Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Adv Funct Mater ; 31(37)2021 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-37745940

RESUMO

Solid tumors are protected from antitumor immune responses due to their hypoxic microenvironments. Weakening hypoxia-driven immunosuppression by hyperoxic breathing of 60% oxygen has shown to be effective in unleashing antitumor immune cells against solid tumors. However, efficacy of systemic oxygenation is limited against solid tumors outside of lungs and has been associated with unwanted side effects. As a result, it is essential to develop targeted oxygenation alternatives to weaken tumor hypoxia as novel approaches to restore immune responses against cancer. Herein, we report on injectable oxygen-generating cryogels (O2-cryogels) to reverse tumor-induced hypoxia. These macroporous biomaterials were designed to locally deliver oxygen, inhibit the expression of hypoxia-inducible genes in hypoxic melanoma cells, and reduce the accumulation of immunosuppressive extracellular adenosine. Our data show that O2-cryogels enhance T cell-mediated secretion of cytotoxic proteins, restoring the killing ability of tumor-specific CTLs, both in vitro and in vivo. In summary, O2-cryogels provide a unique and safe platform to supply oxygen as a co-adjuvant in hypoxic tumors and have the potential to improve cancer immunotherapies.

2.
Eur J Immunol ; 43(3): 655-66, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23208786

RESUMO

Hypoxia-adenosinergic suppression and redirection of the immune response has been implicated in the regulation of antipathogen and antitumor immunity, with hypoxia-inducible factor 1α (HIF-1α) playing a major role. In this study, we investigated the role of isoform I.1, a quantitatively minor alternative isoform of HIF-1α, in antibacterial immunity and sepsis survival. By using the cecal ligation and puncture model of bacterial peritonitis, we studied the function of I.1 isoform in T cells using mice with total I.1 isoform deficiency and mice with T-cell-targeted I.1 knockdown. We found that genetic deletion of the I.1 isoform resulted in enhanced resistance to septic lethality, significantly reduced bacterial load in peripheral blood, increased M1 macrophage polarization, augmented levels of proinflammatory cytokines in serum, and significantly decreased levels of the anti-inflammatory cytokine IL-10. Our data suggest a previously unrecognized immunosuppressive role for the I.1 isoform in T cells during bacterial sepsis. We interpret these data as indicative that the activation-inducible isoform I.1 hinders the contribution of T cells to the antibacterial response by affecting M1/M2 macrophage polarization and microbicidal function.


Assuntos
Infecções Bacterianas/genética , Infecções Bacterianas/imunologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Peritonite/genética , Peritonite/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Animais , Infecções Bacterianas/mortalidade , Modelos Animais de Doenças , Subunidade alfa do Fator 1 Induzível por Hipóxia/deficiência , Ativação Linfocitária/imunologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Knockout , Especificidade de Órgãos/genética , Peritonite/mortalidade , Isoformas de Proteínas , Receptores de Antígenos de Linfócitos T/metabolismo , Sepse/genética , Sepse/imunologia , Sepse/mortalidade , Ativação Transcricional
3.
J Immunol ; 184(1): 154-63, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19949104

RESUMO

Hypoxia-inducible factor (HIF)-1alpha plays a central role in oxygen homeostasis and energy supply by glycolysis in many cell types. We previously reported that an HIF-1alpha gene deficiency caused abnormal B cell development and autoimmunity. In this study we show that HIF-1alpha-enabled glycolysis during B cell development is required in a developmental stage-specific manner. Supporting this conclusion are observations that the glycolytic pathway in HIF-1alpha-deficient B220(+) bone marrow cells is much less functionally effective than in wild-type control cells. The expression of genes encoding the glucose transporters and the key glycolytic enzyme, 6-phosphofructo-2-kinase/fructose-2,6-bishosphatase 3, was greatly reduced in HIF-1alpha-deficient cells. The compensatory adaptation to the defect of glycolysis was reflected in higher levels of expression of respiratory chain-related genes and TCA cycle-related genes in HIF-1alpha-deficient cells than in wild-type cells. In agreement with these findings, HIF-1alpha-deficient cells used pyruvate more efficiently than wild-type cells. The key role of HIF-1alpha-enabled glycolysis in bone marrow B cells was also demonstrated by glucose deprivation during in vitro bone marrow cell culture and by using a glycolysis inhibitor in the bone marrow cell culture. Taken together, these findings indicate that glucose dependency differs at different B cell developmental stages and that HIF-1alpha plays an important role in B cell development.


Assuntos
Linfócitos B/citologia , Linfócitos B/metabolismo , Diferenciação Celular/fisiologia , Glicólise/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/imunologia , Animais , Linfócitos B/imunologia , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Citometria de Fluxo , Expressão Gênica , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
J Biol Chem ; 285(50): 39271-88, 2010 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-20926384

RESUMO

The A2A and A2B adenosine receptors (A2AR and A2BR) are implicated in many physiological processes. However, the mechanisms of their intracellular maturation and trafficking are poorly understood. In comparative studies of A2AR versus A2BR expression in transfected cells, we noticed that the levels of cell surface expression of A2BR were significantly lower than those of A2AR. A large portion of the A2BR was degraded by the proteasome. Studies of cell surface expression of A2BR chimeric molecules in transfectants suggested that A2BR does not have the dominant forward transport signal for export from the endoplasmic reticulum to the cell surface. A2BR surface expression was increased in A2BR chimeras where the A2BR carboxyl terminus (CT) was replaced or fused with the A2AR CT. Co-transfection of A2AR with A2BR enhanced surface expression of A2BR though the F(X)(6)LL motif in the A2AR CT. The requirements of A2AR expression for better A2BR cell surface expression was not only established in transfectants but also confirmed by observations of much lower levels of A2BR-induced intracellular cAMP accumulation in response to A2BR-activating ligand in splenocytes from A2AR(-/-) mice than in wild type mice. The results of mechanistic studies suggested that poor A2BR expression at the cell surface might be accounted for mainly by the lack of a dominant forward transport signal from the endoplasmic reticulum to the plasma membrane; it is likely that A2BR forms a hetero-oligomer complex for better function.


Assuntos
Regulação da Expressão Gênica , Receptor A2A de Adenosina/fisiologia , Receptor A2B de Adenosina/fisiologia , Animais , Transporte Biológico , Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Complexo de Endopeptidases do Proteassoma/metabolismo , Estrutura Terciária de Proteína , Receptor A2A de Adenosina/biossíntese , Receptor A2B de Adenosina/biossíntese , Transdução de Sinais
7.
Cancer Discov ; 10(1): 16-19, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31919119

RESUMO

In this issue of Cancer Discovery, Fong and colleagues describe the encouraging observations of tumor regression, disease control, and survival of patients with otherwise refractory renal cell cancer with progressive disease after treatment with the conceptually novel oral antagonist of the A2A adenosine receptor (A2AR), ciforadenant. A2AR antagonists may represent the until now missing but critically important part of more effective immunotherapies of cancer, because they prevent the inhibition of tumor-reactive T and natural killer cells by blocking the immunosuppressive hypoxia-A2A-adenosinergic signaling, which represents an emerging immunosuppressive hallmark of tumors that are the most resistant to therapies.See related article by Fong et al., p. 40.


Assuntos
Carcinoma de Células Renais , Adenosina , Humanos , Imunoterapia , Antagonistas de Receptores Purinérgicos P1 , Receptor A2A de Adenosina
8.
Curr Opin Pharmacol ; 53: 98-100, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32861959

RESUMO

The anti-hypoxia-A2-Adenosinergic immunotherapies of cancer emerged as the only available now approach to enable the tumor rejection in those progressing cancer patients that are refractory to all other current treatments. Several different classes of drugs are offered to inhibit the Hypoxia-HIF-1alpha-mediated and extracellular adenosine-A2A adenosine receptor-mediated immunosuppressive signaling in tumor microenvironment. It is suggested that the most promising treatments must include the blockade of cAMP-elevating A2A adenosine receptors and the elimination of hypoxia in tumors by oxygenation agents and hyperoxic breathing. The observations in ongoing clinical trials support this conclusion.


Assuntos
Antagonistas do Receptor A2 de Adenosina/uso terapêutico , Adenosina/imunologia , Imunoterapia , Neoplasias/terapia , Receptor A2A de Adenosina/imunologia , Linfócitos T/imunologia , Hipóxia Tumoral/efeitos dos fármacos , Animais , Humanos , Neoplasias/imunologia
9.
J Clin Invest ; 130(11): 5629-5637, 2020 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-32870821

RESUMO

Hypoxia/HIF-1α- and extracellular adenosine/A2 adenosine receptor-mediated immunosuppression protects tissues from collateral damage by antipathogen immune cells. However, this mechanism also protects cancerous tissues by inhibiting antitumor immune cells in hypoxic and extracellular adenosine-rich tumors that are the most resistant to current therapies. Here, we explain a potentially novel, antiimmunosuppressive reasoning to justify strategies using respiratory hyperoxia and oxygenation agents in cancer treatment. Earlier attempts to use oxygenation of tumors as a monotherapy or to improve radiotherapy have failed because oxygenation protocols were not combined with immunotherapies of cancer. In contrast, the proposal for therapeutic use of antihypoxic oxygenation described here was motivated by the need to prevent the hypoxia/HIF-1α-driven accumulation of extracellular adenosine to (a) unleash antitumor immune cells from inhibition by intracellular cAMP and (b) prevent immunosuppressive transcription of cAMP response element- and hypoxia response element-containing immunosuppressive gene products (e.g., TGF-ß). Use of oxygenation agents together with inhibitors of the A2A adenosine receptor may be required to enable the most effective cancer immunotherapy. The emerging outcomes of clinical trials of cancer patients refractory to all other treatments provide support for the molecular and immunological mechanism-based approach to cancer immunotherapy described here.


Assuntos
Antineoplásicos/uso terapêutico , Hiperóxia , Imunoterapia , Neoplasias , Oxigênio/uso terapêutico , Animais , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/terapia
10.
Clin Cancer Res ; 14(19): 5947-52, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18829471

RESUMO

Cancerous tissue protection from tumor-recognizing CD8(+) and CD4(+) T cells (antitumor T cells) limits the therapeutic potential of immunotherapies. We propose that tumor protection is to a large extent due to (a) inhibition of antitumor T cells by hypoxia-driven accumulation of extracellular adenosine in local tumor microenvironment and due to (b) T regulatory cell-produced extracellular adenosine. The adenosine triggers the immunosuppressive signaling via intracellular cyclic AMP-elevating A2A adenosine receptors (A2AR) on antitumor T cells. In addition, the activated antitumor T cells in hypoxic tumor microenvironment could be inhibited by elevated levels of immunosuppressive hypoxia-inducible factor-1alpha. Complete rejection or tumor growth retardation was observed when A2AR has been genetically eliminated or antagonized with synthetic drug or with natural A2AR antagonist 1,3,7-trimethylxanthine (caffeine). The promising strategy may be in combining the anti-hypoxia-adenosinergic treatment that prevents inhibition of antitumor T cells by tumor-produced and T regulatory cell-produced adenosine with targeting of other negative regulators, such as CTL antigen-4 blockade. Observations of tumor rejection in mice and massive prospective epidemiologic studies support the feasibility of anti-hypoxia-adenosinergic combined immunotherapy.


Assuntos
Hipóxia , Imunossupressores/farmacologia , Neoplasias/metabolismo , Receptor A2A de Adenosina/metabolismo , Linfócitos T Reguladores/metabolismo , Adenosina/metabolismo , Animais , Linfócitos T CD8-Positivos/metabolismo , Cafeína/farmacologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Imunoterapia/métodos , Camundongos , Modelos Biológicos , Neoplasias/imunologia , Linfócitos T Citotóxicos/metabolismo
11.
PLoS Biol ; 3(6): e174, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15857155

RESUMO

Acute respiratory distress syndrome (ARDS) usually requires symptomatic supportive therapy by intubation and mechanical ventilation with the supplemental use of high oxygen concentrations. Although oxygen therapy represents a life-saving measure, the recent discovery of a critical tissue-protecting mechanism predicts that administration of oxygen to ARDS patients with uncontrolled pulmonary inflammation also may have dangerous side effects. Oxygenation may weaken the local tissue hypoxia-driven and adenosine A2A receptor (A2AR)-mediated anti-inflammatory mechanism and thereby further exacerbate lung injury. Here we report experiments with wild-type and adenosine A2AR-deficient mice that confirm the predicted effects of oxygen. These results also suggest the possibility of iatrogenic exacerbation of acute lung injury upon oxygen administration due to the oxygenation-associated elimination of A2AR-mediated lung tissue-protecting pathway. We show that this potential complication of clinically widely used oxygenation procedures could be completely prevented by intratracheal injection of a selective A2AR agonist to compensate for the oxygenation-related loss of the lung tissue-protecting endogenous adenosine. The identification of a major iatrogenic complication of oxygen therapy in conditions of acute lung inflammation attracts attention to the need for clinical and epidemiological studies of ARDS patients who require oxygen therapy. It is proposed that oxygen therapy in patients with ARDS and other causes of lung inflammation should be combined with anti-inflammatory measures, e.g., with inhalative application of A2AR agonists. The reported observations may also answer the long-standing question as to why the lungs are the most susceptible to inflammatory injury and why lung failure usually precedes multiple organ failure.


Assuntos
Inflamação/fisiopatologia , Consumo de Oxigênio , Oxigênio/toxicidade , Síndrome do Desconforto Respiratório/fisiopatologia , Animais , Progressão da Doença , Enterotoxinas/toxicidade , Humanos , Hipóxia/fisiopatologia , Inflamação/imunologia , Lipopolissacarídeos/toxicidade , Camundongos , Síndrome do Desconforto Respiratório/induzido quimicamente , Síndrome do Desconforto Respiratório/imunologia
12.
PLoS One ; 12(11): e0187314, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29155844

RESUMO

Human cancers are known to downregulate Major Histocompatibility Complex (MHC) class I expression thereby escaping recognition and rejection by anti-tumor T cells. Here we report that oxygen tension in the tumor microenvironment (TME) serves as an extrinsic cue that regulates antigen presentation by MHC class I molecules. In support of this view, hypoxia is shown to negatively regulate MHC expression in a HIF-dependent manner as evidenced by (i) lower MHC expression in the hypoxic TME in vivo and in hypoxic 3-dimensional (3D) but not 2-dimensional (2D) tumor cell cultures in vitro; (ii) decreased MHC in human renal cell carcinomas with constitutive expression of HIF due to genetic loss of von Hippel-Lindau (VHL) function as compared with isogenically paired cells with restored VHL function, and iii) increased MHC in tumor cells with siRNA-mediated knockdown of HIF. In addition, hypoxia downregulated antigen presenting proteins like TAP 1/2 and LMP7 that are known to have a dominant role in surface display of peptide-MHC complexes. Corroborating oxygen-dependent regulation of MHC antigen presentation, hyperoxia (60% oxygen) transcriptionally upregulated MHC expression and increased levels of TAP2, LMP2 and 7. In conclusion, this study reveals a novel mechanism by which intra-tumoral hypoxia and HIF can potentiate immune escape. It also suggests the use of hyperoxia to improve tumor cell-based cancer vaccines and for mining novel immune epitopes. Furthermore, this study highlights the advantage of 3D cell cultures in reproducing hypoxia-dependent changes observed in the TME.


Assuntos
Hipóxia Celular/imunologia , Genes MHC Classe I/imunologia , Fator 1 Induzível por Hipóxia/genética , Neoplasias Renais/imunologia , Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/genética , Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/imunologia , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/patologia , Regulação Neoplásica da Expressão Gênica , Humanos , Fator 1 Induzível por Hipóxia/imunologia , Neoplasias Renais/genética , Neoplasias Renais/patologia , Oxigênio/metabolismo , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/imunologia , Linfócitos T/imunologia , Ativação Transcricional/genética , Ativação Transcricional/imunologia , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia , Proteína Supressora de Tumor Von Hippel-Lindau/genética , Proteína Supressora de Tumor Von Hippel-Lindau/imunologia
13.
Cell Rep ; 21(13): 3672-3680, 2017 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-29281817

RESUMO

Rapidly evolving pathogens such as HIV or influenza can quickly mutate their antigenic profiles, reducing the efficacy of conventional vaccines. Despite this challenge, functionally required epitopes are highly conserved among heterologous viral strains and represent a key vulnerability that could be targeted during vaccine development. As the antigenicity of these conserved epitopes is frequently subdominant, there is a critical need for innovative vaccination strategies designed to target these neutralizing epitopes. Here, we immunized mice with antigens containing discrete immunodominant and subdominant moieties and show that treatment with soluble heterologous antigen bearing only the immunodominant epitope selectively suppresses these germinal center (GC) B cells. By exploiting this intrinsic tolerance mechanism, we promote the expansion of subdominant B cells in the GC and the subsequent long-lived components of the humoral response. We propose that this strategy may be applied to elicit preferential expansion of subdominant B cells that recognize weakly immunogenic epitopes on microbial pathogens.


Assuntos
Linfócitos B/metabolismo , Centro Germinativo/metabolismo , Epitopos Imunodominantes/metabolismo , Animais , Formação de Anticorpos , Contagem de Células , Células Clonais , Camundongos Endogâmicos C57BL , Nitrofenóis/química , Ovalbumina/imunologia , Fenilacetatos/química , Plasmócitos/metabolismo , Solubilidade
14.
Cancer Res ; 76(11): 3319-31, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-26988986

RESUMO

Proteasome-regulated NF-κB has been shown to be important for cell survival in T-cell lymphoma and Hodgkin lymphoma models. Several new small-molecule proteasome inhibitors are under various stages of active preclinical and clinical development. We completed a comprehensive preclinical examination of the efficacy and associated biologic effects of a second-generation proteasome inhibitor, ixazomib, in T-cell lymphoma and Hodgkin lymphoma cells and in vivo SCID mouse models. We demonstrated that ixazomib induced potent cell death in all cell lines at clinically achievable concentrations. In addition, it significantly inhibited tumor growth and improved survival in T-cell lymphoma and Hodgkin lymphoma human lymphoma xenograft models. Through global transcriptome analyses, proteasomal inhibition showed conserved overlap in downregulation of cell cycle, chromatin modification, and DNA repair processes in ixazomib-sensitive lymphoma cells. The predicted activity for tumor suppressors and oncogenes, the impact on "hallmarks of cancer," and the analysis of key significant genes from global transcriptome analysis for ixazomib strongly favored tumor inhibition via downregulation of MYC and CHK1, its target genes. Furthermore, in ixazomib-treated lymphoma cells, we identified that CHK1 was involved in the regulation of MYC expression through chromatin modification involving histone H3 acetylation via chromatin immunoprecipitation. Finally, using pharmacologic and RNA silencing of CHK1 or the associated MYC-related mechanism, we demonstrated synergistic cell death in combination with antiproteasome therapy. Altogether, ixazomib significantly downregulates MYC and induces potent cell death in T-cell lymphoma and Hodgkin lymphoma, and we identified that combinatorial therapy with anti-CHK1 treatment represents a rational and novel therapeutic approach. Cancer Res; 76(11); 3319-31. ©2016 AACR.


Assuntos
Apoptose/efeitos dos fármacos , Compostos de Boro/farmacologia , Quinase 1 do Ponto de Checagem/metabolismo , Glicina/análogos & derivados , Doença de Hodgkin/patologia , Linfoma de Células T/patologia , Complexo de Endopeptidases do Proteassoma/química , Inibidores de Proteassoma/farmacologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Proliferação de Células/efeitos dos fármacos , Quinase 1 do Ponto de Checagem/genética , Imunoprecipitação da Cromatina , Perfilação da Expressão Gênica , Glicina/farmacologia , Doença de Hodgkin/tratamento farmacológico , Doença de Hodgkin/metabolismo , Humanos , Linfoma de Células T/tratamento farmacológico , Linfoma de Células T/metabolismo , Camundongos , Camundongos SCID , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Blood ; 111(12): 5424-5, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18544695
16.
Sci Transl Med ; 7(277): 277ra30, 2015 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-25739764

RESUMO

Antitumor T cells either avoid or are inhibited in hypoxic and extracellular adenosine-rich tumor microenvironments (TMEs) by A2A adenosine receptors. This may limit further advances in cancer immunotherapy. There is a need for readily available and safe treatments that weaken the hypoxia-A2-adenosinergic immunosuppression in the TME. Recently, we reported that respiratory hyperoxia decreases intratumoral hypoxia and concentrations of extracellular adenosine. We show that it also reverses the hypoxia-adenosinergic immunosuppression in the TME. This, in turn, stimulates (i) enhanced intratumoral infiltration and reduced inhibition of endogenously developed or adoptively transfered tumor-reactive CD8 T cells, (ii) increased proinflammatory cytokines and decreased immunosuppressive molecules, such as transforming growth factor-ß (TGF-ß), (iii) weakened immunosuppression by regulatory T cells, and (iv) improved lung tumor regression and long-term survival in mice. Respiratory hyperoxia also promoted the regression of spontaneous metastasis from orthotopically grown breast tumors. These effects are entirely T cell- and natural killer cell-dependent, thereby justifying the testing of supplemental oxygen as an immunological coadjuvant to combine with existing immunotherapies for cancer.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Oxigênio/uso terapêutico , Adenosina/metabolismo , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Hiperóxia/complicações , Hiperóxia/patologia , Hipóxia/complicações , Hipóxia/imunologia , Hipóxia/patologia , Terapia de Imunossupressão , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Metástase Neoplásica , Neoplasias/patologia , Oxigênio/farmacologia , Indução de Remissão , Respiração/efeitos dos fármacos , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Microambiente Tumoral/efeitos dos fármacos
17.
Int J Biochem Cell Biol ; 35(4): 410-4, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12565702

RESUMO

G protein-coupled adenosine receptors are the subject of intense study as immunomodulators of inflammation especially since the recent demonstration that the A2a receptor acts to down-regulate inflammation and inhibit tissue damage in vivo [Nature 414 (6866) (2001) 916]. The adverse effects of overactive inflammation are evident in diseases e.g. sepsis, rheumatoid arthritis, and multiple sclerosis underscoring the importance of inhibiting inflammation or selectively enhancing inflammatory processes. It has been shown recently that the A2a adenosine receptor is a critical component of an endogenous "immunosuppressive loop" in which extracellular adenosine that accumulates due to local hypoxia caused by inflammatory insult signals through cAMP-elevating A2a receptors in a delayed negative feedback manner. Understanding how tissues regulate inflammation will provide the information necessary to allow for the engineering, or selective targeting, of endogenous inflammatory pathways. Recognition of A2a receptors as "natural" or endogenous brakes of inflammation provides the intellectual scaffolding needed to pursue these goals.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Inflamação/metabolismo , Receptores Purinérgicos P1/metabolismo , Animais , AMP Cíclico/imunologia , AMP Cíclico/metabolismo , Regulação para Baixo/imunologia , Proteínas de Ligação ao GTP/imunologia , Humanos , Terapia de Imunossupressão , Inflamação/imunologia , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Linfócitos/imunologia , Linfócitos/metabolismo , Camundongos , Receptores Purinérgicos P1/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo
18.
Microbes Infect ; 5(6): 515-26, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12758281

RESUMO

Adenosine can be described as a retaliatory metabolite, the production and release of which is usually enhanced under adverse environmental conditions. Binding via specific receptors, adenosine activates endogenous protective mechanisms aiming at the restoration of tissue homeostasis. While adenosinergic downregulation of tissue damage is beneficial in acute inflammation, chronic suppression of the immune system by adenosine may account for immunoparalysis in long-term septic patients.


Assuntos
Doenças Transmissíveis/imunologia , Doenças Transmissíveis/patologia , Receptores Purinérgicos P1/metabolismo , Adenosina/metabolismo , Animais , Doenças Transmissíveis/metabolismo , Humanos , Imunidade Inata , Inflamação/imunologia , Inflamação/patologia , Receptor A2A de Adenosina , Sepse/imunologia , Sepse/metabolismo
19.
Curr Pharm Des ; 9(23): 1827-32, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12871187

RESUMO

The immune system protects organisms from pathogens. The immune cells, in particular T- and B-lymphocytes, develop and acquire effector functions in specialized tissues called the lymphoid organs. The lymphoid organs exhibit lower oxygen tensions than the blood or the atmosphere. Furthermore, inflammatory and tumor environments where lymphocytes execute effector functions also have very low oxygen tensions. These findings led to the hypothesis that lymphocytes may have evolved adaptive mechanisms to function under hypoxic conditions. Cellular responses to hypoxia are triggered by the hypoxia inducible factor-1 alpha (HIF-1 alpha). In this paper we review the development and function of T- and B-lymphocytes in the absence HIF-1 alpha. Our studies suggest that HIF-1 alpha deficiency depresses the function of cytotoxic T-lymphocytes and blocks B-cell development in the bone marrow. B1 lymphocytes of fetal origin, on the other hand, accumulate and may produce auto-antibodies and autoimmunity.


Assuntos
Linfócitos B/fisiologia , Linfócitos T/fisiologia , Fatores de Transcrição/deficiência , Animais , Autoimunidade , Diferenciação Celular , Proteínas de Ligação a DNA/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia , Camundongos , Camundongos Transgênicos
20.
Biochem Pharmacol ; 65(4): 493-501, 2003 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-12566076

RESUMO

Inflammation must be inhibited in order to treat, e.g., sepsis or autoimmune diseases or must be selectively enhanced to improve, for example, immunotherapies of tumors or the development of vaccines. Predictable enhancement of inflammation depends upon the knowledge of the "natural" pathways by which it is down-regulated in vivo. Extracellular adenosine and A(2A) adenosine (purinergic) receptors were identified recently as anti-inflammatory signals and as sensors of excessive inflammatory tissue damage, respectively (Ohta A and Sitkovsky M, Nature 2001;414:916-20). These molecules may function as an important part of a physiological "metabolic switch" mechanism, whereby the inflammatory stimuli-produced local tissue damage and hypoxia cause adenosine accumulation and signaling through cyclic AMP-elevating A(2A) adenosine receptors in a delayed negative feedback manner. Patterns of A(2A) receptor expression are activation- and differentiation-dependent, thereby allowing for the "acquisition" of an immunosuppressive "OFF button" and creation of a time-window for immunomodulation. Identification of A(2A) adenosine receptors as "natural" brakes of inflammation provided a useful framework for understanding how tissues regulate inflammation and how to enhance or decrease (engineer) inflammation by targeting this endogenous anti-inflammatory pathway. These findings point to the need of more detailed testing of anti-inflammatory agonists of A(2A) receptors and create a previously unrecognized strategy to enhance inflammation and targeted tissue damage by using antagonists of A(2A) receptors. It is important to further identify the contributions of different types of immune cells at different stages of the inflammatory processes in different tissues to enable the "tailored" treatments with drugs that modulate the signaling through A(2A) purinergic receptors.


Assuntos
Inflamação/metabolismo , Receptores Purinérgicos P1/fisiologia , Adenosina/metabolismo , Adjuvantes Imunológicos/fisiologia , Animais , Anti-Inflamatórios/metabolismo , AMP Cíclico/metabolismo , Humanos , Receptor A2A de Adenosina , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA