Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
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
2.
Mol Cancer ; 20(1): 165, 2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34906138

RESUMO

BACKGROUND: Stem Cell leukemia/lymphoma syndrome (SCLL) presents as a myeloproliferative disease which can progress to acute myeloid leukemia and is associated with the coincident development of B-cell and T-cell lymphomas. SCLL is driven by the constitutive activation of fibroblast growth factor receptor-1 (FGFR1) as a result of chromosome translocations with poor outcome. Mouse models have been developed which faithfully recapitulate the human disease and have been used to characterize the molecular genetic events that are associated with development and progression of the disease. METHODS: CRISPR/Cas9 approaches were used to generate SCLL cells null for Interleukin receptor associated kinase 1 (IRAK1) and interferon gamma (IFNG) which were introduced into syngeneic hosts through tail vein injection. Development of the disease and changes in immune cell composition and activity were monitored using flow cytometry. Bead-based immunoassays were used to compare the cytokine and chemokine profiles of control and knock out (KO) cells. Antibody mediated, targeted depletion of T cell and MDSCs were performed to evaluate their role in antitumor immune responses. RESULTS: In SCLL, FGFR1 activation silences miR-146b-5p through DNMT1-mediated promoter methylation, which derepresses the downstream target IRAK1. IRAK1 KO SCLL cells were xenografted into immunocompetent syngeneic mice where the typical rapid progression of disease was lost and the mice remained disease free. IRAK1 in this system has no effect on cell cycle progression or apoptosis and robust growth of the KO cells in immunodeficient mice suggested an effect on immune surveillance. Depletion of T-cells in immunocompetent mice restored leukemogenesis of the KO cells, and tumor killing assays confirmed the role of T cells in tumor clearance. Analysis of the immune cell profile in mice transplanted with the IRAK1 expressing mock control (MC) cells shows that there is an increase in levels of myeloid-derived suppressor cells (MDSCs) with a concomitant decrease in CD4+/CD8+ T-cell levels. MDSC suppression assays and depletion experiments showed that these MDSCs were responsible for suppression of the T cell mediated leukemia cell elimination. Immuno-profiling of a panel of secreted cytokines and chemokines showed that activation of IFN-γ is specifically impaired in the KO cells. In vitro and in vivo expression assays and engraftment with interferon gamma receptor-1 (IFNGR1) null mice and IFNG KO SCLL cells, showed the leukemia cells produced IFN-γ directly participating in the induction of MDSCs to establish immune evasion. Inhibition of IRAK1 using pacritinib suppresses leukemogenesis with impaired induction of MDSCs and attenuated suppression of CD4+/CD8+ T-cells. CONCLUSIONS: IRAK1 orchestrates a previously unknown FGFR1-directed immune escape mechanism in SCLL, through induction of MDSCs via regulation of IFN-γ signaling from leukemia cells, and targeting IRAK1 may provide a means of suppressing tumor growth in this syndrome by restoring immune surveillance.


Assuntos
Neoplasias Hematológicas/etiologia , Neoplasias Hematológicas/metabolismo , Evasão da Resposta Imune , Interferon gama/metabolismo , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Células Supressoras Mieloides/imunologia , Células Supressoras Mieloides/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Biomarcadores , Suscetibilidade a Doenças , Regulação Neoplásica da Expressão Gênica , Neoplasias Hematológicas/patologia , Humanos , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais
3.
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
4.
J Immunother Cancer ; 9(6)2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34117113

RESUMO

BACKGROUND: The indoleamine 2,3-dioxygenase (IDO) pathway is a key counter-regulatory mechanism that, in cancer, is exploited by tumors to evade antitumor immunity. Indoximod is a small-molecule IDO pathway inhibitor that reverses the immunosuppressive effects of low tryptophan (Trp) and high kynurenine (Kyn) that result from IDO activity. In this study, indoximod was used in combination with a checkpoint inhibitor (CPI) pembrolizumab for the treatment for advanced melanoma. METHODS: Patients with advanced melanoma were enrolled in a single-arm phase II clinical trial evaluating the addition of indoximod to standard of care CPI approved for melanoma. Investigators administered their choice of CPI including pembrolizumab (P), nivolumab (N), or ipilimumab (I). Indoximod was administered continuously (1200 mg orally two times per day), with concurrent CPI dosed per US Food and Drug Administration (FDA)-approved label. RESULTS: Between July 2014 and July 2017, 131 patients were enrolled. (P) was used more frequently (n=114, 87%) per investigator's choice. The efficacy evaluable population consisted of 89 patients from the phase II cohort with non-ocular melanoma who received indoximod combined with (P).The objective response rate (ORR) for the evaluable population was 51% with confirmed complete response of 20% and disease control rate of 70%. Median progression-free survival was 12.4 months (95% CI 6.4 to 24.9). The ORR for Programmed Death-Ligand 1 (PD-L1)-positive patients was 70% compared with 46% for PD-L1-negative patients. The combination was well tolerated, and side effects were similar to what was expected from single agent (P). CONCLUSION: In this study, the combination of indoximod and (P) was well tolerated and showed antitumor efficacy that is worth further evaluation in selected patients with advanced melanoma.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Imunoterapia/métodos , Melanoma/tratamento farmacológico , Triptofano/análogos & derivados , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Monoclonais Humanizados/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Triptofano/farmacologia , Triptofano/uso terapêutico
5.
Immunology ; 158(4): 353-361, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31557322

RESUMO

Reagents that activate the signaling adaptor stimulator of interferon genes (STING) suppress experimentally induced autoimmunity in murine models of multiple sclerosis and arthritis. In this study, we evaluated STING agonists as potential reagents to inhibit spontaneous autoimmune type I diabetes (T1D) onset in non-obese diabetic (NOD) female mice. Treatments with DNA nanoparticles (DNPs), which activate STING when cargo DNA is sensed, delayed T1D onset and reduced T1D incidence when administered before T1D onset. DNP treatment elevated indoleamine 2,3 dioxygenase (IDO) activity, which regulates T-cell immunity, in spleen, pancreatic lymph nodes and pancreas of NOD mice. Therapeutic responses to DNPs were partially reversed by inhibiting IDO and DNP treatment synergized with insulin therapy to further delay T1D onset and reduce T1D incidence. Treating pre-diabetic NOD mice with cyclic guanyl-adenyl dinucleotide (cGAMP) to activate STING directly delayed T1D onset and stimulated interferon-αß (IFN-αß), while treatment with cyclic diguanyl nucleotide (cdiGMP) did not delay T1D onset or induce IFN-αß in NOD mice. DNA sequence analyses revealed that NOD mice possess a STING polymorphism that may explain differential responses to cGAMP and cdiGMP. In summary, STING agonists attenuate T1D progression and DNPs enhance therapeutic responses to insulin therapy.


Assuntos
DNA/uso terapêutico , Diabetes Mellitus Tipo 1/tratamento farmacológico , Insulina/uso terapêutico , Proteínas de Membrana/agonistas , Nanopartículas/uso terapêutico , Linfócitos T/imunologia , Animais , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , DNA/química , Modelos Animais de Doenças , Sinergismo Farmacológico , Feminino , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos NOD , Nanopartículas/química , Nucleotídeos Cíclicos/metabolismo , Polimorfismo Genético , Regulação para Cima
6.
Int J Mol Sci ; 19(2)2018 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-29443941

RESUMO

Anaplastic thyroid carcinoma (ATC) is almost universally fatal. Elevated keratin-8 (KRT8) protein expression is an established diagnostic cancer biomarker in several epithelial cancers (but not ATC). Several keratins, including KRT8, have been suggested to have a role in cell biology beyond that of structural cytoskeletal proteins. Here, we provide evidence that KRT8 plays a direct role in the growth of ATCs. Genomic and transcriptomic analysis of >5000 patients demonstrates that KRT8 mutation and copy number amplification are frequently evident in epithelial-derived cancers. Carcinomas arising from diverse tissues exhibit KRT8 mRNA and protein overexpression when compared to normal tissue levels. Similarly, in a panel of patient-derived ATC cell lines and patient tumors, KRT8 expression shows a similar pattern. sh-RNA-mediated KRT8 knockdown in these cell lines increases apoptosis, whereas forced overexpression of KRT8 confers resistance to apoptosis under peroxide-induced cell stress conditions. We further show that KRT8 protein binds to annexin A2, a protein known to mediate apoptosis as well as the redox pathway.


Assuntos
Biomarcadores Tumorais/genética , Carcinoma/metabolismo , Queratina-8/genética , Neoplasias da Glândula Tireoide/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Anexina A2/metabolismo , Apoptose , Biomarcadores Tumorais/metabolismo , Carcinoma/genética , Carcinoma/patologia , Linhagem Celular Tumoral , Feminino , Dosagem de Genes , Humanos , Queratina-8/metabolismo , Masculino , Pessoa de Meia-Idade , Mutação , Ligação Proteica , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/patologia , Regulação para Cima
7.
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
8.
J Immunol Methods ; 449: 7-14, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28645528

RESUMO

Dendritic cells (DCs) are specialized antigen-presenting cells that play a pivotal role in the pathogenesis of periodontitis. The use of animal models to study the role of DCs in periodontitis has been limited by lack of a method for sustained depletion of DCs. Hence, the objectives of this study were to validate the zDC-DTR knockin mouse model of conventional DCs (cDCs) depletion, as well as to investigate whether this depletion could be sustained long enough to induce alveolar bone loss in this model. zDC-DTR mice were treated with different dose regimens of diphtheria toxin (DT) to determine survival rate. A loading DT dose of 20ng/bw, followed and maintained with doses of 10ng/bm every 3days for up to 4weeks demonstrated 80% survival. Animals were weighed weekly and peripheral blood was obtained to confirm normal neutrophil counts. Five animals per group were euthanized at baseline, 24h, 1 and 4weeks. Bone marrow (BM), spleen (SP) and gingival tissue (GT) were harvested, and cells were isolated, separated and stained for Pre-DCs precursors (CD45R-MHCII+CD11c+Flt3+CD172a+) in BM, cDCs (CD11c+MHCII+CD209+) in spleen, and DCs in GT (CD45R+MHCII+CD11c+ DC-SIGN/CD209+). Pre-DCs in BM were significantly depleted at 24h and depletion maintained for up to 4weeks, as compared to blank (PBS) controls. Circulating cDCs in spleen demonstrated a non-significant trend to deplete in 1week with high variability among mice. GT also showed a similar non-significant trend to deplete in 24h. The zDC-DTR model seems to be viable for evaluating the role of DCs immune homeostasis disruption and alveolar bone loss pathogenesis in response to long-term oral infection.


Assuntos
Células Dendríticas/imunologia , Modelos Animais de Doenças , Periodontite/imunologia , Animais , Células da Medula Óssea/imunologia , Células Dendríticas/patologia , Toxina Diftérica/administração & dosagem , Toxina Diftérica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Baço/citologia , Baço/imunologia , Fatores de Tempo
9.
Methods Mol Biol ; 707: 39-44, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21287327

RESUMO

Generation of regulatory T cells (or Treg) derived hybridomas offers a tool to study their antigen specificity. T cells hybridomas are produced by fusing TCR α-ß-thymoma BW5147 with highly dividing T cell population. In vitro anergy of Tregs is an obstacle in generation of highly dividing Treg population for their fusion. In this chapter, we describe a simple and efficient method to generate large number of blasting Treg and their successful fusion with thymoma BW5147. The resultant hybridomas lose Treg-specific transcription factor FoxP3, respond to antigenic stimulation by producing IL-2, and thus allow the evaluation of antigen specific, Tregs-derived TCRs.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Hibridomas/citologia , Linfócitos T Reguladores/citologia , Animais , Antígenos/imunologia , Camundongos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Timoma/metabolismo
10.
Proc Natl Acad Sci U S A ; 106(25): 10266-71, 2009 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-19509335

RESUMO

MicroRNAs (miRNAs) are a class of evolutionarily conserved small noncoding RNAs that are increasingly being recognized as important regulators of gene expression. The ribonuclease III enzyme Dicer is essential for the processing of miRNAs. CD1d-restricted invariant natural killer T (iNKT) cells are potent regulators of diverse immune responses. The role of Dicer-generated miRNAs in the development and function of immune regulatory iNKT cells is unknown. Here, we generated a mouse strain with a tissue-specific disruption of Dicer, and showed that lack of miRNAs after the deletion of Dicer by Tie2-Cre (expressed in hematopoietic cells and endothelial cells) interrupted the development and maturation of iNKT cells in the thymus and significantly decreased the number of iNKT cells in different immune organs. Thymic and peripheral iNKT cell compartments were changed in miRNA-deficient mice, with a significantly increased frequency of CD4(+)CD8(+) iNKT cells in the thymus and a significantly decreased frequency of CD4(+) iNKT cells in the spleen. MiRNA-deficient iNKT cells display profound defects in alpha-GalCer-induced activation and cytokine production. Bone marrow (BM) from miRNA-deficient mice poorly reconstituted iNKT cells compared to BM from WT mice. Also, using a thymic iNKT cell transfer model, we found that iNKT cell homeostasis was impaired in miRNA-deficient recipient mice. Our data indicate that miRNAs expressed in hematopoietic cells and endothelial cells are potent regulators of iNKT cell development, function, and homeostasis.


Assuntos
RNA Helicases DEAD-box/metabolismo , Endorribonucleases/metabolismo , Ativação Linfocitária , MicroRNAs/metabolismo , Células T Matadoras Naturais/imunologia , Animais , Antígenos CD4/imunologia , Antígenos CD8/imunologia , RNA Helicases DEAD-box/genética , Endorribonucleases/genética , Células Endoteliais/enzimologia , Células-Tronco Hematopoéticas/enzimologia , Ativação Linfocitária/genética , Camundongos , Camundongos Transgênicos , Células T Matadoras Naturais/enzimologia , Receptor TIE-2/genética , Ribonuclease III , Timo/enzimologia , Timo/imunologia
11.
Immunity ; 25(2): 249-59, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16879995

RESUMO

Foxp3(+)CD4(+)CD25(+) regulatory T cells can differentiate from Foxp3(-)CD4(+) medullary thymocytes and Foxp3(-)CD4(+) naive T cells. However, the impact of these two processes on size and composition of the peripheral repertoire of regulatory T cells is unclear. Here we followed the fate of individual Foxp3(+)CD4(+)CD25(+) thymocytes and T cells in vivo in T cell receptor (TCR) transgenic mice that express a restricted but polyclonal repertoire of TCRs. By utilizing high-throughput single-cell analysis, we showed that Foxp3(+)CD4(+) peripheral T cells were derived from thymic precursors that expressed a different TCRs than Foxp3(-)CD4(+) medullary thymocytes and Foxp3(-)CD4(+) T cells. Furthermore, the diversity of TCRs on Foxp3(+)CD4(+) regulatory T cells exceeded the diversity of TCRs on Foxp3(-)CD4(+) naive T cells, even in mice that lack expression of tissue-specific antigens. Our results imply that higher TCR diversity on Foxp3(+) regulatory T cells helps these cells to match the specificities of autoreactive and naive T cells.


Assuntos
Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/metabolismo , Linhagem da Célula , Fatores de Transcrição Forkhead/metabolismo , Receptores de Interleucina-2/metabolismo , Sequência de Aminoácidos , Animais , Autoantígenos/imunologia , Linfócitos T CD4-Positivos/imunologia , Diferenciação Celular , Células Cultivadas , Fatores de Transcrição Forkhead/química , Fatores de Transcrição Forkhead/imunologia , Camundongos , Dados de Sequência Molecular , Peptídeos/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Interleucina-2/imunologia , Timo/citologia , Timo/imunologia , Timo/metabolismo
12.
Proc Natl Acad Sci U S A ; 99(23): 15012-7, 2002 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-12411579

RESUMO

By using dendritic cells (DCs) transduced with retroviruses encoding covalent A(b)beta/peptide fusion proteins tagged with fluorescent proteins, we followed the relocation of class II MHC molecules loaded with agonist or null peptides during the onset of activation of naive and effector CD4(+) T cells. Clusters of T cell receptor (TCR)/CD3 complex formed in parallel with clusters of agonist class II MHC/peptide complexes on the surface of DCs. However, activation of naive but not effector T cells was accompanied by expulsion of the null class II MHC/peptide complexes from the T cell-DC interface. These effects were perturbed in the presence of exogenously supplied antagonist peptide. These results suggest that interference with selective relocation of agonist and null MHC/peptide complexes in the immunological synapse contributes to the inhibitory effect of antagonist peptides on the response of naive CD4(+) T cells to agonist ligands.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Antígenos de Histocompatibilidade Classe II/genética , Oligopeptídeos/farmacologia , Sequência de Aminoácidos , Animais , Antígenos de Diferenciação de Linfócitos B/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Transporte Biológico , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Cálcio/metabolismo , Columbidae , Grupo dos Citocromos c/genética , Grupo dos Citocromos c/imunologia , Células Dendríticas/fisiologia , Citometria de Fluxo , Cinética , Proteínas Luminescentes/genética , Proteínas Luminescentes/imunologia , Linfonodos/imunologia , Ativação Linfocitária , Camundongos , Camundongos Knockout , Dados de Sequência Molecular
13.
J Immunol ; 168(2): 613-20, 2002 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-11777953

RESUMO

The CD4(+)CD25(+) regulatory T cells can be found in the thymus, but their need to undergo positive and negative selection has been questioned. Instead, it has been hypothesized that CD4(+)CD25(+) cells mature following TCR binding to MHC backbone, to low abundant MHC/peptide complexes, or to class II MHC loaded with peripheral autoantigens. In all these circumstances, processes that are distinct from positive and negative selection would govern the provenance of CD4(+)CD25(+) cells in the thymus. By comparing the development of CD4(+)CD25(-) and CD4(+)CD25(+) cells in mice expressing class II MHC molecules bound with one or many peptide(s), we show that the CD4(+)CD25(+) cells appear during natural selection of CD4(+) T cells. The proportion of CD4(+)CD25(+) cells in the population of CD4(+) thymocytes remains constant, and their total number reflects the complexity of selecting class II MHC/peptide complexes. Hence, thymic development of CD4(+)CD25(+) cells does not exclusively depend on the low-density, high-affinity MHC/peptide complexes or thymic presentation of peripheral self-Ags, but, rather, these cells are selected as a portion of the natural repertoire of CD4(+) T cells. Furthermore, while resistant to deletion mediated by endogenous superantigen(s), these cells were negatively selected on class II MHC/peptide complexes. We postulate that while the CD4(+)CD25(+) thymocytes are first detectable in the thymic medulla, their functional commitment occurs in the thymic cortex.


Assuntos
Antígenos CD4/biossíntese , Epitopos de Linfócito T/imunologia , Peptídeos/imunologia , Receptores de Interleucina-2/biossíntese , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Timo/citologia , Timo/imunologia , Animais , Autoantígenos/biossíntese , Autoantígenos/metabolismo , Diferenciação Celular/imunologia , Divisão Celular/imunologia , Células Cultivadas , Deleção Clonal , Técnicas de Cocultura , Antígenos de Histocompatibilidade Classe II/biossíntese , Antígenos de Histocompatibilidade Classe II/metabolismo , Homeostase/imunologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Peptídeos/metabolismo , Ligação Proteica/imunologia , Receptores de Antígenos de Linfócitos T/biossíntese , Subpopulações de Linfócitos T/metabolismo , Timo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA