Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Immunity ; 54(10): 2354-2371.e8, 2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34614413

RESUMO

Monocytic-lineage inflammatory Ly6c+CD103+ dendritic cells (DCs) promote antitumor immunity, but these DCs are infrequent in tumors, even upon chemotherapy. Here, we examined how targeting pathways that inhibit the differentiation of inflammatory myeloid cells affect antitumor immunity. Pharmacologic inhibition of Bruton's tyrosine kinase (BTK) and the tryptophan-degrading enzyme indoleamine 2,3-dioxygenase (IDO) or deletion of Btk or Ido1 allowed robust differentiation of inflammatory Ly6c+CD103+ DCs during chemotherapy, promoting antitumor T cell responses and inhibiting tumor growth. Immature Ly6c+c-kit+ precursor cells had epigenetic profiles similar to conventional DC precursors; deletion of Btk or Ido1 promoted differentiation of these cells. Mechanistically, a BTK-IDO axis inhibited a tryptophan-sensitive differentiation pathway driven by GATOR2 and mTORC1, and disruption of the GATOR2 in monocyte-lineage precursors prevented differentiation into inflammatory DCs in vivo. IDO-expressing DCs and monocytic cells were present across a range of human tumors. Thus, a BTK-IDO axis represses differentiation of inflammatory DCs during chemotherapy, with implications for targeted therapies.


Assuntos
Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Neoplasias/imunologia , Linfócitos T/imunologia , Tirosina Quinase da Agamaglobulinemia/imunologia , Tirosina Quinase da Agamaglobulinemia/metabolismo , Animais , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Feminino , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Masculino , Camundongos , Monócitos/citologia , Monócitos/imunologia , Monócitos/metabolismo , Transdução de Sinais/imunologia , Linfócitos T/metabolismo , Serina-Treonina Quinases TOR/imunologia , Serina-Treonina Quinases TOR/metabolismo
2.
Immunity ; 48(1): 91-106.e6, 2018 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-29343444

RESUMO

CD103+ dendritic cells are critical for cross-presentation of tumor antigens. Here we have shown that during immunotherapy, large numbers of cells expressing CD103 arose in murine tumors via direct differentiation of Ly6c+ monocytic precursors. These Ly6c+CD103+ cells could derive from bone-marrow monocytic progenitors (cMoPs) or from peripheral cells present within the myeloid-derived suppressor cell (MDSC) population. Differentiation was controlled by inflammation-induced activation of the transcription factor p53, which drove upregulation of Batf3 and acquisition of the Ly6c+CD103+ phenotype. Mice with a targeted deletion of p53 in myeloid cells selectively lost the Ly6c+CD103+ population and became unable to respond to multiple forms of immunotherapy and immunogenic chemotherapy. Conversely, increasing p53 expression using a p53-agonist drug caused a sustained increase in Ly6c+CD103+ cells in tumors during immunotherapy, which markedly enhanced the efficacy and duration of response. Thus, p53-driven differentiation of Ly6c+CD103+ monocytic cells represents a potent and previously unrecognized target for immunotherapy.


Assuntos
Células Apresentadoras de Antígenos/fisiologia , Monócitos/fisiologia , Células Mieloides/metabolismo , Neoplasias/imunologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Células Apresentadoras de Antígenos/imunologia , Antígenos CD/metabolismo , Antígenos Ly/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem Celular , Citometria de Fluxo , Humanos , Imunoterapia/métodos , Cadeias alfa de Integrinas/metabolismo , Camundongos , Monócitos/imunologia , Células Mieloides/fisiologia
3.
Cancer Immunol Immunother ; 66(8): 1049-1058, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28488123

RESUMO

The tumor microenvironment is profoundly immunosuppressive. This creates a major barrier for attempts to combine immunotherapy with conventional chemotherapy or radiation, because the tumor antigens released by these cytotoxic agents are not cross-presented in an immunogenic fashion. In this Focused Research Review, we focus on mouse preclinical studies exploring the role of immunosuppressive Tregs expressing the PTEN lipid phosphatase, and the links between PTEN+ Tregs and the immunoregulatory enzyme indoleamine 2,3-dioxygenase (IDO). IDO has received attention because it can be expressed by a variety of human tumor types in vivo, but IDO can also be induced in host immune cells of both humans and mice in response to inflammation, infection or dying (apoptotic) cells. Mechanistically, IDO and PTEN+ Tregs are closely connected, with IDO causing activation of the PTEN pathway in Tregs. Genetic ablation or pharmacologic inhibition of PTEN in mouse Tregs destabilizes their suppressive phenotype, and this prevents transplantable and autochthonous tumors from creating their normal immunosuppressive microenvironment. Genetic ablation of either IDO or PTEN+ Tregs in mice results in a fundamental defect in the ability to maintain tolerance to antigens associated with apoptotic cells, including dying tumor cells. Consistent with this, pharmacologic inhibitors of either pathway show synergy when combined with cytotoxic agents such as chemotherapy or radiation. Thus, we propose that IDO and PTEN+ Tregs represent closely linked checkpoints that can influence the choice between immune activation versus tolerance to dying tumor cells.


Assuntos
Apresentação de Antígeno , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Neoplasias/imunologia , PTEN Fosfo-Hidrolase/metabolismo , Linfócitos T Reguladores/imunologia , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Microambiente Tumoral
4.
Pediatr Blood Cancer ; 64(3)2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27616578

RESUMO

BACKGROUND: Effective networking and mentorship are critical determinants of career satisfaction and success in academic medicine. The American Society of Pediatric Hematology/Oncology (ASPHO) mentoring program was developed to support Early Career (EC) members. Herein, the authors report on the initial 2-year outcomes of this novel program. PROCEDURE: Mentees selected mentors with expertise in different subspecialties within the field from mentor profiles at the ASPHO Web site. Of 23 enrolled pairs, 19 mentors and 16 mentees completed electronic program feedback evaluations. The authors analyzed data collected between February 2013 and December 2014. The authors used descriptive statistics for categorical data and thematic analysis for qualitative data. RESULTS: The overall response rate was 76% (35/46). At the initiation of the relationship, career development and research planning were the most commonly identified goals for both mentors and mentees. Participants communicated by phone, e-mail, or met in-person at ASPHO annual meetings. Most mentor-mentee pairs were satisfied with the mentoring relationship, considered it a rewarding experience that justified their time and effort, achieved their goals in a timely manner with objective work products, and planned to continue the relationship. However, time constraints and infrequent communications remained a challenge. CONCLUSIONS: Participation in the ASPHO mentoring program suggests a clear benefit to a broad spectrum of ASPHO EC members with diverse personal and professional development needs. Efforts to expand the mentoring program are ongoing and focused on increasing enrollment of mentors to cover a wider diversity of career tracks/subspecialties and evaluating career and academic outcomes more objectively.


Assuntos
Escolha da Profissão , Oncologia/educação , Tutoria , Pediatria/educação , Feminino , Humanos , Masculino , Mentores , Satisfação Pessoal , Projetos Piloto , Avaliação de Programas e Projetos de Saúde
5.
Pediatr Transplant ; 21(4)2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28322484

RESUMO

Pediatric RCC is a rare pediatric neoplasm and is distinctly different compared to adult RCC, often demonstrating translocation morphology evidenced by unique histopathological features and TFE3 or TFEB nuclear expression. We report three cases of pediatric TFE3 positive RCC (TFE3-RCC) occurring in the setting of chronic kidney disease and long-term pharmacological immunosuppression, including two cases that developed in the native kidney following kidney transplantation. Together, these cases suggest that the kidney microenvironment in combination with immune dysregulation is likely contributing factors in the pathogenesis of some pediatric RCC, warranting further study. Long-term post-transplant surveillance may warrant screening for RCC.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Biomarcadores Tumorais/metabolismo , Carcinoma de Células Renais/diagnóstico , Falência Renal Crônica/cirurgia , Neoplasias Renais/diagnóstico , Transplante de Rim , Complicações Pós-Operatórias/diagnóstico , Carcinoma de Células Renais/etiologia , Carcinoma de Células Renais/metabolismo , Criança , Evolução Fatal , Feminino , Humanos , Imunossupressores/efeitos adversos , Lactente , Neoplasias Renais/etiologia , Neoplasias Renais/metabolismo , Masculino , Complicações Pós-Operatórias/etiologia , Complicações Pós-Operatórias/metabolismo , Microambiente Tumoral
6.
Adv Exp Med Biol ; 1036: 91-104, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29275467

RESUMO

In certain settings, chemotherapy can trigger an immunogenic form of tumor cell death. More often, however, tumor cell death after chemotherapy is not immunogenic, and may be actively tolerizing. However, even in these settings the dying tumor cells may be much more immunogenic than previously recognized, if key suppressive immune checkpoints such as indoleamine 2,3-dioxygenase (IDO) can be blocked. This is an important question, because a robust immune response to dying tumor cells could potentially augment the efficacy of conventional chemotherapy, or enhance the strength and duration of response to other immunologic therapies. Recent findings using preclinical models of self-tolerance and autoimmunity suggest that IDO and related downstream pathways may play a fundamental role in the decision between tolerance versus immune activation in response to dying cells. Thus, in the period of tumor cell death following chemotherapy or immunotherapy, the presence of IDO may help dictate the choice between dominant immunosuppression versus inflammation, antigen cross-presentation, and epitope spreading. The IDO pathway thus differs from other checkpoint-blockade strategies, in that it affects early immune responses, at the level of inflammation, activation of antigen-presenting cells, and initial cross-presentation of tumor antigens. This "up-stream" position may make IDO a potent target for therapeutic inhibition.


Assuntos
Tolerância Imunológica , Imunoterapia/métodos , Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Proteínas de Neoplasias/imunologia , Neoplasias , Microambiente Tumoral/imunologia , Animais , Morte Celular , Humanos , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/terapia
7.
J Immunol ; 192(1): 84-91, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24259502

RESUMO

Hemophagocytic lymphohistiocytosis (HLH) is an inborn disorder of immune regulation caused by mutations affecting perforin-dependent cytotoxicity. Defects in this pathway impair negative feedback between cytotoxic lymphocytes and APCs, leading to prolonged and pathologic activation of T cells. Etoposide, a widely used chemotherapeutic drug that inhibits topoisomerase II, is the mainstay of treatment for HLH, although its therapeutic mechanism remains unknown. We used a murine model of HLH, involving lymphocytic choriomeningitis virus infection of perforin-deficient mice, to study the activity and mechanism of etoposide for treating HLH and found that it substantially alleviated all symptoms of murine HLH and allowed prolonged survival. This therapeutic effect was relatively unique among chemotherapeutic agents tested, suggesting distinctive effects on the immune response. We found that the therapeutic mechanism of etoposide in this model system involved potent deletion of activated T cells and efficient suppression of inflammatory cytokine production. This effect was remarkably selective; etoposide did not exert a direct anti-inflammatory effect on macrophages or dendritic cells, and it did not cause deletion of quiescent naive or memory T cells. Finally, etoposide's immunomodulatory effects were similar in wild-type and perforin-deficient animals. Thus, etoposide treats HLH by selectively eliminating pathologic, activated T cells and may have usefulness as a novel immune modulator in a broad array of immunopathologic disorders.


Assuntos
Etoposídeo/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Linfo-Histiocitose Hemofagocítica/tratamento farmacológico , Linfo-Histiocitose Hemofagocítica/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Animais , Apresentação de Antígeno/efeitos dos fármacos , Apresentação de Antígeno/imunologia , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Proteínas de Ligação a DNA/genética , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Modelos Animais de Doenças , Etoposídeo/administração & dosagem , Interferon gama/biossíntese , Vírus da Coriomeningite Linfocítica , Linfo-Histiocitose Hemofagocítica/genética , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Camundongos Knockout , Inibidores da Topoisomerase II/administração & dosagem , Inibidores da Topoisomerase II/farmacologia
8.
Neuro Oncol ; 26(2): 348-361, 2024 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-37715730

RESUMO

BACKGROUND: Recurrent brain tumors are the leading cause of cancer death in children. Indoleamine 2,3-dioxygenase (IDO) is a targetable metabolic checkpoint that, in preclinical models, inhibits anti-tumor immunity following chemotherapy. METHODS: We conducted a phase I trial (NCT02502708) of the oral IDO-pathway inhibitor indoximod in children with recurrent brain tumors or newly diagnosed diffuse intrinsic pontine glioma (DIPG). Separate dose-finding arms were performed for indoximod in combination with oral temozolomide (200 mg/m2/day x 5 days in 28-day cycles), or with palliative conformal radiation. Blood samples were collected at baseline and monthly for single-cell RNA-sequencing with paired single-cell T cell receptor sequencing. RESULTS: Eighty-one patients were treated with indoximod-based combination therapy. Median follow-up was 52 months (range 39-77 months). Maximum tolerated dose was not reached, and the pediatric dose of indoximod was determined as 19.2 mg/kg/dose, twice daily. Median overall survival was 13.3 months (n = 68, range 0.2-62.7) for all patients with recurrent disease and 14.4 months (n = 13, range 4.7-29.7) for DIPG. The subset of n = 26 patients who showed evidence of objective response (even a partial or mixed response) had over 3-fold longer median OS (25.2 months, range 5.4-61.9, p = 0.006) compared to n = 37 nonresponders (7.3 months, range 0.2-62.7). Four patients remain free of active disease longer than 36 months. Single-cell sequencing confirmed emergence of new circulating CD8 T cell clonotypes with late effector phenotype. CONCLUSIONS: Indoximod was well tolerated and could be safely combined with chemotherapy and radiation. Encouraging preliminary evidence of efficacy supports advancing to Phase II/III trials for pediatric brain tumors.


Assuntos
Neoplasias Encefálicas , Neoplasias do Tronco Encefálico , Humanos , Criança , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Temozolomida , Triptofano , Fatores Imunológicos , Imunoterapia , Neoplasias do Tronco Encefálico/patologia
9.
Immunol Invest ; 41(6-7): 765-97, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23017145

RESUMO

Indoleamine 2,3-dioxygenase (IDO) is a natural mechanism of creating acquired tolerance in a variety of physiological settings. This endogenous tolerogenic pathway has important functions in regulating the magnitude of immune responses in settings of infection, pregnancy, tissue transplantation, mucosal interfaces and others. Whether for angiogenesis, stromal formation or immunologic tolerance, tumors often rely on recruiting host mechanisms. IDO is one such potent endogenous mechanism that appears to be frequently hijacked by tumors to establish systemic immune tolerance to tumor antigens. IDO can be expressed by tumors themselves, but, in addition, its natural site of expression is the host immune cells recruited by the tumor (particularly dendritic cells and macrophages). Therapeutic strategies that target the IDO pathway have been shown to synergize with standard chemotherapy and experimental immunotherapies to break tumor-induced tolerance. When such strategies target IDO expressed in host cells, they may be able to disrupt tolerance without creating intrinsic tumor cell drug resistance.


Assuntos
Células Dendríticas/imunologia , Tolerância Imunológica , Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Macrófagos/imunologia , Neoplasias/imunologia , Linfócitos T Reguladores/imunologia , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/enzimologia , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/antagonistas & inibidores , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Cinurenina/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Neoplasias/patologia , Transdução de Sinais , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/patologia , Triptofano/metabolismo
10.
Clin Dev Immunol ; 2012: 937253, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22312408

RESUMO

Central nervous system tumors take advantage of the unique immunology of the CNS and develop exquisitely complex stromal networks that promote growth despite the presence of antigen-presenting cells and tumor-infiltrating lymphocytes. It is precisely this immunological paradox that is essential to the survival of the tumor. We review the evidence for functional CNS immune privilege and the impact it has on tumor tolerance. In this paper, we place an emphasis on the role of tumor-infiltrating myeloid cells in maintaining stromal and vascular quiescence, and we underscore the importance of indoleamine 2,3-dioxygenase activity as a myeloid-driven tumor tolerance mechanism. Much remains to be discovered regarding the tolerogenic mechanisms by which CNS tumors avoid immune clearance. Thus, it is an open question whether tumor tolerance in the brain is fundamentally different from that of peripheral sites of tumorigenesis or whether it simply stands as a particularly strong example of such tolerance.


Assuntos
Neoplasias do Sistema Nervoso Central/imunologia , Tolerância Imunológica , Animais , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/patologia , Criança , Pré-Escolar , Humanos , Camundongos
11.
Cancer Res ; 78(18): 5191-5199, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-30181177

RESUMO

Regulatory T cells (Tregs) are an important contributor to the immunosuppressive tumor microenvironment. To date, however, they have been difficult to target for therapy. One emerging new aspect of Treg biology is their apparent functional instability in the face of certain acute proinflammatory signals such as IL6 and IFNγ. Under the right conditions, these signals can cause a rapid loss of suppressor activity and reprogramming of the Tregs into a proinflammatory phenotype. In this review, we propose the hypothesis that this phenotypic modulation does not reflect infidelity to the Treg lineage, but rather represents a natural, physiologic response of Tregs during beneficial inflammation. In tumors, however, this inflammation-induced Treg destabilization is actively opposed by dominant stabilizing factors such as indoleamine 2,3-dioxygenase and the PTEN phosphatase pathway in Tregs. Under such conditions, tumor-associated Tregs remain highly suppressive and inhibit cross-presentation of tumor antigens released by dying tumor cells. Interrupting these Treg stabilizing pathways can render tumor-associated Tregs sensitive to rapid destabilization during immunotherapy, or during the wave of cell death following chemotherapy or radiation, thus enhancing antitumor immune responses. Understanding the emerging pathways of Treg stabilization and destabilization may reveal new molecular targets for therapy. Cancer Res; 78(18); 5191-9. ©2018 AACR.


Assuntos
Regulação Neoplásica da Expressão Gênica , Imunoterapia/métodos , Neoplasias/metabolismo , Linfócitos T Reguladores/imunologia , Animais , Linhagem Celular Tumoral , Linhagem da Célula , Regulação para Baixo , Humanos , Sistema Imunitário , Inflamação , Interferon gama/metabolismo , Interleucina-6/metabolismo , Neoplasias/terapia , PTEN Fosfo-Hidrolase/metabolismo , Fenótipo , Receptores de Antígenos de Linfócitos T/imunologia , Pesquisa Translacional Biomédica , Microambiente Tumoral/imunologia
12.
J Immunother Cancer ; 2: 21, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25054064

RESUMO

BACKGROUND: Indoleamine 2,3-dioxygenase (IDO) is an enzyme with immune-suppressive properties that is commonly exploited by tumors to evade immune destruction. Anti-tumor T cell responses can be initiated in solid tumors, but are immediately suppressed by compensatory upregulation of immunological checkpoints, including IDO. In addition to these known effects on the adaptive immune system, we previously showed widespread, T cell-dependent complement deposition during allogeneic fetal rejection upon maternal treatment with IDO-blockade. We hypothesized that IDO protects glioblastoma from the full effects of chemo-radiation therapy by preventing vascular activation and complement-dependent tumor destruction. METHODS: To test this hypothesis, we utilized a syngeneic orthotopic glioblastoma model in which GL261 glioblastoma tumor cells were stereotactically implanted into the right frontal lobes of syngeneic mice. These mice were treated with IDO-blocking drugs in combination with chemotherapy and radiation therapy. RESULTS: Pharmacologic inhibition of IDO synergized with chemo-radiation therapy to prolong survival in mice bearing intracranial glioblastoma tumors. We now show that pharmacologic or genetic inhibition of IDO allowed chemo-radiation to trigger widespread complement deposition at sites of tumor growth. Chemotherapy treatment alone resulted in collections of perivascular leukocytes within tumors, but no complement deposition. Adding IDO-blockade led to upregulation of VCAM-1 on vascular endothelium within the tumor microenvironment, and further adding radiation in the presence of IDO-blockade led to widespread deposition of complement. Mice genetically deficient in complement component C3 lost all of the synergistic effects of IDO-blockade on chemo-radiation-induced survival. CONCLUSIONS: Together these findings identify a novel mechanistic link between IDO and complement, and implicate complement as a major downstream effector mechanism for the beneficial effect of IDO-blockade after chemo-radiation therapy. We speculate that this represents a fundamental pathway by which the tumor regulates intratumoral vascular activation and protects itself from immune-mediated tumor destruction.

13.
J Immunol Methods ; 364(1-2): 1-13, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21110979

RESUMO

Hemophagocytic lymphohistiocytosis is a life-threatening multi-system hyperinflammatory disorder characterized by dysfunctional cytolytic lymphocyte responses, hypercytokinemia, and widespread lymphohistiocytic tissue infiltration and destruction. Diagnosis and definitive therapy are often delayed as clinical efforts are directed toward treatment of presumed overwhelming infection. Sporadic cases occur in association with underlying immune dysfunction related to autoimmune disease, malignancy, or severe infection. However, familial cases predominate with remarkable associations between underlying genetic defects and dysregulation of immune responses. Here, we review the genetic and immunologic basis of contemporary diagnostic methods for hemophagocytic lymphohistiocytosis.


Assuntos
Doenças Autoimunes/diagnóstico , Infecções/diagnóstico , Linfo-Histiocitose Hemofagocítica/diagnóstico , Neoplasias/diagnóstico , Patologia Molecular , Doenças Autoimunes/complicações , Autoimunidade , Citocinas/imunologia , Predisposição Genética para Doença , Humanos , Infecções/complicações , Linfo-Histiocitose Hemofagocítica/etiologia , Neoplasias/complicações , Patologia Molecular/métodos , Polimorfismo Genético , Linfócitos T Citotóxicos/imunologia
14.
Science ; 297(5588): 1867-70, 2002 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-12228717

RESUMO

Antigen-presenting cells (APCs) can induce tolerance or immunity. We describe a subset of human APCs that express indoleamine 2,3-dioxygenase (IDO) and inhibit T cell proliferation in vitro. IDO-positive APCs constituted a discrete subset identified by coexpression of the cell-surface markers CD123 and CCR6. In the dendritic cell (DC) lineage, IDO-mediated suppressor activity was present in fully mature as well as immature CD123+ DCs. IDO+ DCs could also be readily detected in vivo, which suggests that these cells may represent a regulatory subset of APCs in humans.


Assuntos
Células Dendríticas/enzimologia , Células Dendríticas/imunologia , Linfócitos T/imunologia , Triptofano Oxigenase/metabolismo , Triptofano/análogos & derivados , Células Apresentadoras de Antígenos/enzimologia , Células Apresentadoras de Antígenos/imunologia , Antígenos CD/análise , Adesão Celular , Linhagem da Célula , Células Cultivadas , Regulação para Baixo , Inibidores Enzimáticos/farmacologia , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase , Interferon gama/farmacologia , Interleucina-10/farmacologia , Subunidade alfa de Receptor de Interleucina-3 , Ativação Linfocitária , Teste de Cultura Mista de Linfócitos , Tecido Linfoide/citologia , Tecido Linfoide/enzimologia , Macrófagos/enzimologia , Receptores CCR6 , Receptores de Quimiocinas/análise , Receptores de Interleucina-3/análise , Triptofano/farmacologia , Triptofano Oxigenase/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA