Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Am J Respir Crit Care Med ; 206(12): 1463-1479, 2022 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-35998281

RESUMO

Rationale: Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and often fatal disorder. Two U.S. Food and Drug Administration-approved antifibrotic drugs, nintedanib and pirfenidone, slow the rate of decline in lung function, but responses are variable and side effects are common. Objectives: Using an in silico data-driven approach, we identified a robust connection between the transcriptomic perturbations in IPF disease and those induced by saracatinib, a selective Src kinase inhibitor originally developed for oncological indications. Based on these observations, we hypothesized that saracatinib would be effective at attenuating pulmonary fibrosis. Methods: We investigated the antifibrotic efficacy of saracatinib relative to nintedanib and pirfenidone in three preclinical models: 1) in vitro in normal human lung fibroblasts; 2) in vivo in bleomycin and recombinant Ad-TGF-ß (adenovirus transforming growth factor-ß) murine models of pulmonary fibrosis; and 3) ex vivo in mice and human precision-cut lung slices from these two murine models as well as patients with IPF and healthy donors. Measurements and Main Results: In each model, the effectiveness of saracatinib in blocking fibrogenic responses was equal or superior to nintedanib and pirfenidone. Transcriptomic analyses of TGF-ß-stimulated normal human lung fibroblasts identified specific gene sets associated with fibrosis, including epithelial-mesenchymal transition, TGF-ß, and WNT signaling that was uniquely altered by saracatinib. Transcriptomic analysis of whole-lung extracts from the two animal models of pulmonary fibrosis revealed that saracatinib reverted many fibrogenic pathways, including epithelial-mesenchymal transition, immune responses, and extracellular matrix organization. Amelioration of fibrosis and inflammatory cascades in human precision-cut lung slices confirmed the potential therapeutic efficacy of saracatinib in human lung fibrosis. Conclusions: These studies identify novel Src-dependent fibrogenic pathways and support the study of the therapeutic effectiveness of saracatinib in IPF treatment.


Assuntos
Fibrose Pulmonar Idiopática , Inibidores de Proteínas Quinases , Animais , Humanos , Camundongos , Bleomicina/efeitos adversos , Fibroblastos/metabolismo , Fibrose , Fibrose Pulmonar Idiopática/tratamento farmacológico , Pulmão/patologia , Inibidores de Proteínas Quinases/uso terapêutico , Quinases da Família src/metabolismo , Fator de Crescimento Transformador beta/metabolismo
2.
Mol Ther ; 21(9): 1727-37, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23857231

RESUMO

Immune responses directed against viral capsid proteins constitute a main safety concern in the use of adeno-associated virus (AAV) as gene transfer vectors in humans. Pharmacological immunosuppression has been proposed as a solution to the problem; however, the approach suffers from several potential limitations. Using MHC class II epitopes initially identified within human IgG, named Tregitopes, we showed that it is possible to modulate CD8+ T cell responses to several viral antigens in vitro. We showed that incubation of peripheral blood mononuclear cells with these epitopes triggers proliferation of CD4+CD25+FoxP3+ T cells that suppress killing of target cells loaded with MHC class I antigens in an antigen-specific fashion, through a mechanism that seems to require cell-to-cell contact. Expression of a construct encoding for the AAV capsid structural protein fused to Tregitopes resulted in reduction of CD8+ T cell reactivity against the AAV capsid following immunization with an adenoviral vector expressing capsid. This was accompanied by an increase in frequency of CD4+CD25+FoxP3+ T cells in spleens and lower levels of inflammatory infiltrates in injected tissues. This proof-of-concept study demonstrates modulation of CD8+ T cell reactivity to an antigen using regulatory T cell epitopes is possible.


Assuntos
Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/imunologia , Proteínas do Capsídeo/imunologia , Dependovirus/imunologia , Epitopos de Linfócito T/imunologia , Vetores Genéticos , Imunoglobulina G/imunologia , Animais , Antígenos Virais/genética , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Capsídeo/imunologia , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Células Cultivadas , Dependovirus/genética , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/metabolismo , Terapia Genética , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Imunoglobulina G/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Baço/imunologia , Linfócitos T Reguladores/imunologia
3.
Clin Immunol ; 149(3): 534-55, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24263283

RESUMO

Protein therapeutics hold a prominent and rapidly expanding place among medicinal products. Purified blood products, recombinant cytokines, growth factors, enzyme replacement factors, monoclonal antibodies, fusion proteins, and chimeric fusion proteins are all examples of therapeutic proteins that have been developed in the past few decades and approved for use in the treatment of human disease. Despite early belief that the fully human nature of these proteins would represent a significant advantage, adverse effects associated with immune responses to some biologic therapies have become a topic of some concern. As a result, drug developers are devising strategies to assess immune responses to protein therapeutics during both the preclinical and the clinical phases of development. While there are many factors that contribute to protein immunogenicity, T cell- (thymus-) dependent (Td) responses appear to play a critical role in the development of antibody responses to biologic therapeutics. A range of methodologies to predict and measure Td immune responses to protein drugs has been developed. This review will focus on the Td contribution to immunogenicity, summarizing current approaches for the prediction and measurement of T cell-dependent immune responses to protein biologics, discussing the advantages and limitations of these technologies, and suggesting a practical approach for assessing and mitigating Td immunogenicity.


Assuntos
Produtos Biológicos/imunologia , Imunidade Celular/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Formação de Anticorpos , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Bioensaio , Produtos Biológicos/administração & dosagem , Biomarcadores Farmacológicos/análise , Citocinas/administração & dosagem , Citocinas/imunologia , Avaliação Pré-Clínica de Medicamentos , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/administração & dosagem , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Simulação de Acoplamento Molecular , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia , Linfócitos T/imunologia
4.
J Clin Immunol ; 33 Suppl 1: S43-9, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22941509

RESUMO

Tregitopes are regulatory T cell epitopes derived from immunoglobulin G (IgG) that stimulate CD25(+) FoxP3(+) T cells to expand. In conjunction with these Tregs, Tregitopes can prevent, treat, and even cure autoimmune disease in mouse models, suppress allo-specific responses in murine transplant models, inhibit CD8(+) T cell responses to recombinant adeno-associated virus (AAV) gene transfer vectors, and induce adaptive Tregs in DO11.10 mice. In this review of recent Tregitope studies, we summarize their effects in vitro and describe recent comparisons between intravenous IgG (IVIG) and Tregitopes in standard in vivo immune tolerance models. Further investigations of the mechanism of action of Tregitopes in the preclinical models described here will lead to clinical trials where Tregitopes may have the potential to alter the treatment of autoimmune disease, transplantation, and allergy, and to improve the efficiency of gene and protein replacement therapies.


Assuntos
Epitopos de Linfócito T/imunologia , Imunoglobulina G/imunologia , Linfócitos T Reguladores/imunologia , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/terapia , Autoimunidade , Humanos , Tolerância Imunológica , Imunoglobulinas Intravenosas/imunologia , Imunoglobulinas Intravenosas/uso terapêutico , Pesquisa/tendências
5.
Biologicals ; 40(5): 364-8, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22770604

RESUMO

Protein therapeutics occupy a very significant position in the biopharmaceutical market. In addition to the preclinical, clinical and post marketing challenges common to other drugs, unwanted immunogenicity is known to affect efficacy and/or safety of most biotherapeutics. A standard set of immunogenicity risk factors are routinely used to inform monitoring strategies in clinical studies. A number of in-silico, in vivo and in vitro approaches have also been employed to predict immunogenicity of biotherapeutics, but with limited success. Emerging data also indicates the role of immune tolerance mechanisms and impact of several product-related factors on modulating host immune responses. Thus, a comprehensive discussion of the impact of innate and adaptive mechanisms and molecules involved in induction of host immune responses on immunogenicity of protein therapeutics is needed. A detailed understanding of these issues is essential in order to fully exploit the therapeutic potential of this class of drugs. This Roundtable Session was designed to provide a common platform for discussing basic immunobiological and pharmacological issues related to the role of biotherapeutic-associated risk factors, as well as host immune system in immunogenicity against protein therapeutics. The session included overview presentations from three speakers, followed by a panel discussion with audience participation.


Assuntos
Imunidade Adaptativa/efeitos dos fármacos , Proteínas/imunologia , Proteínas/uso terapêutico , Imunidade Adaptativa/fisiologia , Animais , Anticorpos/análise , Anticorpos/imunologia , Formação de Anticorpos/efeitos dos fármacos , Produtos Biológicos/imunologia , Produtos Biológicos/uso terapêutico , Antagonismo de Drogas , Composição de Medicamentos/efeitos adversos , Contaminação de Medicamentos , Humanos , Imunoglobulinas/química , Imunoglobulinas/imunologia , Imunoglobulinas/uso terapêutico , Camundongos , Preparações Farmacêuticas , Controle de Qualidade , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/uso terapêutico
6.
Sci Rep ; 11(1): 20827, 2021 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-34675338

RESUMO

Non-alcoholic steatohepatitis (NASH) is a rising health challenge, with no approved drugs. We used a computational drug repositioning strategy to uncover a novel therapy for NASH, identifying a GABA-B receptor agonist, AZD3355 (Lesogaberan) previously evaluated as a therapy for esophageal reflux. AZD3355's potential efficacy in NASH was tested in human stellate cells, human precision cut liver slices (hPCLS), and in vivo in a well-validated murine model of NASH. In human stellate cells AZD3355 significantly downregulated profibrotic gene and protein expression. Transcriptomic analysis of these responses identified key regulatory nodes impacted by AZD3355, including Myc, as well as MAP and ERK kinases. In PCLS, AZD3355 down-regulated collagen1α1, αSMA and TNF-α mRNAs as well as secreted collagen1α1. In vivo, the drug significantly improved histology, profibrogenic gene expression, and tumor development, which was comparable to activity of obeticholic acid in a robust mouse model of NASH, but awaits further testing to determine its relative efficacy in patients. These data identify a well-tolerated clinical stage asset as a novel candidate therapy for human NASH through its hepatoprotective, anti-inflammatory and antifibrotic mechanisms of action. The approach validates computational methods to identify novel therapies in NASH in uncovering new pathways of disease development that can be rapidly translated into clinical trials.


Assuntos
Reposicionamento de Medicamentos , Agonistas dos Receptores de GABA-B/uso terapêutico , Fígado/efeitos dos fármacos , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Ácidos Fosfínicos/uso terapêutico , Propilaminas/uso terapêutico , Adulto , Idoso , Animais , Linhagem Celular , Modelos Animais de Doenças , Feminino , Agonistas dos Receptores de GABA-B/farmacologia , Humanos , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Ácidos Fosfínicos/farmacologia , Propilaminas/farmacologia
7.
Clin Cancer Res ; 12(2): 569-76, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16428502

RESUMO

PURPOSE: Cancer immunotherapy has been limited by anergy of patient T cells, inadequate numbers of precursor tumor-specific CTL, and difficulty in producing therapeutic doses of CTL. To overcome these limitations, bispecific antibodies have been used to create artificial antibody receptors that direct polyclonal activated T cells (ATC) to target tumor antigens. Studies reported herein were designed to characterize bispecific antibody-armed ATC functions during multiple rounds of targeted cell stimulation. EXPERIMENTAL DESIGN: ATCs were generated from human peripheral blood mononuclear cells (PBMC) by culture with anti-CD3 and interleukin 2 for 14 days and armed with anti-CD3 x anti-Her2 bispecific antibody (Her2Bi). In vitro, Her2Bi-armed ATC were examined for a range of functions after repeated stimulation with the Her2/neu-expressing breast cancer cell line SK-BR-3. PBMC isolated from cancer patients treated with Her2Bi-armed ATC were tested ex vivo for cytotoxicity against SK-BR-3. RESULTS: In vitro, armed ATC divided, maintained surface Her2Bi, and expressed a range of activities for extended periods of time. Perforin-mediated cytotoxic activity by armed ATC continued for at least 336 hours, and cytokines and chemokines (i.e., IFN-gamma and regulated on activation, normal T-cell expressed and secreted protein [RANTES]) were secreted during successive rounds of stimulation. Furthermore, PBMC isolated from patients over their courses of immunotherapy exhibited significant cytolytic activity against SK-BR-3 as a function of Her2Bi-armed ATC infusions. CONCLUSIONS: These studies show that armed ATC are specific, durable, and highly functional T-cell populations in vitro. These previously unappreciated broad and long-term functions of armed ATC are encouraging for their therapeutic use in treating cancer.


Assuntos
Anticorpos Biespecíficos/farmacologia , Neoplasias da Mama/terapia , Complexo CD3/imunologia , Citotoxicidade Imunológica , Receptor ErbB-2/imunologia , Linfócitos T/imunologia , Neoplasias da Mama/imunologia , Linhagem Celular Tumoral , Quimiocina CCL5/metabolismo , Feminino , Humanos , Imunoterapia , Interferon gama/metabolismo , Interleucina-2/imunologia , Ativação Linfocitária , Glicoproteínas de Membrana/farmacologia , Perforina , Proteínas Citotóxicas Formadoras de Poros
8.
Cancer Chemother Pharmacol ; 54(3): 249-58, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15173956

RESUMO

Taurolidine (TRD) was designed in the 1970s as a broad-spectrum antibiotic and is used clinically at high doses without systemic toxicity. We have found that this agent possesses cytotoxic activity in human tumor cell lines and antineoplastic activity in mice bearing i.p. human tumor xenografts. We now report the mechanism by which TRD induces cell death in DU145 human prostate tumor cells. The IC50 (3 days) of TRD in this model was 16.8+/-1.1 microM. Cytotoxicity was associated with DNA debris and increased membrane phosphatidylserine externalization, both suggesting the induction of apoptosis. This was confirmed by the ability of TRD to induce PARP cleavage in these cells, an effect prevented by coexposure to the pan-caspase inhibitor zVAD-FMK. TRD exposure also resulted in the appearance of cytochrome c in the cytoplasm, procaspase 9 activation within 2 h of drug exposure and procaspase 8 activation 4 h after exposure. Parallel experiments revealed that cytochrome c appearance in the cytoplasm was not blocked by preexposure to zVAD-FMK, while activation of both procaspase 9 and procaspase 8 was prevented. Finally, antineoplastic activity was assessed in mice bearing subcutaneous xenografts of DU145 cells. Initial studies quantitated the toxicity of three i.p. injections of TRD, administered as one injection on three alternate days per week, at doses ranging from 500 to 700 mg/kg per injection. The 500 mg/kg dose produced about 7% mortality after three cycles and effectively inhibited tumor growth. Thus, TRD induced mitochondrial-mediated apoptosis in DU145 human prostate tumor cells and this effect could be exploited for therapeutic advantage.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Próstata/tratamento farmacológico , Taurina/análogos & derivados , Taurina/farmacologia , Tiadiazinas/farmacologia , Animais , Antineoplásicos/administração & dosagem , Humanos , Injeções Intraperitoneais , Masculino , Camundongos , Camundongos Nus , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Neoplasias Experimentais , Neoplasias da Próstata/patologia , Neoplasias da Próstata/veterinária , Taurina/administração & dosagem , Tiadiazinas/administração & dosagem , Transplante Heterólogo , Células Tumorais Cultivadas
9.
Autoimmun Rev ; 12(3): 436-43, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22944299

RESUMO

In the course of screening immunoglobulin G (IgG) sequences for T cell epitopes, we identified novel Treg epitope peptides, now called Tregitopes, contained in the highly conserved framework regions of Fab and Fc. Tregitopes may provide one explanation for the expansion and stimulation of Treg cells following intravenous immunoglobulin (IVIg) therapy. Their distinguishing characteristics include in silico signatures that suggest high-affinity binding to multiple human HLA class II DR and conservation across IgG isotypes and mammalian species with only minor amino acid modifications. Tregitopes induce expansion of CD4(+)/CD25(hi)/FoxP3(+) T cells and suppress immune responses to co-incubated antigens in vitro. By comparing the human IgG Tregitopes (hTregitopes 167 and 289, located in the IgG CH1 and CH2 domains) and Fab to murine sequences, we identified class II-restricted murine Tregitope homologs (mTregitopes). In vivo, mTregitopes suppress inflammation and reproducibly induce Tregs to expand. In vitro studies suggest that the Tregitope mechanism of action is to induce Tregs to respond, leading to production of regulatory signals, followed by modulation of dendritic cell phenotype. The identification of Treg epitopes in IgG suggests that additional Tregitopes may also be present in other autologous proteins; methods for identifying and validating such peptides are described here. The discovery of Tregitopes in IgG and other autologous proteins may lead to the development of new insights as to the role of Tregs in autoimmune diseases.


Assuntos
Epitopos de Linfócito T/imunologia , Imunoglobulinas Intravenosas/imunologia , Linfócitos T Reguladores/imunologia , Animais , Mapeamento de Epitopos , Epitopos de Linfócito T/metabolismo , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Imunoglobulinas Intravenosas/metabolismo , Camundongos , Ligação Proteica
10.
J Diabetes Res ; 2013: 621693, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23710469

RESUMO

HLA class II-restricted regulatory T cell (Treg) epitopes in IgG (also called "Tregitopes") have been reported to suppress immune responses to coadministered antigens by stimulating the expansion of natural Tregs (nTregs). Here we evaluate their impact on human immune responses to islet cell antigens ex vivo and on the modulation of type 1 diabetes (T1D) in a murine model in vivo. Co-administration of Tregitopes and T1D antigens delayed development of hyperglycemia and reduced the incidence of diabetes in NOD mice. Suppression of diabetes could be observed even following onset of disease. To measure the impact of Tregitope treatment on T cell responses, we evaluated the effect of Tregitope treatment in DO11.10 mice. Upregulation of FoxP3 in KJ1-26-stained OVA-specific CD4(+) T cells was observed following treatment of DO11.10 mice with Tregitopes, along with reductions in anti-OVA Ig and T effector responses. In ex vivo studies of human T cells, peripheral blood mononuclear cells' (PBMC) responses to GAD65 epitopes in the presence and absence of Tregitope were variable. Suppression of immune responses to GAD65 epitopes ex vivo by Tregitope appeared to be more effective in assays using PBMC from a newly diagnosed diabetic subject than for other more established diabetic subjects, and correlation of the degree of suppression with predicted HLA restriction of the Tregitopes was confirmed. Implementation of these defined regulatory T cell epitopes for therapy of T1D and other autoimmune diseases may lead to a paradigm shift in disease management.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Epitopos de Linfócito T/imunologia , Tolerância Imunológica/imunologia , Imunoglobulina G/imunologia , Linfócitos T Reguladores/imunologia , Imunidade Adaptativa/imunologia , Alelos , Animais , Proliferação de Células , Diabetes Mellitus Tipo 1/genética , Antígeno HLA-DR4/genética , Antígeno HLA-DR4/imunologia , Humanos , Imunofenotipagem , Camundongos , Camundongos Endogâmicos NOD
11.
Hum Vaccin Immunother ; 8(10): 1459-64, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23095864

RESUMO

Babies born with Pompe disease require life-long treatment with enzyme-replacement therapy (ERT). Despite the human origin of the therapy, recombinant human lysosomal acid α glucosidase (GAA, rhGAA), ERT unfortunately leads to the development of high titers of anti-rhGAA antibody, decreased effectiveness of ERT, and a fatal outcome for a significant number of children who have Pompe disease. The severity of disease, anti-drug antibody (ADA) development, and the consequences thereof are directly related to the degree of the enzyme deficiency. Babies born with a complete deficiency GAA are said to have cross-reactive immunologic material (CRIM)-negative Pompe disease and are highly likely to develop GAA ADA. Less frequently, GAA ADA develop in CRIM-positive individuals. Currently, GAA-ADA sero-positive babies are treated with a combination of immunosuppressive drugs to induce immunological tolerance to ERT, but the long-term effect of these regimens is unknown. Alternative approaches that might redirect the immune response toward antigen-specific tolerance without immunosuppressive agents are needed. Methods leading to the induction of antigen-specific regulatory T cells (Tregs), using peptides such as Tregitopes (T regulatory cell epitopes) are under consideration for the future treatment of CRIM-negative Pompe disease. Tregitopes are natural T cell epitopes derived from immunoglobulin G (IgG) that cause the expansion and activation of regulatory T cells (Treg). Teaching the immune system to tolerate GAA by co-delivering GAA with Tregitope peptides might dramatically improve the lives of CRIM-negative babies and could be applied to other enzyme replacement therapies to which ADA have been induced.


Assuntos
Terapia de Reposição de Enzimas/métodos , Doença de Depósito de Glicogênio Tipo II/terapia , Doença de Depósito de Glicogênio Tipo II/tratamento farmacológico , Doença de Depósito de Glicogênio Tipo II/imunologia , Humanos , Imunossupressores/uso terapêutico , Lisossomos/enzimologia , Modelos Biológicos , alfa-Glucosidases/imunologia , alfa-Glucosidases/uso terapêutico
12.
J Biol Chem ; 281(26): 17707-17, 2006 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-16636048

RESUMO

Stat3 and its isoforms belong to a family of cytoplasmic transcription factors that affect the synthesis of various proteins. Caspases are cysteinyl-aspartate proteases that function under apoptotic and non-apoptotic conditions. We now report that, in addition to transcriptional splicing, Stat3 fragmentation can be mediated by caspases. Caspase activation in DU145 cells was achieved by staurosporine (STS) exposure, and Western analysis revealed a reduction in full-length Stat3 (fl-Stat3) expression that was caspase-mediated. This proteolytic relationship was further studied by exposing purified Stat3 protein to a mixture of active caspases under cell-free conditions. This demonstrated that caspases directly cleaved Stat3 and Stat3 cleavage was accompanied by the apparent formation of cleavage fragment(s). Stat3 cleavage fragments, reflecting multiple caspase cleavage sites, also were observed in vitro following STS exposure in DU145 cells and in HEK293T cells transfected to express Stat3 truncation mutants. The impact of cleavage on Stat3 transcriptional activity next was assessed and revealed that cleavage of fl-Stat3 was accompanied by reductions in Stat3-DNA binding, Stat3-driven reporter protein (luciferase) activity, and the expression of selected Stat3-dependent genes. Further, reduced Stat3 expression correlated with increased sensitivity to apoptotic stimuli. In concomitant experiments, reporter activity was assessed in Stat3 truncation mutant-expressing HEK293T cells and revealed that, under non-apoptotic conditions, expression of different Stat3 fragments induced differential effects on Stat3-driven luciferase activity. These findings demonstrate that fl-Stat3 undergoes proteolytic processing by caspases that reduces its expression and leads to the formation of cleavage fragments that may modulate Stat3 transcriptional activity.


Assuntos
Apoptose/fisiologia , Caspases/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Ativação Transcricional/fisiologia , Clorometilcetonas de Aminoácidos/farmacologia , Apoptose/efeitos dos fármacos , Inibidores de Caspase , Inibidores de Cisteína Proteinase/farmacologia , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/fisiologia , Células HeLa , Humanos , Mutagênese , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Estaurosporina/farmacologia
13.
J Immunol ; 174(1): 320-7, 2005 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-15611255

RESUMO

There is a growing appreciation for how cells integrate and appropriately respond to competing signals for proliferation and apoptosis. The studies described in this report examined the effects of exposure to the cytokine IFN-alpha (IFN-alpha-2a) on sensitivity of the human cell lines H9 and SKW6.4 to Fas ligand (FasL)-induced apoptosis. In a concentration-dependent manner, FasL induced apoptosis, as shown by Western blot analysis of procaspase 8 and poly(ADP-ribose) polymerase cleavage after 3-h exposure and by cytofluorometric analysis of sub-G(0)-G(1) cellular DNA content after 24-h exposure. H9 and SKW6.4 cell lines responded to 10,000 IU/ml IFN-alpha-2a, as evidenced by decreased cell proliferation and tyrosine phosphorylation of Stat1 and Stat3 proteins without significant cytotoxicity. The effects of cytokine exposure on apoptosis were examined; incubation in medium containing 10,000 IU/ml IFN-alpha-2a for 1 h before FasL treatment significantly reduced all above-mentioned hallmarks of apoptosis. Surprisingly, these antiapoptotic effects of IFN-alpha-2a were independent of de novo protein synthesis, because they occurred in both the absence and the presence of cycloheximide. However, chemical inhibitors of JAK completely abrogated the effects of IFN-alpha-2a on FasL-induced apoptosis, indicating a direct role for JAK-mediated protein phosphorylation in modulating sensitivity to apoptosis. Together, these data suggest a novel mechanism, independent of protein synthesis, by which cytokine signals through JAKs can interact with and attenuate this receptor-mediated apoptotic process.


Assuntos
Apoptose/efeitos dos fármacos , Glicoproteínas de Membrana/farmacologia , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais/fisiologia , Apoptose/imunologia , Western Blotting , Proliferação de Células/efeitos dos fármacos , Cicloeximida/farmacologia , Proteínas de Ligação a DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta a Droga , Proteína Ligante Fas , Humanos , Interferon-alfa/imunologia , Interferon-alfa/farmacologia , Janus Quinase 1 , Glicoproteínas de Membrana/imunologia , Fosforilação , Inibidores da Síntese de Proteínas/farmacologia , Proteínas Tirosina Quinases/imunologia , Fator de Transcrição STAT1 , Fator de Transcrição STAT3 , Transativadores/efeitos dos fármacos , Transativadores/metabolismo , Células Tumorais Cultivadas , Tirosina/efeitos dos fármacos , Tirosina/metabolismo
14.
J Immunol ; 168(1): 308-15, 2002 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-11751975

RESUMO

Most bacteria that enter the bloodstream are taken up by the liver. Previously, we reported that such organisms are initially bound extracellularly and subsequently killed by immigrating neutrophils, not Kupffer cells as widely presumed in the literature. Rather, the principal functions of Kupffer cells demonstrated herein are to clear bacteria from the peripheral blood and to promote accumulation of bactericidal neutrophils at the principal site of microbial deposition in the liver, i.e., the Kupffer cell surface. In a mouse model of listeriosis, uptake of bacteria by the liver at 10 min postinfection i.v. was reduced from approximately 60% of the inoculum in normal mice to approximately 15% in mice rendered Kupffer cell deficient. Immunocytochemical analysis of liver sections derived from normal animals at 2 h postinfection revealed the massive immigration of neutrophils and their colocalization with Kupffer cells. Photomicrographs of the purified nonparenchymal liver cell population derived from these infected mice demonstrated listeriae inside neutrophils and neutrophils within Kupffer cells. Complementary adhesion molecules promoted the interaction between these two cell populations. Pretreatment of mice with mAbs specific for CD11b/CD18 (type 3 complement receptor) or its counter-receptor, CD54, inhibited the accumulation of neutrophils in the liver and the elimination of listeriae. Complement was not a factor; complement depletion affected neither the clearance of listeriae by Kupffer cells nor the antimicrobial activity expressed by infiltrating neutrophils.


Assuntos
Molécula 1 de Adesão Intercelular/fisiologia , Células de Kupffer/imunologia , Listeriose/imunologia , Fígado/imunologia , Antígeno de Macrófago 1/fisiologia , Neutrófilos/imunologia , Animais , Anticorpos/farmacologia , Proteínas do Sistema Complemento/fisiologia , Feminino , Imuno-Histoquímica , Molécula 1 de Adesão Intercelular/imunologia , Cinética , Células de Kupffer/citologia , Listeria monocytogenes/isolamento & purificação , Listeriose/microbiologia , Listeriose/patologia , Fígado/microbiologia , Fígado/patologia , Antígeno de Macrófago 1/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos , Neutrófilos/microbiologia , Peroxidase/metabolismo , Sepse/imunologia , Sepse/microbiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa