Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Cell Death Dis ; 5: e1102, 2014 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-24603330

RESUMO

Systemic inflammatory response syndromes (SIRS) may be caused by both infectious and sterile insults, such as trauma, ischemia-reperfusion or burns. They are characterized by early excessive inflammatory cytokine production and the endogenous release of several toxic and damaging molecules. These are necessary to fight and resolve the cause of SIRS, but often end up progressively damaging cells and tissues, leading to life-threatening multiple organ dysfunction syndrome (MODS). As inflammasome-dependent cytokines such as interleukin-1ß are critically involved in the development of MODS and death in SIRS, and ATP is an essential activator of inflammasomes in vitro, we decided to analyze the ability of ATP removal to prevent excessive tissue damage and mortality in a murine LPS-induced inflammation model. Our results indeed indicate an important pro-inflammatory role for extracellular ATP. However, the effect of ATP is not restricted to inflammasome activation at all. Removing extracellular ATP with systemic apyrase treatment not only prevented IL-1ß accumulation but also the production of inflammasome-independent cytokines such as TNF and IL-10. In addition, ATP removal also prevented systemic evidence of cellular disintegration, mitochondrial damage, apoptosis, intestinal barrier disruption and even mortality. Although blocking ATP receptors with the broad-spectrum P2 purinergic receptor antagonist suramin imitated certain beneficial effects of apyrase treatment, it could not prevent morbidity or mortality at all. We conclude that removal of systemic extracellular ATP could be a valuable strategy to dampen systemic inflammatory damage and toxicity in SIRS.


Assuntos
Trifosfato de Adenosina/metabolismo , Mediadores da Inflamação/metabolismo , Síndrome de Resposta Inflamatória Sistêmica/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Apoptose , Apirase/farmacologia , Citocinas/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Endotoxinas , Feminino , Hidrólise , Inflamassomos/imunologia , Inflamassomos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/imunologia , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Antagonistas do Receptor Purinérgico P2/farmacologia , Transdução de Sinais , Suramina/farmacologia , Síndrome de Resposta Inflamatória Sistêmica/induzido quimicamente , Síndrome de Resposta Inflamatória Sistêmica/imunologia , Síndrome de Resposta Inflamatória Sistêmica/patologia , Síndrome de Resposta Inflamatória Sistêmica/prevenção & controle , Fatores de Tempo
2.
Cell Death Dis ; 3: e437, 2012 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-23190609

RESUMO

Necrostatin-1 (Nec-1) is widely used in disease models to examine the contribution of receptor-interacting protein kinase (RIPK) 1 in cell death and inflammation. We studied three Nec-1 analogs: Nec-1, the active inhibitor of RIPK1, Nec-1 inactive (Nec-1i), its inactive variant, and Nec-1 stable (Nec-1s), its more stable variant. We report that Nec-1 is identical to methyl-thiohydantoin-tryptophan, an inhibitor of the potent immunomodulatory enzyme indoleamine 2,3-dioxygenase (IDO). Both Nec-1 and Nec-1i inhibited human IDO, but Nec-1s did not, as predicted by molecular modeling. Therefore, Nec-1s is a more specific RIPK1 inhibitor lacking the IDO-targeting effect. Next, although Nec-1i was ∼100 × less effective than Nec-1 in inhibiting human RIPK1 kinase activity in vitro, it was only 10 times less potent than Nec-1 and Nec-1s in a mouse necroptosis assay and became even equipotent at high concentrations. Along the same line, in vivo, high doses of Nec-1, Nec-1i and Nec-1s prevented tumor necrosis factor (TNF)-induced mortality equally well, excluding the use of Nec-1i as an inactive control. Paradoxically, low doses of Nec-1 or Nec-1i, but not Nec -1s, even sensitized mice to TNF-induced mortality. Importantly, Nec-1s did not exhibit this low dose toxicity, stressing again the preferred use of Nec-1s in vivo. Our findings have important implications for the interpretation of Nec-1-based data in experimental disease models.


Assuntos
Imidazóis/administração & dosagem , Imidazóis/química , Indóis/administração & dosagem , Indóis/química , Síndrome de Resposta Inflamatória Sistêmica/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Modelos Animais de Doenças , Tratamento Farmacológico , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/química , Feminino , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Estrutura Molecular , Proteína Serina-Treonina Quinases de Interação com Receptores/antagonistas & inibidores , Especificidade da Espécie
3.
Cell Death Differ ; 18(8): 1316-25, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21311566

RESUMO

Anthracycline antibiotics are inducers of an immunogenic form of apoptosis that has immunostimulatory properties because of the release of damage-associated molecular patterns. To study the mechanisms used by the innate immune system to sense this immunogenic form of cell death, we established an in vivo model of cell death induced by intraperitoneal injection of doxorubicin, a prototype of anthracyclines. The acute sterile inflammation in this model is characterized by rapid influx of neutrophils and increased levels of IL-6 and monocyte chemotactic protein-1. We demonstrate that acute inflammation induced by doxorubicin is associated with apoptosis of monocytes/macrophages and that it is specific for doxorubicin, an immunogenic chemotherapeutic. Further, the inflammatory response is significantly reduced in mice deficient in myeloid differentiation primary response gene 88 (MyD88), TLR-2 or TLR-9. Importantly, a TLR-9 antagonist reduces the recruitment of neutrophils induced by doxorubicin. By contrast, the acute inflammatory response is not affected in TRIF(Lps2) mutant mice and in TLR-3, TLR-4 and caspase-1 knockout mice, which shows that the inflammasome does not have a major role in doxorubicin-induced acute inflammation. Our findings provide important new insights into how the innate immune system senses immunogenic apoptotic cells and clearly demonstrate that the TLR-2/TLR-9-MyD88 signaling pathways have a central role in initiating the acute inflammatory response to this immunogenic form of apoptosis.


Assuntos
Apoptose/imunologia , Doxorrubicina/farmacologia , Inflamação/induzido quimicamente , Inflamação/imunologia , Receptor 2 Toll-Like/imunologia , Receptor Toll-Like 9/imunologia , Animais , Antibióticos Antineoplásicos/farmacologia , Caspase 1/genética , Caspase 1/imunologia , Feminino , Macrófagos/citologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/citologia , Monócitos/imunologia , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/imunologia , Transdução de Sinais/imunologia , Receptor 2 Toll-Like/genética , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia , Receptor Toll-Like 9/antagonistas & inibidores , Receptor Toll-Like 9/genética
4.
Libyan Journal of Medicine ; 3(1): 1-6, 2008.
Artigo em Inglês | AIM (África) | ID: biblio-1265032

RESUMO

Sepsis is a systemic inflammatory response syndrome in the presence of suspected or proven infection; and it may progress to or encompass organ failure (severe sepsis) and hypotension (septic shock). Clinicians possess an arsenal of supportive measures to combat severe sepsis and septic shock; and some success; albeit controversial; has been achieved by using low doses of corticosteroids or recombinant human activated protein C. However; a truly effective mediator-directed specific treatment has not been developed yet. Treatment with low doses of corticosteroids or with recombinant human activated protein C remains controversial and its success very limited. Attempts to treat shock by blocking LPS; TNF or IL-1 were unsuccessful; as were attempts to use interferon-gamma or granulocyte colony stimulating factor. Inhibiting nitric oxide synthases held promise but met with considerable difficulties. Scavenging excess nitric oxide or targeting molecules downstream of inducible nitric oxide synthase; such as soluble guanylate cyclase or potassium channels; might offer other alternatives


Assuntos
Corticosteroides , Óxido Nítrico , Proteína C , Sepse/terapia
5.
Libyan j. med ; 3(1)2008.
Artigo em Inglês | AIM (África) | ID: biblio-1265068

RESUMO

Sepsis is a systemic inflammatory response syndrome in the presence of suspected or proven infection; and it may progress to or encompass organ failure (severe sepsis) and hypotension (septic shock). Clinicians possess an arsenal of supportive measures to combat severe sepsis and septic shock; and some success; albeit controversial; has been achieved by using low doses of corticosteroids or recombinant human activated protein C. However; a truly effective mediator-directed specific treatment has not been developed yet. Treatment with low doses of corticosteroids or with recombinant human activated protein C remains controversial and its success very limited. Attempts to treat shock by blocking LPS; TNF or IL-1 were unsuccessful; as were attempts to use interferon-gamma or granulocyte colony stimulating factor. Inhibiting nitric oxide synthases held promise but met with considerable difficulties. Scavenging excess nitric oxide or targeting molecules downstream of inducible nitric oxide synthase; such as soluble guanylate cyclase or potassium channels; might offer other alternatives


Assuntos
Óxido Nítrico , Proteína C , Sepse
6.
Kidney Int ; 72(5): 557-65, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17538569

RESUMO

Refractory hypotension with end-organ hypoperfusion and failure is an ominous feature of shock. Distributive shock is caused by severe infections (septic shock) or severe systemic allergic reactions (anaphylactic shock). In 1986, it was concluded that nitric oxide (NO) is the endothelium-derived relaxing factor that had been discovered 6 years earlier. Since then, NO has been shown to be important for the physiological and pathological control of vascular tone. Nevertheless, although inhibition of NO synthesis restores blood pressure, NO synthase (NOS) inhibition cannot improve outcome, on the contrary. This implies that NO acts as a double-edged sword during septic shock. Consequently, the focus has shifted towards selective inducible NOS (iNOS) inhibitors. The contribution of NO to anaphylactic shock seems to be more straightforward, as NOS inhibition abrogates shock in conscious mice. Surprisingly, however, this shock-inducing NO is not produced by the inducible iNOS, but by the so-called constitutive enzyme endothelial NOS. This review summarizes the contribution of NO to septic and anaphylactic shock. Although NOS inhibition may be promising for the treatment of anaphylactic shock, the failure of a phase III trial indicates that other approaches are required for the successful treatment of septic shock. Amongst these, high hopes are set for selective iNOS inhibitors. But it might also be necessary to shift gears and focus on downstream cardiovascular targets of NO or on other vasodilating phenomena.


Assuntos
Anafilaxia/tratamento farmacológico , Óxido Nítrico/fisiologia , Anafilaxia/epidemiologia , Anafilaxia/etiologia , Animais , Inibidores Enzimáticos/uso terapêutico , Humanos , Hipotensão/tratamento farmacológico , Hipotensão/etiologia , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Choque Séptico/epidemiologia , Choque Séptico/etiologia
7.
Cell Mol Life Sci ; 62(14): 1632-40, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15990956

RESUMO

Tumor necrosis factor (TNF) is involved in pathologies like septic shock, inflammatory bowel disease and rheumatoid arthritis. TNF and lipopolysaccharide can incite lethal shock, in which cardiovascular collapse is centrally orchestrated by the vasodilating free radical nitric oxide (NO). However, NO synthase (NOS) inhibition causes increased morbidity and/or mortality, suggesting a dual role for NO. To investigate the potential protective role of NO during TNF shock, we treated mice with TNF with or without NOS inhibition. Experiments in endothelial- NOS- and inducible NOS-deficient mice identified inducible NOS as the source of protective NO. Distinctive TNF-induced lipid peroxidation, especially in liver and kidney, was aggravated by NOS inhibition. In addition, various antioxidant treatments and a phospholipase A2 (PLA2) inhibitor prevented sensitization by NOS inhibition. Together, these in vivo results indicate that induced NO not only causes hemodynamic collapse, but is also essential for curbing TNF-induced oxidative stress, which appears to hinge on PLA2-dependent mechanisms.


Assuntos
Óxido Nítrico/metabolismo , Estresse Oxidativo , Choque/metabolismo , Fatores de Necrose Tumoral/farmacologia , Aldeídos/análise , Animais , Antioxidantes/farmacologia , Hidroxianisol Butilado/farmacologia , Catalase/farmacologia , Óxidos N-Cíclicos/farmacologia , Inibidores Enzimáticos/farmacologia , Feminino , Imuno-Histoquímica , Injeções Intravenosas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/fisiologia , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , Fosfolipases A/antagonistas & inibidores , Fosfolipases A2 , Espécies Reativas de Oxigênio/metabolismo , Choque/etiologia , Choque/mortalidade , Marcadores de Spin , Superóxido Dismutase/farmacologia , Taxa de Sobrevida , Fatores de Necrose Tumoral/toxicidade
8.
Immunity ; 13(2): 223-31, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10981965

RESUMO

Hypotension and shock observed in sepsis, SIRS, and tumor necrosis factor (TNF) or cytokine-based cancer treatment are the consequence of excessive nitric oxide (NO) production and subsequent soluble guanylate cyclase (sGC)-mediated vascular smooth muscle relaxation. We demonstrate here that, while NO synthase (NOS) inhibitors exacerbated toxicity, inhibitors of sGC activation protected against TNF-induced lethality, bradycardia, and hypotension. Importantly, sGC inhibition did not interfere with the antitumor activity of TNF. Using NOS inhibitors or iNOS-deficient animals, we furthermore observed that no protection against TNF toxicity could be obtained in the absence of NO. These data imply that iNOS- (and not eNOS-) derived NO is an endogenous protective molecule indispensable to survive a TNF challenge and exerting this beneficial effect via sGC-independent mechanisms.


Assuntos
Inibidores Enzimáticos/farmacologia , Guanilato Ciclase/antagonistas & inibidores , Óxido Nítrico Sintase/metabolismo , Choque/metabolismo , Choque/prevenção & controle , Fator de Necrose Tumoral alfa/toxicidade , Animais , Inibidores Enzimáticos/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo II
9.
J Immunol ; 162(8): 4762-72, 1999 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-10202018

RESUMO

Murine experimental meningitis models induced by either Escherichia coli LPS, live Streptococcus pneumoniae, or Listeria monocytogenes were used to study the origin and potential function of soluble CD14 (sCD14) in the brain during bacterial meningitis. Whereas intracerebral infection caused only a minor and/or transient increase of sCD14 levels in the serum, dramatically elevated concentrations of sCD14 were detected in the cerebrospinal fluid. Reverse-transcriptase PCR and FACS analysis of the leukocytes invading the subarachnoid compartment revealed an active amplification of CD14 transcription and concomitant surface expression. These findings were confirmed by in situ hybridization and immunohistochemical analysis. In contrast, parenchymal astrocytes and microglial cells were shown not to significantly contribute to the elevated levels of sCD14. Simultaneous intracerebral inoculation of rsCD14 and S. pneumoniae resulted in a markedly increased local cytokine response. Taken together, these data provide the first evidence that sCD14 can act as an inflammatory co-ligand in vivo. Thus, during bacterial meningitis, sCD14 is massively released by intrathecal leukocytes, and the sCD14 found in the cerebrospinal fluid can play an important role in the pathogenesis of this disease.


Assuntos
Receptores de Lipopolissacarídeos/fisiologia , Meningites Bacterianas/imunologia , Animais , Astrócitos/metabolismo , Encéfalo/imunologia , Encéfalo/metabolismo , Criança , Modelos Animais de Doenças , Infecções por Escherichia coli/sangue , Infecções por Escherichia coli/líquido cefalorraquidiano , Infecções por Escherichia coli/imunologia , Feminino , Humanos , Injeções Subcutâneas , Leucócitos/imunologia , Leucócitos/metabolismo , Receptores de Lipopolissacarídeos/sangue , Receptores de Lipopolissacarídeos/líquido cefalorraquidiano , Receptores de Lipopolissacarídeos/genética , Listeriose/sangue , Listeriose/líquido cefalorraquidiano , Listeriose/imunologia , Meningites Bacterianas/sangue , Meningites Bacterianas/líquido cefalorraquidiano , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Infecções Pneumocócicas/sangue , Infecções Pneumocócicas/líquido cefalorraquidiano , Infecções Pneumocócicas/imunologia , RNA Mensageiro/biossíntese , Proteínas Recombinantes/administração & dosagem , Solubilidade
10.
J Bacteriol ; 181(4): 1126-33, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9973337

RESUMO

The Escherichia coli Pst system belongs to the family of ABC transporters. It is part of a phosphate (PHO) regulon which is regulated by extracellular phosphate. Under conditions of phosphate limitation, the response regulator PhoB is phosphorylated by the histidine kinase PhoR and binds to promoters that share a consensus PHO box. Under conditions of phosphate excess, PhoR, Pst, and PhoU downregulate the PHO regulon. Screening of a library of pneumococcal mutants with defects in exported proteins revealed a putative two-component regulatory system, PnpR-PnpS, and a downstream ABC transporter, similar to the Pst system in E. coli including a gene encoding a PhoU protein. Similar to E. coli, mutagenesis of the ATP-binding cassette gene, pstB, resulted in decreased uptake of phosphate. The effects of the loss of the pneumococcal Pst system extended to decreased transformation and lysis. Withdrawal of phosphate led to transformation deficiency in the parent strain R6x but not to penicillin tolerance, suggesting that reduced bacterial death was independent of phosphate. None of these phenotypes was observed in the pneumococcal loss-of-function mutant phoU. By using a lacZ reporter construct, it was demonstrated that expression of the two-component regulatory system PnpR-PnpS was not influenced by different concentrations of phosphate. These results suggest a more complex role of the Pst system in pneumococcal physiology than in that of E. coli.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Proteínas de Bactérias/genética , Genes Bacterianos , Fosfatos/metabolismo , Fosfotransferases , Streptococcus pneumoniae/genética , Dados de Sequência Molecular , Mutação , N-Acetil-Muramil-L-Alanina Amidase/biossíntese , Transdução de Sinais/genética
11.
Infect Immun ; 65(8): 3255-60, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9234783

RESUMO

The cell wall is a key inflammatory agent of gram-positive bacteria. Possible receptors mediating cell wall-induced inflammation include CD14 and platelet-activating factor (PAF) receptor. To delineate the conditions under which these various receptors might be used, human monocytic THP-1 cells and heparinized whole human blood were stimulated with lipopolysaccharide (LPS), intact Streptococcus pneumoniae bacteria, or purified pneumococcal cell wall. THP-1 culture supernatant or cell-free plasma was analyzed for the presence of tumor necrosis factor, interleukin-1beta (IL-1beta), and IL-6. For the cultured monocytes, anti-CD14 inhibited induction of the inflammatory cytokines by the cell wall and LPS but not by intact pneumococcal bacteria. Despite the difference in CD-14 usage, the intracellular pathways induced by the three agents demonstrated similarities, as revealed in the presence of specific signal transduction inhibitors such as cholera toxin, pertussis toxin, and genistein. Cytokine production in whole human blood indicated that anti-CD14 failed to block responses to cell wall and intact pneumococci, whereas while LPS-induced responses were inhibited. PAF receptor antagonist had no effect under any conditions in both assays. These results indicate that although cell walls bind to both CD14 and PAF receptor, only CD14 appears to engender a cytokine response under restricted conditions. Furthermore, host cell responses to intact pneumococci are consistently independent of CD14 and PAF receptor.


Assuntos
Citocinas/biossíntese , Bactérias Gram-Positivas/fisiologia , Receptores de Lipopolissacarídeos/fisiologia , Monócitos/fisiologia , Receptores de Superfície Celular , Receptores Acoplados a Proteínas G , Humanos , Glicoproteínas da Membrana de Plaquetas/fisiologia , Células Tumorais Cultivadas
12.
J Immunol ; 156(12): 4686-90, 1996 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-8648113

RESUMO

Human TNF, a specific agonist of murine (m)TNF-R55, is fairly harmless in healthy mice, but readily induces lethality in animals sensitized by a malignant tumor or a bacterial infection. We here report that IL-12 is both a necessary and sufficient mediator in Calmette-Guérin bacillus-induced sensitization to the lethal effects of hTNF. Evidence for this involvement is provided by the protection observed after anti-mIL-12 Ab treatment and by the fact that Calmette-Guérin bacillus-induced sensitization can be mimicked by repetitive mIL-12 administration. IL-12 exerts this effect by inducing IFN-gamma in NK cells. Tumor-induced sensitization, however, does not seem to involve mIL-12.


Assuntos
Interleucina-12/fisiologia , Mycobacterium bovis/imunologia , Fator de Necrose Tumoral alfa/toxicidade , Animais , Antígenos CD/fisiologia , Feminino , Humanos , Interferon gama/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Interferon/fisiologia , Tuberculose/imunologia , Receptor de Interferon gama
13.
J Immunol ; 154(6): 2753-63, 1995 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-7876546

RESUMO

In healthy mice, murine (m) TNF is fairly lethal, whereas human (h) TNF (a selective murine TNF-R55 agonist) is rather harmless. However, we and others observed that mice suffering from a bacterial infection, such as Bacillus Calmette-Guérin (BCG), or bearing i.m. some types of tumor, develop a hypersensitivity to the IL-6-inducing and lethal properties of hTNF. This is a cardinal problem as it severely limits the potential use of hTNF-R55-specific agonists for systemic treatment of human cancer. Using mice carrying a targeted disruption in the gene encoding the IFN-gamma receptor (IFN-gamma Ro/o), we here report that endogenous IFN-gamma plays a crucial role in the development of TNF hypersensitivity during BCG infection. Indeed, both the lethality and the IL-6 induced by hTNF were drastically reduced in IFN-gamma Ro/o mice as compared with control mice. These results demonstrate that the enhancement of TNF effects is at least an equally important mechanism by which IFN-gamma contributes to BCG-induced hypersensitivity as the previously described augmentation of TNF production. Experiments in athymic nude mice, either depleted of NK cells or not, revealed that the latter cell population is an important source of the sensitizing IFN-gamma during BCG infection. In contrast, IFN-gamma Ro/o mice were as susceptible as control mice to the sensitizing effects of tumors. mTNF, which interacts with both mTNF-R55 and mTNF-R75 and causes lethality on its own, is as toxic in IFN-gamma Ro/o mice as in wt control mice; this means that TNF-induced IFN-gamma does not play a role in mTNF-induced lethality.


Assuntos
Interferon gama/fisiologia , Mycobacterium bovis/imunologia , Neoplasias Experimentais/imunologia , Tuberculose/imunologia , Fator de Necrose Tumoral alfa/toxicidade , Animais , Feminino , Interleucina-6/biossíntese , Células Matadoras Naturais/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Neoplasias Experimentais/mortalidade , Proteínas Recombinantes/toxicidade , Linfócitos T/imunologia , Fator de Necrose Tumoral alfa/imunologia
14.
J Inflamm ; 47(1-2): 18-26, 1995.
Artigo em Inglês | MEDLINE | ID: mdl-8913926

RESUMO

The extension of the positive results obtained with tumor necrosis factor (TNF) in the locoregional treatment of cancer to systemic treatments requires the selective inhibition of its shock-inducing properties. In this paper, recent data regarding the mechanisms by which infections and tumors render mice extremely sensitive to the lethal effects of TNF as well as regarding the inhibition of the dose-limiting toxicities, hypotension and hepatotoxicity, are summarized. An interleukin-12 (IL-12) driven induction of interferon-gamma (IFN-gamma), probably in synergism with endogenous TNF, was found to mediate infection-induced sensitization. The sensitization induced by tumors develops independent of the IL-12/IFN-gamma axis but ultimately leads to a common step, which can be inhibited by alpha-CD11a and is specific for sensitization. Hypotension can be inhibited by methylene blue (MB), an inhibitor of the nitric oxide (NO)-induced activation of the cytosolic guanylate cyclase, without the indispensable protective properties of NO being affected. Finally, two acute phase proteins, alpha 1-acid-glycoprotein (AGP) and alpha 1-antitrypsin (AT), were able to protect against the TNF-induced liver failure. None of these three inhibitors seems to affect the antitumor effects of TNF.


Assuntos
Doenças Transmissíveis/tratamento farmacológico , Neoplasias Experimentais/tratamento farmacológico , Neoplasias/tratamento farmacológico , Fator de Necrose Tumoral alfa/efeitos adversos , Animais , Citocinas/uso terapêutico , Interações Medicamentosas , Humanos , Camundongos , Fator de Necrose Tumoral alfa/uso terapêutico
15.
Eur J Immunol ; 24(9): 2237-42, 1994 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-7522168

RESUMO

Whether interleukin (IL)-6 contributes to tumor necrosis factor (TNF)-induced lethal shock or whether, on the contrary, it is part of a protective feedback system, remains unresolved. Here, we report experiments with IL-6 gene-disrupted mice (IL-6(0/0)). We have tested the susceptibility of these to TNF-induced metabolic changes and lethality in different models, and compared the results with those obtained with IL-6+/+ wild-type mice. We studied the response to TNF in three different models: (i) murine TNF administration; (ii) TNF in galactosamine (GalN)-sensitized mice; (iii) TNF in Bacillus Calmette-Guérin-sensitized mice. We observed no significant difference between the two types of mice in any of the three models. Furthermore, IL-6(0/0) mice could be equally well desensitized (by IL-1) to TNF/GalN-induced lethality and tolerized to TNF-induced shock as IL-6+/+ mice. We also observed that, in response to turpentine, TNF or IL-1, IL-6(0/0) mice produced significantly less acute phase proteins (APP) than IL-6+/+ mice. In IL-6(0/0) mice, less corticosterone was induced by TNF than in the control mice, while the response to adrenocorticotropic hormone was the same. The results indicate that IL-6 is not contributing in a major way to the pathogenesis leading to TNF-induced shock, and that neither IL-6 nor the APP studied are essential for a protective feedback system.


Assuntos
Interleucina-6/fisiologia , Fator de Necrose Tumoral alfa/toxicidade , Proteínas de Fase Aguda/biossíntese , Animais , Vacina BCG/imunologia , Corticosterona/biossíntese , Galactosamina/toxicidade , Tolerância Imunológica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Terebintina/toxicidade
16.
Circ Shock ; 43(4): 185-90, 1994 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-7895324

RESUMO

The use of TNF-mutants that are selective agonists of the TNF-R55 is one strategy that is being explored to broaden the therapeutic margin of TNF. Several problems still have to be overcome before they can be used in clinical trials. Regarding the sensitizing effect of some infections and some tumours, we identified IFN-gamma as a mediator in BCG- but not in tumour-induced sensitization. In both models, the vessel wall is most probably the key tissue as alpha-LFA-1 antibodies could protect against lethality. Studies in primates showed that an unexpected feature, namely, the longer half-life of such mutants, might interfere with this strategy. Recent observations also indicate that the mechanism of tolerance-induction, another way to separate antitumour and toxic effects of TNF, might reside in the functional ablation of the TNF-R75. Using IL-60/0 knockout mice, we could not find any causal role for IL-6 in TNF-mediated lethality, this in contrast to results obtained previously with neutralizing antibodies. Finally, we identified the acute phase protein alpha 1-acid glycoprotein as a protein with protective properties towards TNF-induced lethality and liver damage.


Assuntos
Antígenos CD , Neoplasias Experimentais/tratamento farmacológico , Receptores do Fator de Necrose Tumoral/agonistas , Fator de Necrose Tumoral alfa/uso terapêutico , Animais , Tolerância a Medicamentos , Mutação , Receptores Tipo I de Fatores de Necrose Tumoral
17.
Immunobiology ; 187(3-5): 317-29, 1993 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8392490

RESUMO

Cytokines are important mediators of the effects observed after the administration of endotoxin. One of them, tumor necrosis factor, is particularly important since it plays a cardinal role in two major endotoxin activities: its antitumor effect and its capacity to induce a systemic inflammatory response syndrome. TNF exerts its activity on a wide variety of target cells by the triggering of two distinct receptor types. TNF-R55 and TNF-R75. They induce distinct intracellular signals but can have cooperative effects. So, their differential triggering or modulation may have clinically relevant consequences. Based upon observations in the mouse, where hTNF does not interact with the TNF-R75 while mTNF triggers both receptor types, we propose that both receptors need to be triggered to obtain lethality after the administration of TNF. Since human TNF has retained antitumor activity, esp. in combination with IFN-gamma, TNF-mutants that are selective agonists for the TNF-R55 might have a broader therapeutic margin. One such human TNF mutant was already shown to be as effective as the wild-type hTNF in a xenograft model. However, several sensitizing agents may mimic TNF-R75 triggering and so make TNF-R55 triggering a lethal challenge. The fact that two such agents, RU38486 and IL-1 have similar effects regarding their kinetics and their capacity to sensitize for the lethality- and IL-6-inducing effect of hTNF may give a hint regarding the mechanism of the sensitizing effect.


Assuntos
Interleucina-1/imunologia , Interleucina-6/imunologia , Receptores de Superfície Celular/imunologia , Fator de Necrose Tumoral alfa/imunologia , Animais , Humanos , Receptores do Fator de Necrose Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...