Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mult Scler ; 15(3): 323-8, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19028832

RESUMO

BACKGROUND: It is unknown whether immunosuppression of patients who have developed interferon-beta (IFN-beta) neutralizing antibodies (NAbs) hastens disappearance of NAbs in the blood. OBJECTIVE: We wanted to test whether immunosuppression with cyclic methylprednisolone (MP) in combination with azathioprine (AZA) for 6 months accelerates recovery of IFN-beta bioactivity in patients with multiple sclerosis (MS) with abolished in-vivo myxovirus resistance protein A (MxA) mRNA response to IFN-beta. METHODS: We included 13 patients with MS with NAbs and a low IFN-beta bioavailability detected by the MxA-mRNA response in a descriptive, non-randomized trial. Another 14 NAb-positive patients with a low MxA-mRNA response served as controls. The primary outcome was the fraction of patients who regained an MxA-mRNA response to IFN-beta. NAbs were measured by means of a clinically validated cytopathic effect assay and a new reporter gene assay. The in-vivo MxA-mRNA response was measured by real-time polymerase chain reaction. RESULTS: A total of 11 patients in the treatment group completed the trial. In all, two of these 11 patients regained an in-vivo MxA-mRNA response as compared to one of 14 patients in the control group. CONCLUSION: Treatment with AZA and cyclic MP for 6 months has little or no effect on IFN-beta bioactivity in NAb-positive patients with MS.


Assuntos
Azatioprina/administração & dosagem , Glucocorticoides/administração & dosagem , Imunossupressores/administração & dosagem , Interferon beta/imunologia , Metilprednisolona/administração & dosagem , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/imunologia , Anticorpos/sangue , Resistência a Medicamentos/imunologia , Quimioterapia Combinada , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/imunologia , Genes Reporter , Humanos , Interferon beta/uso terapêutico , Proteínas de Resistência a Myxovirus , RNA Mensageiro/metabolismo , Falha de Tratamento
2.
J Interferon Cytokine Res ; 28(6): 393-404, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18593334

RESUMO

Development of neutralizing antibodies (NAbs) to interferons (IFNs) can reduce the clinical response to IFN therapy. As current cell-based assays for quantifying NAbs have limitations, a highly sensitive and reproducible assay was developed, using division-arrested frozen human U937 cells transfected with the luciferase reportergene controlled by an IFN-responsive chimeric promoter, which allows IFN activity to be determined with precision within hours. Assay-ready PIL5 cells can be stored frozen for >3 years without loss of IFN sensitivity or the need for cell propagation. The assay is highly IFN sensitive (detecting <1.0 IU/mL), reproducible (SE +/- 15%) over concentrations from <1.0 to 100 IU/mL and able to measure different IFN subtypes and their pegylated variants. The use of this assay has shown that NAbs from patients treated with IFN-alpha2 exhibited markedly lower titers against 10 LU/mL of low specific activity IFNs, namely, IFN-alpha1, PEG-Intron(TM) (Schering-Plough, Levallois-Perret,France), or Pegasys(TM) (Hoffmann-La Roche, Neuilly-sur-Seine, France, than against 10 LU/mL IFN-alpha2. Similarly, NAbs from patients treated with IFN-beta1a exhibit lower titers against 10 LU/mL of low specific activity IFN-beta1b than against IFN-beta1a. The combination of the use of division-arrested, IFN-responsive human cells transfected with the luciferase reporter-gene makes the rapid PIL5 assay for NAbs highly advantageous.


Assuntos
Anticorpos/imunologia , Imunoensaio , Interferon Tipo I/imunologia , Interferon-alfa/imunologia , Anticorpos/sangue , Divisão Celular , Epitopos , Genes Reporter , Humanos , Imunoterapia Ativa , Interferon alfa-2 , Interferon-alfa/uso terapêutico , Luciferases , Polietilenoglicóis , Regiões Promotoras Genéticas , Proteínas Recombinantes , Células U937
3.
Br J Biomed Sci ; 65(1): 13-7, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18476489

RESUMO

Methicillin-resistant Staphylococcus aureus (MRSA) continues to cause major problems, both in hospitals and the community. Microbiology departments need to review their methodology regularly to ensure that they are contributing in the most appropriate manner to the battle against MRSA. Media employing chromogenic enzymes to aid the isolation and identification of MRSA is a relatively new approach. In this study, 192 swabs from 112 different patients were inoculated on two chromogen-containing media and four other commonly used solid MRSA media to determine which gave the appropriate combination of sensititivity, specificity and speed of result. Methicillin-resistant S. aureus was isolated on at least one of the six media from 102 of the 192 swabs. Both chromogenic media proved to be statistically significantly more sensitive than the other media after overnight incubation and had a sensitivity of 96% after 48 hours' incubation. The recent introduction of chromogen-containing MRSA media offers microbiology laboratories the opportunity to isolate and confirm the majority of MRSA infections/colonisations in 24 hours, which should result in better patient care. The possible slight increase in costs should not provide a valid excuse for using inferior methodologies.


Assuntos
Compostos Cromogênicos , Meios de Cultura , Resistência a Meticilina , Staphylococcus aureus/isolamento & purificação , Antibacterianos/farmacologia , Reações Falso-Positivas , Humanos , Testes de Sensibilidade Microbiana/métodos , Distribuição Aleatória , Sensibilidade e Especificidade , Infecções Estafilocócicas/economia , Infecções Estafilocócicas/epidemiologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/crescimento & desenvolvimento , Fatores de Tempo , Reino Unido/epidemiologia
4.
Transplant Proc ; 39(6): 1761-4, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17692605

RESUMO

Pentoxifylline (PTX) has been shown to protect the liver against normothermic ischemia-reperfusion (I-R) injury. The aims of this study were to investigate the action of PTX on tumor necrosis factor alpha (TNFalpha) gene transcription following normothermic liver I-R as well as to evaluate the resulting effects on liver function and survival. A segmental normothermic liver ischemia was induced for 90 minutes. Rats were divided into three groups: group 1, control, Ringer lactate administration; group 2, PTX treatment; group 3, sham-operated control rats. PTX (50 mg/kg) was injected intravenously 30 minutes before induction of ischemia and 30 minutes before reperfusion. The nonischemic liver lobes were resected at the end of ischemia. Survival rates were compared and serum activities of TNFalpha, aspartate aminotransferase, alanine aminotransferase, and lactate dehydrogenase were measured. Liver histology was assessed 6 hours after reperfusion. Liver TNFalpha mRNA was assessed by polymerase chain reaction amplification at different times after reperfusion. PTX treatment significantly decreased serum activities of TNFalpha and inhibited liver expression of TNFalpha mRNA. The extent of liver necrosis and serum levels of liver enzymes were significantly decreased by PTX treatment, resulting in a significant increase in 7-day survival compared with nontreated control rats. In conclusion, PTX inhibits liver TNFalpha gene transcription, decreases serum TNFalpha levels, and reduces liver injury following normothermic I-R.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Circulação Hepática , Pentoxifilina/farmacologia , Traumatismo por Reperfusão/prevenção & controle , Transcrição Gênica/efeitos dos fármacos , Fator de Necrose Tumoral alfa/genética , Animais , Circulação Hepática/efeitos dos fármacos , Masculino , Modelos Animais , Ratos , Ratos Long-Evans , Vasodilatadores/farmacologia
5.
Leukemia ; 21(4): 714-24, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17375123

RESUMO

Type I interferon (IFN) is shown to control the reversible quiescence of a primitive human bone marrow mesenchymal stem cell (MSC) subpopulation. A 24 h pre-treatment of Stro1+/GlycoA- or CD45-/GlycoA- subpopulations with a monoclonal antibody (mAb) against the IFNAR1 chain of the human type I IFN receptor (64G12), or with a polyclonal anti-IFNalpha antibody, resulted in a marked increase in the number of very large colonies (CFU-F >3000 cells) obtained in the presence of low, but necessary, concentrations of bFGF. Over a 2-month culture period, this short activation promoted a faster and greater amplification of mesenchymal progenitors for adipocytes and osteoblasts. Activation correlated with inhibition of STAT1 and STAT2 phosphorylation and of STAT1 nuclear translocation. A non-neutralizing anti-IFNAR1 mAb was ineffective. We demonstrate that control and activated MSCs express ST3GAL3, a sialyltransferase necessary to produce the embryonic antigens SSEA-3 and -4. Interestingly, activated MSC progeny expressed SSEA-3 and -4 at a higher level than control cultures, but this was not correlated with a significant expression of other embryonic markers. As MSCs represent an essential tool in tissue regeneration, the use of 64G12, which rapidly recruits a higher number of primitive cells, might increase amplification safety for cell therapy.


Assuntos
Interferon-alfa/fisiologia , Interferon beta/fisiologia , Células-Tronco Mesenquimais/citologia , Células da Medula Óssea/citologia , Técnicas de Cultura de Células , Diferenciação Celular , Divisão Celular/efeitos dos fármacos , Ensaio de Unidades Formadoras de Colônias , Primers do DNA , Matriz Extracelular/fisiologia , Humanos , Imunofenotipagem , Cinética , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/imunologia , Reação em Cadeia da Polimerase , Fator de Crescimento Transformador beta1/farmacologia
6.
Oncogene ; 26(3): 328-38, 2007 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-16832344

RESUMO

To characterize the mechanisms underlying apoptosis induced by viral infection, transcriptional activation of genes encoding members of the 'BH3-only' family of proteins was analysed during the course of virus infection. Among these genes, only NOXA is transcriptionally activated by vesicular stomatitis virus (VSV), sendai virus (SV), measles virus, herpes simplex virus, or dsRNA and required for efficient apoptosis of cells. Transcriptional activation of NOXA by VSV or SV is independent of p53, but requires the presence of interferon regulatory factor 1 (IRF-1), IRF-3 and cAMP-responsive element binding protein (CREB). Binding to and transactivation of the NOXA promoter by each of these transcription factors is governed by post-translational modification involving different pathways for each factor. Thus, SV infection activates IRF-3 and CREB by phosphorylation triggered by Toll like receptor 3 signalling, and a pathway involving calcium-independent phopholipase A2, respectively. In addition transactivation induced by IRF-1 during viral infection correlates with a 10 kDa increase in its molecular weight, suggesting a covalent linkage with a previously unknown regulatory polypeptide.


Assuntos
Apoptose , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Fator Regulador 1 de Interferon/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Vírus de RNA/fisiologia , RNA Viral/genética , Proteína Supressora de Tumor p53/metabolismo , Northern Blotting , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Cálcio/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Vírus do Sarampo/fisiologia , Fosfolipases A/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Vírus de RNA/classificação , Vírus Sendai/fisiologia , Transdução de Sinais , Simplexvirus/fisiologia , Receptor 3 Toll-Like/metabolismo , Transcrição Gênica , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Vírus da Estomatite Vesicular Indiana/fisiologia
7.
Med Hypotheses ; 60(2): 293-7, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12606250

RESUMO

Whether retroviral integration is a phenomenon specific to properties of the surrounding genomic region is a widely debated question. In this paper we attempt to enlight the involvement of genomic regions prone to DNA double strand breaks in such process, as well as the more general concept of genome plasticity concerning repair, recombination, transposition events. While performing a differential display analysis of the promonocytic cell line U937 and clone U42 HIV infected counterpart, we found, out of about 15 highly dysregulated genes, expected according to our previous proteomic analysis, two dysregulated cellular transcripts that are shown in the present study to colocalize on band 22q11. The LB14 transcript maps within the DiGeorge critical region. Whereas the AG46 transcript encodes the immunoglobulin-lambda like polypeptide 1 (IGLL1) 4.7Mb apart from LB14. The 22q11 band is remarkable for its high plasticity involving DNA double strand breaks, that may lead to translocations, large deletions, and immunoglobulin rearrangements, frequently observed in this region. We suggest that provirus integration preferentially occurs in such genomic regions and that the subsequent insertional mutagenesis leads to the present observations. Finally, we stress out the possibility that the small size of chromosome 22 is associated with this physical property of the genome.


Assuntos
Genoma Humano , Retroviridae/metabolismo , Integração Viral , Cromossomos Humanos Par 22 , Dano ao DNA , Perfilação da Expressão Gênica , HIV/patogenicidade , Humanos , Retroviridae/genética , Células U937
8.
Leukemia ; 16(6): 1135-42, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12040445

RESUMO

Affinity purified, freshly isolated CD34+ progenitors were shown to express low levels of type I interferon (IFN) receptors (740 +/- 60 binding sites/cell, K(d) 0.7 +/- 0.04 nM) determined by Scatchard's analysis using a radiolabelled, neutralizing, monoclonal antibody directed against the IFNAR1 chain of the human type I IFN receptor. Treatment of freshly isolated (day 0), highly purified (>95% pure) CD34+ cells with recombinant IFN-alpha resulted in rapid tyrosine phosphorylation and activation of STAT1, Tyk2 and JAK1 as shown by Western immunoblotting. Similarly, IFN treatment was shown by confocal microscopy to result in rapid nuclear localization of the transcription factors IRF1 and STAT2, demonstrating the presence of functional IFN receptors on freshly isolated (day 0) CD34+ cells. The number of specific type I IFN receptor binding sites expressed on hematopoietic progenitor cells increased to some 1440 +/- 40 per cell after 11 days of cultivation of CD34+ cells in vitrosuggesting that receptor expression increases with cell differentiation. IFN-mediated signal transduction and the inhibitory effect of IFN-alpha on 7 or 14 days CFU-GM and BFU-E colony formation was abrogated in the presence of the anti-IFNAR1 mAb, indicating that IFN-alpha acts directly on the proliferation of human hematopoietic progenitor cells via receptor activated signal transduction without excluding the induction of other cytokines or growth factors by residual accessory cells.


Assuntos
Antígenos CD34/análise , Células-Tronco Hematopoéticas/metabolismo , Receptores de Interferon/fisiologia , Transdução de Sinais , Transporte Ativo do Núcleo Celular , Anticorpos Monoclonais/farmacologia , Divisão Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Hematopoéticas/química , Humanos , Fator Regulador 1 de Interferon , Interferon-alfa/antagonistas & inibidores , Janus Quinase 1 , Cinética , Proteínas de Membrana , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Tirosina Quinases/metabolismo , Proteínas/metabolismo , Receptor de Interferon alfa e beta , Receptores de Interferon/antagonistas & inibidores , Receptores de Interferon/imunologia , Fator de Transcrição STAT1 , Fator de Transcrição STAT2 , TYK2 Quinase , Transativadores/metabolismo
9.
J Interferon Cytokine Res ; 21(8): 575-81, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11559435

RESUMO

Intraperitoneal (i.p.) administration of 20,000 IU recombinant murine IFN-alpha (rMuIFN-alpha) was highly effective in protecting mice challenged i.p. with doses of encephalomyocarditis virus (EMCV) ranging from 44 to 440 LD(50) (p<0.001). Oromucosal (o.m.) IFN therapy was also found to be effective in protecting mice challenged with a lethal dose of EMCV. Thus, 40% of animals infected with 44 LD(50) of EMCV and treated o.m. with 20,000 IU rMuIFN-alpha survived infection with a mean survival time of 12.0 +/- 2.46 days relative to a mean of 6.11 +/- 0.38 days in the control group (p<0.05). Oromucosal IFN therapy was found to be ineffective, however, in animals infected with higher doses of EMCV (88-440 LD(50)), even though intraperitoneal administration of the same dose of rMuIFN-alpha resulted in the survival of 90%, 50%, and 60% of animals infected with 88, 220, and 440 LD(50) of EMCV, respectively. These results suggest that oromucosal IFN therapy is effective at relatively low viral load only and that the mechanism of action of oromucosal IFN therapy may be different from that of parenterally administered IFN. Our results suggest that oromucosal IFN therapy may be most effective in chronic viral infections as an alternative to parenterally administered IFN, which is clinically effective but poorly tolerated.


Assuntos
Antivirais/administração & dosagem , Infecções por Cardiovirus/tratamento farmacológico , Infecções por Cardiovirus/virologia , Interferon Tipo I/administração & dosagem , Carga Viral , Administração Intranasal , Animais , Antivirais/uso terapêutico , Infecções por Cardiovirus/mortalidade , Vírus da Encefalomiocardite/efeitos dos fármacos , Injeções Intraperitoneais , Interferon Tipo I/uso terapêutico , Dose Letal Mediana , Masculino , Camundongos , Mucosa Bucal/efeitos dos fármacos , Mucosa Bucal/virologia , Orofaringe , Proteínas Recombinantes , Taxa de Sobrevida
10.
J Interferon Cytokine Res ; 21(8): 583-93, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11559436

RESUMO

Oromucosal (o.m.) administration of interferon-alpha (IFN-alpha) during either allergic sensitization (days 0-6) or the hypersensitive response (days 11 and 12) or both periods caused a dose-dependent reduction in allergen-specific IgE production and allergen-induced eosinophil recruitment in mice sensitized to ragweed pollen, a common allergen in humans. Treatment during the hypersensitive response period alone appeared to be most effective. Oromucosal treatment was as effective as intraperitoneal (i.p.) treatment, with maximum inhibition of both allergen-specific IgE production and allergen-induced eosinophil recruitment observed at a dose of a 1000 IU IFN-alpha. Treatment of animals with up to 10(5) IU murine IFN-alpha/beta (MuIFN-alpha/beta) by either the om. or i.p. route did not inhibit significantly allergen-specific IgG production, which may even have been increased at certain doses of IFN. Treatment of animals with up to 10(5) IU MuIFN-alpha/beta by either the o.m. or i.p. route did not affect significantly total serum IgE or IgG levels. Oromucosal administration of IFN-alpha reduced allergen-specific IgE production and allergen-induced eosinophil recruitment in the absence of detectable toxicity, the induction of H(2) antigen expression, and 2',5'-oligoadenylate synthetase activity associated with parenteral administration of IFN-alpha and thus may find application for the treatment of asthma and associated viral infections.


Assuntos
Alérgenos/imunologia , Hipersensibilidade Imediata/imunologia , Imunização , Interferon Tipo I/administração & dosagem , Pólen/imunologia , 2',5'-Oligoadenilato Sintetase/metabolismo , Administração Intranasal , Alérgenos/administração & dosagem , Animais , Asma/enzimologia , Asma/imunologia , Asma/patologia , Movimento Celular/imunologia , Eosinófilos/imunologia , Eosinófilos/patologia , Epitopos/imunologia , Antígenos de Histocompatibilidade Classe I/biossíntese , Humanos , Hidrocortisona/administração & dosagem , Hipersensibilidade Imediata/enzimologia , Hipersensibilidade Imediata/patologia , Imunoglobulina E/sangue , Imunoglobulina G/sangue , Injeções Intraperitoneais , Interferon Tipo I/uso terapêutico , Interleucina-12/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes
11.
J Interferon Cytokine Res ; 21(7): 539-45, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11506749

RESUMO

Interferons (IFN) have been shown to be effective in protecting animals against lethal viral infections when administered systemically in relatively high doses. Intraperitoneal (i.p.) injection of mice with encephalomyocarditis virus (EMCV) gives rise to a rapidly progressive fatal disease characterized by central nervous system involvement and encephalitis. IFN-alpha has been shown to be effective in protecting mice against lethal EMCV infection when given via parenteral and oral/sublingual routes. The current study was designed to explore the ability of orally/sublingually and intranasally (i.n.) administered IFN-alpha to treat mice infected with EMCV in support of a planned clinical trial to evaluate efficacy of oral IFN-alpha in human viral infections. The primary objective of the study was to determine the efficacy of recombinant murine IFN-alpha (rMuIFN-alpha) in the treatment of mice infected with 100 LD(50) EMCV following oral, i.n., and i.p. administration at doses of 20,000 and 100,000 IU. The results of the current experiment did not indicate protection from infection with EMCV in mice that received IFN by the i.n. or oral/sublingual routes. The negative controls, infection of mice with 100 LD(50) of EMCV followed by treatment with excipient via all three routes, resulted in death of nearly all mice, as expected. The positive control, treatment of EMCV-infected (100 LD(50)) mice with rMuIFN-alpha via the i.p. route, was successful in protecting a significant number of mice from death compared with matched controls. This study points out the need to determine the optimum conditions for administration of oral/sublingual or i.n. IFN to insure maximum efficacy against viral infections.


Assuntos
Antivirais/administração & dosagem , Antivirais/efeitos adversos , Infecções por Cardiovirus/tratamento farmacológico , Vírus da Encefalomiocardite/efeitos dos fármacos , Interferon Tipo I/administração & dosagem , Interferon Tipo I/efeitos adversos , Administração Intranasal , Administração Oral , Animais , Antivirais/uso terapêutico , Infecções por Cardiovirus/mortalidade , Avaliação Pré-Clínica de Medicamentos/métodos , Feminino , Injeções Intraperitoneais , Interferon Tipo I/uso terapêutico , Dose Letal Mediana , Camundongos , Distribuição Aleatória , Proteínas Recombinantes , Titulometria
12.
Proc Natl Acad Sci U S A ; 98(10): 5752-7, 2001 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-11320211

RESUMO

To understand how virulent mycobacteria subvert host immunity and establish disease, we examined the differential response of mice to infection with various human outbreak Mycobacterium tuberculosis clinical isolates. One clinical isolate, HN878, was found to be hypervirulent, as demonstrated by unusually early death of infected immune-competent mice, compared with infection with other clinical isolates. The differential effect on survival required lymphocyte function because severe combined immunodeficiency (SCID) mice infected with HN878 or other clinical isolates all died at the same rate. The hypervirulence of HN878 was associated with failure to induce M. tuberculosis-specific proliferation and IFN-gamma production by spleen and lymph node cells from infected mice. In addition, 2- to 4-fold lower levels of tumor necrosis factor-alpha (TNF-alpha), IL-6, IL-12, and IFN-gamma mRNAs were observed in lungs of HN878-infected mice. IL-10, IL-4, and IL-5 mRNA levels were not significantly elevated in lungs of HN878 infected mice. In contrast, IFN-alpha mRNA levels were significantly higher in lungs of these mice. To further investigate the role of Type 1 IFNs, mice infected with HN878 were treated intranasally with purified IFN-alpha/beta. The treatment resulted in increased lung bacillary loads and even further reduced survival. These results suggest that the hypervirulence of HN878 may be due to failure of this strain to stimulate Th1 type immunity. In addition, the lack of development of Th1 immunity in response to HN878 appears to be associated with increased induction of Type 1 IFNs.


Assuntos
Interferon-alfa/biossíntese , Interferon beta/biossíntese , Mycobacterium tuberculosis/patogenicidade , Células Th1/imunologia , Animais , Divisão Celular , Feminino , Interferon-alfa/genética , Interferon beta/genética , Pulmão/metabolismo , Camundongos , Camundongos SCID , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tuberculose Pulmonar/imunologia , Virulência
13.
J Interferon Cytokine Res ; 21(12): 1031-8, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11798460

RESUMO

The specific IgM and IgG antibody responses to subcutaneous (s.c.) treatment of mice with recombinant human IFN-alpha2a (rHuIFN-alpha2a) or IFN-beta were inhibited in a dose-dependent manner by prior oromucosal (o.m.) administration of rHuIFN-alpha2a or IFN-beta, respectively. Pretreatment of animals once a day for 7 days by the o.m. route with the highest dose of IFN-alpha2a tested (10(7) IU) resulted in complete inhibition of the peak IFN-alpha2a-specific IgG antibody response detected 28 days after subsequent s.c. injection of IFN-alpha2a (p < 0.001). Similarly, prior o.m. administration of 1-10 microg rHuGM-CSF per day for 7 days resulted in a statistically significant (p < 0.001) inhibition of the peak GM-CSF-specific IgG antibody response detected 28 days after s.c. administration of GM-CSF. In contrast, prior o.m. treatment with a quantity of bovine serum albumin (BSA) (100 microg) or human serum albumin (HSA) (10 microg) equivalent, respectively, to the protein content of the highest dose of IFN-alpha2a or GM-CSF administered by the o.m. route, did not affect significantly the IFN-alpha2a-specific or GM-CSF-specific IgG antibody responses detected on subsequent s.c. administration of IFN-alpha2a or GM-CSF. Oromucosal administration of IFN-alpha2a, IFN-beta, or GM-CSF alone did not induce detectable IFN-alpha2a-specific, IFN-beta-specific, or GM-CSF-specific IgM or IgG antibody responses at any of the time points tested. These results suggest that short-term o.m. administration of a recombinant protein is an effective means of inducing peripheral tolerance to subsequent parenteral administration of a therapeutic protein.


Assuntos
Tolerância a Medicamentos , Proteínas Recombinantes/farmacologia , Administração Intranasal , Animais , Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Imunoglobulina G/biossíntese , Imunoglobulina M/biossíntese , Injeções Subcutâneas , Interferon Tipo I/administração & dosagem , Interferon Tipo I/farmacologia , Interferon alfa-2 , Interferon-alfa/administração & dosagem , Interferon-alfa/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Mucosa Nasal , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/uso terapêutico
14.
Eur Cytokine Netw ; 11(4): 560-73, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11125298

RESUMO

Interferons (IFNs) in common with other cytokines activate Janus tyrosine kinases and latent STAT transcription factors upon binding to their cell surface receptor. Type I IFNs bind to a receptor composed of two transmembrane polypeptides, IFNAR1 and IFNAR2, which belong to the class II cytokine receptor family that also includes the cellular receptors for IFN-gamma, interleukin-10 and coagulation protease factor VII (tissue factor). The extracellular domain of the type I IFN receptor chain IFNAR1, has four fibronectin type-III sub-domains. Human IFNAR1 has intrinsic weak affinity for type I IFNs and plays an essential role in transmembrane signaling, formation of a high affinity complex with IFN and the modulation of ligand specificity. In order to characterise the ligand binding site on IFNAR1 we analysed the epitope recognized by the anti-IFNAR1 mAb, 64G12, which inhibits the binding and biological activities of both IFN-alpha and IFN-beta. The target peptide recognized by the 64G12 mAb was determined by screening a set of 48 overlapping peptides covering the first two subdomains (residues 23-229) of the extracellular region of IFNAR1. The results of this study show that the peptide (FSSLKLNVY), localized within the first sub-domain (residues 89-97) of IFNAR1, which is recognized by the 64G12 mAb, most likely overlaps a site to which both IFN-alpha and IFN-beta bind in the ligand-receptor complex. Thus, since the 64G12 mAb can neutralize the biological activities of all the type I IFNs tested, we suggest that the target peptide recognized by the 64G12 mAb, is a possible anchorage point on IFNAR1, common to binding of both IFN-alpha and IFN-beta.


Assuntos
Mapeamento de Epitopos , Interferon Tipo I/metabolismo , Oligopeptídeos/metabolismo , Receptores de Interferon/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Células CHO , Células COS , Bovinos , Cricetinae , Proteínas de Ligação a DNA/metabolismo , Humanos , Fragmentos Fab das Imunoglobulinas/imunologia , Interferon Tipo I/imunologia , Interferon-alfa/metabolismo , Interferon-alfa/farmacologia , Janus Quinase 1 , Proteínas de Membrana , Dados de Sequência Molecular , Oligopeptídeos/imunologia , Fosforilação , Fosfotirosina/metabolismo , Proteínas Tirosina Quinases/metabolismo , Receptor de Interferon alfa e beta , Receptores de Interferon/imunologia , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes/imunologia , Fator de Transcrição STAT1 , Fator de Transcrição STAT2 , Transdução de Sinais , Transativadores/metabolismo , Células Tumorais Cultivadas , Vírus da Estomatite Vesicular Indiana/crescimento & desenvolvimento , Replicação Viral
15.
Genomics ; 70(1): 140-9, 2000 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11087671

RESUMO

We have previously reported a transcript of a novel mouse gene (Scrg1) with increased expression in transmissible spongiform encephalopathies and the cloning of the human mRNA analogue. In this paper, we present the genomic organization of the mouse and human SCRG1 loci, which exhibit a high degree of conservation. The genes are composed of three exons; the two downstream exons contain the protein coding region. The mouse gene is expressed in brain tissue essentially as a 0.7-kb message but also as a minor 2.6-kb mRNA. We have sequenced 20 kb of DNA at the mouse Scrg1 locus and found that the longer transcript is the prolongation of the 0.7-kb mRNA to a polyadenylation site located about 2 kb further downstream. Sequencing revealed that the mouse Scrg1 gene is physically linked to Sap30, a gene that encodes a protein of the histone deacetylase complex, and genetic linkage mapping assigned the localization of Scrg1 to chromosome 8 between Ant1 and Hmg2. Northern blot analysis showed that Scrg1 is under strict developmental control in mouse embryo and is expressed by cells of neuronal origin in vitro. Comparison of the rat, mouse, and human SCRG1 proteins identified a box of 35 identical contiguous amino acids and a characteristic cysteine distribution pattern defining a new protein signature.


Assuntos
Ligação Genética , Histona Desacetilases/genética , Proteínas do Tecido Nervoso/genética , Neurônios/química , Sequência de Aminoácidos , Animais , Sequência de Bases , Células Cultivadas , Mapeamento Cromossômico , Embrião de Mamíferos , Biblioteca Genômica , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Neurônios/citologia , Proteínas de Plantas/genética , RNA Mensageiro , Ratos , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Distribuição Tecidual
17.
Brain Res Mol Brain Res ; 76(1): 173-9, 2000 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-10719228

RESUMO

The expression of the mRNA of nine scrapie responsive genes was analyzed in the brains of FVB/N mice infected with bovine spongiform encephalopathy (BSE). The RNA transcripts of eight genes were overexpressed to a comparable extent in both BSE-infected and scrapie-infected mice, indicating a common series of pathogenic events in the two transmissible spongiform encephalopathies (TSEs). In contrast, the serine proteinase inhibitor spi 2, an analogue of the human alpha-1 antichymotrypsin gene, was overexpressed to a greater extent in the brains of scrapie-infected animals than in animals infected with BSE, reflecting either an agent specific or a mouse strain specific response. The levels of spi 2 mRNA were increased during the course of scrapie prior to the onset of clinical signs of the disease and the increase reached 11 to 45 fold relative to uninfected controls in terminally ill mice. Spi 2, in common with four of the other scrapie responsive genes studied, is known to be associated with pro-inflammatory processes. These observations underline the importance of cell reactivity in TSE. In addition, scrg2 mRNA the level of which is enhanced in TSE-infected mouse brain, was identified as a previously unrecognized long transcript of the murine aldolase C gene. However, the level of the principal aldolase C mRNA is unaffected in TSE. The increased representation of the longer transcript in the late stage of the disease may reflect changes in mRNA processing and/or stability in reactive astrocytes or in damaged Purkinje cells.


Assuntos
Encéfalo/metabolismo , Encefalopatia Espongiforme Bovina/metabolismo , RNA Mensageiro/metabolismo , Scrapie/genética , Animais , Sequência de Bases , Complemento C1q/genética , Complemento C1q/metabolismo , Encefalopatia Espongiforme Bovina/enzimologia , Frutose-Bifosfato Aldolase/genética , Frutose-Bifosfato Aldolase/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Scrapie/enzimologia , Scrapie/metabolismo , Inibidores de Serina Proteinase/genética , Inibidores de Serina Proteinase/metabolismo
18.
Nucleic Acids Res ; 27(22): 4483-90, 1999 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-10536159

RESUMO

Expression of the interleukin-6 (IL-6) gene is usually tightly controlled and may be induced in specific tissues only after treatment with appropriate stimuli. The molecular mechanisms responsible for IL-6 gene repression in specific tissues or cell lines remain poorly defined. In order to address this question we have studied two human breast carcinoma cell lines, MDA-MB-231, in which the IL-6 gene is expressed, and MCF-7, in which it is not. The promoter region of the IL-6 gene was analysed in both cell lines with reference to two different parameters: (i) DNase I hypersensitivity; (ii) the in vivo pattern of DNA-protein interactions. We show herein that the mechanism responsible for silencing IL-6 gene expression in MCF-7 cells most probably involves a modification of chromatin structure, as suggested by a decreased sensitivity of the IL-6 promoter to DNase I relative to the IL-6-expressing cell line MDA-MB-231. Moreover, we show that a 'closed' nucleosomal structure in MCF-7 cells does not inhibit the binding of nuclear proteins to IL-6 gene regulatory sequences in vivo. We suggest, therefore, that, in non-expressing cells, local chromatin remodelling at the proximal promoter is inhibited by negative regulators, as suggested by two specific hallmarks of nuclear factor binding that are not observed in expressing cells: an additional in vivo footprint spanning positions -135/-119 and an additional DNase I hypersensitive site far upstream, around position -1400. Furthermore, a specific factor binding in vitro to the -140/-116 region of the IL-6 promoter is found in MCF-7 cells.


Assuntos
Cromatina/fisiologia , Regulação da Expressão Gênica , Interleucina-6/genética , Sequência de Bases , Cromatina/química , Pegada de DNA , Desoxirribonuclease I/metabolismo , Humanos , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Ésteres do Ácido Sulfúrico/metabolismo , Células Tumorais Cultivadas
19.
J Interferon Cytokine Res ; 19(8): 911-21, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10476938

RESUMO

Mucosal administration of the Th1 stimulatory cytokines interleukin-2 (IL-2), IL-12, IL-15, IL-18, or granulocyte-macrophage colony-stimulating factor (GM-CSF) induced antiviral activity in mice challenged systemically with a lethal dose of encephalomyocarditis virus (EMCV) similar to that observed following parenteral administration. In contrast, mucosal administration of the Th2 stimulatory cytokines IL-4, IL-5, IL-10, or IL-13 did not affect significantly the survival of EMCV-infected animals. Mucosal administration of IL-2 or IL-12 also exerted a marked antitumor activity in mice inoculated intravenously with Friend erythroleukemia cells. Recombinant IL-2 and IL-18, but none of the other recombinant cytokines tested, induced low levels of IFN in vitro. Polyclonal antibodies to both mouse and human interferon-alpha/beta (IFN-alpha/beta) abrogated the antiviral activity of IL-2 in vivo, even though the anti-human IFN-alpha/beta antibody did not neutralize mouse IFN-alpha/beta, and neither antibody bound to IL-2. IL-15 did not exhibit antiviral activity in IFN-alpha/beta R-/- mice, which are deficient in natural killer (NK) cell activity. These results suggest that mucosal Th1 cytokine therapy induces a soluble factor or activates a specific cell population in the lymphoid or epithelial tissue of the oropharyngeal cavity, which potentiates elimination of virus-infected or neoplasic cells systemically.


Assuntos
Antineoplásicos/uso terapêutico , Antivirais/uso terapêutico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Interferons/uso terapêutico , Interleucinas/uso terapêutico , Animais , Vírus da Leucemia Murina de Friend/efeitos dos fármacos , Humanos , Dose Letal Mediana , Leucemia Experimental/tratamento farmacológico , Leucemia Experimental/patologia , Camundongos , Mucosa/efeitos dos fármacos , Transplante de Neoplasias , Proteínas Recombinantes/uso terapêutico , Taxa de Sobrevida , Viroses/tratamento farmacológico , Replicação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...