Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Elife ; 122023 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-38127070

RESUMO

Proper activation of cytotoxic T cells via the T cell receptor and the costimulatory receptor CD28 is essential for adaptive immunity against viruses, intracellular bacteria, and cancers. Through biochemical analysis of RNA:protein interactions, we uncovered a non-coding RNA circuit regulating activation and differentiation of cytotoxic T cells composed of the long non-coding RNA Malat1 (Metastasis Associated Lung Adenocarcinoma Transcript 1) and the microRNA family miR-15/16. miR-15/16 is a widely and highly expressed tumor suppressor miRNA family important for cell proliferation and survival. miR-15/16 play important roles in T cell responses to viral infection, including the regulation of antigen-specific T cell expansion and memory. Comparative Argonaute-2 high-throughput sequencing of crosslinking immunoprecipitation (AHC) combined with gene expression profiling in normal and miR-15/16-deficient mouse T cells revealed a large network of hundreds of direct miR-15/16 target mRNAs, many with functional relevance for T cell activation, survival and memory formation. Among these targets, Malat1 contained the largest absolute magnitude miR-15/16-dependent AHC peak. This binding site was among the strongest lncRNA:miRNA interactions detected in the T cell transcriptome. We used CRISPR targeting with homology directed repair to generate mice with a 5-nucleotide mutation in the miR-15/16-binding site in Malat1. This mutation interrupted Malat1:miR-15/16 interaction, and enhanced the repression of other miR-15/16 target genes, including CD28. Interrupting Malat1 interaction with miR-15/16 decreased cytotoxic T cell activation, including the expression of interleukin 2 (IL-2) and a broader CD28-responsive gene program. Accordingly, Malat1 mutation diminished memory cell persistence in mice following LCMV Armstrong and Listeria monocytogenes infection. This study marks a significant advance in the study of long non-coding RNAs in the immune system by ascribing cell-intrinsic, sequence-specific in vivo function to Malat1. These findings have implications for T cell-mediated autoimmune diseases, antiviral and anti-tumor immunity, as well as lung adenocarcinoma and other malignancies where Malat1 is overexpressed.


Assuntos
Células T de Memória , MicroRNAs , RNA Longo não Codificante , Linfócitos T Citotóxicos , Animais , Camundongos , Antígenos CD28 , MicroRNAs/genética , RNA Longo não Codificante/genética
2.
Cell Rep ; 42(8): 112928, 2023 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-37542720

RESUMO

Identifying molecular circuits that control adipose tissue macrophage (ATM) function is necessary to understand how ATMs contribute to tissue homeostasis and obesity-induced insulin resistance. In this study, we find that mice with a myeloid-specific knockout of the miR-23-27-24 clusters of microRNAs (miRNAs) gain less weight on a high-fat diet but exhibit worsened glucose and insulin tolerance. Analysis of ATMs from these mice shows selectively reduced numbers and proliferation of a recently reported subset of lipid-associated CD9+Trem2+ ATMs (lipid-associated macrophages [LAMs]). Leveraging the role of miRNAs to control networks of genes, we use RNA sequencing (RNA-seq), functional screens, and biochemical assays to identify candidate target transcripts that regulate proliferation-associated signaling. We determine that miR-23 directly targets the mRNA of Eif4ebp2, a gene that restricts protein synthesis and proliferation in macrophages. Altogether, our study demonstrates that control of proliferation of a protective subset of LAMs by noncoding RNAs contributes to protection against diet-induced obesity metabolic dysfunction.


Assuntos
Resistência à Insulina , MicroRNAs , Camundongos , Animais , Tecido Adiposo/metabolismo , Obesidade/genética , Obesidade/metabolismo , Macrófagos/metabolismo , Resistência à Insulina/fisiologia , MicroRNAs/genética , MicroRNAs/metabolismo , Dieta Hiperlipídica , Lipídeos , Proliferação de Células , Camundongos Endogâmicos C57BL , Inflamação/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores Imunológicos/metabolismo
3.
bioRxiv ; 2023 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-37547023

RESUMO

Proper activation of cytotoxic T cells via the T cell receptor and the costimulatory receptor CD28 is essential for adaptive immunity against viruses, many intracellular bacteria and cancers. Through biochemical analysis of RNA:protein interactions, we uncovered a non-coding RNA circuit regulating activation and differentiation of cytotoxic T cells composed of the long non-coding RNA Malat1 (Metastasis Associated Lung Adenocarcinoma Transcript 1) and the microRNA family miR-15/16. miR-15/16 is a widely and highly expressed tumor suppressor miRNA family important for cell proliferation and survival. miR-15/16 also play important roles in T cell responses to viral infection, including the regulation of antigen-specific T cell expansion and T cell memory. Comparative Argonaute-2 high throughput sequencing of crosslinking immunoprecipitation (Ago2 HITS-CLIP, or AHC) combined with gene expression profiling in normal and miR-15/16-deficient T cells revealed a large network of several hundred direct miR-15/16 target mRNAs, many with functional relevance for T cell activation, survival and memory formation. Among these targets, the long non-coding RNA Malat1 contained the largest absolute magnitude miR-15/16-dependent AHC peak in T cells. This binding site was also among the strongest lncRNA:miRNA interactions detected in the T cell transcriptome. We used CRISPR targeting with homology directed repair to generate mice with a 5-nucleotide mutation in the miR-15/16 binding site in Malat1. This mutation interrupted Malat1:miR-15/16 interaction, and enhanced the repression of other miR-15/16 target genes, including CD28. Interrupting Malat1 interaction with miR-15/16 decreased cytotoxic T cell activation, including the expression of IL-2 and a broader CD28-responsive gene program. Accordingly, Malat1 mutation diminished memory cell persistence following LCMV Armstrong and Listeria monocytogenes infection. This study marks a significant advance in the study of long noncoding RNAs in the immune system by ascribing cell-intrinsic, sequence-specific in vivo function to Malat1. These findings have implications for T cell-mediated autoimmune diseases, antiviral and anti-tumor immunity, as well as lung adenocarcinoma and other malignancies where Malat1 is overexpressed.

4.
Am J Hum Genet ; 110(7): 1068-1085, 2023 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-37352860

RESUMO

ERI1 is a 3'-to-5' exoribonuclease involved in RNA metabolic pathways including 5.8S rRNA processing and turnover of histone mRNAs. Its biological and medical significance remain unclear. Here, we uncover a phenotypic dichotomy associated with bi-allelic ERI1 variants by reporting eight affected individuals from seven unrelated families. A severe spondyloepimetaphyseal dysplasia (SEMD) was identified in five affected individuals with missense variants but not in those with bi-allelic null variants, who showed mild intellectual disability and digital anomalies. The ERI1 missense variants cause a loss of the exoribonuclease activity, leading to defective trimming of the 5.8S rRNA 3' end and a decreased degradation of replication-dependent histone mRNAs. Affected-individual-derived induced pluripotent stem cells (iPSCs) showed impaired in vitro chondrogenesis with downregulation of genes regulating skeletal patterning. Our study establishes an entity previously unreported in OMIM and provides a model showing a more severe effect of missense alleles than null alleles within recessive genotypes, suggesting a key role of ERI1-mediated RNA metabolism in human skeletal patterning and chondrogenesis.


Assuntos
Exorribonucleases , Histonas , Humanos , Exorribonucleases/genética , Histonas/genética , Mutação de Sentido Incorreto/genética , RNA Ribossômico 5,8S , RNA , RNA Mensageiro/genética
5.
bioRxiv ; 2023 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-36993421

RESUMO

The miR-15/16 family is a highly expressed group of tumor suppressor miRNAs that target a large network of genes in T cells to restrict their cell cycle, memory formation and survival. Upon T cell activation, miR-15/16 are downregulated, allowing rapid expansion of differentiated effector T cells to mediate a sustained immune response. Here, using conditional deletion of miR-15/16 in immunosuppressive regulatory T cells (Tregs) that express FOXP3, we identify new functions of the miR-15/16 family in T cell immunity. miR-15/16 are indispensable to maintain peripheral tolerance by securing efficient suppression by a limited number of Tregs. miR-15/16-deficiency alters Treg expression of critical functional proteins including FOXP3, IL2Rα/CD25, CTLA4, PD-1 and IL7Rα/CD127, and results in accumulation of functionally impaired FOXP3loCD25loCD127hi Tregs. Excessive proliferation in the absence of miR-15/16 inhibition of cell cycle programs shifts Treg diversity and produces an effector Treg phenotype characterized by low expression of TCF1, CD25 and CD62L, and high expression of CD44. These Tregs fail to control immune activation of CD4+ effector T cells, leading to spontaneous multi-organ inflammation and increased allergic airway inflammation in a mouse model of asthma. Together, our results demonstrate that miR-15/16 expression in Tregs is essential to maintain immune tolerance.

6.
Proc Natl Acad Sci U S A ; 119(17): e2106083119, 2022 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-35446623

RESUMO

CD8 T cells mediate protection against intracellular pathogens and tumors. However, persistent antigen during chronic infections or cancer leads to T cell exhaustion, suboptimal functionality, and reduced protective capacity. Despite considerable work interrogating the transcriptional regulation of exhausted CD8 T cells (TEX), the posttranscriptional control of TEX remains poorly understood. Here, we interrogated the role of microRNAs (miRs) in CD8 T cells responding to acutely resolved or chronic viral infection and identified miR-29a as a key regulator of TEX. Enforced expression of miR-29a improved CD8 T cell responses during chronic viral infection and antagonized exhaustion. miR-29a inhibited exhaustion-driving transcriptional pathways, including inflammatory and T cell receptor signaling, and regulated ribosomal biogenesis. As a result, miR-29a fostered a memory-like CD8 T cell differentiation state during chronic infection. Thus, we identify miR-29a as a key regulator of TEX and define mechanisms by which miR-29a can divert exhaustion toward a more beneficial memory-like CD8 T cell differentiation state.


Assuntos
MicroRNAs , Neoplasias , Linfócitos T CD8-Positivos , Humanos , Imunoterapia/métodos , MicroRNAs/genética , MicroRNAs/metabolismo , Neoplasias/metabolismo , Infecção Persistente
7.
JCI Insight ; 6(5)2021 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-33682796

RESUMO

IL-13-induced goblet cell metaplasia contributes to airway remodeling and pathological mucus hypersecretion in asthma. miRNAs are potent modulators of cellular responses, but their role in mucus regulation is largely unexplored. We hypothesized that airway epithelial miRNAs play roles in IL-13-induced mucus regulation. miR-141 is highly expressed in human and mouse airway epithelium, is altered in bronchial brushings from asthmatic subjects at baseline, and is induced shortly after airway allergen exposure. We established a CRISPR/Cas9-based protocol to target miR-141 in primary human bronchial epithelial cells that were differentiated at air-liquid-interface, and goblet cell hyperplasia was induced by IL-13 stimulation. miR-141 disruption resulted in decreased goblet cell frequency, intracellular MUC5AC, and total secreted mucus. These effects correlated with a reduction in a goblet cell gene expression signature and enrichment of a basal cell gene expression signature defined by single cell RNA sequencing. Furthermore, intranasal administration of a sequence-specific mmu-miR-141-3p inhibitor in mice decreased Aspergillus-induced secreted mucus and mucus-producing cells in the lung and reduced airway hyperresponsiveness without affecting cellular inflammation. In conclusion, we have identified a miRNA that regulates pathological airway mucus production and is amenable to therapeutic manipulation through an inhaled route.


Assuntos
Remodelação das Vias Aéreas , Asma , Células Caliciformes , Interleucina-13/metabolismo , Pulmão , MicroRNAs/metabolismo , Muco/metabolismo , Animais , Aspergillus , Asma/metabolismo , Asma/patologia , Proteína 9 Associada à CRISPR , Diferenciação Celular , Células Cultivadas , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Células Caliciformes/metabolismo , Células Caliciformes/patologia , Humanos , Pulmão/citologia , Pulmão/metabolismo , Pulmão/patologia , Masculino , Metaplasia , Camundongos Endogâmicos C57BL , Mucina-5AC/metabolismo
8.
Artigo em Inglês | MEDLINE | ID: mdl-31844841

RESUMO

MicroRNAs (miRNAs) are a class of short noncoding RNAs that play critical roles in the regulation of a broad range of biological processes. Like transcription factors, miRNAs exert their effects by modulating the expression of networks of genes that operate in common or convergent pathways. CD8+ T cells are critical agents of the adaptive immune system that provide protection from infection and cancer. Here, we review the important roles of miRNAs in the regulation of CD8+ T cell biology and provide perspectives on the broader emerging principles of miRNA function.

9.
Cell Rep ; 28(11): 2795-2806.e3, 2019 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-31509743

RESUMO

The 3' UTR (UTR) of human mRNAs plays a critical role in controlling protein expression and function. Importantly, 3' UTRs of human messages are not invariant for each gene but rather are shaped by alternative polyadenylation (APA) in a cell state-dependent manner, including in response to T cell activation. However, the proteins and mechanisms driving APA regulation remain poorly understood. Here we show that the RNA-binding protein CELF2 controls APA of its own message in a signal-dependent manner by competing with core enhancers of the polyadenylation machinery for binding to RNA. We further show that CELF2 binding overlaps with APA enhancers transcriptome-wide, and almost half of 3' UTRs that undergo T cell signaling-induced APA are regulated in a CELF2-dependent manner. These studies thus reveal CELF2 to be a critical regulator of 3' UTR identity in T cells and demonstrate an additional mechanism for CELF2 in regulating polyadenylation site choice.


Assuntos
Proteínas CELF/metabolismo , Regulação da Expressão Gênica/genética , Proteínas do Tecido Nervoso/metabolismo , Poliadenilação/genética , Proteínas de Ligação a RNA/metabolismo , Regiões 3' não Traduzidas , Proteínas CELF/genética , Linhagem Celular Tumoral , Fator de Especificidade de Clivagem e Poliadenilação/genética , Fator de Especificidade de Clivagem e Poliadenilação/metabolismo , Elementos Facilitadores Genéticos , Humanos , Íntrons/genética , Proteínas do Tecido Nervoso/genética , Ligação Proteica , Fatores de Processamento de RNA/genética , Fatores de Processamento de RNA/metabolismo , RNA-Seq , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais/genética , Fator de Processamento U2AF/genética , Fator de Processamento U2AF/metabolismo , Transcriptoma
10.
Cell Rep ; 28(8): 2169-2181.e4, 2019 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-31433990

RESUMO

Coordinate control of T cell proliferation, survival, and differentiation are essential for host protection from pathogens and cancer. Long-lived memory cells, whose precursors are formed during the initial immunological insult, provide protection from future encounters, and their generation is the goal of many vaccination strategies. microRNAs (miRNAs) are key nodes in regulatory networks that shape effective T cell responses through the fine-tuning of thousands of genes. Here, using compound conditional mutant mice to eliminate miR-15/16 family miRNAs in T cells, we show that miR-15/16 restrict T cell cycle, survival, and memory T cell differentiation. High throughput sequencing of RNA isolated by cross-linking immunoprecipitation of AGO2 combined with gene expression analysis in miR-15/16-deficient T cells indicates that these effects are mediated through the direct inhibition of an extensive network of target genes within pathways critical to cell cycle, survival, and memory.


Assuntos
Ciclo Celular , Diferenciação Celular , Memória Imunológica , MicroRNAs/metabolismo , Linfócitos T/citologia , Linfócitos T/imunologia , Animais , Antígenos/metabolismo , Ciclo Celular/genética , Diferenciação Celular/genética , Sobrevivência Celular/genética , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Loci Gênicos , Vírus da Coriomeningite Linfocítica/fisiologia , Camundongos Transgênicos , MicroRNAs/genética
11.
Immunohorizons ; 3(8): 389-401, 2019 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-31427364

RESUMO

The germinal center (GC) is the anatomical site where humoral immunity evolves. B cells undergo cycles of proliferation and selection to produce high-affinity Abs against Ag. Direct linkage of a TLR9 agonist (CpG) to a T-dependent Ag increases the number of GC B cells. We used a T-dependent Ag complexed with CpG and a genetic model for ablating the TLR9 signaling adaptor molecule MyD88 specifically in B cells (B-MyD88- mice) together with transcriptomics to determine how this innate pathway positively regulates the GC. GC B cells from complex Ag-immunized B-MyD88- mice were defective in inducing gene expression signatures downstream of c-Myc and mTORC1. In agreement with the latter gene signature, ribosomal protein S6 phosphorylation was increased in GC B cells from wild-type mice compared with B-MyD88- mice. However, GC B cell expression of a c-Myc protein reporter was enhanced by CpG attached to Ag in both wild-type and B-MyD88- mice, indicating a B cell-extrinsic effect on c-Myc protein expression combined with a B cell-intrinsic enhancement of gene expression downstream of c-Myc. Both mTORC1 activity and c-Myc are directly induced by T cell help, indicating that TLR9 signaling in GC B cells either enhances their access to T cell help or directly influences these pathways to further enhance the effect of T cell help. Taken together, these findings indicate that TLR9 signaling in the GC could provide a surrogate prosurvival stimulus, "TLR help," thus lowering the threshold for selection and increasing the magnitude of the GC response.


Assuntos
Antígenos/química , Linfócitos B/imunologia , Centro Germinativo/metabolismo , Ligantes , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Receptor Toll-Like 9/agonistas , Animais , Antígenos/metabolismo , Ativação Linfocitária/imunologia , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Camundongos , Camundongos Transgênicos , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Transcriptoma , gama-Globulinas/imunologia
12.
Cell Rep ; 26(4): 933-944.e4, 2019 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-30673615

RESUMO

Extracellular RNAs (exRNAs) can be released by numerous cell types in vitro, are often protected within vesicles, and can modify recipient cell function. To determine how the composition and cellular sources of exRNAs and the extracellular vesicles (EVs) that carry them change in vivo during tissue inflammation, we analyzed bronchoalveolar lavage fluid (BALF) from mice before and after lung allergen challenge. In the lung, extracellular microRNAs (ex-miRNAs) had a composition that was highly correlated with airway-lining epithelium. Using cell type-specific membrane tagging and single vesicle flow, we also found that 80% of detected vesicles were of epithelial origin. After the induction of allergic airway inflammation, miRNAs selectively expressed by immune cells, including miR-223 and miR-142a, increased and hematopoietic-cell-derived EVs also increased >2-fold. These data demonstrate that infiltrating immune cells release ex-miRNAs and EVs in inflamed tissues to alter the local extracellular environment.


Assuntos
Asma/metabolismo , Líquido da Lavagem Broncoalveolar , Pulmão/metabolismo , MicroRNAs/metabolismo , Animais , Camundongos , Camundongos Transgênicos
13.
J Exp Med ; 214(12): 3627-3643, 2017 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-29122948

RESUMO

MicroRNAs (miRNAs) exert powerful effects on immunity through coordinate regulation of multiple target genes in a wide variety of cells. Type 2 innate lymphoid cells (ILC2s) are tissue sentinel mediators of allergic inflammation. We established the physiological requirements for miRNAs in ILC2 homeostasis and immune function and compared the global miRNA repertoire of resting and activated ILC2s and T helper type 2 (TH2) cells. After exposure to the natural allergen papain, mice selectively lacking the miR-17∼92 cluster in ILC2s displayed reduced lung inflammation. Moreover, miR-17∼92-deficient ILC2s exhibited defective growth and cytokine expression in response to IL-33 and thymic stromal lymphopoietin in vitro. The miR-17∼92 cluster member miR-19a promoted IL-13 and IL-5 production and inhibited expression of several targets, including SOCS1 and A20, signaling inhibitors that limit IL-13 and IL-5 production. These findings establish miRNAs as important regulators of ILC2 biology, reveal overlapping but nonidentical miRNA-regulated gene expression networks in ILC2s and TH2 cells, and reinforce the therapeutic potential of targeting miR-19 to alleviate pathogenic allergic responses.


Assuntos
Homeostase/genética , Hipersensibilidade/genética , Hipersensibilidade/imunologia , Imunidade Inata/genética , Inflamação/patologia , Linfócitos/metabolismo , MicroRNAs/metabolismo , Animais , Proliferação de Células , Citocinas/biossíntese , Regulação da Expressão Gênica , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Análise de Sequência de RNA , Proteína 1 Supressora da Sinalização de Citocina/metabolismo , Células Th2/metabolismo , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/metabolismo
14.
Proc Natl Acad Sci U S A ; 113(31): 8765-70, 2016 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-27432971

RESUMO

Type 2 inflammation occurs in a large subgroup of asthmatics, and novel cytokine-directed therapies are being developed to treat this population. In mouse models, interleukin-33 (IL-33) activates lung resident innate lymphoid type 2 cells (ILC2s) to initiate airway type 2 inflammation. In human asthma, which is chronic and difficult to model, the role of IL-33 and the target cells responsible for persistent type 2 inflammation remain undefined. Full-length IL-33 is a nuclear protein and may function as an "alarmin" during cell death, a process that is uncommon in chronic stable asthma. We demonstrate a previously unidentified mechanism of IL-33 activity that involves alternative transcript splicing, which may operate in stable asthma. In human airway epithelial cells, alternative splicing of the IL-33 transcript is consistently present, and the deletion of exons 3 and 4 (Δ exon 3,4) confers cytoplasmic localization and facilitates extracellular secretion, while retaining signaling capacity. In nonexacerbating asthmatics, the expression of Δ exon 3,4 is strongly associated with airway type 2 inflammation, whereas full-length IL-33 is not. To further define the extracellular role of IL-33 in stable asthma, we sought to determine the cellular targets of its activity. Comprehensive flow cytometry and RNA sequencing of sputum cells suggest basophils and mast cells, not ILC2s, are the cellular sources of type 2 cytokines in chronic asthma. We conclude that IL-33 isoforms activate basophils and mast cells to drive type 2 inflammation in chronic stable asthma, and novel IL-33 inhibitors will need to block all biologically active isoforms.


Assuntos
Processamento Alternativo , Asma/genética , Inflamação/genética , Interleucina-33/genética , Adulto , Asma/metabolismo , Basófilos/metabolismo , Linhagem Celular , Células Epiteliais/metabolismo , Feminino , Perfilação da Expressão Gênica/métodos , Humanos , Inflamação/metabolismo , Proteína 1 Semelhante a Receptor de Interleucina-1/genética , Proteína 1 Semelhante a Receptor de Interleucina-1/metabolismo , Interleucina-33/metabolismo , Masculino , Mastócitos/metabolismo , Pessoa de Meia-Idade , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia , Escarro/citologia , Escarro/metabolismo , Adulto Jovem
15.
Cancer Cell ; 27(3): 409-25, 2015 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-25759025

RESUMO

Studying 830 pre-B ALL cases from four clinical trials, we found that human ALL can be divided into two fundamentally distinct subtypes based on pre-BCR function. While absent in the majority of ALL cases, tonic pre-BCR signaling was found in 112 cases (13.5%). In these cases, tonic pre-BCR signaling induced activation of BCL6, which in turn increased pre-BCR signaling output at the transcriptional level. Interestingly, inhibition of pre-BCR-related tyrosine kinases reduced constitutive BCL6 expression and selectively killed patient-derived pre-BCR(+) ALL cells. These findings identify a genetically and phenotypically distinct subset of human ALL that critically depends on tonic pre-BCR signaling. In vivo treatment studies suggested that pre-BCR tyrosine kinase inhibitors are useful for the treatment of patients with pre-BCR(+) ALL.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Regulação Neoplásica da Expressão Gênica , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Células Precursoras de Linfócitos B/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Ensaios Clínicos como Assunto , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Dados de Sequência Molecular , Fosfatidilinositol 3-Quinase/metabolismo , Fator de Transcrição 1 de Leucemia de Células Pré-B , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-bcl-6 , Transdução de Sinais , Quinase Syk , Regulação para Cima , Quinases da Família src/metabolismo
16.
Immunity ; 42(2): 265-278, 2015 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-25680272

RESUMO

During persistent antigen stimulation, CD8(+) T cells show a gradual decrease in effector function, referred to as exhaustion, which impairs responses in the setting of tumors and infections. Here we demonstrate that the transcription factor NFAT controls the program of T cell exhaustion. When expressed in cells, an engineered form of NFAT1 unable to interact with AP-1 transcription factors diminished T cell receptor (TCR) signaling, increased the expression of inhibitory cell surface receptors, and interfered with the ability of CD8(+) T cells to protect against Listeria infection and attenuate tumor growth in vivo. We defined the genomic regions occupied by endogenous and engineered NFAT1 in primary CD8(+) T cells and showed that genes directly induced by the engineered NFAT1 overlapped with genes expressed in exhausted CD8(+) T cells in vivo. Our data show that NFAT promotes T cell anergy and exhaustion by binding at sites that do not require cooperation with AP-1.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Anergia Clonal/genética , Fatores de Transcrição NFATC/fisiologia , Proteínas Recombinantes/farmacologia , Fator de Transcrição AP-1/metabolismo , Animais , Células Cultivadas , Anergia Clonal/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Listeria monocytogenes/imunologia , Listeriose/imunologia , Listeriose/microbiologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Transgênicos , Fatores de Transcrição NFATC/genética , Neoplasias/imunologia , Regiões Promotoras Genéticas/genética , Receptores de Antígenos de Linfócitos T/imunologia , Proteínas Recombinantes/genética
17.
Nat Immunol ; 15(8): 777-88, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24997565

RESUMO

A characteristic feature of asthma is the aberrant accumulation, differentiation or function of memory CD4(+) T cells that produce type 2 cytokines (TH2 cells). By mapping genome-wide histone modification profiles for subsets of T cells isolated from peripheral blood of healthy and asthmatic individuals, we identified enhancers with known and potential roles in the normal differentiation of human TH1 cells and TH2 cells. We discovered disease-specific enhancers in T cells that differ between healthy and asthmatic individuals. Enhancers that gained the histone H3 Lys4 dimethyl (H3K4me2) mark during TH2 cell development showed the highest enrichment for asthma-associated single nucleotide polymorphisms (SNPs), which supported a pathogenic role for TH2 cells in asthma. In silico analysis of cell-specific enhancers revealed transcription factors, microRNAs and genes potentially linked to human TH2 cell differentiation. Our results establish the feasibility and utility of enhancer profiling in well-defined populations of specialized cell types involved in disease pathogenesis.


Assuntos
Asma/genética , Asma/imunologia , Predisposição Genética para Doença , Células Th1/imunologia , Células Th2/imunologia , Adolescente , Adulto , Idoso , Sítios de Ligação/genética , Sítios de Ligação/imunologia , Diferenciação Celular/imunologia , Células Cultivadas , Subunidade alfa 3 de Fator de Ligação ao Core/genética , Metilação de DNA/genética , Epigenômica , Feminino , Fator de Transcrição GATA3/genética , Estudo de Associação Genômica Ampla , Histonas/genética , Histonas/imunologia , Humanos , Memória Imunológica/imunologia , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas/genética , Ligação Proteica/genética , Ligação Proteica/imunologia , Análise de Sequência de RNA , Proteínas com Domínio T/genética , Adulto Jovem
18.
Curr Opin Immunol ; 28: 6-11, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24530656

RESUMO

The generation of germinal centers (GCs) is a hallmark feature of the adaptive immune response, resulting in the production of high-affinity antibodies that neutralize pathogens and confer protection upon reinfection. The GC response requires interactions between different immune cell types, and the coordination of complex and dynamic gene expression networks within these cells. Here we provide deeper insights into how microRNAs, small endogenously expressed RNAs, regulate the cellular processes involved in the differentiation and function of T follicular helper cells and germinal center B cells, the two main players of the T cell-dependent humoral immune response.


Assuntos
Centro Germinativo/imunologia , MicroRNAs/imunologia , Animais , Linfócitos B/imunologia , Transformação Celular Neoplásica/imunologia , Regulação da Expressão Gênica , Humanos , Linfócitos T Auxiliares-Indutores/imunologia
20.
Immunity ; 38(3): 596-605, 2013 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-23499493

RESUMO

T follicular helper (Tfh) cells provide help to B cells and are crucial for establishment of germinal center (GC) reactions, including production of high-affinity antibodies and generation of memory B cells and long-lived plasma cells. Here we report that the magnitude of the Tfh cell response was dictated by the amount of antigen and directly correlated with the magnitude of the GC B cell response. In addition, maintenance of the Tfh cell phenotype required sustained antigenic stimulation by GC B cells. In lymphopenic conditions, a strong and prolonged Tfh cell response led to bystander B cell activation, hypergammaglobulinemia, and production of poly- and self-reactive antibodies. These data demonstrate that antigen dose determines the size and duration of the Tfh cell response and GC reaction, highlight the transient nature of the Tfh cell phenotype, and suggest a link between overstimulation of Tfh cells and the development of dysregulated humoral immune responses.


Assuntos
Antígenos/imunologia , Linfócitos B/imunologia , Centro Germinativo/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Anticorpos/imunologia , Linfócitos B/metabolismo , Complexo CD3/genética , Complexo CD3/imunologia , Complexo CD3/metabolismo , Antígenos CD40/imunologia , Antígenos CD40/metabolismo , Ligante de CD40/imunologia , Ligante de CD40/metabolismo , Linhagem Celular Tumoral , Citometria de Fluxo , Centro Germinativo/metabolismo , Humanos , Imunização/métodos , Imunofenotipagem , Ligante Coestimulador de Linfócitos T Induzíveis/imunologia , Ligante Coestimulador de Linfócitos T Induzíveis/metabolismo , Proteína Coestimuladora de Linfócitos T Induzíveis/imunologia , Proteína Coestimuladora de Linfócitos T Induzíveis/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Contagem de Linfócitos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Ovalbumina/administração & dosagem , Ovalbumina/imunologia , Ligação Proteica/imunologia , Receptores CXCR5/genética , Receptores CXCR5/imunologia , Receptores CXCR5/metabolismo , Linfócitos T Auxiliares-Indutores/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA