Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 23(12): 1763-1776, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36316474

RESUMEN

The nuclear corepressors NCOR1 and NCOR2 interact with transcription factors involved in B cell development and potentially link these factors to alterations in chromatin structure and gene expression. Herein, we demonstrate that Ncor1/2 deletion limits B cell differentiation via impaired recombination, attenuates pre-BCR signaling and enhances STAT5-dependent transcription. Furthermore, NCOR1/2-deficient B cells exhibited derepression of EZH2-repressed gene modules, including the p53 pathway. These alterations resulted in aberrant Rag1 and Rag2 expression and accessibility. Whole-genome sequencing of Ncor1/2 DKO B cells identified increased number of structural variants with cryptic recombination signal sequences. Finally, deletion of Ncor1 alleles in mice facilitated leukemic transformation, whereas human leukemias with less NCOR1 correlated with worse survival. NCOR1/2 mutations in human leukemia correlated with increased RAG expression and number of structural variants. These studies illuminate how the corepressors NCOR1/2 regulate B cell differentiation and provide insights into how NCOR1/2 mutations may promote B cell transformation.


Asunto(s)
Hematopoyesis , Transducción de Señal , Ratones , Humanos , Animales , Proteínas Co-Represoras , Núcleo Celular , Genómica , Co-Represor 2 de Receptor Nuclear/genética , Co-Represor 1 de Receptor Nuclear/genética
2.
Nat Immunol ; 20(2): 195-205, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30643267

RESUMEN

The developmental programs that generate a broad repertoire of regulatory T cells (Treg cells) able to respond to both self antigens and non-self antigens remain unclear. Here we found that mature Treg cells were generated through two distinct developmental programs involving CD25+ Treg cell progenitors (CD25+ TregP cells) and Foxp3lo Treg cell progenitors (Foxp3lo TregP cells). CD25+ TregP cells showed higher rates of apoptosis and interacted with thymic self antigens with higher affinity than did Foxp3lo TregP cells, and had a T cell antigen receptor repertoire and transcriptome distinct from that of Foxp3lo TregP cells. The development of both CD25+ TregP cells and Foxp3lo TregP cells was controlled by distinct signaling pathways and enhancers. Transcriptomics and histocytometric data suggested that CD25+ TregP cells and Foxp3lo TregP cells arose by coopting negative-selection programs and positive-selection programs, respectively. Treg cells derived from CD25+ TregP cells, but not those derived from Foxp3lo TregP cells, prevented experimental autoimmune encephalitis. Our findings indicate that Treg cells arise through two distinct developmental programs that are both required for a comprehensive Treg cell repertoire capable of establishing immunotolerance.


Asunto(s)
Diferenciación Celular/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Células Progenitoras Linfoides/fisiología , Linfocitos T Reguladores/fisiología , Timo/crecimiento & desarrollo , Animales , Autoantígenos/inmunología , Colitis/inmunología , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Adyuvante de Freund/administración & dosificación , Adyuvante de Freund/inmunología , Humanos , Tolerancia Inmunológica/inmunología , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Células Progenitoras Linfoides/trasplante , Ratones , Ratones Transgénicos , Mycobacterium tuberculosis/inmunología , Glicoproteína Mielina-Oligodendrócito/administración & dosificación , Glicoproteína Mielina-Oligodendrócito/inmunología , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/inmunología , Transducción de Señal , Organismos Libres de Patógenos Específicos , Timo/citología , Timo/inmunología
3.
Nat Immunol ; 18(6): 694-704, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28369050

RESUMEN

The transcription factor STAT5 has a critical role in B cell acute lymphoblastic leukemia (B-ALL). How STAT5 mediates this effect is unclear. Here we found that activation of STAT5 worked together with defects in signaling components of the precursor to the B cell antigen receptor (pre-BCR), including defects in BLNK, BTK, PKCß, NF-κB1 and IKAROS, to initiate B-ALL. STAT5 antagonized the transcription factors NF-κB and IKAROS by opposing regulation of shared target genes. Super-enhancers showed enrichment for STAT5 binding and were associated with an opposing network of transcription factors, including PAX5, EBF1, PU.1, IRF4 and IKAROS. Patients with a high ratio of active STAT5 to NF-κB or IKAROS had more-aggressive disease. Our studies indicate that an imbalance of two opposing transcriptional programs drives B-ALL and suggest that restoring the balance of these pathways might inhibit B-ALL.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Linfocitos B , Regulación Neoplásica de la Expresión Génica , Factor de Transcripción Ikaros/genética , Receptores de Células Precursoras de Linfocitos B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Factor de Transcripción STAT5/metabolismo , Agammaglobulinemia Tirosina Quinasa , Animales , Inmunoprecipitación de Cromatina , Citometría de Flujo , Humanos , Factores Reguladores del Interferón/genética , Ratones , Reacción en Cadena de la Polimerasa Multiplex , Subunidad p50 de NF-kappa B/genética , Factor de Transcripción PAX5/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/mortalidad , Pronóstico , Proteína Quinasa C beta/genética , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Tasa de Supervivencia , Transactivadores/genética
4.
Nat Immunol ; 15(5): 473-81, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24633226

RESUMEN

Regulatory T cells (Treg cells) express members of the tumor-necrosis factor (TNF) receptor superfamily (TNFRSF), but the role of those receptors in the thymic development of Treg cells is undefined. We found here that Treg cell progenitors had high expression of the TNFRSF members GITR, OX40 and TNFR2. Expression of those receptors correlated directly with the signal strength of the T cell antigen receptor (TCR) and required the coreceptor CD28 and the kinase TAK1. The neutralization of ligands that are members of the TNF superfamily (TNFSF) diminished the development of Treg cells. Conversely, TNFRSF agonists enhanced the differentiation of Treg cell progenitors by augmenting responsiveness of the interleukin 2 receptor (IL-2R) and transcription factor STAT5. Costimulation with the ligand of GITR elicited dose-dependent enrichment for cells of lower TCR affinity in the Treg cell repertoire. In vivo, combined inhibition of GITR, OX40 and TNFR2 abrogated the development of Treg cells. Thus, expression of members of the TNFRSF on Treg cell progenitors translated strong TCR signals into molecular parameters that specifically promoted the development of Treg cells and shaped the Treg cell repertoire.


Asunto(s)
Receptor Cross-Talk , Receptores de Antígenos de Linfocitos T/agonistas , Linfocitos T Reguladores/inmunología , Timo/inmunología , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo , Animales , Antígenos CD28/genética , Antígenos CD28/metabolismo , Diferenciación Celular/genética , Células Cultivadas , Proteína Relacionada con TNFR Inducida por Glucocorticoide/genética , Proteína Relacionada con TNFR Inducida por Glucocorticoide/metabolismo , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor Cross-Talk/inmunología , Receptores OX40/genética , Receptores OX40/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Factor de Transcripción STAT5/metabolismo , Transducción de Señal/genética , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/genética
5.
J Allergy Clin Immunol ; 154(1): 143-156, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38185418

RESUMEN

BACKGROUND: Dedicator of cytokinesis 8 (DOCK8)-deficient patients have severe eczema, elevated IgE, and eosinophilia, features of atopic dermatitis (AD). OBJECTIVE: We sought to understand the mechanisms of eczema in DOCK8 deficiency. METHODS: Skin biopsy samples were characterized by histology, immunofluorescence microscopy, and gene expression. Skin barrier function was measured by transepidermal water loss. Allergic skin inflammation was elicited in mice by epicutaneous sensitization with ovalbumin (OVA) or cutaneous application of Staphylococcus aureus. RESULTS: Skin lesions of DOCK8-deficient patients exhibited type 2 inflammation, and the patients' skin was colonized by Saureus, as in AD. Unlike in AD, DOCK8-deficient patients had a reduced FOXP3:CD4 ratio in their skin lesions, and their skin barrier function was intrinsically intact. Dock8-/- mice exhibited reduced numbers of cutaneous T regulatory (Treg) cells and a normal skin barrier. Dock8-/- and mice with an inducible Dock8 deletion in Treg cells exhibited increased allergic skin inflammation after epicutaneous sensitization with OVA. DOCK8 was shown to be important for Treg cell stability at sites of allergic inflammation and for the generation, survival, and suppressive activity of inducible Treg cells. Adoptive transfer of wild-type, but not DOCK8-deficient, OVA-specific, inducible Treg cells suppressed allergic inflammation in OVA-sensitized skin of Dock8-/- mice. These mice developed severe allergic skin inflammation and elevated serum IgE levels after topical exposure to Saureus. Both were attenuated after adoptive transfer of WT but not DOCK8-deficient Treg cells. CONCLUSION: Treg cell dysfunction increases susceptibility to allergic skin inflammation in DOCK8 deficiency and synergizes with cutaneous exposure to Saureus to drive eczema in DOCK8 deficiency.


Asunto(s)
Eccema , Factores de Intercambio de Guanina Nucleótido , Ratones Noqueados , Piel , Staphylococcus aureus , Linfocitos T Reguladores , Animales , Linfocitos T Reguladores/inmunología , Factores de Intercambio de Guanina Nucleótido/deficiencia , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/inmunología , Eccema/inmunología , Staphylococcus aureus/inmunología , Humanos , Ratones , Piel/inmunología , Piel/patología , Femenino , Masculino , Ratones Endogámicos C57BL , Dermatitis Atópica/inmunología
6.
Blood ; 140(4): 335-348, 2022 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-35275990

RESUMEN

Patients with acute lymphoblastic leukemia have experienced significantly improved outcomes due to the advent of chimeric antigen receptor (CAR) T cells and bispecific T-cell engagers, although a proportion of patients still relapse despite these advances. T-cell exhaustion has been recently suggested to be an important driver of relapse in these patients. Indeed, phenotypic exhaustion of CD4+ T cells is predictive of relapse and poor overall survival in B-cell acute lymphoblastic leukemia (B-ALL). Thus, therapies that counter T-cell exhaustion, such as immune checkpoint blockade, may improve leukemia immunosurveillance and prevent relapse. Here, we used a murine model of Ph+ B-ALL as well as human bone marrow biopsy samples to assess the fundamental nature of CD4+ T-cell exhaustion and the preclinical therapeutic potential for combining anti-PD-L1 based checkpoint blockade with tyrosine kinase inhibitors targeting the BCR-ABL oncoprotein. Single-cell RNA-sequence analysis revealed that B-ALL induces a unique subset of CD4+ T cells with both cytotoxic and helper functions. Combination treatment with the tyrosine kinase inhibitor nilotinib and anti-PD-L1 dramatically improves long-term survival of leukemic mice. Depletion of CD4+ T cells prior to therapy completely abrogates the survival benefit, implicating CD4+ T cells as key drivers of the protective anti-leukemia immune response. Indeed, treatment with anti-PD-L1 leads to clonal expansion of leukemia-specific CD4+ T cells with the aforementioned helper/cytotoxic phenotype as well as reduced expression of exhaustion markers. These findings support efforts to use PD1/PD-L1 checkpoint blockade in clinical trials and highlight the importance of CD4+ T-cell dysfunction in limiting the endogenous anti-leukemia response.


Asunto(s)
Antineoplásicos , Leucemia de Células B , Leucemia-Linfoma Linfoblástico de Células Precursoras , Animales , Antígeno B7-H1 , Linfocitos T CD4-Positivos , Humanos , Ratones , Pirimidinas , Recurrencia
7.
J Immunol ; 209(7): 1300-1313, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-36038290

RESUMEN

Recent studies have demonstrated that regulatory T cells (Tregs) develop in the thymus via two pathways involving distinct Treg progenitors (TregP): CD25+FOXP3- (CD25+ TregP) and CD25-FOXP3lo (FOXP3lo TregP) Treg progenitors. To examine this process in more detail, we carried out single-cell RNA sequencing (scRNA-Seq) and TCR-Seq on sorted murine CD4+CD8+ double-positive (DP) thymocytes, CD4+ single-positive (CD4SP) thymocytes, CD25+FOXP3-CD73- TregP, CD25-FOXP3loCD73- TregP, newly generated mature CD25+FOXP3+CD73- Tregs, and FOXP3+CD73+ recirculating/long-term resident Tregs (RT-Tregs). Sorted populations were individually hashtagged and then combined into one scRNA-Seq/TCR-Seq library before sequencing and subsequent analysis. We found that both CD25+ TregP and FOXP3lo TregP arise via an initial agonist-activated state that gives rise to a second transitional stage before differentiating into mature Tregs Using both scRNA-Seq and bulk RNA-Seq on sorted thymocyte subsets, we demonstrate that CD25+ TregP are significantly enriched for Il2 production, suggesting that they are the major source of IL-2 needed to convert TregP into mature Tregs Using TCR-Seq, we found that several TCRs were clearly biased in favor of the conventional or Treg lineages, but that a large fraction of TCRs were found in both these lineages. Finally, we found that RT-Tregs in the thymus are not monomorphic but are composed of multiple distinct subsets and that these RT-Tregs express the most diverse TCR repertoire of all CD4SP thymocytes. Thus, our studies define multiple stages of Treg differentiation within the murine thymus and serve as a resource for future studies on CD4+ thymocyte development and Treg differentiation.


Asunto(s)
Interleucina-2 , Linfocitos T Reguladores , Animales , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Interleucina-2/metabolismo , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Ratones , Receptores de Antígenos de Linfocitos T/metabolismo , Análisis de Secuencia de ARN , Linfocitos T Reguladores/metabolismo , Timo/metabolismo
8.
Proc Natl Acad Sci U S A ; 116(25): 12442-12451, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31147469

RESUMEN

Tumor-associated macrophages contribute to tumor progression and therapeutic resistance in breast cancer. Within the tumor microenvironment, tumor-derived factors activate pathways that modulate macrophage function. Using in vitro and in vivo models, we find that tumor-derived factors induce activation of the Janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3) pathway in macrophages. We also demonstrate that loss of STAT3 in myeloid cells leads to enhanced mammary tumorigenesis. Further studies show that macrophages contribute to resistance of mammary tumors to the JAK/STAT inhibitor ruxolitinib in vivo and that ruxolitinib-treated macrophages produce soluble factors that promote resistance of tumor cells to JAK inhibition in vitro. Finally, we demonstrate that STAT3 deletion and JAK/STAT inhibition in macrophages increases expression of the protumorigenic factor cyclooxygenase-2 (COX-2), and that COX-2 inhibition enhances responsiveness of tumors to ruxolitinib. These findings define a mechanism through which macrophages promote therapeutic resistance and highlight the importance of understanding the impact of targeted therapies on the tumor microenvironment.


Asunto(s)
Carcinogénesis , Inhibidores de las Cinasas Janus/farmacología , Macrófagos/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Humanos , Macrófagos/enzimología , Ratones , Nitrilos , Pirazoles/farmacología , Pirimidinas , Microambiente Tumoral
9.
Genes Dev ; 28(12): 1337-50, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24939936

RESUMEN

Loss-of-function mutations in hematopoietic transcription factors including PAX5 occur in most cases of B-progenitor acute lymphoblastic leukemia (B-ALL), a disease characterized by the accumulation of undifferentiated lymphoblasts. Although PAX5 mutation is a critical driver of B-ALL development in mice and humans, it remains unclear how its loss contributes to leukemogenesis and whether ongoing PAX5 deficiency is required for B-ALL maintenance. Here we used transgenic RNAi to reversibly suppress endogenous Pax5 expression in the hematopoietic compartment of mice, which cooperates with activated signal transducer and activator of transcription 5 (STAT5) to induce B-ALL. In this model, restoring endogenous Pax5 expression in established B-ALL triggers immunophenotypic maturation and durable disease remission by engaging a transcriptional program reminiscent of normal B-cell differentiation. Notably, even brief Pax5 restoration in B-ALL cells causes rapid cell cycle exit and disables their leukemia-initiating capacity. These and similar findings in human B-ALL cell lines establish that Pax5 hypomorphism promotes B-ALL self-renewal by impairing a differentiation program that can be re-engaged despite the presence of additional oncogenic lesions. Our results establish a causal relationship between the hallmark genetic and phenotypic features of B-ALL and suggest that engaging the latent differentiation potential of B-ALL cells may provide new therapeutic entry points.


Asunto(s)
Diferenciación Celular/genética , Factor de Transcripción PAX5/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Células Precursoras de Linfocitos B/citología , Animales , Línea Celular Tumoral , Células Cultivadas , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Genes myc/genética , Humanos , Ratones , Ratones Transgénicos , Factor de Transcripción PAX5/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal
10.
Nat Immunol ; 10(6): 655-64, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19448632

RESUMEN

Coordinated recombination of homologous antigen receptor loci is thought to be important for allelic exclusion. Here we show that homologous immunoglobulin alleles pair in a stage-specific way that mirrors the recombination patterns of these loci. The frequency of homologous immunoglobulin pairing was much lower in the absence of the RAG-1-RAG-2 recombinase and was restored in Rag1-/- developing B cells with a transgene expressing a RAG-1 active-site mutant that supported DNA binding but not cleavage. The introduction of DNA breaks on one immunoglobulin allele induced ATM-dependent repositioning of the other allele to pericentromeric heterochromatin. ATM activated by the cleaved allele acts in trans on the uncleaved allele to prevent biallelic recombination and chromosome breaks or translocations.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas de Unión al ADN/genética , Proteínas de Homeodominio/genética , Inmunoglobulinas/genética , Proteínas Serina-Treonina Quinasas/genética , Recombinación Genética , Proteínas Supresoras de Tumor/genética , Alelos , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Linfocitos B/metabolismo , Células Cultivadas , Roturas del ADN , Reordenamiento Génico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , VDJ Recombinasas/metabolismo
11.
J Immunol ; 203(8): 2031-2041, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31591259

RESUMEN

Development of a comprehensive regulatory T (Treg) cell compartment in the thymus is required to maintain immune homeostasis and prevent autoimmunity. In this study, we review cellular and molecular determinants of Treg cell development in the thymus. We focus on the evidence for a self-antigen-focused Treg cell repertoire as well as the APCs responsible for presenting self-antigens to developing thymocytes. We also cover the contribution of different cytokines to thymic Treg development and the cellular populations that produce these cytokines. Finally, we update the originally proposed "two-step" model of thymic Treg differentiation by incorporating new evidence demonstrating that Treg cells develop from two Treg progenitor populations and discuss the functional importance of Treg cells generated via either progenitor pathway.


Asunto(s)
Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Timo/citología , Timo/inmunología , Animales , Diferenciación Celular/inmunología , Homeostasis/inmunología , Humanos
12.
J Immunol ; 200(12): 3926-3933, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29728511

RESUMEN

The cytokine IL-2 is critical for promoting the development, homeostasis, and function of regulatory T (Treg) cells. The cellular sources of IL-2 that promote these processes remain unclear. T cells, B cells, and dendritic cells (DCs) are known to make IL-2 in peripheral tissues. We found that T cells and DCs in the thymus also make IL-2. To identify cellular sources of IL-2 in Treg cell development and homeostasis, we used Il2FL/FL mice to selectively delete Il2 in T cells, B cells, and DCs. Because IL-15 can partially substitute for IL-2 in Treg cell development, we carried out the majority of these studies on an Il15-/- background. Deletion of Il2 in B cells, DCs, or both these subsets had no effect on Treg cell development, either in wild-type (WT) or Il15-/- mice. Deletion of Il2 in T cells had minimal effects in WT mice but virtually eliminated developing Treg cells in Il15-/- mice. In the spleen and most peripheral lymphoid organs, deletion of Il2 in B cells, DCs, or both subsets had no effect on Treg cell homeostasis. In contrast, deletion of Il2 in T cells led to a significant decrease in Treg cells in either WT or Il15-/- mice. The one exception was the mesenteric lymph nodes where significantly fewer Treg cells were observed when Il2 was deleted in both T cells and DCs. Thus, T cells are the sole source of IL-2 needed for Treg cell development, but DCs can contribute to Treg cell homeostasis in select organs.


Asunto(s)
Homeostasis/inmunología , Interleucina-2/inmunología , Linfocitos T Reguladores/inmunología , Animales , Linfocitos B/inmunología , Células Dendríticas/inmunología , Ganglios Linfáticos/inmunología , Linfocitos/inmunología , Ratones , Bazo/inmunología
13.
Dev Biol ; 428(1): 232-244, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28606561

RESUMEN

Macrophages are required for proper mammary gland development and maintaining tissue homeostasis. However, the mechanisms by which macrophages regulate this process remain unclear. Here, we identify STAT5 as an important regulator of macrophage function in the developing mammary gland. Analysis of mammary glands from mice with STAT5-deficient macrophages demonstrates delayed ductal elongation, enhanced ductal branching and increased epithelial proliferation. Further analysis reveals that STAT5 deletion in macrophages leads to enhanced expression of proliferative factors such as Cyp19a1/aromatase and IL-6. Mechanistic studies demonstrate that STAT5 binds directly to the Cyp19a1 promoter in macrophages to suppress gene expression and that loss of STAT5 results in enhanced stromal expression of aromatase. Finally, we demonstrate that STAT5 deletion in macrophages cooperates with oncogenic initiation in mammary epithelium to accelerate the formation of estrogen receptor (ER)-positive hyperplasias. These studies establish the importance of STAT5 in macrophages during ductal morphogenesis in the mammary gland and demonstrate that altering STAT5 function in macrophages can affect the development of tissue-specific disease.


Asunto(s)
Activación de Macrófagos/genética , Macrófagos/citología , Glándulas Mamarias Animales/embriología , Morfogénesis/genética , Organogénesis/genética , Factor de Transcripción STAT5/genética , Animales , Aromatasa/biosíntesis , Aromatasa/genética , Línea Celular , Proliferación Celular/genética , Activación Enzimática/genética , Células Epiteliales/citología , Femenino , Regulación de la Expresión Génica/genética , Interleucina-6/biosíntesis , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptores de Estrógenos/metabolismo
15.
PLoS Comput Biol ; 13(7): e1005482, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28683103

RESUMEN

Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL) is characterized by a very poor prognosis and a high likelihood of acquired chemo-resistance. Although tyrosine kinase inhibitor (TKI) therapy has improved clinical outcome, most ALL patients relapse following treatment with TKI due to the development of resistance. We developed an in vitro model of Nilotinib-resistant Ph+ leukemia cells to investigate whether low dose radiation (LDR) in combination with TKI therapy overcome chemo-resistance. Additionally, we developed a mathematical model, parameterized by cell viability experiments under Nilotinib treatment and LDR, to explain the cellular response to combination therapy. The addition of LDR significantly reduced drug resistance both in vitro and in computational model. Decreased expression level of phosphorylated AKT suggests that the combination treatment plays an important role in overcoming resistance through the AKT pathway. Model-predicted cellular responses to the combined therapy provide good agreement with experimental results. Augmentation of LDR and Nilotinib therapy seems to be beneficial to control Ph+ leukemia resistance and the quantitative model can determine optimal dosing schedule to enhance the effectiveness of the combination therapy.


Asunto(s)
Quimioradioterapia/métodos , Modelos Biológicos , Leucemia-Linfoma Linfoblástico de Células Precursoras/fisiopatología , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirimidinas/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Línea Celular Tumoral , Simulación por Computador , Resistencia a Antineoplásicos/efectos de la radiación , Ratones , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Resultado del Tratamiento
16.
J Immunol ; 196(11): 4793-804, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27183622

RESUMEN

Checkpoint blockade-based immunotherapies are effective in cancers with high numbers of nonsynonymous mutations. In contrast, current paradigms suggest that such approaches will be ineffective in cancers with few nonsynonymous mutations. To examine this issue, we made use of a murine model of BCR-ABL(+) B-lineage acute lymphoblastic leukemia. Using a principal component analysis, we found that robust MHC class II expression, coupled with appropriate costimulation, correlated with lower leukemic burden. We next assessed whether checkpoint blockade or therapeutic vaccination could improve survival in mice with pre-established leukemia. Consistent with the low mutation load in our leukemia model, we found that checkpoint blockade alone had only modest effects on survival. In contrast, robust heterologous vaccination with a peptide derived from the BCR-ABL fusion (BAp), a key driver mutation, generated a small population of mice that survived long-term. Checkpoint blockade strongly synergized with heterologous vaccination to enhance overall survival in mice with leukemia. Enhanced survival did not correlate with numbers of BAp:I-A(b)-specific T cells, but rather with increased expression of IL-10, IL-17, and granzyme B and decreased expression of programmed death 1 on these cells. Our findings demonstrate that vaccination to key driver mutations cooperates with checkpoint blockade and allows for immune control of cancers with low nonsynonymous mutation loads.


Asunto(s)
Puntos de Control del Ciclo Celular/fisiología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Vacunación , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
17.
J Immunol ; 195(8): 4028-37, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26378075

RESUMEN

BCR-ABL(+) acute lymphoblastic leukemia patients have transient responses to current therapies. However, the fusion of BCR to ABL generates a potential leukemia-specific Ag that could be a target for immunotherapy. We demonstrate that the immune system can limit BCR-ABL(+) leukemia progression although ultimately this immune response fails. To address how BCR-ABL(+) leukemia escapes immune surveillance, we developed a peptide: MHC class II tetramer that labels endogenous BCR-ABL-specific CD4(+) T cells. Naive mice harbored a small population of BCR-ABL-specific T cells that proliferated modestly upon immunization. The small number of naive BCR-ABL-specific T cells was due to negative selection in the thymus, which depleted BCR-ABL-specific T cells. Consistent with this observation, we saw that BCR-ABL-specific T cells were cross-reactive with an endogenous peptide derived from ABL. Despite this cross-reactivity, the remaining population of BCR-ABL reactive T cells proliferated upon immunization with the BCR-ABL fusion peptide and adjuvant. In response to BCR-ABL(+) leukemia, BCR-ABL-specific T cells proliferated and converted into regulatory T (Treg) cells, a process that was dependent on cross-reactivity with self-antigen, TGF-ß1, and MHC class II Ag presentation by leukemic cells. Treg cells were critical for leukemia progression in C57BL/6 mice, as transient Treg cell ablation led to extended survival of leukemic mice. Thus, BCR-ABL(+) leukemia actively suppresses antileukemia immune responses by converting cross-reactive leukemia-specific T cells into Treg cells.


Asunto(s)
Presentación de Antígeno , Neoplasias Experimentales/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Linfocitos T Reguladores/inmunología , Animales , Reacciones Cruzadas , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/inmunología , Ratones , Ratones Noqueados , Neoplasias Experimentales/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Linfocitos T Reguladores/patología , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/inmunología
18.
Clin Immunol ; 161(2): 260-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26319414

RESUMEN

IL-7 is known to be vital for T cell homeostasis but has previously been presumed to be dispensable for TCR-induced activation. Here, we show that IL-7 is critical for the initial activation of CD4(+) T cells in that it provides some of the necessary early signaling components, such as activated STAT5 and Akt. Accordingly, short-term in vivo IL-7Rα blockade inhibited the activation and expansion of autoantigen-specific CD4(+) T cells and, when used to treat experimental autoimmune encephalomyelitis (EAE), prevented and ameliorated disease. Our studies demonstrate that IL-7 signaling is a prerequisite for optimal CD4(+) T cell activation and that IL-7R antagonism may be effective in treating CD4(+) T cell-mediated neuroinflammation and other autoimmune inflammatory conditions.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Interleucina-7/inmunología , Activación de Linfocitos/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Proliferación Celular , Citocinas/inmunología , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/metabolismo , Citometría de Flujo , Humanos , Interleucina-7/deficiencia , Interleucina-7/genética , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Ratones Transgénicos , Fosforilación/inmunología , Proteínas Proto-Oncogénicas c-akt/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Interleucina-7/inmunología , Receptores de Interleucina-7/metabolismo , Factor de Transcripción STAT2/inmunología , Factor de Transcripción STAT2/metabolismo , Transducción de Señal/inmunología
19.
J Immunol ; 188(5): 2276-84, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22279106

RESUMEN

Stage-specific rearrangement of Ig H and L chain genes poses an enigma because both processes use the same recombinatorial machinery, but the H chain locus is accessible at the pro-B cell stage, whereas the L chain loci become accessible at the pre-B cell stage. Transcription factor STAT5 is a positive-acting factor for rearrangement of distal V(H) genes, but attenuation of IL-7 signaling and loss of activated STAT5 at the pre-B cell stage corresponds with Igκ locus accessibility and rearrangement, suggesting that STAT5 plays an inhibitory role at this locus. Indeed, loss of IL-7 signaling correlates with increased activity at the Igκ intron enhancer. However, the κE3' enhancer must also be regulated as this enhancer plays a role in Igκ rearrangement. We show in this study that STAT5 can repress κE3' enhancer activity. We find that STAT5 binds to a site that overlaps the κE3' PU.1 binding site. We observed reciprocal binding by STAT5 and PU.1 to the κE3' enhancer in primary bone marrow cells, STAT5 and PU.1 retrovirally transduced pro-B cell lines, or embryonic stem cells induced to differentiate into B lineage cells. Binding by STAT5 corresponded with low occupancy of other enhancer binding proteins, whereas PU.1 binding corresponded with recruitment of IRF4 and E2A to the κE3' enhancer. We also find that IRF4 expression can override the repressive activity of STAT5. We propose a novel PU.1/STAT5 displacement model during B cell development, and this, coupled with increased IRF4 and E2A activity, regulates κE3' enhancer function.


Asunto(s)
Diferenciación Celular/inmunología , Proteínas de Unión al ADN/fisiología , Elementos de Facilitación Genéticos/inmunología , Cadenas kappa de Inmunoglobulina/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Animales , Subgrupos de Linfocitos B/citología , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Unión Competitiva/genética , Unión Competitiva/inmunología , Diferenciación Celular/genética , Línea Celular Tumoral , Células Cultivadas , Técnicas de Cocultivo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Elementos de Facilitación Genéticos/genética , Cadenas kappa de Inmunoglobulina/metabolismo , Cadenas kappa de Inmunoglobulina/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Moleculares , Células 3T3 NIH , Unión Proteica/genética , Unión Proteica/inmunología , Proteínas Proto-Oncogénicas/fisiología , Factor de Transcripción STAT5/fisiología , Transactivadores/fisiología
20.
J Immunol ; 189(1): 28-32, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22623329

RESUMEN

To ensure immune tolerance, regulatory T cell (Treg) numbers must be maintained by cell division. This process has been thought to be strictly dependent on the Treg TCR interacting with MHC class II. In this study, we report that Treg division does not absolutely require cell-autonomous TCR signaling in vivo, depending on the degree of IL-2-mediated stimulation provided. At steady state IL-2 levels, Tregs require cell-autonomous TCR signaling to divide. However, when given exogenous IL-2 or when STAT5 is selectively activated in Tregs, Treg division can occur independently of MHC class II and TCR signaling. Thus, depending on the amount of IL-2R stimulation, a wide range of TCR signals supports Treg division, which may contribute to preservation of a diverse repertoire of Treg TCR specificities. These findings also have therapeutic implications, as TCR signaling by Tregs may not be required when using IL-2 to increase Treg numbers for treatment of inflammatory disorders.


Asunto(s)
Proliferación Celular , Interleucina-2/fisiología , Receptores de Antígenos de Linfocitos T/fisiología , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Traslado Adoptivo , Animales , Presentación de Antígeno/genética , Presentación de Antígeno/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/trasplante , División Celular/genética , División Celular/inmunología , Homeostasis/genética , Homeostasis/inmunología , Ratones , Ratones Noqueados , Ratones Transgénicos , Receptores de Interleucina-2/fisiología , Transducción de Señal/genética , Linfocitos T Reguladores/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA