Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 212(7): 1129-1141, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38363226

RESUMO

In contrast to the "helper" activities of most CD4+ T effector subsets, CD4+ cytotoxic T lymphocytes (CD4-CTLs) perform functions normally associated with CD8+ T and NK cells. Specifically, CD4-CTLs secrete cytotoxic molecules and directly target and kill compromised cells in an MHC class II-restricted fashion. The functions of these cells have been described in diverse immunological contexts, including their ability to provide protection during antiviral and antitumor responses, as well as being implicated in autoimmunity. Despite their significance to human health, the complete mechanisms that govern their programming remain unclear. In this article, we identify the Ikaros zinc finger transcription factor Eos (Ikzf4) as a positive regulator of CD4-CTL differentiation during murine immune responses against influenza virus infection. We find that the frequency of Eos+ cells is elevated in lung CD4-CTL populations and that the cytotoxic gene program is compromised in Eos-deficient CD4+ T cells. Consequently, we observe a reduced frequency and number of lung-residing, influenza virus-responsive CD4-CTLs in the absence of Eos. Mechanistically, we determine that this is due, at least in part, to reduced expression of IL-2 and IL-15 cytokine receptor subunits on the surface of Eos-deficient CD4+ T cells, both of which support the CD4-CTL program. Finally, we find that Aiolos, a related Ikaros family member and known CD4-CTL antagonist, represses Eos expression by antagonizing STAT5-dependent activation of the Ikzf4 promoter. Collectively, our findings reveal a mechanism wherein Eos and Aiolos act in opposition to regulate cytotoxic programming of CD4+ T cells.


Assuntos
Antineoplásicos , Linfócitos T CD4-Positivos , Camundongos , Humanos , Animais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Linfócitos T Citotóxicos , Diferenciação Celular , Citocinas/metabolismo , Antineoplásicos/metabolismo
2.
Nat Commun ; 14(1): 5923, 2023 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-37740028

RESUMO

Treatment of osteoporosis commonly diminishes osteoclast number which suppresses bone formation thus compromising fracture prevention. Bone formation is not suppressed, however, when bone degradation is reduced by retarding osteoclast functional resorptive capacity, rather than differentiation. We find deletion of deubiquitinase, BRCA1-associated protein 1 (Bap1), in myeloid cells (Bap1∆LysM), arrests osteoclast function but not formation. Bap1∆LysM osteoclasts fail to organize their cytoskeleton which is essential for bone degradation consequently increasing bone mass in both male and female mice. The deubiquitinase activity of BAP1 modifies osteoclast function by metabolic reprogramming. Bap1 deficient osteoclast upregulate the cystine transporter, Slc7a11, by enhanced H2Aub occupancy of its promoter. SLC7A11 controls cellular reactive oxygen species levels and redirects the mitochondrial metabolites away from the tricarboxylic acid cycle, both being necessary for osteoclast function. Thus, in osteoclasts BAP1 appears to regulate the epigenetic-metabolic axis and is a potential target to reduce bone degradation while maintaining osteogenesis in osteoporotic patients.


Assuntos
Osteoclastos , Osteogênese , Animais , Feminino , Humanos , Masculino , Camundongos , Densidade Óssea , Ciclo do Ácido Cítrico , Enzimas Desubiquitinantes , Osteogênese/genética , Proteínas Supressoras de Tumor/genética , Ubiquitina Tiolesterase/genética
3.
Cancers (Basel) ; 15(3)2023 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-36765663

RESUMO

Loss of cytotoxicity and defective metabolism are linked to glycogen synthase kinase 3 beta (GSK3ß) overexpression in natural killer (NK) cells from patients with acute myeloid leukemia or from healthy donors after expansion ex vivo with IL-15. Drug inhibition of GSK3ß in these NK cells improves their maturation and cytotoxic activity, but the mechanisms of GSK3ß-mediated dysfunction have not been well studied. Here, we show that expansion of NK cells with feeder cells expressing membrane-bound IL-21 maintained normal GSK3ß levels, allowing us to study GSK3ß function using CRISPR gene editing. We deleted GSK3B and expanded paired-donor knockout and wild-type (WT) NK cells and then assessed transcriptional and functional alterations induced by loss of GSK3ß. Surprisingly, our data showed that deletion of GSK3B did not alter cytotoxicity, cytokine production, or maturation (as determined by CD57 expression). However, GSK3B-KO cells demonstrated significant changes in expression of genes related to rRNA processing, cell proliferation, and metabolic function, suggesting possible metabolic reprogramming. Next, we found that key genes downregulated in GSK3B-KO NK cells were upregulated in GSK3ß-overexpressing NK cells from AML patients, confirming this correlation in a clinical setting. Lastly, we measured cellular energetics and observed that GSK3B-KO NK cells exhibited 150% higher spare respiratory capacity, a marker of metabolic fitness. These findings suggest a role for GSK3ß in regulating NK cell metabolism.

4.
Annu Rev Immunol ; 40: 387-411, 2022 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-35119910

RESUMO

Cell identity and function largely rely on the programming of transcriptomes during development and differentiation. Signature gene expression programs are orchestrated by regulatory circuits consisting of cis-acting promoters and enhancers, which respond to a plethora of cues via the action of transcription factors. In turn, transcription factors direct epigenetic modifications to revise chromatin landscapes, and drive contacts between distal promoter-enhancer combinations. In immune cells, regulatory circuits for effector genes are especially complex and flexible, utilizing distinct sets of transcription factors and enhancers, depending on the cues each cell type receives during an infection, after sensing cellular damage, or upon encountering a tumor. Here, we review major players in the coordination of gene regulatory programs within innate and adaptive immune cells, as well as integrative omics approaches that can be leveraged to decipher their underlying circuitry.


Assuntos
Cromatina , Redes Reguladoras de Genes , Animais , Regulação da Expressão Gênica , Humanos , Regiões Promotoras Genéticas , Fatores de Transcrição/genética
5.
Cell Rep ; 34(7): 108749, 2021 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-33596420

RESUMO

Aberrant expression of nuclear transporters and deregulated subcellular localization of their cargo proteins are emerging as drivers and therapeutic targets of cancer. Here, we present evidence that the nuclear exporter exportin-6 and its cargo profilin-1 constitute a functionally important and frequently deregulated axis in cancer. Exportin-6 upregulation occurs in numerous cancer types and is associated with poor patient survival. Reducing exportin-6 level in breast cancer cells triggers antitumor effects by accumulating nuclear profilin-1. Mechanistically, nuclear profilin-1 interacts with eleven-nineteen-leukemia protein (ENL) within the super elongation complex (SEC) and inhibits the ability of the SEC to drive transcription of numerous pro-cancer genes including MYC. XPO6 and MYC are positively correlated across diverse cancer types including breast cancer. Therapeutically, exportin-6 loss sensitizes breast cancer cells to the bromodomain and extra-terminal (BET) inhibitor JQ1. Thus, exportin-6 upregulation is a previously unrecognized cancer driver event by spatially inhibiting nuclear profilin-1 as a tumor suppressor.


Assuntos
Carioferinas/metabolismo , Neoplasias/metabolismo , Profilinas/antagonistas & inibidores , Profilinas/metabolismo , Animais , Linhagem Celular Tumoral , Feminino , Xenoenxertos , Humanos , Carioferinas/genética , Células MCF-7 , Camundongos , Camundongos Nus , Neoplasias/genética , Profilinas/genética , Análise de Sobrevida , Regulação para Cima
6.
Sci Transl Med ; 12(532)2020 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-32102931

RESUMO

Atopic dermatitis (AD) is a widespread, chronic skin disease associated with aberrant allergic inflammation. Current treatments involve either broad or targeted immunosuppression strategies. However, enhancing the immune system to control disease remains untested. We demonstrate that patients with AD harbor a blood natural killer (NK) cell deficiency that both has diagnostic value and improves with therapy. Multidimensional protein and RNA profiling revealed subset-level changes associated with enhanced NK cell death. Murine NK cell deficiency was associated with enhanced type 2 inflammation in the skin, suggesting that NK cells play a critical immunoregulatory role in this context. On the basis of these findings, we used an NK cell-boosting interleukin-15 (IL-15) superagonist and observed marked improvement in AD-like disease in mice. These findings reveal a previously unrecognized application of IL-15 superagonism, currently in development for cancer immunotherapy, as an immunotherapeutic strategy for AD.


Assuntos
Dermatite Atópica , Deficiência de GATA2 , Animais , Dermatite Atópica/terapia , Modelos Animais de Doenças , Humanos , Imunoterapia , Células Matadoras Naturais , Camundongos
7.
Cell ; 176(1-2): 348-360.e12, 2019 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-30595449

RESUMO

Natural killer (NK) cells develop from common progenitors but diverge into distinct subsets, which differ in cytokine production, cytotoxicity, homing, and memory traits. Given their promise in adoptive cell therapies for cancer, a deeper understanding of regulatory modules controlling clinically beneficial NK phenotypes is of high priority. We report integrated "-omics" analysis of human NK subsets, which revealed super-enhancers associated with gene cohorts that may coordinate NK functions and localization. A transcription factor-based regulatory scheme also emerged, which is evolutionarily conserved and shared by innate and adaptive lymphocytes. For both NK and T lineages, a TCF1-LEF1-MYC axis dominated the regulatory landscape of long-lived, proliferative subsets that traffic to lymph nodes. In contrast, effector populations circulating between blood and peripheral tissues shared a PRDM1-dominant landscape. This resource defines transcriptional modules, regulated by feedback loops, which may be leveraged to enhance phenotypes for NK cell-based therapies.


Assuntos
Células Matadoras Naturais/classificação , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/fisiologia , Citocinas/imunologia , Citocinas/metabolismo , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Humanos , Fenótipo
8.
Blood Adv ; 2(19): 2467-2477, 2018 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-30266822

RESUMO

Additional sex comb-like 1 (ASXL1) mutations are commonly associated with myeloid malignancies and are markers of aggressive disease. The fact that ASXL1 is necessary for myeloid differentiation raises the possibility it also regulates osteoclasts. We find deletion of ASXL1 in myeloid cells results in bone loss with increased abundance of osteoclasts. Because ASXL1 is an enhancer of trithorax and polycomb (ETP) protein, we asked if it modulates osteoclast differentiation by maintaining balance between positive and negative epigenetic regulators. In fact, loss of ASXL1 induces concordant loss of inhibitory H3K27me3 with gain of H3K4me3 at key osteoclast differentiation genes, including nuclear factor for activated T cells 1 (NFATc1) and itgb3 In the setting of ASXL1 deficiency, increased NFATc1 binds to the Blimp1 (Prdm1) promoter thereby enhancing expression of this pro-osteoclastogenic gene. The global reduction of K27 trimethylation in ASXL1-deficient osteoclasts is also attended by a 40-fold increase in expression of the histone demethylase Jumonji domain-containing 3 (Jmjd3). Jmjd3 knockdown in ASXL1-deficient osteoclast precursors increases H3K27me3 on the NFATc1 promoter and impairs osteoclast formation. Thus, in addition to promoting myeloid malignancies, ASXL1 controls epigenetic reprogramming of osteoclasts to regulate bone resorption and mass.


Assuntos
Diferenciação Celular/genética , Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição NFATC/genética , Osteoclastos/metabolismo , Proteínas Repressoras/genética , Animais , Sítios de Ligação , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Linhagem Celular , Deleção de Genes , Histonas/metabolismo , Humanos , Metilação , Camundongos , Camundongos Knockout , Modelos Biológicos , Células Mieloides , Osteoclastos/citologia , Osteogênese/genética , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Repressoras/metabolismo
9.
Proc Natl Acad Sci U S A ; 112(27): 8367-72, 2015 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-26100872

RESUMO

Genome stability relies on epigenetic mechanisms that enforce repression of endogenous retroviruses (ERVs). Current evidence suggests that distinct chromatin-based mechanisms repress ERVs in cells of embryonic origin (histone methylation dominant) vs. more differentiated cells (DNA methylation dominant). However, the latter aspect of this model has not been tested. Remarkably, and in contrast to the prevailing model, we find that repressive histone methylation catalyzed by the enzyme SETDB1 is critical for suppression of specific ERV families and exogenous retroviruses in committed B-lineage cells from adult mice. The profile of ERV activation in SETDB1-deficient B cells is distinct from that observed in corresponding embryonic tissues, despite the loss of repressive chromatin modifications at all ERVs. We provide evidence that, on loss of SETDB1, ERVs are activated in a lineage-specific manner depending on the set of transcription factors available to target proviral regulatory elements. These findings have important implications for genome stability in somatic cells, as well as the interface between epigenetic repression and viral latency.


Assuntos
Linfócitos B/metabolismo , Retrovirus Endógenos/genética , Histona-Lisina N-Metiltransferase/genética , Retroviridae/genética , Animais , Linfócitos B/virologia , Western Blotting , Células da Medula Óssea/metabolismo , Células da Medula Óssea/virologia , Células Cultivadas , Metilação de DNA , Retrovirus Endógenos/metabolismo , Epigênese Genética , Citometria de Fluxo , Perfilação da Expressão Gênica , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Interações Hospedeiro-Patógeno , Lisina/metabolismo , Metilação , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Knockout , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Células Precursoras de Linfócitos B/metabolismo , Células Precursoras de Linfócitos B/virologia , Retroviridae/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sequências Repetidas Terminais/genética , Ativação Viral/genética
10.
Arthritis Rheum ; 63(9): 2606-16, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21618198

RESUMO

OBJECTIVE: Low-dose methotrexate (MTX) is an effective therapy for rheumatoid arthritis (RA), yet its mechanism of action is incompletely understood. The aim of this study was to explore the induction of apoptosis by MTX. METHODS: Flow cytometry was performed to assess changes in the levels of intracellular proteins, reactive oxygen species (ROS), and apoptosis. Quantitative polymerase chain reaction was performed to assess changes in the transcript levels of select target genes in response to MTX. RESULTS: MTX did not directly induce apoptosis but rather "primed" cells for markedly increased sensitivity to apoptosis via either mitochondrial or death receptor pathways, by a JNK-dependent mechanism. Increased sensitivity to apoptosis was mediated, at least in part, by MTX-dependent production of ROS, JNK activation, and JNK-dependent induction of genes whose protein products promote apoptosis. Supplementation with tetrahydrobiopterin blocked these MTX-induced effects. Patients with RA who were receiving low-dose MTX therapy expressed elevated levels of the JNK target gene, jun. CONCLUSION: Our results support a model whereby MTX inhibits reduction of dihydrobiopterin to tetrahydrobiopterin, resulting in increased production of ROS, increased JNK activity, and increased sensitivity to apoptosis. The finding of increased jun levels in patients with RA receiving low-dose MTX supports the notion that this pathway is activated by MTX in vivo and may contribute to the efficacy of MTX in inflammatory disease.


Assuntos
Antirreumáticos/uso terapêutico , Apoptose/efeitos dos fármacos , Artrite Reumatoide/tratamento farmacológico , Expressão Gênica/efeitos dos fármacos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Metotrexato/uso terapêutico , Adulto , Antirreumáticos/farmacologia , Apoptose/fisiologia , Artrite Reumatoide/metabolismo , Biopterinas/análogos & derivados , Biopterinas/farmacologia , Linhagem Celular Tumoral , Humanos , Metotrexato/farmacologia , Pessoa de Meia-Idade , Espécies Reativas de Oxigênio/metabolismo , Células Tumorais Cultivadas
11.
Immunity ; 33(2): 254-65, 2010 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-20691614

RESUMO

Follicular (FO) and marginal zone (MZ) B cells are maintained in distinct locations within the spleen, but the genetic basis for this separation is still enigmatic. We now report that B cell sequestration requires lineage-specific regulation of migratory receptors by the transcription factor Klf2. Moreover, using gene-targeted mice we show that altered splenic B cell migration confers a significant in vivo gain-of-function phenotype to FO B cells, including the ability to quickly respond to MZ-associated antigens and pathogens in a T cell-dependent manner. This work demonstrates that in wild-type animals, naive FO B cells are actively removed from the MZ, thus restricting their capacity to respond to blood-borne pathogens.


Assuntos
Linfócitos B/citologia , Linfócitos B/imunologia , Movimento Celular , Imunidade Humoral , Baço/citologia , Baço/imunologia , Animais , Antígenos CD19/genética , Antígenos CD19/imunologia , Antígenos T-Independentes/genética , Antígenos T-Independentes/imunologia , Medula Óssea/imunologia , Diferenciação Celular , Células Cultivadas , Fatores de Transcrição Kruppel-Like/deficiência , Fatores de Transcrição Kruppel-Like/imunologia , Camundongos , Camundongos Knockout , Receptores CCR/imunologia
12.
J Immunol ; 185(3): 1492-501, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20574006

RESUMO

Genes, such as IFNG, which are expressed in multiple cell lineages of the immune system, may employ a common set of regulatory elements to direct transcription in multiple cell types or individual regulatory elements to direct expression in individual cell lineages. By employing a bacterial artificial chromosome transgenic system, we demonstrate that IFNG employs unique regulatory elements to achieve lineage-specific transcriptional control. Specifically, a one 1-kb element 30 kb upstream of IFNG activates transcription in T cells and NKT cells but not in NK cells. This distal regulatory element is a Runx3 binding site in Th1 cells and is needed for RNA polymerase II recruitment to IFNG, but it is not absolutely required for histone acetylation of the IFNG locus. These results support a model whereby IFNG uses cis-regulatory elements with cell type-restricted function.


Assuntos
Linhagem da Célula/genética , Linhagem da Célula/imunologia , Regulação da Expressão Gênica/imunologia , Loci Gênicos/imunologia , Interferon gama/biossíntese , Interferon gama/genética , Animais , Células Cultivadas , Sequência Conservada/genética , Sequência Conservada/imunologia , Subunidade alfa 3 de Fator de Ligação ao Core/metabolismo , Humanos , Células Matadoras Naturais/enzimologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células T Matadoras Naturais/enzimologia , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/metabolismo , Transporte Proteico/genética , Transporte Proteico/imunologia , RNA Polimerase II/metabolismo , Elementos Reguladores de Transcrição/imunologia , Células Th1/enzimologia , Células Th1/imunologia , Células Th1/metabolismo , Sítio de Iniciação de Transcrição
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA