Epitopos/imunologia , Proteínas de Membrana/imunologia , Pneumonia/imunologia , Células Th2/imunologia , Animais , Anticorpos Monoclonais/imunologia , Asma/tratamento farmacológico , Asma/genética , Asma/imunologia , Asma/patologia , Células Cultivadas , Citocinas/imunologia , Mapeamento de Epitopos , Epitopos/genética , Feminino , Humanos , Pulmão/imunologia , Pulmão/patologia , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina/imunologia , Ovalbumina/toxicidade , Pneumonia/tratamento farmacológico , Pneumonia/genética , Pneumonia/patologia , Estrutura Terciária de Proteína/genética , Locos de Características Quantitativas/genética , Locos de Características Quantitativas/imunologia , Células Th2/patologia
18.
J Immunol ; 177(4): 2671-80, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16888029

RESUMO

Constitutive overexpression of B cell-activating factor belonging to the TNF family (BAFF) promotes development of systemic lupus erythematosus (SLE), and treatment of SLE mice with BAFF antagonists ameliorates disease. To determine whether SLE can develop de novo in BAFF-deficient hosts, BAFF-deficient New Zealand Mixed (NZM) 2328 (NZM.Baff(-/-)) mice were generated. In NZM.Baff(-/-) mice, spleen B cells (including CD5(+) B1a and CD5(-) B1b B cells), germinal centers, Ig-secreting cells, and T cells were reduced in comparison to NZM.Baff(+/+) mice. Serum total Ig and autoantibody levels were reduced at 4-6 mo but approached wild-type levels with increasing age, indicating that autoreactive B cells can survive and secrete autoantibodies despite the complete absence of BAFF. At least some of these autoantibodies are nephrophilic in that glomerular deposition of total IgG and IgG1 (but not of IgG2a, IgG2b, or C3) was substantial in NZM.Baff(-/-) mice by 12-13 mo of age. Despite proliferative glomerulonephritis, highlighted by widespread glomerular hyaline thrombi, being common among NZM.Baff(-/-) mice by 6-7 mo of age, severe proteinuria and mortality were greatly attenuated. These results demonstrate that the lifelong absence of BAFF does not protect NZM 2328 mice from serological autoimmunity and renal pathology. Nevertheless, the character of the renal pathology is altered, and the mice are largely spared from clinically overt disease (severe proteinuria and premature death). These observations may have profound ramifications for the use of BAFF antagonists in human SLE and related diseases.


Assuntos
Autoanticorpos/sangue , Predisposição Genética para Doença , Rim/imunologia , Rim/patologia , Nefrite Lúpica/genética , Nefrite Lúpica/imunologia , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Fator de Necrose Tumoral alfa/deficiência , Fator de Necrose Tumoral alfa/genética , Animais , Autoanticorpos/biossíntese , Fator Ativador de Células B , Feminino , Nefrite Lúpica/mortalidade , Nefrite Lúpica/patologia , Masculino , Proteínas de Membrana/sangue , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NZB , Camundongos Knockout
19.
EMBO J ; 25(24): 5826-39, 2006 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-17124496

RESUMO

Inflammation participates in tissue repair through multiple mechanisms including directly regulating the cell fate of resident progenitor cells critical for successful regeneration. Upon surveying target cell types of the TNF ligand TWEAK, we observed that TWEAK binds to all progenitor cells of the mesenchymal lineage and induces NF-kappaB activation and the expression of pro-survival, pro-proliferative and homing receptor genes in the mesenchymal stem cells, suggesting that this pro-inflammatory cytokine may play an important role in controlling progenitor cell biology. We explored this potential using both the established C2C12 cell line and primary mouse muscle myoblasts, and demonstrated that TWEAK promoted their proliferation and inhibited their terminal differentiation. By generating mice deficient in the TWEAK receptor Fn14, we further showed that Fn14-deficient primary myoblasts displayed significantly reduced proliferative capacity and altered myotube formation. Following cardiotoxin injection, a known trigger for satellite cell-driven skeletal muscle regeneration, Fn14-deficient mice exhibited reduced inflammatory response and delayed muscle fiber regeneration compared with wild-type mice. These results indicate that the TWEAK/Fn14 pathway is a novel regulator of skeletal muscle precursor cells and illustrate an important mechanism by which inflammatory cytokines influence tissue regeneration and repair. Coupled with our recent demonstration that TWEAK potentiates liver progenitor cell proliferation, the expression of Fn14 on all mesenchymal lineage progenitor cells supports a broad involvement of this pathway in other tissue injury and disease settings.


Assuntos
Células-Tronco Mesenquimais/citologia , Músculo Esquelético/fisiologia , Receptores do Fator de Necrose Tumoral/metabolismo , Regeneração , Fatores de Necrose Tumoral/metabolismo , Animais , Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Proteínas Cardiotóxicas de Elapídeos/farmacologia , Citocina TWEAK , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Inflamação , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Modelos Biológicos , Desenvolvimento Muscular/efeitos dos fármacos , Músculo Esquelético/citologia , Músculo Esquelético/efeitos dos fármacos , Mioblastos/citologia , Mioblastos/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores do Fator de Necrose Tumoral/deficiência , Receptores do Fator de Necrose Tumoral/genética , Regeneração/efeitos dos fármacos , Receptor de TWEAK , Fatores de Necrose Tumoral/genética
20.
J Immunol ; 175(1): 319-28, 2005 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15972664

RESUMO

DeltaBAFF is a novel splicing isoform of the regulator B cell-activating factor (BAFF, BLyS), a TNF family protein with powerful immunoregulatory effects. Overexpression of BAFF leads to excessive B cell accumulation, activation, autoantibodies, and lupus-like disease, whereas an absence of BAFF causes peripheral B cell immunodeficiency. Based on the ability of DeltaBAFF to multimerize with full-length BAFF and to limit BAFF proteolytic shedding from the cell surface, we previously proposed a role for DeltaBAFF in restraining the effects of BAFF and in regulating B lymphocyte homeostasis. To test these ideas we generated mice transgenic for DeltaBAFF under the control of human CD68 regulatory elements, which target expression to myeloid and dendritic cells. We also generated in parallel BAFF transgenic mice using the same expression elements. Analysis of the transgenic mice revealed that DeltaBAFF and BAFF had opposing effects on B cell survival and marginal zone B cell numbers. DeltaBAFF transgenic mice had reduced B cell numbers and T cell-dependent Ab responses, but normal preimmune serum Ig levels. In contrast, BAFF transgenic mice had extraordinarily elevated Ig levels and increases in subsets of B cells. Unexpectedly, both BAFF and DeltaBAFF appeared to modulate the numbers of B-1 phenotype B cells.


Assuntos
Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/genética , Processamento Alternativo , Animais , Antígenos CD/genética , Antígenos de Diferenciação Mielomonocítica/genética , Fator Ativador de Células B , Subpopulações de Linfócitos B/citologia , Subpopulações de Linfócitos B/imunologia , Sequência de Bases , DNA Complementar/genética , Feminino , Humanos , Imunoglobulinas/sangue , Masculino , Proteínas de Membrana/deficiência , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Linfócitos T/imunologia , Fator de Necrose Tumoral alfa/deficiência , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA