Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
2.
J Biomed Sci ; 28(1): 41, 2021 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-34082769

RESUMO

Lysine-specific demethylase 1 (LSD1) targets mono- or di-methylated histone H3K4 and H3K9 as well as non-histone substrates and functions in the regulation of gene expression as a transcriptional repressor or activator. This enzyme plays a pivotal role in various physiological processes, including development, differentiation, inflammation, thermogenesis, neuronal and cerebral physiology, and the maintenance of stemness in stem cells. LSD1 also participates in pathological processes, including cancer as the most representative disease. It promotes oncogenesis by facilitating the survival of cancer cells and by generating a pro-cancer microenvironment. In this review, we discuss the role of LSD1 in several aspects of cancer, such as hypoxia, epithelial-to-mesenchymal transition, stemness versus differentiation of cancer stem cells, as well as anti-tumor immunity. Additionally, the current understanding of the involvement of LSD1 in various other pathological processes is discussed.


Assuntos
Histona Desmetilases/genética , Homeostase/genética , Neoplasias/genética , Animais , Diferenciação Celular/genética , Transição Epitelial-Mesenquimal/genética , Histona Desmetilases/imunologia , Histona Desmetilases/metabolismo , Homeostase/imunologia , Humanos , Hipóxia/enzimologia , Hipóxia/genética , Hipóxia/imunologia , Camundongos , Neoplasias/enzimologia , Neoplasias/imunologia , Células-Tronco Neoplásicas/fisiologia , Evasão Tumoral/genética
3.
Cell Rep ; 35(2): 108966, 2021 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-33852868

RESUMO

Persistent virus infections can cause pathogenesis that is debilitating or lethal. During these infections, virus-specific T cells fail to protect due to weakened antiviral activity or failure to persist. These outcomes are governed by histone modifications, although it is unknown which enzymes contribute to T cell loss or impaired function over time. In this study, we show that T cell receptor-stimulated CD8+ T cells increase their expression of UTX (ubiquitously transcribed tetratricopeptide repeat, X chromosome) to enhance gene expression. During chronic lymphocytic choriomeningitis virus (LCMV) infection in mice, UTX binds to enhancers and transcription start sites of effector genes, allowing for improved cytotoxic T lymphocyte (CTL)-mediated protection, independent of its trimethylation of histone 3 lysine 27 (H3K27me3) demethylase activity. UTX also limits the frequency and durability of virus-specific CD8+ T cells, which correspond to increased expression of inhibitory receptors. Thus, UTX guides gene expression patterns in CD8+ T cells, advancing early antiviral defenses while reducing the longevity of CD8+ T cell responses.


Assuntos
Citotoxicidade Imunológica/genética , Histona Desmetilases/genética , Memória Imunológica/genética , Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Receptor Celular 2 do Vírus da Hepatite A/genética , Receptor Celular 2 do Vírus da Hepatite A/imunologia , Histona Desmetilases/deficiência , Histona Desmetilases/imunologia , Histonas/genética , Histonas/imunologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Coriomeningite Linfocítica/patologia , Vírus da Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/crescimento & desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/imunologia , Transdução de Sinais , Linfócitos T Citotóxicos/virologia , Carga Viral/genética , Carga Viral/imunologia , Proteína do Gene 3 de Ativação de Linfócitos
4.
PLoS Pathog ; 17(3): e1009476, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33788902

RESUMO

Infectious and inflammatory diseases in the intestine remain a serious threat for patients world-wide. Reprogramming of the intestinal epithelium towards a protective effector state is important to manage inflammation and immunity and can be therapeutically targeted. The role of epigenetic regulatory enzymes within these processes is not yet defined. Here, we use a mouse model that has an intestinal-epithelial specific deletion of the histone demethylase Lsd1 (cKO mice), which maintains the epithelium in a fixed reparative state. Challenge of cKO mice with bacteria-induced colitis or a helminth infection model both resulted in increased pathogenesis. Mechanistically, we discovered that LSD1 is important for goblet cell maturation and goblet-cell effector molecules such as RELMß. We propose that this may be in part mediated by directly controlling genes that facilitate cytoskeletal organization, which is important in goblet cell biology. This study therefore identifies intestinal-epithelial epigenetic regulation by LSD1 as a critical element in host protection from infection.


Assuntos
Infecções por Enterobacteriaceae/imunologia , Células Caliciformes/imunologia , Histona Desmetilases/imunologia , Mucosa Intestinal/metabolismo , Tricuríase/imunologia , Animais , Citrobacter rodentium , Células Caliciformes/metabolismo , Histona Desmetilases/metabolismo , Mucosa Intestinal/imunologia , Camundongos , Camundongos Knockout , Trichuris
5.
Int J Mol Sci ; 22(3)2021 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-33498655

RESUMO

Women represent 80% of people affected by autoimmune diseases. Although, many studies have demonstrated a role for sex hormone receptor signaling, particularly estrogens, in the direct regulation of innate and adaptive components of the immune system, recent data suggest that female sex hormones are not the only cause of the female predisposition to autoimmunity. Besides sex steroid hormones, growing evidence points towards the role of X-linked genetic factors. In female mammals, one of the two X chromosomes is randomly inactivated during embryonic development, resulting in a cellular mosaicism, where about one-half of the cells in a given tissue express either the maternal X chromosome or the paternal one. X chromosome inactivation (XCI) is however not complete and 15 to 23% of genes from the inactive X chromosome (Xi) escape XCI, thereby contributing to the emergence of a female-specific heterogeneous population of cells with bi-allelic expression of some X-linked genes. Although the direct contribution of this genetic mechanism in the female susceptibility to autoimmunity still remains to be established, the cellular mosaicism resulting from XCI escape is likely to create a unique functional plasticity within female immune cells. Here, we review recent findings identifying key immune related genes that escape XCI and the relationship between gene dosage imbalance and functional responsiveness in female cells.


Assuntos
Doenças Autoimunes/genética , Imunidade/genética , Inativação do Cromossomo X , Animais , Cromossomos Humanos X , Feminino , Histona Desmetilases/genética , Histona Desmetilases/imunologia , Histona Desmetilases/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Masculino , Polimorfismo de Nucleotídeo Único , RNA Longo não Codificante/genética , RNA Longo não Codificante/imunologia , Receptor 7 Toll-Like/genética , Receptor 7 Toll-Like/imunologia , Receptor 8 Toll-Like/genética , Receptor 8 Toll-Like/imunologia
6.
Front Immunol ; 12: 811364, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35046962

RESUMO

Chimeric antigen receptor (CAR) T cells targeting CD19 antigen have produced remarkable clinical outcomes for cancer patients. However, identifying measures to enhance effector function remains one of the most challenging issues in CD19-targeted immunotherapy. Here, we report a novel approach in which a microRNA (miRNA) or short-hairpin RNA (shRNA) cassette was integrated into CAR-expressing retroviral vectors. Using this system, we generated anti-CD19 CAR-T cells co-expressing miR155 or LSD1 shRNA and found that anti-CD19 CAR-T cells with miR155 upregulation or LSD1 downregulation exhibited increased anti-tumor functions in vitro and in vivo. Transcriptional profiling analysis by RNA sequencing revealed the targets of miR155 and LSD1 in anti-CD19 CAR-T cells. Our experiments indicated that introduction of miRNA or shRNA expression into anti-CD19 CAR T-cells might be an effective strategy to improve the anti-tumor effects of CAR-T cell therapy.


Assuntos
Histona Desmetilases/imunologia , Imunoterapia Adotiva/métodos , MicroRNAs/imunologia , Receptores de Antígenos Quiméricos/imunologia , Animais , Antígenos CD19/imunologia , Humanos , Camundongos , Neoplasias Experimentais/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Front Immunol ; 11: 2196, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33042135

RESUMO

Cell therapies such as chimeric-antigen receptor (CAR) T-cells and NK cells are cutting-edge methods for treating cancer and other diseases. There is high interest in optimizing drug treatment regimens to best work together with emerging cell therapies, such as targeting epigenetic enzymes to stimulate recognition of tumor cells by immune cells. Herein, we uncover new mechanisms of the histone demethylase LSD1, and various inhibitors targeting unique domains of LSD1, in the function of NK cells grown for cell therapy. Catalytic inhibitors (tranylcypromine and the structural derivatives GSK LSD1 and RN-1) can irreversibly block the demethylase activity of LSD1, while scaffolding inhibitors (SP-2509 and clinical successor SP-2577, also known as seclidemstat) disrupt epigenetic complexes that include LSD1. Relevant combinations of LSD1 inhibitors with cell therapy infusions and immune checkpoint blockade have shown efficacy in pre-clinical solid tumor models, reinforcing a need to understand how these drugs would impact T- and NK cells. We find that scaffolding LSD1 inhibitors potently reduce oxidative phosphorylation and glycolysis of NK cells, and higher doses induce mitochondrial reactive oxygen species and depletion of the antioxidant glutathione. These effects are unique to scaffolding inhibitors compared to catalytic, to NK cells compared to T-cells, and importantly, can fully ablate the lytic capacity of NK cells. Supplementation with biologically achievable levels of glutathione rescues NK cell cytolytic function but not NK cell metabolism. Our results suggest glutathione supplementation may reverse NK cell activity suppression in patients treated with seclidemstat.


Assuntos
Glutationa , Histona Desmetilases , Imunidade Celular , Células Matadoras Naturais , Glutationa/imunologia , Glutationa/metabolismo , Histona Desmetilases/imunologia , Histona Desmetilases/metabolismo , Humanos , Células Matadoras Naturais/enzimologia , Células Matadoras Naturais/imunologia
9.
J Immunol ; 204(1): 159-168, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31748348

RESUMO

Severe disease following respiratory syncytial virus (RSV) infection has been linked to enhanced proinflammatory cytokine production that promotes a Th2-type immune environment. Epigenetic regulation in immune cells following viral infection plays a role in the inflammatory response and may result from upregulation of key epigenetic modifiers. In this study, we show that RSV-infected bone marrow-derived dendritic cells (BMDC) as well as pulmonary dendritic cells (DC) from RSV-infected mice upregulated the expression of Kdm6b/Jmjd3 and Kdm6a/Utx, H3K27 demethylases. KDM6-specific chemical inhibition (GSK J4) in BMDC led to decreased production of chemokines and cytokines associated with the inflammatory response during RSV infection (i.e., CCL-2, CCL-3, CCL-5, IL-6) as well as decreased MHC class II and costimulatory marker (CD80/86) expression. RSV-infected BMDC treated with GSK J4 altered coactivation of T cell cytokine production to RSV as well as a primary OVA response. Airway sensitization of naive mice with RSV-infected BMDCs exacerbate a live challenge with RSV infection but was inhibited when BMDCs were treated with GSK J4 prior to sensitization. Finally, in vivo treatment with the KDM6 inhibitor, GSK J4, during RSV infection reduced inflammatory DC in the lungs along with IL-13 levels and overall inflammation. These results suggest that KDM6 expression in DC enhances proinflammatory innate cytokine production to promote an altered Th2 immune response following RSV infection that leads to more severe immunopathology.


Assuntos
Histona Desmetilases/imunologia , Inflamação/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Regulação para Cima , Animais , Linhagem Celular Tumoral , Células Dendríticas/imunologia , Células Dendríticas/patologia , Feminino , Humanos , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Infecções por Vírus Respiratório Sincicial/patologia
10.
J Immunol ; 203(7): 1867-1881, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31492745

RESUMO

Marginal zone B cells (MZB) are a mature B cell subset that rapidly respond to blood-borne pathogens. Although the transcriptional changes that occur throughout MZB development are known, the corresponding epigenetic changes and epigenetic modifying proteins that facilitate these changes are poorly understood. The histone demethylase LSD1 is an epigenetic modifier that promotes plasmablast formation, but its role in B cell development has not been explored. In this study, a role for LSD1 in the development of B cell subsets was examined. B cell-conditional deletion of LSD1 in mice resulted in a decrease in MZB whereas follicular B cells and bone marrow B cell populations were minimally affected. LSD1 repressed genes in MZB that were normally upregulated in the myeloid and follicular B cell lineages. Correspondingly, LSD1 regulated chromatin accessibility at the motifs of transcription factors known to regulate splenic B cell development, including NF-κB motifs. The importance of NF-κB signaling was examined through an ex vivo MZB development assay, which showed that both LSD1-deficient and NF-κB-inhibited transitional B cells failed to undergo full MZB development. Gene expression and chromatin accessibility analyses of in vivo- and ex vivo-generated LSD1-deficient MZB indicated that LSD1 regulated the downstream target genes of noncanonical NF-κB signaling. Additionally LSD1 was found to interact with the noncanonical NF-κB transcription factor p52. Together, these data reveal that the epigenetic modulation of the noncanonical NF-κB signaling pathway by LSD1 is an essential process during the development of MZB.


Assuntos
Subpopulações de Linfócitos B/metabolismo , Epigênese Genética/imunologia , Histona Desmetilases/imunologia , Subunidade p52 de NF-kappa B/imunologia , Transdução de Sinais/imunologia , Animais , Subpopulações de Linfócitos B/citologia , Histona Desmetilases/genética , Camundongos , Camundongos Knockout , Subunidade p52 de NF-kappa B/genética , Transdução de Sinais/genética
11.
J Immunol ; 201(9): 2799-2811, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30232138

RESUMO

B cells undergo epigenetic remodeling as they differentiate into Ab-secreting cells (ASC). LSD1 is a histone demethylase known to decommission active enhancers and cooperate with the ASC master regulatory transcription factor Blimp-1. The contribution of LSD1 to ASC formation is poorly understood. In this study, we show that LSD1 is necessary for proliferation and differentiation of mouse naive B cells (nB) into plasmablasts (PB). Following LPS inoculation, LSD1-deficient hosts exhibited a 2-fold reduction of splenic PB and serum IgM. LSD1-deficient PB exhibited derepression and superinduction of genes involved in immune system processes; a subset of these being direct Blimp-1 target-repressed genes. Cell cycle genes were globally downregulated without LSD1, which corresponded to a decrease in the proliferative capacity of LSD1-deficient activated B cells. PB lacking LSD1 displayed increased histone H3 lysine 4 monomethylation and chromatin accessibility at nB active enhancers and the binding sites of transcription factors Blimp-1, PU.1, and IRF4 that mapped to LSD1-repressed genes. Together, these data show that LSD1 is required for normal in vivo PB formation, distinguish LSD1 as a transcriptional rheostat and epigenetic modifier of B cell differentiation, and identify LSD1 as a factor responsible for decommissioning nB active enhancers.


Assuntos
Linfócitos B/citologia , Diferenciação Celular/imunologia , Histona Desmetilases/imunologia , Plasmócitos/citologia , Animais , Linfócitos B/imunologia , Proliferação de Células/fisiologia , Camundongos , Plasmócitos/imunologia
12.
Nat Commun ; 9(1): 2720, 2018 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-30006524

RESUMO

To explain the excess cancer rate in males, several candidates for "escape from X-inactivation tumor-suppressor" (EXITS) were recently identified. In this report we provide direct experimental evidence supporting UTX's role as an EXITS gene. Using a mouse lymphoma model, we show clear dosage effect of UTX copy number during tumorigenesis, which strongly supports the EXITS theory. Importantly, UTX deletion not only accelerates lymphomagenesis, it also strongly promotes tumor progression. UTX-knockout tumors are more aggressive, showing enhanced brain dissemination and formation of blood vessels. Efnb1 is overexpressed in UTX KO tumors and can lead to such phenotypes. In human patients, lymphomas with low UTX expression also express high levels of Efnb1, and cause significantly poor survival. Lastly, we show that UTX deficiency renders lymphoma sensitive to cytarabine treatment. Taken together, these data highlight UTX loss's profound impacts on tumor initiation and drug response.


Assuntos
Neoplasias Encefálicas/genética , Carcinogênese/genética , Efrina-B1/genética , Regulação Neoplásica da Expressão Gênica , Histona Desmetilases/genética , Linfoma de Células B/genética , Animais , Antimetabólitos Antineoplásicos/farmacologia , Linfócitos B/imunologia , Linfócitos B/patologia , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/secundário , Carcinogênese/imunologia , Carcinogênese/patologia , Citarabina/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Resistencia a Medicamentos Antineoplásicos/imunologia , Efrina-B1/imunologia , Feminino , Dosagem de Genes , Histona Desmetilases/deficiência , Histona Desmetilases/imunologia , Humanos , Linfoma de Células B/imunologia , Linfoma de Células B/patologia , Masculino , Camundongos , Camundongos Knockout , Neoplasias Experimentais , Fatores Sexuais , Transdução de Sinais , Análise de Sobrevida , Inativação do Cromossomo X
13.
Hematol Oncol ; 36(1): 328-335, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28695659

RESUMO

Extranodal natural killer (NK)/T-cell lymphoma, nasal type (ENKTL) is an extranodal aggressive T or NK-cell lymphoma that is characteristically associated with Epstein-Barr virus (EBV) infection and cytotoxic tissue-destructive features. Although ENKTL is described as a distinct entity according to the 2008 WHO classification, a considerable complexity is associated with the differential diagnosis of other T-cell lymphomas with respect to tumour cell origins, locations, and the presence of EBV infection, as well as molecular and cytogenetic abnormalities. Here, we report a rare case of EBV-negative ENKTL, where the absence of EBV in the true NK-lineage cells was confirmed by extensive phenotypic and genotypic analyses. Furthermore, using the next-generation sequencing approach, we identified mutations in the tumour suppressor genes KDM6A and TP53. The clinicopathological characteristics were almost similar to those of EBV-positive ENKTL, except for the absence of EBV and histologically apparent angioinvasiveness. This is the first reported ENKTL case with mutations in the KDM6A gene. KDM6A is one of the histone-modifying genes that are mutated in many human diseases including haematological cancers. Epigenetic regulation of gene expression has recently been demonstrated in ENKTL, and a similar pathway is thought to play an oncogenic role in EBV-negative ENKTL. Our report shows the extent of comprehensive examination required before making a definitive diagnosis for NK- and T-cell neoplasms and broadens the therapeutic options for potential targets.


Assuntos
Histona Desmetilases/imunologia , Linfoma Extranodal de Células T-NK/imunologia , Proteínas Nucleares/imunologia , Feminino , Humanos , Linfoma Extranodal de Células T-NK/tratamento farmacológico , Linfoma Extranodal de Células T-NK/patologia , Pessoa de Meia-Idade , Mutação
14.
Biochem Biophys Res Commun ; 493(2): 1122-1128, 2017 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-28887033

RESUMO

Atmospheric particulates, especially PM2.5, not only damage the respiratory system, but also play important roles in pulmonary immunity. China is influenced by atmospheric diffusion conditions, industrial manufacturers, and heating and discharging. PM2.5 levels in the air rise substantially in the winter, which is also a period of flu high-incidence. Although an epidemiological link exists between PM2.5 and flu, we do not understand how long-term PM2.5 inhalation affects pulmonary immunity and the influenza virus response. Our study has prepared an in vivo PM2.5 mouse pharyngeal wall drop-in model and has found that PM2.5 exposure leads to mouse inflammatory injuries and furthers influenza A infection. Our results suggest that short-term exposure to PM2.5 significantly enhances the survival rate of influenza A-contaminated mice, while long-term PM2.5 inhalation lowers the capacity of pulmonary macrophages to secrete IL-6 and IFN-ß. A disorder in the pulmonary innate defense system results in increased death rates following influenza infection. On a macromolecular level, this mechamism involves Kdm6a down-regulation after long-term exposure to PM 2.5 and a resultant increase in H3K4 and H3K9 methylation in IL-6 and IFN-ß promoter regions. In summary, PM2.5 causes injuries of lung tissue cells and downregulates immune defense mechanisms in the lung.


Assuntos
Histona Desmetilases/genética , Vírus da Influenza A/fisiologia , Influenza Humana/etiologia , Interferon beta/genética , Interleucina-6/genética , Macrófagos Alveolares/patologia , Material Particulado/efeitos adversos , Animais , Modelos Animais de Doenças , Regulação para Baixo , Código das Histonas , Histona Desmetilases/imunologia , Humanos , Imunidade Inata , Vírus da Influenza A/imunologia , Influenza Humana/genética , Influenza Humana/imunologia , Influenza Humana/patologia , Interferon beta/imunologia , Interleucina-6/imunologia , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Pulmão/virologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/virologia , Camundongos , Infecções por Orthomyxoviridae/etiologia , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/patologia , Material Particulado/imunologia , Regiões Promotoras Genéticas
15.
J Virol ; 91(1)2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27795407

RESUMO

Neuroinvasive herpesviruses have evolved to efficiently infect and establish latency in neurons. The nervous system has limited capability to regenerate, so immune responses therein are carefully regulated to be nondestructive, with dependence on atypical intrinsic and innate defenses. In this article we review studies of some of these noncanonical defense pathways and how herpesvirus gene products counter them, highlighting the contributions that primary neuronal in vitro models have made to our understanding of this field.


Assuntos
Inativação Gênica , Herpesviridae/crescimento & desenvolvimento , Evasão da Resposta Imune , Neurônios/virologia , Latência Viral/imunologia , Autofagia/genética , Autofagia/imunologia , Transporte Axonal , Proteínas Correpressoras/genética , Proteínas Correpressoras/imunologia , Herpesviridae/imunologia , Histona Desacetilases/genética , Histona Desacetilases/imunologia , Histona Desmetilases/genética , Histona Desmetilases/imunologia , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/imunologia , Imunidade Inata , Interferons/genética , Interferons/imunologia , MicroRNAs/genética , MicroRNAs/imunologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/imunologia , Neurônios/imunologia , Transdução de Sinais , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/imunologia
16.
Sci Rep ; 6: 21166, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26876951

RESUMO

The invertebrate's innate immune system was reported to show some form of adaptive features, termed trained immunity. However, the memory characteristics of innate immune system and the mechanisms behind such phenomena remain unclear. Using the invertebrate model Artemia, we verified the possibility or impossibility of trained immunity, examining the presence or absence of enduring memory against homologous and heterologous antigens (Vibrio spp.) during a transgenerational study. We also determined the mechanisms behind such phenomenon. Our results showed the occurrence of memory and partial discrimination in Artemia's immune system, as manifested by increased resistance, for three successive generations, of the progenies of Vibrio-exposed ancestors towards a homologous bacterial strain, rather than to a heterologous strain. This increased resistance phenotype was associated with elevated levels of hsp70 and hmgb1 signaling molecules and alteration in the expression of key innate immunity-related genes. Our results also showed stochastic pattern in the acetylation and methylation levels of H4 and H3K4me3 histones, respectively, in the progenies whose ancestors were challenged. Overall results suggest that innate immune responses in invertebrates have the capacity to be trained, and epigenetic reprogramming of (selected) innate immune effectors is likely to have central place in the mechanisms leading to trained immunity.


Assuntos
Imunidade Adaptativa/imunologia , Artemia/imunologia , Histonas/imunologia , Imunidade Inata/genética , Acetilação , Imunidade Adaptativa/genética , Animais , Antígenos de Bactérias/imunologia , Artemia/genética , Proteínas de Choque Térmico HSP70 , Histona Desmetilases/genética , Histona Desmetilases/imunologia , Histonas/genética , Modelos Biológicos , Vibrio/imunologia , Vibrio/patogenicidade
17.
Int J Biochem Cell Biol ; 67: 148-57, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26193001

RESUMO

Gene expression is epigenetically regulated through DNA methylation and covalent chromatin modifications, such as acetylation, phosphorylation, ubiquitination, sumoylation, and methylation of histones. Histone methylation state is dynamically regulated by different groups of histone methyltransferases and demethylases. The trimethylation of histone 3 (H3K4) at lysine 4 is usually associated with the activation of gene expression, whereas trimethylation of histone 3 at lysine 27 (H3K27) is associated with the repression of gene expression. The polycomb repressive complex contains the H3K27 methyltransferase Ezh2 and controls dimethylation and trimethylation of H3K27 (H3K27me2/3). The Jumonji domain containing-3 (Jmjd3, KDM6B) and ubiquitously transcribed X-chromosome tetratricopeptide repeat protein (UTX, KDM6A) have been identified as H3K27 demethylases that catalyze the demethylation of H3K27me2/3. The role and mechanisms of both JMJD3 and UTX have been extensively studied for their involvement in development, cell plasticity, immune system, neurodegenerative disease, and cancer. In this review, we will focus on recent progresses made on understanding JMJD3 in the regulation of gene expression in development and diseases. This article is part of a Directed Issue entitled: Epigenetics dynamics in development and disease.


Assuntos
Epigênese Genética , Histona Desmetilases/genética , Histona Desmetilases com o Domínio Jumonji/genética , Neoplasias/genética , Doenças Neurodegenerativas/genética , Proteínas Nucleares/genética , Complexo Repressor Polycomb 2/genética , Animais , Cromatina/química , Cromatina/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste , Regulação da Expressão Gênica no Desenvolvimento , Camadas Germinativas/citologia , Camadas Germinativas/crescimento & desenvolvimento , Camadas Germinativas/metabolismo , Histona Desmetilases/imunologia , Histonas/genética , Histonas/metabolismo , Humanos , Imunidade Inata , Histona Desmetilases com o Domínio Jumonji/imunologia , Lisina/metabolismo , Camundongos , Neoplasias/imunologia , Neoplasias/patologia , Doenças Neurodegenerativas/imunologia , Doenças Neurodegenerativas/patologia , Proteínas Nucleares/imunologia , Complexo Repressor Polycomb 2/imunologia
18.
J Biomol Screen ; 17(1): 18-26, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21965113

RESUMO

In the past years, a lot of attention has been given to the identification and characterization of selective and potent inhibitors of chromatin-modifying enzymes to better understand their specific role in transcriptional regulation. As aberrant histone methylation is involved in different pathological processes, the search for methyltransferase and demethylase inhibitors has emerged as a crucial issue in current medicinal chemistry research. High-throughput in vitro assays are important tools for the identification of new methyltransferase or demethylase inhibitors. These usually use oligopeptide substrates derived from histone sequences, although in many cases, they are not good substrates for these enzymes. Here, the authors report about the setup and establishment of in vitro assays that use native core histones as substrates, enabling an assay environment that better resembles native conditions. They have applied these substrates for the known formaldehyde dehydrogenase assay for the histone demethylase LSD1 and have established two new antibody-based assays. For LSD1, a heterogeneous assay format was set up, and a homogeneous assay was used for the characterization of the arginine methyltransferase PRMT1. Validation of the system was achieved with reference inhibitors in each case.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Epigênese Genética , Ensaios de Triagem em Larga Escala/métodos , Histonas/metabolismo , Aldeído Oxirredutases/metabolismo , Anticorpos/metabolismo , Relação Dose-Resposta a Droga , Histona Desmetilases/antagonistas & inibidores , Histona Desmetilases/imunologia , Histona Desmetilases/metabolismo , Proteína-Arginina N-Metiltransferases/antagonistas & inibidores , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/metabolismo , Reprodutibilidade dos Testes
19.
Int J Cancer ; 128(9): 2114-24, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21105039

RESUMO

The nuclear protein PLU-1/JARID1B/KDM5 is widely expressed in breast cancers while showing highly restricted expression in normal adult tissues. To investigate whether JARID1B is a potential target antigen for immunotherapy of breast cancer, we have analyzed the responses of CD8(+) T cells to JARID1B HLA-A*0201 peptides in vitro and used peptide multimers to detect the presence of JARID1B reactive T cells in the circulation of breast cancer patients. Peptides were selected using two web-based algorithms: criteria for inclusion being a high score in both prediction algorithms, and nonhomology with retinoblastoma binding protein-2 (RBP2/JARID1A/KDM5A). A 65-peptide panel was selected and assayed for binding strength by competition assay to obtain the IC(50). The immunogenicity in vitro of these peptides was assessed by T cell stimulation experiments, using autologous dendritic cells as APCs in the first rounds followed by autologous lymphoblasts. Fourteen of the peptides assayed produced cultures having >2% of the CD8(+) cells being IFN-γ(+) after 3-6 rounds of stimulation. An HLA-A*0201 cell line could activate the specific T cells if pulsed with peptide, but endogenous peptide levels were insufficient for activation. Nevertheless, multimer staining of circulating T cells from breast cancer patients showed a significantly higher percentage of multimer positive CD8(+) T cells, as compared to healthy adults for two of three JARID1B epitopes tested. One of these, peptide 73 (QLYALPCVL), was analyzed for memory phenotype, and found to have a significantly higher proportion of central memory T cells than the control group, demonstrating a previous exposure to the peptide.


Assuntos
Neoplasias da Mama/imunologia , Linfócitos T CD8-Positivos/imunologia , Antígenos HLA-A/imunologia , Histona Desmetilases/imunologia , Histona Desmetilases com o Domínio Jumonji/imunologia , Ativação Linfocitária , Células Neoplásicas Circulantes/imunologia , Proteínas Nucleares/imunologia , Proteínas Repressoras/imunologia , Adulto , Antígenos de Neoplasias/imunologia , Neoplasias da Mama/sangue , Neoplasias da Mama/patologia , Separação Celular , Epitopos de Linfócito T/imunologia , Feminino , Citometria de Fluxo , Antígeno HLA-A2 , Histona Desmetilases/biossíntese , Humanos , Hibridização In Situ , Histona Desmetilases com o Domínio Jumonji/biossíntese , Estadiamento de Neoplasias , Proteínas Nucleares/biossíntese , Peptídeos/imunologia , Proteínas Repressoras/biossíntese
20.
Immunol Rev ; 238(1): 233-46, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20969596

RESUMO

Current research suggests that a number of newly identified T-helper cell subsets retain a degree of context-dependent plasticity in their signature cytokine expression patterns. To understand this process, a major challenge is to determine the molecular mechanisms by which lineage-defining transcription factors regulate gene expression profiles in T-helper cells. This mechanistic information will aid in our interpretation of whether a T-helper cell state that expresses or retains the capacity to re-express a combination of lineage-defining transcription factors will have a stable or more flexible gene expression profile. Studies examining the developmental T-box transcription factor T-bet demonstrate the powerful information that is gained from combining in vivo analysis with basic biochemical and molecular mechanism approaches. Significantly, T-bet's ability to physically recruit epigenetic modifying complexes, in particular a Jmjd3 H3K27-demethylase and a Set7/9 H3K4-methyltransferase complex, to its target genes allows T-bet to effectively reverse and establish new epigenetic states. This observation suggests that until T-bet is permanently extinguished, T-helper cells will retain some plasticity toward a T-helper 1-like program. Therefore, insight into the complexity of T-helper cell commitment decisions will be aided by determining the molecular mechanisms for lineage-defining transcription factors.


Assuntos
Diferenciação Celular/imunologia , Histona Desmetilases/imunologia , Histona-Lisina N-Metiltransferase/imunologia , Proteínas com Domínio T/imunologia , Células Th1/imunologia , Animais , Linhagem da Célula , Epigênese Genética/imunologia , Regulação da Expressão Gênica no Desenvolvimento/imunologia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA