Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
J Immunol ; 208(3): 571-581, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34965962

RESUMO

IDO2 is one of two closely related tryptophan catabolizing enzymes induced under inflammatory conditions. In contrast to the immunoregulatory role defined for IDO1 in cancer models, IDO2 has a proinflammatory function in models of autoimmunity and contact hypersensitivity. In humans, two common single-nucleotide polymorphisms have been identified that severely impair IDO2 enzymatic function, such that <25% of individuals express IDO2 with full catalytic potential. This, together with IDO2's relatively weak enzymatic activity, suggests that IDO2 may have a role outside of its function in tryptophan catabolism. To determine whether the enzymatic activity of IDO2 is required for its proinflammatory function, we used newly generated catalytically inactive IDO2 knock-in mice together with established models of contact hypersensitivity and autoimmune arthritis. Contact hypersensitivity was attenuated in catalytically inactive IDO2 knock-in mice. In contrast, induction of autoimmune arthritis was unaffected by the absence of IDO2 enzymatic activity. In pursuing this nonenzymatic IDO2 function, we identified GAPDH, Runx1, RANbp10, and Mgea5 as IDO2-binding proteins that do not interact with IDO1, implicating them as potential mediators of IDO2-specific function. Taken together, our findings identify a novel function for IDO2, independent of its tryptophan catabolizing activity, and suggest that this nonenzymatic function could involve multiple signaling pathways. These data show that the enzymatic activity of IDO2 is required only for some inflammatory immune responses and provide, to our knowledge, the first evidence of a nonenzymatic role for IDO2 in mediating autoimmune disease.


Assuntos
Artrite/imunologia , Autoimunidade/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Animais , Antígenos de Neoplasias/metabolismo , Linhagem Celular , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Técnicas de Introdução de Genes , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Células HEK293 , Humanos , Inflamação/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/metabolismo , Polimorfismo de Nucleotídeo Único/genética
2.
J Immunol ; 196(11): 4487-97, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27183624

RESUMO

Mechanistic insight into how adaptive immune responses are modified along the self-nonself continuum may offer more effective opportunities to treat autoimmune disease, cancer, and other sterile inflammatory disorders. Recent genetic studies in the KRN mouse model of rheumatoid arthritis demonstrate that the immunomodulatory molecule IDO2 modifies responses to self-antigens; however, the mechanisms involved are obscure. In this study, we show that IDO2 exerts a critical function in B cells to support the generation of autoimmunity. In experiments with IDO2-deficient mice, adoptive transplant experiments demonstrated that IDO2 expression in B cells was both necessary and sufficient to support robust arthritis development. IDO2 function in B cells was contingent on a cognate, Ag-specific interaction to exert its immunomodulatory effects on arthritis development. We confirmed a similar requirement in an established model of contact hypersensitivity, in which IDO2-expressing B cells are required for a robust inflammatory response. Mechanistic investigations showed that IDO2-deficient B cells lacked the ability to upregulate the costimulatory marker CD40, suggesting IDO2 acts at the T-B cell interface to modulate the potency of T cell help needed to promote autoantibody production. Overall, our findings revealed that IDO2 expression by B cells modulates autoimmune responses by supporting the cross talk between autoreactive T and B cells.


Assuntos
Autoimunidade/imunologia , Linfócitos B/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Linfócitos T/imunologia , Animais , Células Cultivadas , Indolamina-Pirrol 2,3,-Dioxigenase/deficiência , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
3.
J Cell Biochem ; 116(11): 2541-51, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25939245

RESUMO

Non-compensated dilated cardiomyopathy (DCM) leading to death from heart failure is rising rapidly in developed countries due to aging demographics, and there is a need for informative preclinical models to guide the development of effective therapeutic strategies to prevent or delay disease onset. In this study, we describe a novel model of heart failure based on cardiac-specific deletion of the prototypical mammalian BAR adapter-encoding gene Bin1, a modifier of age-associated disease. Bin1 deletion during embryonic development causes hypertrophic cardiomyopathy and neonatal lethality, but there is little information on how Bin1 affects cardiac function in adult animals. Here we report that cardiomyocyte-specific loss of Bin1 causes age-associated dilated cardiomyopathy (DCM) beginning by 8-10 months of age. Echocardiographic analysis showed that Bin1 loss caused a 45% reduction in ejection fraction during aging. Younger animals rapidly developed DCM if cardiac pressure overload was created by transverse aortic constriction. Heterozygotes exhibited an intermediate phenotype indicating Bin1 is haplo-insufficient to sustain normal heart function. Bin1 loss increased left ventricle (LV) volume and diameter during aging, but it did not alter LV volume or diameter in hearts from heterozygous mice nor did it affect LV mass. Bin1 loss increased interstitial fibrosis and mislocalization of the voltage-dependent calcium channel Cav 1.2, and the lipid raft scaffold protein caveolin-3, which normally complexes with Bin1 and Cav 1.2 in cardiomyocyte membranes. Our findings show how cardiac deficiency in Bin1 function causes age- and stress-associated heart failure, and they establish a new preclinical model of this terminal cardiac disease.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/deficiência , Envelhecimento/genética , Cardiomiopatia Dilatada/genética , Miócitos Cardíacos/patologia , Proteínas do Tecido Nervoso/deficiência , Proteínas Supressoras de Tumor/deficiência , Animais , Cardiomiopatia Dilatada/fisiopatologia , Modelos Animais de Doenças , Camundongos , Camundongos Knockout , Miócitos Cardíacos/metabolismo , Especificidade de Órgãos , Volume Sistólico
4.
Int Immunol ; 26(7): 357-67, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24402311

RESUMO

IDO2 is implicated in tryptophan catabolism and immunity but its physiological functions are not well established. Here we report the characterization of mice genetically deficient in IDO2, which develop normally but exhibit defects in IDO-mediated T-cell regulation and inflammatory responses. Construction of this strain was prompted in part by our discovery that IDO2 function is attenuated in macrophages from Ido1 (-/-) mice due to altered message splicing, generating a functional mosaic with implications for interpreting findings in Ido1 (-/-) mice. No apparent defects were observed in Ido2 (-/-) mice in embryonic development or hematopoietic differentiation, with wild-type profiles documented for kynurenine in blood serum and for immune cells in spleen, lymph nodes, peritoneum, thymus and bone marrow of naive mice. In contrast, upon immune stimulation we determined that IDO1-dependent T regulatory cell generation was defective in Ido2 (-/-) mice, supporting Ido1-Ido2 genetic interaction and establishing a functional role for Ido2 in immune modulation. Pathophysiologically, both Ido1 (-/-) and Ido2 (-/-) mice displayed reduced skin contact hypersensitivity responses, but mechanistic distinctions were apparent, with only Ido2 deficiency associated with a suppression of immune regulatory cytokines that included GM-CSF, G-CSF, IFN-γ, TNF-α, IL-6 and MCP-1/CCL2. Different contributions to inflammation were likewise indicated by the finding that Ido2 (-/-) mice did not phenocopy Ido1 (-/-) mice in the reduced susceptibility of the latter to inflammatory skin cancer. Taken together, our results offer an initial glimpse into immune modulation by IDO2, revealing its genetic interaction with IDO1 and distinguishing its non-redundant contributions to inflammation.


Assuntos
Regulação da Expressão Gênica/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Papiloma/imunologia , Neoplasias Cutâneas/imunologia , Linfócitos T Reguladores/imunologia , 9,10-Dimetil-1,2-benzantraceno , Animais , Carcinógenos , Citocinas/biossíntese , Células Dendríticas/imunologia , Células Dendríticas/patologia , Feminino , Indolamina-Pirrol 2,3,-Dioxigenase/deficiência , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Cinurenina , Teste de Cultura Mista de Linfócitos , Masculino , Camundongos , Camundongos Knockout , Papiloma/induzido quimicamente , Papiloma/genética , Papiloma/patologia , Transdução de Sinais , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Linfócitos T Reguladores/patologia , Acetato de Tetradecanoilforbol/análogos & derivados , Equilíbrio Th1-Th2
6.
Nat Rev Cancer ; 6(8): 613-25, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16862192

RESUMO

Cancer immunotherapy has been predominantly focused on biologically based intervention strategies. However, recent advances in the understanding of tumour-host interactions at the molecular level have revealed targets that might be amenable to intervention with small-molecule inhibitors. In particular, key effectors of tumoral immune escape have been identified that contribute to a dominant toleragenic state that is suspected of limiting the successful implementation of treatment strategies that rely on boosting immune function. Within the context of the pathophysiology of cancer-associated immune tolerance, this Review delineates potential molecular targets for therapeutic intervention and the progress that has been made in developing small-molecule inhibitors.


Assuntos
Antineoplásicos/farmacologia , Tolerância Imunológica/efeitos dos fármacos , Neoplasias/imunologia , Antineoplásicos/uso terapêutico , Arginase/antagonistas & inibidores , Inibidores de Ciclo-Oxigenase 2/farmacologia , Citocinas/antagonistas & inibidores , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Neoplasias/fisiopatologia , Óxido Nítrico Sintase/antagonistas & inibidores , Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores
7.
Cancer Immunol Immunother ; 63(7): 721-35, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24711084

RESUMO

Genetic and pharmacological studies of indoleamine 2,3-dioxygenase (IDO) have established this tryptophan catabolic enzyme as a central driver of malignant development and progression. IDO acts in tumor, stromal and immune cells to support pathogenic inflammatory processes that engender immune tolerance to tumor antigens. The multifaceted effects of IDO activation in cancer include the suppression of T and NK cells, the generation and activation of T regulatory cells and myeloid-derived suppressor cells, and the promotion of tumor angiogenesis. Mechanistic investigations have defined the aryl hydrocarbon receptor, the master metabolic regulator mTORC1 and the stress kinase Gcn2 as key effector signaling elements for IDO, which also exerts a non-catalytic role in TGF-ß signaling. Small-molecule inhibitors of IDO exhibit anticancer activity and cooperate with immunotherapy, radiotherapy or chemotherapy to trigger rapid regression of aggressive tumors otherwise resistant to treatment. Notably, the dramatic antitumor activity of certain targeted therapeutics such as imatinib (Gleevec) in gastrointestinal stromal tumors has been traced in part to IDO downregulation. Further, antitumor responses to immune checkpoint inhibitors can be heightened safely by a clinical lead inhibitor of the IDO pathway that relieves IDO-mediated suppression of mTORC1 in T cells. In this personal perspective on IDO as a nodal mediator of pathogenic inflammation and immune escape in cancer, we provide a conceptual foundation for the clinical development of IDO inhibitors as a novel class of immunomodulators with broad application in the treatment of advanced human cancer.


Assuntos
Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Inflamação/imunologia , Neoplasias/enzimologia , Neoplasias/imunologia , Evasão Tumoral , Animais , Humanos , Tolerância Imunológica , Imunoterapia , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Inflamação/enzimologia , Neoplasias/patologia , Transdução de Sinais
8.
Cancer J ; 29(1): 34-37, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36693156

RESUMO

ABSTRACT: Vaccine strategies for cancer differ from infectious disease in focusing mainly on clearing rather than preventing disease. Here we survey general vaccine strategies and combination therapy concepts being investigated for cancer treatment, with a focus on tumor antigens rather than cancer-inducing viruses or microorganisms. Many tumor antigens are "altered-self" and tend to arouse weaker immune responses than "foreign" antigens expressed by infectious agents. Further, unlike an infectious disease patient, a cancer patient's immune system is damaged, suppressed, or senescent and mainly tolerant of their disease. Thus, vaccine efficacy in a cancer patient will rely upon adjuvant or combination treatments that correct the inflammatory tumor microenvironment and degrade tumoral immunosuppression that dominates patient immunity. This brief overview is aimed at new researchers in cancer immunology seeking an overview of vaccine concepts to eradicate malignancy by provoking a selective immune attack.


Assuntos
Vacinas Anticâncer , Neoplasias , Humanos , Vacinas Anticâncer/uso terapêutico , Neoplasias/terapia , Antígenos de Neoplasias , Microambiente Tumoral
9.
Front Oncol ; 13: 1165298, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37182174

RESUMO

In parallel with the genetic and epigenetic changes that accumulate in tumor cells, chronic tumor-promoting inflammation establishes a local microenvironment that fosters the development of malignancy. While knowledge of the specific factors that distinguish tumor-promoting from non-tumor-promoting inflammation remains inchoate, nevertheless, as highlighted in this series on the 'Hallmarks of Cancer', it is clear that tumor-promoting inflammation is essential to neoplasia and metastatic progression making identification of specific factors critical. Studies of immunometabolism and inflamometabolism have revealed a role for the tryptophan catabolizing enzyme IDO1 as a core element in tumor-promoting inflammation. At one level, IDO1 expression promotes immune tolerance to tumor antigens, thereby helping tumors evade adaptive immune control. Additionally, recent findings indicate that IDO1 also promotes tumor neovascularization by subverting local innate immunity. This newly recognized function for IDO1 is mediated by a unique myeloid cell population termed IDVCs (IDO1-dependent vascularizing cells). Initially identified in metastatic lesions, IDVCs may exert broader effects on pathologic neovascularization in various disease settings. Mechanistically, induction of IDO1 expression in IDVCs by the inflammatory cytokine IFNγ blocks the antagonistic effect of IFNγ on neovascularization by stimulating the expression of IL6, a powerful pro-angiogenic cytokine. By contributing to vascular access, this newly ascribed function for IDO1 aligns with its involvement in other cancer hallmark functionalities, (tumor-promoting inflammation, immune escape, altered cellular metabolism, metastasis), which may stem from an underlying role in normal physiological functions such as wound healing and pregnancy. Understanding the nuances of how IDO1 involvement in these cancer hallmark functionalities varies between different tumor settings will be crucial to the future development of successful IDO1-directed therapies.

10.
Nat Med ; 11(3): 312-9, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15711557

RESUMO

Immune escape is a crucial feature of cancer progression about which little is known. Elevation of the immunomodulatory enzyme indoleamine 2,3-dioxygenase (IDO) in tumor cells can facilitate immune escape. Not known is how IDO becomes elevated or whether IDO inhibitors will be useful for cancer treatment. Here we show that IDO is under genetic control of Bin1, which is attenuated in many human malignancies. Mouse knockout studies indicate that Bin1 loss elevates the STAT1- and NF-kappaB-dependent expression of IDO, driving escape of oncogenically transformed cells from T cell-dependent antitumor immunity. In MMTV-Neu mice, an established breast cancer model, we show that small-molecule inhibitors of IDO cooperate with cytotoxic agents to elicit regression of established tumors refractory to single-agent therapy. Our findings suggest that Bin1 loss promotes immune escape in cancer by deregulating IDO and that IDO inhibitors may improve responses to cancer chemotherapy.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas do Tecido Nervoso/genética , Triptofano Oxigenase/antagonistas & inibidores , Triptofano Oxigenase/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Transformação Celular Neoplásica , Proteínas de Ligação a DNA/fisiologia , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Indolamina-Pirrol 2,3,-Dioxigenase , Indóis/farmacologia , Indóis/uso terapêutico , Interferon gama/farmacologia , Neoplasias Mamárias Experimentais/tratamento farmacológico , Camundongos , Dados de Sequência Molecular , NF-kappa B/farmacologia , Proteínas do Tecido Nervoso/fisiologia , Paclitaxel/uso terapêutico , Ratos , Fator de Transcrição STAT1 , Tioidantoínas/farmacologia , Tioidantoínas/uso terapêutico , Transativadores/fisiologia , Triptofano Oxigenase/biossíntese , Evasão Tumoral/fisiologia , Proteínas Supressoras de Tumor/fisiologia
11.
Dig Dis Sci ; 57(7): 1813-21, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22526583

RESUMO

BACKGROUND: Inflammatory bowel disease (IBD) is associated with defects in intestinal barriers that rely upon cellular tight junctions. Thus, identifying genes that could be targeted to enforce tight junctions and improve barrier function may lead to new treatment strategies for IBD. AIMS: This preclinical study aimed to evaluate an hypothesized role for the tumor suppressor gene Bin1 as a modifier of the severity of experimental colitis. METHODS: We ablated the Bin1 gene in a mosaic mouse model to evaluate its effects on experimental colitis and intestinal barrier function. Gross pathology, histology and inflammatory cytokine expression patterns were characterized and ex vivo physiology determinations were conducted to evaluate barrier function in intact colon tissue. RESULTS: Bin1 attenuation limited experimental colitis in a sexually dimorphic manner with stronger protection in female subjects. Colitis suppression was associated with an increase in basal transepithelial electrical resistance (TER) and a decrease in paracellular transepithelial flux, compared to control wild-type animals. In contrast, Bin1 attenuation did not affect short circuit current, nor did it alter the epithelial barrier response to non-inflammatory permeability enhancers in the absence of inflammatory stimuli. CONCLUSIONS: Bin1 is a genetic modifier of experimental colitis that controls the paracellular pathway of transcellular ion transport regulated by cellular tight junctions. Our findings offer a preclinical validation of Bin1 as a novel therapeutic target for IBD treatment.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/deficiência , Permeabilidade da Membrana Celular/fisiologia , Colite/prevenção & controle , Mucosa Intestinal/fisiologia , Proteínas do Tecido Nervoso/deficiência , Proteínas Supressoras de Tumor/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Colite/induzido quimicamente , Colite/patologia , Sulfato de Dextrana/efeitos adversos , Modelos Animais de Doenças , Feminino , Mucosa Intestinal/patologia , Masculino , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Índice de Gravidade de Doença , Caracteres Sexuais , Junções Íntimas/fisiologia , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/fisiologia
12.
Immunol Rev ; 222: 206-21, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18364004

RESUMO

Indoleamine 2, 3-dioxygenase (IDO) degrades the essential amino acid tryptophan in mammals, catalyzing the initial and rate-limiting step in the de novo biosynthesis nicotinamide adenine dinucleotide (NAD). Broad evidence implicates IDO and the tryptophan catabolic pathway in generation of immune tolerance to foreign antigens in tissue microenvironments. In particular, recent findings have established that IDO is overexpressed in both tumor cells and antigen-presenting cells in tumor-draining lymph nodes, where it promotes the establishment of peripheral immune tolerance to tumor antigens. In the normal physiologic state, IDO is important in creating an environment that limits damage to tissues due to an overactive immune system. However, by fostering immune suppression, IDO can facilitate the survival and growth of tumor cells expressing unique antigens that would be recognized normally as foreign. In preclinical studies, small-molecule inhibitors of IDO can reverse this mechanism of immunosuppression, complementing classical cytotoxic cancer chemotherapeutic agents' ability to trigger regression of treatment-resistant tumors. These results have encouraged the clinical translation of IDO inhibitors, the first of which entered phase I clinical trials in the fall of 2007. In this article, we survey the work defining IDO as an important mediator of peripheral tolerance, review evidence of IDO dysregulation in cancer cells, and provide an overview of the development of IDO inhibitors as a new immunoregulatory treatment modality for clinical trials.


Assuntos
Tolerância Imunológica/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Neoplasias/imunologia , Linfócitos T/imunologia , Evasão Tumoral/imunologia , Animais , Apresentação de Antígeno , Ensaios Clínicos Fase I como Assunto , Células Dendríticas/imunologia , Indução Enzimática , Inibidores Enzimáticos/uso terapêutico , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/antagonistas & inibidores , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Neoplasias/patologia , Linfócitos T/enzimologia , Triptofano/metabolismo
13.
Am J Pathol ; 176(5): 2082-7, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20228228

RESUMO

Chronic inflammation drives the development of many cancers, but a genetic definition of what constitutes 'cancer-associated' inflammation has not been determined. Recently, a mouse genetic study revealed a critical role for the immune escape mediator indoleamine 2,3-dioxygenase (IDO) in supporting inflammatory skin carcinogenesis. IDO is generally regarded as being immunosuppressive; however, there was no discernable difference in generalized inflammatory processes in IDO-null mice under conditions where tumor development was significantly suppressed, implicating IDO as key to establishing the pathogenic state of 'cancer-associated' inflammation. Here we review recent findings and their potential implications to understanding the relationship between immune escape and inflammation in cancer. Briefly, we propose that genetic pathways of immune escape in cancer are synonymous with pathways that define 'cancer-associated' inflammation and that these processes may be identical rather than distinct, as generally presumed, in terms of their genetic definition.


Assuntos
Regulação Neoplásica da Expressão Gênica , Inflamação , Animais , Progressão da Doença , Epigênese Genética , Humanos , Sistema Imunitário , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Linfonodos/patologia , Camundongos , Modelos Biológicos , Modelos Genéticos , Neoplasias/metabolismo , Neoplasias/patologia , Splicing de RNA , Neoplasias Cutâneas/patologia
14.
J Immunol ; 182(12): 7509-17, 2009 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-19494274

RESUMO

Rheumatoid arthritis (RA) is a chronic and debilitating inflammatory autoimmune disease of unknown etiology. As with a variety of autoimmune disorders, evidence of elevated tryptophan catabolism has been detected in RA patients indicative of activation of the immunomodulatory enzyme IDO. However, the role that IDO plays in the disease process is not well understood. The conceptualization that IDO acts solely to suppress effector T cell activation has led to the general assumption that inhibition of IDO activity should exacerbate autoimmune disorders. Recent results in cancer models, however, suggest a more complex role for IDO as an integral component of the inflammatory microenvironment necessary for supporting tumor outgrowth. This has led us to investigate the involvement of IDO in the pathological inflammation associated with RA. Using the K/BxN murine RA model and IDO inhibitor 1-methyl-tryptophan, we found that inhibiting IDO activity had the unexpected consequence of ameliorating, rather than exacerbating arthritis symptoms. 1-Methyl tryptophan treatment led to decreased autoantibody titers, reduced levels of inflammatory cytokines, and an attenuated disease course. This alleviation of arthritis was not due to an altered T cell response, but rather resulted from a diminished autoreactive B cell response, thus demonstrating a previously unappreciated role for IDO in stimulating B cell responses. Our findings raise the question of how an immunosuppressive enzyme can paradoxically drive autoimmunity. We suggest that IDO is not simply immunosuppressive, but rather plays a more complex role in modulating inflammatory responses, in particular those that are driven by autoreactive B cells.


Assuntos
Autoimunidade/imunologia , Linfócitos B/enzimologia , Linfócitos B/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Animais , Anticorpos/imunologia , Artrite/patologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Glucose-6-Fosfato Isomerase/imunologia , Glucose-6-Fosfato Isomerase/metabolismo , Interleucina-17/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Fatores de Tempo , Triptofano/análogos & derivados , Triptofano/farmacologia
15.
Proc Natl Acad Sci U S A ; 105(44): 17073-8, 2008 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-18952840

RESUMO

Topical application of phorbol myristate acetate (PMA) elicits intense local inflammation that facilitates outgrowth of premalignant lesions in skin after carcinogen exposure. The inflammatory response to PMA treatment activates immune stimulatory mechanisms. However, we show here that PMA exposure also induces plasmacytoid dendritic cells (pDCs) in local draining lymph nodes (dLNs) to express indoleamine 2,3 dioxygenase (IDO), which confers T cell suppressor activity on pDCs. The induced IDO-mediated inhibitory activity in this subset of pDCs was potent, dominantly suppressing the T cell stimulatory activity of other DCs that comprise the major fraction of dLN DCs. IDO induction in pDCs depended on inflammatory signaling by means of IFN type I and II receptors, the TLR/IL-1 signaling adaptor MyD88, and on cellular stress responses to amino acid withdrawal by means of the integrated stress response kinase GCN2. Consistent with the hypothesis that T cell suppressive, IDO(+) pDCs elicited by PMA exposure create local immune privilege that favors tumor development, IDO-deficient mice exhibited a robust tumor-resistant phenotype in the standard DMBA/PMA 2-stage carcinogenesis model of skin papilloma formation. Thus, IDO is a key immunosuppressive factor that facilitates tumor progression in this setting of chronic inflammation driven by repeated topical PMA exposure.


Assuntos
Dermatite de Contato/enzimologia , Tolerância Imunológica/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Papiloma/imunologia , Neoplasias Cutâneas/imunologia , Animais , Células Dendríticas/citologia , Células Dendríticas/enzimologia , Células Dendríticas/imunologia , Dermatite de Contato/imunologia , Dermatite de Contato/patologia , Progressão da Doença , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Linfonodos/enzimologia , Linfonodos/imunologia , Linfonodos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Papiloma/patologia , Transdução de Sinais/imunologia , Neoplasias Cutâneas/patologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Acetato de Tetradecanoilforbol/farmacologia
16.
Cancer Immunol Res ; 9(5): 514-528, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33622713

RESUMO

In addition to immunosuppression, it is generally accepted that myeloid-derived suppressor cells (MDSC) also support tumor angiogenesis. The tryptophan-catabolizing enzyme indoleamine 2,3-dioxygenase (IDO1) has been implicated in promoting neovascularization through its positioning as a key regulatory node between the inflammatory cytokines IFNγ and IL6. Here, we report that within the heterogeneous expanse of Gr-1+ MDSCs, both IDO1 expression and the ability to elicit neovascularization in vivo were associated with a minor subset of autofluorescent, CD11blo cells. IDO1 expression was further restricted to a discrete, CD11c and asialo-GM1 double-positive subpopulation of these cells, designated here as IDVCs (IDO1-dependent vascularizing cells), due to the dominant role that IDO1 activity in these cells was found to play in promoting neovascularization. Mechanistically, the induction of IDO1 in IDVCs provided a negative-feedback constraint on the antiangiogenic effect of host IFNγ by intrinsically signaling for the production of IL6 through general control nonderepressible 2 (GCN2)-mediated activation of the integrated stress response. These findings reveal fundamental molecular and cellular insights into how IDO1 interfaces with the inflammatory milieu to promote neovascularization.


Assuntos
Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Inflamação/metabolismo , Interferon gama/metabolismo , Interleucina-6/metabolismo , Neovascularização Patológica/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Inflamação/patologia , Interferon gama/genética , Interleucina-6/genética , Camundongos Endogâmicos BALB C , Camundongos Knockout , Metástase Neoplásica , Neoplasias/etiologia , Neoplasias/metabolismo , Neoplasias/patologia , Neovascularização Patológica/genética , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais
17.
Biochim Biophys Acta ; 1795(1): 25-36, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18930786

RESUMO

The evolutionarily conserved amphiphysin-like genes Bin1 and Bin3 function in membrane and actin dynamics, cell polarity, and stress signaling. Recent genetic studies in mice discriminate non-essential roles in endocytic processes commonly ascribed to amphiphysins from essential roles in cancer suppression. Bin1 acts in default pathways of apoptosis and senescence that are triggered by the Myc and Raf oncogenes in primary cells, and Bin1 gene products display a 'moonlighting function' in the nucleus where a variety of other 'endocytic' proteins are also found. Together, genetic investigations in yeast, flies, and mice suggest that amphiphysin-like adapter proteins may suppress cancer by helping integrate cell polarity signals generated by actin and vesicle dynamics with central regulators of cell cycle arrest, apoptosis, and immune surveillance.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Genes Supressores de Tumor/fisiologia , Neoplasias/genética , Proteínas do Tecido Nervoso/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Polaridade Celular/genética , Humanos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/fisiologia , Modelos Biológicos , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Estresse Fisiológico/genética , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/fisiologia
18.
Cancer Immunol Immunother ; 59(11): 1655-63, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20640572

RESUMO

Indoleamine 2,3-dioxygenase (IDO) is generally considered to be immunosuppressive but recent findings suggest this characterization oversimplifies its role in disease pathogenesis. Recently, we showed that IDO is essential for tumor outgrowth in the classical two-stage model of inflammatory skin carcinogenesis. Here, we report that IDO loss did not exacerbate classical inflammatory responses. Rather, IDO induction could be elicited by environmental signals and tumor promoters as an integral component of the inflammatory tissue microenvironment even in the absence of cancer. IDO loss had limited impact on tumor outgrowth in carcinogenesis models that lacked an explicit inflammatory tumor promoter. In the context of inflammatory carcinogenesis where IDO was critical to tumor development, the most important source of IDO was radiation-resistant non-hematopoietic cells, consistent with evidence that loss of the IDO regulatory tumor suppressor gene Bin1 in transformed skin cells facilitates IDO-mediated immune escape by a cell autonomous mechanism. Taken together, our results identify IDO as an integral component of 'cancer-associated' inflammation that tilts the immune system toward tumor support. More generally, they promote the concept that mediators of immune escape and cancer-associated inflammation may be genetically synonymous.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Tolerância Imunológica , Indolamina-Pirrol 2,3,-Dioxigenase/fisiologia , Inflamação/patologia , Proteínas do Tecido Nervoso/fisiologia , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Proteínas Supressoras de Tumor/fisiologia , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/enzimologia , Medula Óssea/efeitos da radiação , Transplante de Medula Óssea , Carcinógenos/toxicidade , Sinergismo Farmacológico , Humanos , Inflamação/metabolismo , Interferon gama/farmacologia , Interleucina-1beta/farmacologia , Queratinócitos/citologia , Queratinócitos/efeitos dos fármacos , Queratinócitos/efeitos da radiação , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Cutâneas/enzimologia , Acetato de Tetradecanoilforbol/farmacologia , Evasão Tumoral , Células U937 , Irradiação Corporal Total
19.
Front Immunol ; 11: 1861, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32973768

RESUMO

Indoleamine-2,3-dioxygenase (IDO)1 and IDO2 are two closely related tryptophan catabolizing enzymes encoded by linked genes. The IDO pathway is also immunomodulatory, with IDO1 well-characterized as a mediator of tumor immune evasion. Due to its homology with IDO1, IDO2 has been proposed to have a similar immunoregulatory function. Indeed, IDO2, like IDO1, is necessary for the differentiation of regulatory T cells in vitro. However, compared to IDO1, in vivo studies demonstrated a contrasting role for IDO2, with experiments in preclinical models of autoimmune arthritis establishing a proinflammatory role for IDO2 in mediating B and T cell activation driving autoimmune disease. Given their potentially opposing roles in inflammatory responses, interpretation of results obtained using IDO1 or IDO2 single knockout mice could be complicated by the expression of the other enzyme. Here we use IDO1 and IDO2 single and double knockout (dko) mice to define the differential roles of IDO1 and IDO2 in B cell-mediated immune responses. Autoreactive T and B cell responses and severity of joint inflammation were decreased in IDO2 ko, but not IDO1 ko arthritic mice. Dko mice had a reduction in the number of autoantibody secreting cells and severity of arthritis: however, percentages of differentiated T cells and their associated cytokines were not reduced compared to IDO1 ko or wild-type mice. These data suggest that autoreactive B cell responses are mediated by IDO2, while autoreactive T cell responses are indirectly affected by IDO1 expression in the IDO2 ko mice. IDO2 also influenced antibody responses in models of influenza infection and immunization with T cell-independent type II antigens. Taken together, these studies provide evidence for the contrasting roles IDO1 and IDO2 play in immune responses, with IDO1 mediating T cell suppressive effects and IDO2 working directly in B cells as a proinflammatory mediator of B cell responses.


Assuntos
Linfócitos B/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Ativação Linfocitária/imunologia , Linfócitos T/imunologia , Animais , Artrite Experimental/imunologia , Humanos , Inflamação/imunologia , Camundongos , Camundongos Knockout , Infecções por Orthomyxoviridae/imunologia
20.
J Immunother Cancer ; 8(2)2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32690770

RESUMO

BACKGROUND: The tryptophan-catabolizing enzyme indoleamine 2,3-dioxygenase 1 (IDO1), which subverts T-cell immunity at multiple levels, is itself subject to inherent T-cell reactivity. This intriguing deviation from central tolerance has been interpreted as counterbalancing IDO1-mediated immunosuppression. Based on this hypothesis, clinical studies employing an IDO1 peptide-based vaccine approach for cancer treatment have been initiated, but there remains a pressing need to further investigate the immunological ramifications of stimulating the anti-IDO1 T-cell response in this manner. METHODS: CT26 colon carcinoma tumors were evaluated for expression of IDO1 protein by western blot analysis, immunofluorescence microscopy and flow cytometry. Mouse IDO1-derived peptides, predicted to bind either major histocompatibility complex (MHC) class I or II of the H2d BALB/c strain, were emulsified in 50% Montanide for prophylactic or therapeutic vaccine treatment of CT26 tumor-bearing mice initiated either 7 days prior to or following tumor cell injection, respectively. In some therapeutic treatment experiments, administration of programmed cell death protein 1-binding antibody (anti-PD1 antibody) or epacadostat was concurrently initiated. Tumor size was determined by caliper measurements and comparative tumor growth suppression was assessed by longitudinal analyses of tumor growth data. For adoptive transfer, T cells from complete responder animals were isolated using paramagnetic beads and fluorescence-activated cell sorting. RESULTS: This study identifies mouse MHC class I-directed and II-directed, IDO1-derived peptides capable of eliciting antitumor responses, despite finding IDO1 expressed exclusively in tumor-infiltrating immune cells. Treatment of established tumors with anti-PD1 antibody and class I-directed but not class II-directed IDO1 peptide vaccines produced an enhanced antitumor response. Likewise, class I-directed and II-directed IDO1 peptides elicited an enhanced combinatorial response, suggesting distinct mechanisms of action. Consistent with this interpretation, adoptive transfer of isolated CD8+ T cells from class I and CD4+ T cells from class II peptide-vaccinated responder mice delayed tumor growth. The class II-directed response was completely IDO1-dependent while the class I-directed response included an IDO1-independent component consistent with antigen spread. CONCLUSIONS: The in vivo antitumor effects demonstrated with IDO1-based vaccines via targeting of the tumor microenvironment highlight the utility of mouse models for further exploration and refinement of this novel vaccine-based approach to IDO1-directed cancer therapy and its potential to improve patient response rates to anti-PD1 therapy.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/uso terapêutico , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Vacinas de Subunidades Antigênicas/uso terapêutico , Animais , Vacinas Anticâncer/farmacologia , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos Transgênicos , Vacinas de Subunidades Antigênicas/farmacologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa