Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
1.
Clin Exp Immunol ; 167(2): 179-87, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22235993

RESUMO

Carbon monoxide (CO) is produced during the catabolism of free haem, catalyzed by haem oxygenase (HO) enzymes, and its physiological roles include vasodilation, neurotransmission, inhibition of platelet aggregation and anti-proliferative effects on smooth muscle. In vivo preclinical studies have shown that exogenously administered quantities of CO may represent an effective treatment for conditions characterized by a dysregulated immune response. The carbon monoxide-releasing molecules (CORMs) represent a group of compounds capable of carrying and liberating controlled quantities of CO in the cellular systems. This review covers the physiological and anti-inflammatory properties of the HO/CO pathway in the central nervous system. It also discusses the effects of CORMs in preclinical models of inflammation. The accumulating data discussed herein support the possibility that CORMs may represent a novel class of drugs with disease-modifying properties in multiple sclerosis.


Assuntos
Boranos/uso terapêutico , Monóxido de Carbono/uso terapêutico , Carbonatos/uso terapêutico , Esclerose Múltipla/tratamento farmacológico , Compostos Organometálicos/uso terapêutico , Animais , Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/uso terapêutico , Autoimunidade/efeitos dos fármacos , Boranos/administração & dosagem , Monóxido de Carbono/administração & dosagem , Monóxido de Carbono/metabolismo , Carbonatos/administração & dosagem , Cardiotônicos/administração & dosagem , Cardiotônicos/uso terapêutico , Citocinas/biossíntese , Avaliação Pré-Clínica de Medicamentos , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/imunologia , Guanilato Ciclase/metabolismo , Heme/metabolismo , Heme Oxigenase (Desciclizante)/fisiologia , Heme Oxigenase-1/deficiência , Heme Oxigenase-1/fisiologia , Humanos , Inflamação/tratamento farmacológico , Esclerose Múltipla/imunologia , Neuroimunomodulação/efeitos dos fármacos , Neuroimunomodulação/fisiologia , Compostos Organometálicos/administração & dosagem , Oxirredução , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais/efeitos dos fármacos , Guanilil Ciclase Solúvel , Vasodilatadores/administração & dosagem , Vasodilatadores/uso terapêutico
2.
Int J Immunopathol Pharmacol ; 24(3): 621-30, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21978694

RESUMO

Breast cancer is more frequent in human nulliparae, whereas its incidence is reduced by early fullterm pregnancy. Rodent studies suggest that chorionic gonadotropin secretion during pregnancy affords protection by inducing breast structure differentiation. Opposite effects, however, have been observed in cancer prone transgenic mice overexpressing the ß subunit of chorionic gonadotropin or pituitary luteinic hormone (LH). Here we assessed the effect of administration of human chorionic gonadotropin (hCG) for 21 days (corresponding to the duration of a mouse pregnancy) in virgin female mice transgenic for the activated rat (r-) ERBB-2 oncogene (BALB-neuT). In these mice, the onset of atypical mammary duct hyperplasia and its progression towards multiple mammary carcinomas is accelerated by hCG. hCG enhances the in vitro proliferation and in vivo metastatization of tumor cells from a BALB-neuT mammary tumor expressing the hCG/LH as well as the ERBB-2 receptors. These findings suggest that hCG favours the growth and progression of hCG/LH and ERBB-2 receptor-positive breast tumors.


Assuntos
Gonadotropina Coriônica/metabolismo , Gonadotropina Coriônica/farmacologia , Hormônio Luteinizante/metabolismo , Neoplasias Mamárias Experimentais/patologia , Receptor ErbB-2/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Progressão da Doença , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Imuno-Histoquímica , Injeções Intravenosas , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos BALB C , Ovariectomia , Ovário/efeitos dos fármacos , Ovário/metabolismo , Sais de Tetrazólio , Tiazóis
3.
Cytokine ; 13(4): 209-19, 2001 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-11237428

RESUMO

Myeloid progenitor inhibitory factor (MPIF)-2 is a beta-chemokine with select and potent activities on eosinophils and myeloid progenitors. In the beta-chemokine family, biological activity is modulated by differential processing of the amino-terminus. Here, for MPIF-2, we describe the biological activities of NH(2)-terminal deletion mutants and compare regions necessary for eosinophil and myeloid progenitor activities. Five MPIF-2 proteins with deletions at the amino-terminus were produced in Escherichia coli and assayed for calcium mobilization, chemotaxis and receptor binding activities on eosinophils, and for their ability to inhibit colony formation of human myeloid bone marrow progenitors. For eosinophils, deletion of the first two amino acids did not markedly alter activity, while subsequent truncations result in a complete loss of activity. One of the MPIF-2 mutants, MPIF-2 (P30-R99) was converted from an agonist to an antagonist of eotaxin, MPIF-2 and MCP-4 functional responses in eosinophil calcium flux and chemotaxis assays. Surprisingly, while displaying a complete loss of agonist activity toward eosinophils, MPIF-2 (P30-R99) retains ability to inhibit human bone marrow myeloid progenitor cell colony formation. In addition, processing at the amino terminus of MPIF-2 in vivo, may result in a chemokine with altered biological activities.


Assuntos
Quimiocinas CC/genética , Eosinófilos/metabolismo , Células Progenitoras Mieloides/metabolismo , Sítios de Ligação , Cálcio/metabolismo , Sinalização do Cálcio , Quimiocina CCL24 , Quimiocinas CC/biossíntese , Quimiocinas CC/fisiologia , Vetores Genéticos/metabolismo , Humanos , Fragmentos de Peptídeos/biossíntese , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/fisiologia , Receptores CCR3 , Receptores de Quimiocinas/metabolismo , Receptores de HIV/metabolismo , Deleção de Sequência
4.
Structure ; 8(9): 927-36, 2000 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-10986460

RESUMO

BACKGROUND: Molecular interactions among cytokines and cytokine receptors form the basis of many cell-signaling pathways relevant to immune function. Interferon-gamma (IFN-gamma) signals through a multimeric receptor complex consisting of two different but structurally related transmembrane chains: the high-affinity receptor-binding subunit (IFN-gammaRalpha) and a species-specific accessory factor (AF-1 or IFN-gammaRbeta). In the signaling complex, the two receptors probably interact with one another through their extracellular domains. Understanding the atomic interactions of signaling complexes enhances the ability to control and alter cell signaling and also provides a greater understanding of basic biochemical processes. RESULTS: The crystal structure of the complex of human IFN-gamma with the soluble, glycosylated extracellular part of IFN-gammaRalpha has been determined at 2.9 A resolution using multiwavelength anomalous diffraction methods. In addition to the expected 2:1 complex, the crystal structure reveals the presence of a third receptor molecule not directly associated with the IFN-gamma dimer. Two distinct intermolecular contacts, involving the edge strands of the C-terminal domains, are observed between this extra receptor and the 2:1 receptor-ligand complex thereby forming a 3:1 complex. CONCLUSIONS: The observed interactions in the 2:1 complex of the high-affinity cell-surface receptor with the IFN-gamma cytokine are similar to those seen in a previously reported structure where the receptor chains were not glycosylated. The formation of beta-sheet packing interactions between pairs of IFN-gammaRalpha receptors in these crystals suggests a possible model for receptor oligomerization of Ralpha and the structurally homologous Rbeta receptors in the fully active IFN-gamma signaling complex.


Assuntos
Interferon gama/química , Receptores de Interferon/química , Sítios de Ligação , Cristalografia por Raios X , Glicosilação , Humanos , Interferon gama/metabolismo , Ligantes , Modelos Moleculares , Fragmentos de Peptídeos/química , Conformação Proteica , Estrutura Secundária de Proteína , Receptores de Interferon/metabolismo , Receptor de Interferon gama
5.
Cytokine ; 12(4): 315-23, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10805211

RESUMO

We have studied the effects of either exogenously-administered interferon (IFN-)gamma or of a nonimmunogenic mouse IFN-gamma receptor-Immunoglobulin (IFN-gamma R-Ig) fusion protein on the development of Concanavalin (Con)A-induced hepatitis in NMRI mice. PBS-treated control mice injected with 20 mg/kg ConA developed classical serological and histological signs of hepatitis with elevation of transaminases in the blood and infiltration of the liver by mononuclear cells and neutrophils. Treating the mice with rat IFN-gamma 24 h prior to and 1 h after ConA-challenge markedly exacerbated these signs of hepatitis in a dose-dependent fashion. Moreover, mice injected with lower, non hepatitogenic, doses of ConA (10, 5 mg/kg) became fully susceptible to develop hepatitis upon similar treatment with IFN-gamma. Concordantly, ConA-induced hepatitis was abrogated by either IFN-gamma R-Ig fusion protein or anti-IFN-gamma mAb. These data provide further evidence for the central pathogenetic role of endogenous IFN-gamma in ConA-induced hepatitis and demonstrate the feasibility to prevent disease development by means of a non immunogenic IFN-gamma R-Ig fusion protein.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/imunologia , Interferon gama/imunologia , Fígado/imunologia , Receptores de Interferon/imunologia , Linfócitos T/imunologia , Animais , Anticorpos Monoclonais/imunologia , Células CHO , Doença Hepática Induzida por Substâncias e Drogas/sangue , Doença Hepática Induzida por Substâncias e Drogas/patologia , Concanavalina A/administração & dosagem , Cricetinae , Citocinas/sangue , Citocinas/imunologia , Imunoglobulinas/genética , Imunoglobulinas/imunologia , Interferon gama/farmacologia , Interleucina-4/imunologia , Fígado/lesões , Fígado/patologia , Masculino , Camundongos , Ratos , Receptores de Interferon/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Receptor de Interferon gama
6.
J Immunol ; 164(6): 3200-6, 2000 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-10706711

RESUMO

The human beta chemokine known as LEC (also called NCC-4, HCC-4, or LMC) displays chemotactic activity for monocytes and dendritic cells. The possibility that its local presence increases tumor immunogenicity is addressed in this paper. TSA parental cells (TSA-pc) are poorly immunogenic adenocarcinoma cells that grow progressively, kill both nu/nu and syngeneic BALB/c mice, and give rise to lung metastases. TSA cells engineered to release LEC (TSA-LEC) are still able to grow in nu/nu mice, but are promptly rejected and display a marginal metastatic phenotype in BALB/c mice. Rejection is associated with a marked T lymphocyte and granulocyte infiltration, along with extensive macrophage and dendritic cell recruitment. NK cells and CD4+ T lymphocytes are uninfluential in TSA-LEC cell rejection, whereas both CD8+ lymphocytes and polymorphonuclear leukocytes play a major role. An antitumor immune memory is established very quickly after rejection, since 6 days later 75% of BALB/c mice were already resistant to a TSA-pc challenge. Spleen cells from rejecting mice display specific cytotoxic activity against TSA-pc and secrete IFN-gamma and IL-2 when restimulated by TSA-pc. The ability of LEC to markedly improve recognition of poorly immunogenic cells by promoting APC-T cell cross-talk suggests that it could be an effective component of antitumor vaccines.


Assuntos
Células Apresentadoras de Antígenos/patologia , Movimento Celular/imunologia , Quimiocinas CC/fisiologia , Rejeição de Enxerto/imunologia , Granulócitos/patologia , Memória Imunológica , Linfócitos/patologia , Neoplasias Mamárias Experimentais/imunologia , Adenocarcinoma/imunologia , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Células Apresentadoras de Antígenos/imunologia , Divisão Celular/imunologia , Quimiocinas CC/metabolismo , Feminino , Rejeição de Enxerto/patologia , Reação Enxerto-Hospedeiro/imunologia , Granulócitos/imunologia , Humanos , Linfócitos/imunologia , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células Tumorais Cultivadas
7.
Eur J Immunol ; 30(2): 438-47, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10671199

RESUMO

Systemic lupus erythematosus (SLE)-prone female MRL-lpr / lpr (MRL-lpr) mice were treated with mouse or rat IFN-gamma under different experimental conditions, both prophylactically in 6- to 8 week-old animals and therapeutically in 12- to 18-week-old SLE-affected mice. It was found that IFN-gamma heterogeneously modulated the course of the disease in MRL-lpr mice. When administered prophylactically, IFN-gamma favorably modulated the histological, serological and clinical signs of the disease. Relative to untreated or PBS-treated control animals, the MRL-lpr mice which received IFN gamma were virtually free of inflammatory infiltration of the kidneys and the lungs, had lower levels of azotemia with reduction of both circulating IgG1, IgG2a and IgG3 and anti-double strand (ds) and single strand (ss) DNA antibodies, milder skin vasculitis, significantly reduced enlargement of their lymph nodes and lower weight of the spleens. IFN-gamma also lowered the rate of mortality of MRL-lpr mice. In contrast to these findings, therapeutically administered IFN-gamma worsened the course of the disease in MRL-lpr mice, which exhibited increased proteinuria, higher levels of IgG2a and IgG3 and anti-ds and -ss DNA antibodies, more aggressive nephritis and died at an earlier age than PBS-treated control mice. The dichotomic effect of IFN-gamma on disease manifestation in MRL-lpr mice offers new insights into the complex role of this cytokine in the regulation of systemic autoimmunity such as SLE.


Assuntos
Autoimunidade , Interferon gama/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Animais , Autoanticorpos/imunologia , Feminino , Interferon gama/farmacologia , Lúpus Eritematoso Sistêmico/etiologia , Camundongos , Camundongos Endogâmicos MRL lpr , Ratos
8.
J Virol ; 73(12): 9944-51, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10559307

RESUMO

Herein, we report that Autographa californica nucleopolyhedrovirus, a member of the Baculoviridae family, is capable of stimulating antiviral activity in mammalian cells. Baculoviruses are not pathogenic to mammalian cells. Nevertheless, live baculovirus is shown here to induce interferons (IFN) from murine and human cell lines and induces in vivo protection of mice from encephalomyocarditis virus infection. Monoclonal antibodies specific for the baculovirus envelope gp67 neutralize baculovirus-dependent IFN production. Moreover, UV treatment of baculovirus eliminates both infectivity and IFN-inducing activity. In contrast, the IFN-inducing activity of the baculovirus was unaffected by DNase or RNase treatment. These data demonstrate that IFN production can be induced in mammalian cells by baculovirus even though the cells fail to serve as a natural host for an active viral infection. Baculoviruses, therefore, provide a novel model in which to study at least one alternative mechanism for IFN induction in mammalian cells.


Assuntos
Interferon-alfa/imunologia , Interferon beta/imunologia , Nucleopoliedrovírus/imunologia , Animais , Anticorpos Monoclonais/imunologia , Células COS , Infecções por Cardiovirus/imunologia , Infecções por Cardiovirus/prevenção & controle , DNA/imunologia , Feminino , Humanos , Interferon-alfa/biossíntese , Interferon beta/biossíntese , Lipopolissacarídeos/imunologia , Mamíferos , Vírus Elberfeld do Camundongo/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Mariposas/virologia , Testes de Neutralização , RNA de Cadeia Dupla/imunologia
9.
J Immunol ; 163(9): 4886-93, 1999 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-10528190

RESUMO

BALB/c mammary adenocarcinoma cells engineered to express TNF-related apoptosis-inducing ligand (TRAIL)/APO-2 ligand (APO-2L) on their membrane (TSA-TRAIL) grow with kinetics similar to that of parental cells (TSA-pc) in vitro and in nu/nu mice. In contrast, TSA-TRAIL cells grow faster than TSA-pc in normal BALB/c mice. In DBA/2 mice, which differ from BALB/c mice at minor histocompatibility Ags, they also grow faster and display a higher percentage of tumor takes than TSA-pc. In fully histoincompatible C57BL/6 (B6) mice, TSA-TRAIL cells form evident tumors that are slowly rejected by most mice, but outgrow in a few. In contrast, TSA-pc cells are rejected at once by B6 mice. Since TRAIL/APO-2L induces apoptosis by interacting with a variety of specific receptors, this rapid growth in both syngeneic and allogeneic mice may be the result of an immunosuppressive mechanism. The following evidence supports this hypothesis: 1) TSA-TRAIL cells overcome the strong immunity against TSA-pc cells elicited in BALB/c mice by preimmunization with TSA cells engineered to release IL-4; 2) their rejection by B6 mice does not prime a CTL-mediated memory; 3) thymidine uptake by T lymphocytes unstimulated or stimulated by allogeneic cells is inhibited when TSA-TRAIL cells are added as third party cells; 4) CTL kill TSA-pc but not TSA-TRAIL cells in 48-h assays; and 5) activated lymphocytes interacting with TSA-TRAIL cells in vivo and in vitro undergo apoptosis.


Assuntos
Adenocarcinoma/imunologia , Regulação Neoplásica da Expressão Gênica/imunologia , Neoplasias Mamárias Experimentais/imunologia , Glicoproteínas de Membrana/genética , Linfócitos T/imunologia , Evasão Tumoral/imunologia , Fator de Necrose Tumoral alfa/genética , Adenocarcinoma/genética , Animais , Proteínas Reguladoras de Apoptose , Divisão Celular/genética , Divisão Celular/imunologia , Citotoxicidade Imunológica/genética , Citotoxicidade Imunológica/imunologia , Feminino , Neoplasias Mamárias Experimentais/genética , Glicoproteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Nus , Transplante de Neoplasias , Engenharia de Proteínas , Especificidade da Espécie , Ligante Indutor de Apoptose Relacionado a TNF , Células Tumorais Cultivadas , Evasão Tumoral/genética , Fator de Necrose Tumoral alfa/biossíntese
10.
Autoimmunity ; 30(2): 71-80, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10435719

RESUMO

Interferon (IFN)-gamma and interleukin (IL)-4 are prototypic type 1 and type 2 cytokines which are known to play pathogenetic and protective roles, respectively, in NOD mouse IDDM. The capacity of male NOD mice to produce more IL-4 and less IFN-gamma within the insulitic lesions than females has been suggested to contribute to their lower incidence of diabetes. In this study we have tested the effects of prolonged prophylactic treatment of male NOD mice with rat IFN-gamma, mouse IFN-gamma, anti-IL-4 monoclonal antibody (mAb) and recombinant murine soluble IL-4 receptor (smIL-4R) on the diabetogenic events leading to insulitis and diabetes. None of these treatments influenced spontaneous and/or cyclophosphamide-induced autoimmune diabetogenesis in male NOD mice. Control mice exhibited comparable histological signs of insulitis and incidence of diabetes to those treated with either mouse/rat IFN-gamma or specific IL-4 inhibitors. On the contrary, both clinical and histological signs of diabetes were suppressed by prophylactic treatment with anti-IFN-gamma mAb. These findings indicate that the autoimmune diathesis of male NOD mice towards IDDM cannot be augmented by manipulation of endogenous IFN-gamma or IL-4.


Assuntos
Doenças Autoimunes/etiologia , Diabetes Mellitus Tipo 1/etiologia , Interferon gama/toxicidade , Interleucina-4/antagonistas & inibidores , Animais , Anticorpos Monoclonais/farmacologia , Autoanticorpos/sangue , Autoanticorpos/imunologia , Autoantígenos/imunologia , Doenças Autoimunes/induzido quimicamente , Doenças Autoimunes/prevenção & controle , Células CHO , Cricetinae , Ciclofosfamida/toxicidade , Diabetes Mellitus Tipo 1/induzido quimicamente , Diabetes Mellitus Tipo 1/prevenção & controle , Feminino , Predisposição Genética para Doença , Imunoglobulina E/sangue , Imunoglobulina E/imunologia , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Interferon gama/antagonistas & inibidores , Interleucina-4/sangue , Masculino , Camundongos , Camundongos Endogâmicos NOD , Ratos , Receptores de Interleucina-4/fisiologia , Proteínas Recombinantes/farmacologia , Células Th2/imunologia
11.
J Leukoc Biol ; 65(6): 822-8, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10380905

RESUMO

We have examined the biological activity of the CC chemokine myeloid progenitor inhibitory factor 1 (MPIF-1) on human dendritic cells. MPIF-1 has chemotactic activity on dendritic cells derived from either peripheral blood monocytes or cord blood CD34+ progenitors. However, chemokine treatment did not induce further cell activation or maturation. In addition, MPIF-1 is constitutively released by monocyte-derived dendritic cells but not macrophages or monocytes (resting or stimulated). The proinflammatory stimuli lipopolysaccharide and tumor necrosis factor alpha, which induced the release of monocyte chemotactic protein-1, macrophage inflammatory protein-1alpha (MIP-1alpha), MIP-1beta, and interleukin-8, did not affect MPIF-1 release. In contrast, CD40 ligation and interferon-gamma treatment, while stimulating the production of the other chemokines, caused a pronounced reduction of MPIF-1 transcript and protein release. Thus, in dendritic cells the regulation of the production and release of MPIF-1 is distinct in comparison to other CC and CXC chemokines.


Assuntos
Quimiocinas CC/fisiologia , Células Dendríticas/efeitos dos fármacos , Antineoplásicos/farmacologia , Humanos , Interferon gama/farmacologia , Monócitos/citologia , Proteínas/metabolismo , Proteínas/farmacologia , Receptores do Fator de Necrose Tumoral/fisiologia , Fator 3 Associado a Receptor de TNF , Dedos de Zinco/fisiologia
12.
Diabetes ; 47(1): 32-8, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9421371

RESUMO

Previous studies have shown that anti-gamma-interferon (IFN-gamma) antibody reduces the frequency of autoimmune IDDM in the DP-BB rat. We tested the effects of systemically administered recombinant rat IFN-gamma in both DP-BB and DR-BB rats. Unexpectedly, IFN-gamma markedly reduced the incidence of IDDM as compared with control rats when administered six times per week at a dosage of 280,000 U between ages 30-35 to 105 days or ages 60-64 to 105 days. A lower dosage (28,000 U on alternate days) was also protective when administered to DP-BB rats between birth and age 60 days. However, long-lasting protection against IDDM development over the 1-year study period was achieved only by the highest dosage of IFN-gamma administered from age 30 to 105 days. Ex vivo production of tumor necrosis factor-alpha from splenic lymphoid cells (SLCs) and peritoneal macrophages of the rats treated with IFN-gamma was comparable with that of controls; however, SLCs from the IFN-gamma-treated animals secreted lower amounts of IFN-gamma after stimulation with concanavalin A. IFN-gamma treatment also markedly reduced the frequency of phenotypically activated SLC-expressing class II antigens and interleukin-2 receptor. Finally, in agreement with the observed antidiabetogenic effects, exogenously administered IFN-gamma induced neither insulitis nor IDDM development in DR-BB rats, a subline of DP-BB rats in which autoimmune diabetes rarely occurs spontaneously but can be induced by administration of polyinosinic-polycytidilic acid.


Assuntos
Diabetes Mellitus Experimental/prevenção & controle , Diabetes Mellitus Tipo 1/prevenção & controle , Hipoglicemiantes/farmacologia , Interferon gama/farmacologia , Envelhecimento/fisiologia , Animais , Concanavalina A/farmacologia , Diabetes Mellitus Experimental/epidemiologia , Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Tipo 1/epidemiologia , Diabetes Mellitus Tipo 1/fisiopatologia , Relação Dose-Resposta a Droga , Feminino , Antígenos de Histocompatibilidade Classe II/análise , Antígenos de Histocompatibilidade Classe II/metabolismo , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/uso terapêutico , Imunossupressores/farmacologia , Incidência , Injeções Intraperitoneais , Interferon gama/administração & dosagem , Interferon gama/metabolismo , Interferon gama/uso terapêutico , Macrófagos Peritoneais/citologia , Macrófagos Peritoneais/metabolismo , Masculino , Fenótipo , Distribuição Aleatória , Ratos , Ratos Endogâmicos BB , Receptores de Interleucina-2/análise , Receptores de Interleucina-2/metabolismo , Proteínas Recombinantes , Baço/citologia , Baço/metabolismo , Tacrolimo/farmacologia , Fator de Necrose Tumoral alfa/metabolismo
13.
J Interferon Cytokine Res ; 17 Suppl 1: S23-7, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9241612

RESUMO

A preclinical evaluation of the immunogenicity of various preparations of interferon-alpha (IFN-alpha) was performed with in vitro and in vivo animal models. The distribution of genes for IFN-alpha 2a, IFN-alpha 2b, and IFN-alpha 2c in various cell populations and the response of human T cell clones to IFN-alpha peptides were investigated. The immunogenicity of IFN-alpha in IFN-alpha 2b transgenic mice and factors that influence the immunogenicity of IFN-alpha in normal mice were also studied. The genes for IFN-alpha 2a and IFN-alpha 2b were found in KG-1 cells, whereas IFN-alpha 2b and IFN-alpha 2c genes were present in Namalwa cells. No difference in proliferation of human T cells, T cell lines, or T cell clones could be obtained with IFN-alpha peptides. In transgenic mice bearing the human IFN-alpha 2b gene, no antibody response was obtained following immunization with either IFN-alpha 2a or IFN-alpha 2b. Normal mice immunized with either IFN-alpha 2a or IFN-alpha 2b produced equivalent titers of antibodies, which cross-reacted with both IFNs. Studies evaluating the relative immunogenicity of IFN-alpha in normal mice demonstrated that a number of treatment and host variables can modulate immunogenicity of IFN-alpha preparations.


Assuntos
Interferon-alfa/imunologia , Sequência de Aminoácidos , Animais , Células Cultivadas , Relação Dose-Resposta Imunológica , Genes , Humanos , Interferon alfa-2 , Interferon beta/imunologia , Ativação Linfocitária , Complexo Principal de Histocompatibilidade , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Peptídeos/imunologia , Ligação Proteica , Proteínas Recombinantes , Relação Estrutura-Atividade , Linfócitos T/imunologia
14.
Infection ; 25(4): 206-12, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9266258

RESUMO

Mice infected with bacteria develop an interferon-gamma (IFN-gamma) dependent hypersensitivity to lipopolysaccharide (LPS) and other bacterial components. The broader aim of this study is to find out whether such hypersensitivity also occurs in patients suffering from bacterial infections. The capacity of stimulated peripheral blood cells from infected, intensive-care patients to produce cytokines (IFN-gamma, tumor necrosis factor-alpha (TNF-alpha) and interleukin-6 (IL-6)) was compared to that of healthy donors. Culturing of the cells was carried out preferentially in whole blood diluted 1:3. Whole blood cultures (WBC) were stimulated with lipopolysaccharide (LPS), whole killed Salmonella typhimurium and Staphylococcus aureus and concanavalin A (ConA), and the cytokine production was determined. Two main findings emerged from this study: The IFN-gamma production by WBC of patients was, compared to healthy donors, markedly suppressed, regardless of stimulus used. Further, patients' WBC exhibited a suppressed TNF-alpha production after stimulation with LPS. Surprisingly, following stimulation with bacteria (S. typhimurium and S. aureus) an elevated TNF-alpha and IL-6 response was obtained. Thus, in severely infected patients the cytokine responses of peripheral blood cells to LPS may be suppressed, while the response to other bacterial components is enhanced.


Assuntos
Bacteriemia/imunologia , Concanavalina A/farmacologia , Citocinas/biossíntese , Lipopolissacarídeos/efeitos adversos , Salmonella typhimurium , Staphylococcus aureus , Adolescente , Adulto , Feminino , Humanos , Hidrocortisona/sangue , Unidades de Terapia Intensiva , Interferon gama/biossíntese , Interleucina-6/biossíntese , Leucócitos/imunologia , Leucócitos/microbiologia , Masculino , Pessoa de Meia-Idade , Radioimunoensaio , Fator de Necrose Tumoral alfa/biossíntese
15.
Clin Exp Immunol ; 109(1): 149-56, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9218838

RESUMO

Upon treatment with HgCl2, H-2s mice, such as B10.S, develop an activation of B lymphocytes that depends, at least partially, on activation of T helper type 2 (Th2) cells and results in increased serum levels of IgG1 and IgE, appearance of IgG autoantibodies, and development of immune glomerulonephritis and vasculitis. Results of previous studies and of experiments presented here indicate that the B cell activation and systemic autoimmune disease fail to develop in MHC-congenic B10.D2 (H-2d) and B10.BR (H-2k) mice treated with HgCl2, although B10.D2 T cells showed signs of activation by and specificity for HgCl2 comparable to those seen in strain B10.S. Here, we report that following HgCl2 injections the antibody response to sheep erythrocytes is normal in B10.S, but suppressed in B10.D2 mice. This suppression was prevented by MoAb to mouse IFN-gamma. Conversely, treatment of B10.D2 mice with murine recombinant IFN-gamma (rIFN-gamma) was able to reproduce the immunosuppression seen after HgCl2 treatment. In B10.S mice, it took administration of both rIFN-gamma and HgCl2 to suppress the anti-sheep erythrocyte response. Although rIFN-gamma diminished the increase in IgE serum levels of HgCl2-treated B10.S mice, it failed to prevent their autoantibody production and immune glomerulonephritis. These findings further strengthen the concept that B10.S mice react to HgCl2 by preferential activation of their Th2 cells producing IL-4, whereas B10.D2 mice react to HgCl2 by preferential activation of their Th1 cells, which produce IFN-gamma and thus suppress antibody responses.


Assuntos
Terapia de Imunossupressão , Interferon gama/genética , Interferon gama/imunologia , Cloreto de Mercúrio/imunologia , Animais , Anticorpos Antinucleares , Anticorpos Bloqueadores/imunologia , Formação de Anticorpos , Autoanticorpos/metabolismo , Eritrócitos/imunologia , Feminino , Glomerulonefrite/imunologia , Imunoglobulina E/análise , Imunoglobulina E/sangue , Imunoglobulina E/metabolismo , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Interferon gama/metabolismo , Interleucina-4/metabolismo , Rim/imunologia , Camundongos , Camundongos Endogâmicos , Camundongos Mutantes , Proteínas Recombinantes/imunologia , Ovinos , Células Th1/imunologia , Células Th1/metabolismo , Células Th2/imunologia , Células Th2/metabolismo
16.
FEBS Lett ; 408(2): 211-6, 1997 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-9187369

RESUMO

We have studied the biological properties of a new human CC chemokine, CKbeta8, consisting of 99 amino acids including six cysteines. CKbeta8 mRNA transcripts were induced in monocytes by IL-1beta and, to a lesser extent, by IFNgamma, and were detected in RNA extracted from normal human liver and gastrointestinal tract. CKbeta8 is chemotactic for monocytes, but is inactive on IL-2 conditioned T lymphocytes, eosinophils and neutrophils. Desensitization experiments indicate that CKbeta8 and MIP-1beta completely share receptors on monocytes and that the CKbeta8 receptor, which appears to differ from the known ones, is also recognized by MCP-1, MCP-2, MCP-3, MCP-4, MIP-1alpha and RANTES.


Assuntos
Quimiocinas CC , Quimiocinas/química , Quimiocinas/farmacologia , Monócitos/fisiologia , Acetilglucosaminidase/metabolismo , Sequência de Aminoácidos , Northern Blotting , Cálcio/análise , Cálcio/metabolismo , Quimiocinas/genética , Quimiotaxia de Leucócito , Clonagem Molecular , Citocalasina B/farmacologia , Eosinófilos/efeitos dos fármacos , Corantes Fluorescentes/metabolismo , Fura-2/metabolismo , Regulação da Expressão Gênica , Humanos , Interleucina-2/farmacologia , Dados de Sequência Molecular , Receptores de Citocinas/metabolismo , Alinhamento de Sequência , Análise de Sequência , Linfócitos T/efeitos dos fármacos , Fatores de Virulência de Bordetella/farmacologia
17.
J Exp Med ; 185(7): 1163-72, 1997 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-9104803

RESUMO

Two novel human beta-chemokines, Ck beta-8 or myeloid progenitor inhibitory factor 1 (MPIF-1), and Ck beta-6 or MPIF-2, were discovered as part of a large scale cDNA sequencing effort. The MPIF-1 and MPIF-2 cDNAs were isolated from aortic endothelium and activated monocyte libraries, respectively. Both of the cDNAs were cloned into a baculovirus vector and expressed in insect cells. The mature recombinant MPIF-1 protein consists of 99 amino acids and is most homologous to macrophage inflammatory protein (MIP)-1alpha, showing 51% identity. It displays chemotactic activity on resting T lymphocytes and monocytes, a minimal but significant activity on neutrophils, and is negative on activated T lymphocytes. MPIF-1 is also a potent suppressor of bone marrow low proliferative potential colony-forming cells, a committed progenitor that gives rise to granulocyte and monocyte lineages. The mature recombinant MPIF-2 has 93 amino acid residues and shows 39 and 42% identity with monocyte chemoattractant protein (MCP)-3 and MIP-1alpha, respectively. It displays chemotactic activity on resting T lymphocytes, a minimal activity on neutrophils, and is negative on monocytes and activated T lymphocytes. On eosinophils, MPIF-2 produces a transient rise of cytosolic Ca2+ and uses the receptor for eotaxin and MCP-4. In hematopoietic assays, MPIF-2 strongly suppressed the colony formation by the high proliferative potential colony-forming cell (HPP-CFC), which represents a multipotential hematopoietic progenitor.


Assuntos
Quimiocinas CC , Quimiocinas/isolamento & purificação , Células-Tronco Hematopoéticas/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Sequência de Bases , Cálcio/metabolismo , Quimiocina CCL24 , Quimiocinas/genética , Quimiocinas/farmacologia , Quimiotaxia de Leucócito , Clonagem Molecular , Citosol/metabolismo , DNA/genética , Relação Dose-Resposta a Droga , Humanos , Camundongos , Dados de Sequência Molecular , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Distribuição Tecidual
18.
Endocrinology ; 138(1): 281-8, 1997 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8977415

RESUMO

The role of endogenous interferon-gamma (IFN gamma) in the development of insulin-dependent diabetes mellitus (IDDM) in diabetes-prone BB rats was evaluated. Several groups of these animals were treated under different, experimental conditions with a purified polyclonal antibody (Ab), antirat IFN gamma. The results show that when administered at doses of 100 or 200 micrograms/week from the 30/33th until the 105th day of age, the anti-IFN gamma Ab reversibly reduced the incidence of IDDM compared to that in control rats treated with either irrelevant rabbit IgG or PBS. Moreover, when given up to the 105th day of age, these doses of anti-IFN gamma Abs exerted comparable preventive effects regardless of whether application started as early as within 24 h after birth or at the end of the prediabetic period (e.g. 70/75 days). In contrast, under none of the above experimental conditions did larger doses of anti-IFN gamma Ab (500 micrograms or 1 mg/week) exert antidiabetogenic effects in the BB rats. Apparently, this was due to the exuberant production of neutralizing Abs elicited by the large amount of the xenogeneic Ab injected. At histoimmunological analyses, the BB rats treated with 200 micrograms/ week anti-IFN gamma Abs from 30-80 days of age exhibited a milder insulitic process along with diminished spleen frequency of activated lymphoid cells (MHC class II and interleukin-2 receptor positive). Taken together, these results provide further in vivo evidence for the central pathogenic role of IFN gamma in BB rat IDDM and anticipate the usefulness of specific IFN gamma inhibitors in the prevention of the disease in the clinical setting. Defining novel and less immunogenic forms of specific IFN gamma inhibitors than xenogeneic Abs is important for improving the efficiency of anti-IFN gamma-oriented approaches.


Assuntos
Anticorpos/uso terapêutico , Diabetes Mellitus Tipo 1/prevenção & controle , Interferon gama/antagonistas & inibidores , Animais , Diabetes Mellitus Tipo 1/imunologia , Feminino , Antígenos HLA-DR/análise , Antígenos de Histocompatibilidade Classe II/análise , Imunoglobulina G/biossíntese , Interferon gama/sangue , Ativação Linfocitária , Masculino , Coelhos , Ratos , Ratos Endogâmicos BB
19.
Cytokine Growth Factor Rev ; 8(3): 189-206, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9462485

RESUMO

With the purification and cloning of the interferon gamma (IFN-gamma) receptor chains the mechanism of IFN-gamma action and the resultant signal transduction events were delineated in remarkable detail. The interferon gamma (IFN-gamma) receptor complex consists of two chains: IFN-gammaR1, the ligand-binding chain, and IFN-gammaR2, the accessory chain. Binding of IFN-gamma causes oligomerization of the two IFN-gamma receptor subunits, IFN-gammaR1 and IFN-gammaR2, which initiates the signal transduction events: activation of Jak1 and Jak2 receptor associated protein tyrosine kinases, phosphorylation of the IFN-gammaR1 intracellular domain on Tyr440 followed by phosphorylation and activation of Stat1alpha, the latent transcriptional factor. With all these steps established, the IFN-gamma receptor complex has provided the basic model for understanding the receptors for other members of the family of class II cytokine receptors.


Assuntos
Interferon gama/metabolismo , Receptores de Interferon/genética , Receptores de Interferon/fisiologia , Animais , Humanos , Modelos Biológicos
20.
Endocrinology ; 137(12): 5567-75, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8940385

RESUMO

Previous studies have shown that in vivo treatment with antiinterferon-gamma (anti-IFNgamma) monoclonal antibodies (mAbs) prevents the development of autoimmune diabetes in NOD mice. Although these findings anticipate that specific anti-IFNgamma therapies may be useful for the prevention/treatment of human insulin-dependent diabetes mellitus, there are several reasons why the use of anti-IFNgamma mAb may be difficult in the clinical setting. With the aim to develop alternative forms of specific anti-IFNgamma therapies, we recently produced a nonimmunogenic form of the soluble IFNgamma receptor (sIFNgammaR) that binds and neutralizes murine IFNgamma with an affinity higher than that of anti-IFNgamma mAb. In this study we compared the efficacy of sIFNgammaR to that of two anti-IFNgamma mAbs (XMG 1.2 and AN-18) in the prevention of spontaneous and accelerated (cyclophosphamide-induced) forms of autoimmune diabetes in NOD mice. The results show that in the spontaneous model, sIFNgammaR could prevent histological and clinical signs of autoimmune diabetes as efficiently as the two mAbs. Under ex vivo conditions, sIFNgammaR exhibited a more powerful modulatory effect than XMG 1.2 mAb on cytokine secretion from splenic lymphoid cells, which resulted in a significant reduction of Concanavalin A-induced IL-2 secretion and an augmented release of both unstimulated and lipopolysaccharide-induced IL-6. Moreover, although both mAbs were immunogenic and elicited formation of high titers of anti-rat IgG, sIFNgammaR did not induce antibody formation. Unexpectedly, in the cyclophosphamide-induced model, sIFNgammaR turned out to be less effective than either of the two anti-IFNgamma mAbs. Taken together, these data support the role of IFNgamma in the pathogenesis of NOD mice, but, more importantly, suggest that a nonimmunogenic approach is possible to the diminution of the effects of IFNgamma in this model.


Assuntos
Diabetes Mellitus Tipo 1/prevenção & controle , Receptores de Interferon/fisiologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Formação de Anticorpos , Ciclofosfamida , Diabetes Mellitus Tipo 1/induzido quimicamente , Diabetes Mellitus Tipo 1/genética , Feminino , Interferon gama/imunologia , Camundongos , Camundongos Endogâmicos NOD , Solubilidade , Linfócitos T Reguladores/fisiologia , Linfócitos T Reguladores/transplante , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...