Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.238
Filtrar
1.
Zhonghua Bing Li Xue Za Zhi ; 53(6): 563-569, 2024 Jun 08.
Artigo em Chinês | MEDLINE | ID: mdl-38825901

RESUMO

Objective: To analyze the clinicopathological features and differential diagnosis of large B-cell lymphoma with IRF4 rearrangement, aiming enhance its recognition and prevent misdiagnosis. Methods: The clinicopathological features, immunophenotype, and fluorescence in situ hybridization (FISH) results of six cases diagnosed with IRF4 rearrangement-positive B-cell lymphoma at the Affiliated Hospital of Xuzhou Medical University from 2015 to 2023 were retrospectively analyzed. Additionally, a comprehensive review of the literature was conducted. Results: Six patients with IRF4 rearrangement-positive large B-cell lymphoma were included. Patients 1 to 5 included three males and two females with a median age of 19 years ranging from 11 to 34 years. Four patients presented with head and neck lesions, while the other one had a breast nodule; all were in clinical Ann Arbor stages I to Ⅱ. Morphologically, entirely diffuse pattern was present in two cases, purely follicular pattern in one case, and diffuse and follicular patterns in other two cases. The tumor cells, predominantly centroblasts mixed with some irregular centrocytes, were of medium to large size, with a starry sky appearance observed in two cases. Immunophenotyping revealed all cases were positive for bcl-6 and MUM1, with a Ki-67 index ranging from 70% to 90%, and CD10 was positive in two cases. IRF4 rearrangement was confirmed in all cases by FISH analysis, with dual IRF4/bcl-6 rearrangements identified in two cases, leading to a diagnosis of LBCL-IRF4. Case 6, a 39-year-old female with a tonsillar mass and classified as clinical Ann Arbor stage Ⅳ, displayed predominantly diffuse large B-cell lymphoma (DLBCL) morphology with 20% high-grade follicular lymphoma characteristics. Immunohistochemistry showed negative CD10 and positive bcl-6/MUM1, with a Ki-67 index of approximately 80%. Triple rearrangements of IRF4/bcl-2/bcl-6 were identified by FISH, leading to a diagnosis of DLBCL with 20% follicular lymphoma (FL). All six patients achieved complete remission after treatment, with no progression or relapse during a follow-up period of 31-100 months. Conclusions: Large B-cell lymphoma with IRF4 rearrangement is a rare entity with pathological features that overlap with those of FL and DLBCL. While IRF4 rearrangement is necessary for diagnosing LBCL-IRF4, it is not specific and requires differentiation from other aggressive B-cell lymphomas with IRF4 rearrangement.


Assuntos
Rearranjo Gênico , Hibridização in Situ Fluorescente , Fatores Reguladores de Interferon , Linfoma Difuso de Grandes Células B , Proteínas Proto-Oncogênicas c-bcl-6 , Humanos , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo , Diagnóstico Diferencial , Feminino , Masculino , Linfoma Difuso de Grandes Células B/genética , Linfoma Difuso de Grandes Células B/patologia , Linfoma Difuso de Grandes Células B/diagnóstico , Linfoma Difuso de Grandes Células B/metabolismo , Adulto , Adolescente , Estudos Retrospectivos , Proteínas Proto-Oncogênicas c-bcl-6/genética , Proteínas Proto-Oncogênicas c-bcl-6/metabolismo , Criança , Adulto Jovem , Imunofenotipagem , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo
2.
Fish Shellfish Immunol ; 150: 109656, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38801844

RESUMO

Interferon regulatory factor 11 (IRF11), a fish specific member of IRF family, is a transcription factor known for its positive role in teleost antiviral defense by regulating IFN expression. Despite its recognized function, the precise mechanism of IRF11 in type I IFNs production remains largely unknown. In this study, we identified IRF11 in Japanese eel, Anguilla japonica, (AjIRF11) and determined its involvement in the later phase of fish IFN production. Our results demonstrate that IRF11-induced IFN production operates through ISRE binding. Mutations in each ISRE site within the promoter of AjIFN2 or AjIFN4 abolished IRF11-mediated activation of IFN promoters. In addition, the overexpression of AjIRF11 does not significantly impact the activation of AjIFN promoters induced by RLR-related signaling pathway proteins. Furthermore, IRF11-knockdown in ZFLs (zebrafish liver cells) has no effect on the RLRs-induced expression of zebrafish IFN-φ1 and IFN-φ3, indicating that IRF11 is not involved in the RLR-mediated IFN production. However, AjIRF11 can form transcription complexes with AjSTAT1 or AjSTAT2, or form homo- or heterodimers with AjIRF1 to stimulate the transcription of type I IFNs. Overall, it is shown in this study that IRF11 can act synergistically with STAT1 and/or STAT2 for the induction of IFN.


Assuntos
Anguilla , Proteínas de Peixes , Interferon Tipo I , Fator de Transcrição STAT1 , Fator de Transcrição STAT2 , Animais , Fator de Transcrição STAT2/genética , Fator de Transcrição STAT2/metabolismo , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Proteínas de Peixes/genética , Proteínas de Peixes/imunologia , Proteínas de Peixes/metabolismo , Anguilla/genética , Anguilla/imunologia , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo , Fatores Reguladores de Interferon/imunologia , Doenças dos Peixes/imunologia , Imunidade Inata/genética , Peixe-Zebra/genética , Peixe-Zebra/imunologia , Regulação da Expressão Gênica/imunologia
3.
Fish Shellfish Immunol ; 150: 109611, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38734119

RESUMO

During virus-host co-evolution, viruses have developed multiple strategies to dampen IFN response and prevent its antiviral activity in host cells. To date, the interactions between host IFN response and the immune evasion strategies exploited by fish iridoviruses still remain largely uncertain. Here, a potential immune evasion protein candidate of Singapore grouper iridovirus (SGIV), VP82 (encoded by SGIV ORF82) was screened and its roles during viral replication were investigated in detail. Firstly, VP82 overexpression dramatically decreased IFN or ISRE promoter activity and the transcription levels of IFN stimulated genes (ISGs) stimulated by grouper cyclic GMP-AMP synthase (EccGAS)/stimulator of interferon genes (EcSTING), TANK-binding kinase 1 (EcTBK1), IFN regulatory factor 3 (EcIRF3)and EcIRF7. Secondly, Co-IP assays indicated that VP82 interacted with EcIRF3 and EcIRF7, but not EcSTING and EcTBK1, which was consistent with the co-localization between VP82 and EcIRF3 or EcIRF7. Furthermore, VP82 promoted the degradation of EcIRF3 and EcIRF7 in a dose-dependent manner via the autophagy pathway. Finally, VP82 overexpression accelerated SGIV replication, evidenced by the increased transcriptions of viral core genes and viral production. Moreover, the antiviral action of EcIRF3 or EcIRF7 was significantly depressed in VP82 overexpressed cells. Together, VP82 was speculated to exert crucial roles for SGIV replication by inhibiting the IFN response via the degradation of IRF3 and IRF7. Our findings provided new insights into understanding the immune evasion strategies utilized by fish iridovirus through IFN regulation.


Assuntos
Infecções por Vírus de DNA , Doenças dos Peixes , Proteínas de Peixes , Fator Regulador 3 de Interferon , Fator Regulador 7 de Interferon , Ranavirus , Proteínas Virais , Animais , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/metabolismo , Fator Regulador 7 de Interferon/imunologia , Proteínas de Peixes/genética , Proteínas de Peixes/imunologia , Proteínas de Peixes/metabolismo , Doenças dos Peixes/imunologia , Doenças dos Peixes/virologia , Infecções por Vírus de DNA/imunologia , Infecções por Vírus de DNA/veterinária , Ranavirus/fisiologia , Proteínas Virais/genética , Proteínas Virais/metabolismo , Imunidade Inata/genética , Interferons/genética , Interferons/imunologia , Interferons/metabolismo , Evasão da Resposta Imune , Bass/imunologia , Bass/genética , Replicação Viral , Proteínas de Peixe-Zebra , Fatores Reguladores de Interferon
4.
J Virol ; 98(6): e0025524, 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38752725

RESUMO

Human herpesvirus 8 (HHV-8), associated with Kaposi sarcoma, primary effusion lymphoma (PEL), and multicentric Castleman disease, encodes four interferon regulatory factor homologs, vIRFs 1-4, that interact with and inhibit various mediators of host-cell defense against virus infection. A cellular protein targeted by all the vIRFs is ubiquitin-specific protease 7 (USP7); while replication-modulatory and latently infected PEL-cell pro-viability phenotypes of USP7 targeting have been identified for vIRFs 1-3, the significance of the interaction of vIRF-4 with USP7 has remained undetermined. Here we show, through genetic ablation of the vIRF-4-USP7 interaction in infected cells, that vIRF-4 association with USP7 is necessary for optimal expression of vIRF-4 and normal HHV-8 replication. Findings from experiments on transfected and infected cells identified ubiquitination of vIRF-4 via K48-linkage and USP7-binding-associated suppression of vIRF-4 ubiquitination and, in infected cells, increased vIRF-4 expression. Analysis of IFN-I induction and associated signaling as a function of vIRF-4 and its interaction with USP7 identified a role of each in innate-immune suppression. Finally, activation via K63-polyubiquitination of the innate-immune signaling mediator TRAF3 was found to be suppressed by vIRF-4 in a USP7-binding-associated manner in infected cells, but not in transfected cells, likely via binding-regulated expression of vIRF-4. Together, our data identify the first examples of vIRF ubiquitination and a vIRF substrate of USP7, enhanced expression of vIRF-4 via its interaction with USP7, and TRAF3-inhibitory activity of vIRF-4. The findings address, for the first time, the biological significance of the interaction of vIRF-4 with USP7 and reveal a mechanism of vIRF-4-mediated innate-immune evasion and pro-replication activity via TRAF3 regulation. IMPORTANCE: HHV-8 homologs of cellular interferon regulatory factors (IRFs), involved in host-cell defense against virus infection, interact in an inhibitory fashion with IRFs and other mediators of antiviral innate immunity. These interactions are of demonstrated or hypothesized importance for successful primary, productive (lytic), and latent (persistent) infection by HHV-8. While HHV-8 vIRF-4 is known to interact physically with USP7 deubiquitinase, a key regulator of various cellular proteins, the functional and biological significance of the interaction has not been addressed. The present study identifies the interaction as important for HHV-8 productive replication and, indeed, for vIRF-4 expression and reveals a new function of vIRF-4 via inhibition of the activity of TRAF3, a pivotal mediator of host-cell antiviral activity through activation of cellular IRFs and induction of type-I interferons. These findings identify potential targets for the development of novel anti-HHV-8 agents, such as those able to disrupt vIRF-4-USP7 interaction or vIRF-4-stabilizing USP7 activity.


Assuntos
Herpesvirus Humano 8 , Fatores Reguladores de Interferon , Peptidase 7 Específica de Ubiquitina , Ubiquitinação , Proteínas Virais , Replicação Viral , Humanos , Herpesvirus Humano 8/fisiologia , Herpesvirus Humano 8/metabolismo , Peptidase 7 Específica de Ubiquitina/metabolismo , Peptidase 7 Específica de Ubiquitina/genética , Fatores Reguladores de Interferon/metabolismo , Fatores Reguladores de Interferon/genética , Proteínas Virais/metabolismo , Proteínas Virais/genética , Células HEK293 , Fator 3 Associado a Receptor de TNF/metabolismo , Fator 3 Associado a Receptor de TNF/genética , Ligação Proteica , Interações Hospedeiro-Patógeno
5.
Mol Biol Cell ; 35(7): ar102, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38809584

RESUMO

Interferon Regulatory Factor 6 (IRF6) is a transcription factor essential for keratinocyte cell-cell adhesions. Previously, we found that recycling of E-cadherin was defective in the absence of IRF6, yet total E-cadherin levels were not altered, suggesting a previously unknown, nontranscriptional function for IRF6. IRF6 protein contains a DNA binding domain (DBD) and a protein binding domain (PBD). The transcriptional function of IRF6 depends on its DBD and PBD, however, whether the PBD is necessary for the interaction with cytoplasmic proteins has yet to be demonstrated. Here, we show that an intact PBD is required for recruitment of cell-cell adhesion proteins at the plasma membrane, including the recycling of E-cadherin. Colocalizations and coimmunoprecipitations reveal that IRF6 forms a complex in recycling endosomes with Rab11, Myosin Vb, and E-cadherin, and that the PBD is required for this interaction. These data indicate that IRF6 is a novel effector of the endosomal recycling of E-cadherin and demonstrate a non-transcriptional function for IRF6 in regulating cell-cell adhesions.


Assuntos
Caderinas , Adesão Celular , Endossomos , Fatores Reguladores de Interferon , Caderinas/metabolismo , Endossomos/metabolismo , Fatores Reguladores de Interferon/metabolismo , Fatores Reguladores de Interferon/genética , Humanos , Adesão Celular/fisiologia , Animais , Queratinócitos/metabolismo , Membrana Celular/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Ligação Proteica , Camundongos , Transporte Proteico , Domínios Proteicos
6.
Arch Microbiol ; 206(6): 249, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38713385

RESUMO

Escherichia coli (E. coli) can induce severe clinical bovine mastitis, which is to blame for large losses experienced by dairy farms. Macrophage polarization into various states is in response to pathogen infections. Lycopene, a naturally occurring hydrocarbon carotenoid, relieved inflammation by controlling M1/M2 status of macrophages. Thus, we wanted to explore the effect of lycopene on polarization states of macrophages in E. coli-induced mastitis. Macrophages were cultivated with lycopene for 24, before E. coli inoculation for 6 h. Lycopene (0.5 µmol/L) significantly enhanced cell viabilities and significantly reduced lactic dehydrogenase (LDH) levels in macrophages, whereas 2 and 3 µmol/L lycopene significantly enhanced LDH activities. Lycopene treatment significantly reduced the increase in LDH release, iNOS, CD86, TNF-α, IL-1ß and phosphatase and tensin homolog (PTEN) expressions in E. coli group. 0.5 µmol/L lycopene significantly increased E. coli-induced downregulation of CD206, arginase I (ARG1), indoleamine 2,3-dioxygenase (IDO), chitinase 3-like 3 (YM1), PI3K, AKT, p-AKT, mammalian target of rapamycin (mTOR), p-mTOR, jumonji domain-containing protein-3 (JMJD3) and interferon regulatory factor 4 (IRF4) levels. Moreover, Ginkgolic acid C17:1 (a specific PTEN inhibitor), 740YPDGFR (a specific PI3K activator), SC79 (a specific AKT activator) or CHPG sodium salt (a specific NF-κB activator) significantly decreased CD206, AGR1, IDO and YM1 expressions in lycopene and E. coli-treated macrophages. Therefore, lycopene increased M2 macrophages via inhibiting NOTCH1-PI3K-mTOR-NF-κB-JMJD3-IRF4 pathway in response to E. coli infection in macrophages. These results contribute to revealing the pathogenesis of E. coli-caused bovine mastitis, providing the new angle of the prevention and management of mastitis.


Assuntos
Infecções por Escherichia coli , Escherichia coli , Licopeno , Macrófagos , Transdução de Sinais , Animais , Bovinos , Feminino , Camundongos , Linhagem Celular , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/imunologia , Fatores Reguladores de Interferon/metabolismo , Fatores Reguladores de Interferon/genética , Licopeno/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/microbiologia , Macrófagos/imunologia , Macrófagos/metabolismo , Mastite Bovina/microbiologia , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositol 3-Quinases/genética , Receptor Notch1/metabolismo , Receptor Notch1/genética , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo
7.
J Immunol Res ; 2024: 8121284, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38799117

RESUMO

Macroalgae are considered healthy food ingredients due to their content in numerous bioactive compounds, and the traditional use of whole macroalgae in Asian cuisine suggests a contribution to longevity. Although much information is available about the bioactivity of pure algal compounds, such as different polyphenols and polysaccharides, documentation of potential effects of whole macroalgae as part of Western diets is limited. Lifestyle- and age-related diseases, which have a high impact on population health, are closely connected to underlying chronic inflammation. Therefore, we have studied crude extracts of green (Ulva fenestrata) and brown (Saccharina latissima) macroalgae, as two of the most promising food macroalgae in the Nordic countries for their effect on inflammation in vitro. Human macrophage-like reporter THP-1 cells were treated with macroalgae extracts and stimulated with lipopolysaccharide (LPS) to induce inflammatory signalling. Effects of the macroalgae extracts were assessed on transcription factor activity of NF-κB and IRF as well as secretion and/or expression of the cytokines TNF-α and IFN-ß and chemokines IL-8 and CXCL10. The crude macroalgae extracts were further separated into polyphenol-enriched and polysaccharide-enriched fractions, which were also tested for their effect on transcription factor activity. Interestingly, we observed a selective activation of NF-κB, when cells were treated with macroalgae extracts. On the other hand, pretreatment with macroalgae extracts selectively repressed IRF activation when inflammatory signaling was subsequently induced by LPS. This effect was consistent for both tested species as well as for polyphenol- and polysaccharide-enriched fractions, of which the latter had more pronounced effects. Overall, this is the first indication of how macroalgae could modulate inflammatory signaling by selective activation and subsequent repression of different pathways. Further in vitro and in vivo studies of this mechanism would be needed to understand how macroalgae consumption could influence the prevention of noncommunicable, lifestyle- and age-related diseases that are highly related to unbalanced inflammatory processes.


Assuntos
Inflamação , Macrófagos , NF-kappa B , Phaeophyceae , Alga Marinha , Transdução de Sinais , Humanos , NF-kappa B/metabolismo , Macrófagos/metabolismo , Macrófagos/imunologia , Macrófagos/efeitos dos fármacos , Inflamação/metabolismo , Inflamação/imunologia , Fatores Reguladores de Interferon/metabolismo , Fatores Reguladores de Interferon/genética , Citocinas/metabolismo , Células THP-1 , Extratos Vegetais/farmacologia , Lipopolissacarídeos , Algas Comestíveis , Laminaria
8.
Nat Commun ; 15(1): 4232, 2024 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-38762479

RESUMO

Toll-like receptor 9 (TLR9) recognizes bacterial, viral and self DNA and play an important role in immunity and inflammation. However, the role of TLR9 in obesity is less well-studied. Here, we generate B-cell-specific Tlr9-deficient (Tlr9fl/fl/Cd19Cre+/-, KO) B6 mice and model obesity using a high-fat diet. Compared with control mice, B-cell-specific-Tlr9-deficient mice exhibited increased fat tissue inflammation, weight gain, and impaired glucose and insulin tolerance. Furthermore, the frequencies of IL-10-producing-B cells and marginal zone B cells were reduced, and those of follicular and germinal center B cells were increased. This was associated with increased frequencies of IFNγ-producing-T cells and increased follicular helper cells. In addition, gut microbiota from the KO mice induced a pro-inflammatory state leading to immunological and metabolic dysregulation when transferred to germ-free mice. Using 16 S rRNA gene sequencing, we identify altered gut microbial communities including reduced Lachnospiraceae, which may play a role in altered metabolism in KO mice. We identify an important network involving Tlr9, Irf4 and Il-10 interconnecting metabolic homeostasis, with the function of B and T cells, and gut microbiota in obesity.


Assuntos
Linfócitos B , Dieta Hiperlipídica , Disbiose , Microbioma Gastrointestinal , Inflamação , Interleucina-10 , Camundongos Knockout , Obesidade , Receptor Toll-Like 9 , Animais , Obesidade/imunologia , Obesidade/microbiologia , Obesidade/metabolismo , Disbiose/imunologia , Disbiose/microbiologia , Receptor Toll-Like 9/metabolismo , Receptor Toll-Like 9/genética , Linfócitos B/imunologia , Linfócitos B/metabolismo , Inflamação/metabolismo , Camundongos , Dieta Hiperlipídica/efeitos adversos , Interleucina-10/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Fatores Reguladores de Interferon
9.
Clin Exp Pharmacol Physiol ; 51(7): e13868, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38745265

RESUMO

Cervical cancer (CC) is a gynaecological malignancy tumour that seriously threatens women's health. Recent evidence has identified that interferon regulatory factor 5 (IRF5), a nucleoplasm shuttling protein, is a pivotal transcription factor regulating the growth and metastasis of various human tumours. This study aimed to investigate the function and molecular basis of IRF5 in CC development. IRF5, protein phosphatase 6 catalytic subunit (PPP6C) and methyltransferase-like 3 (METTL3) mRNA levels were evaluated by quantitative real-time (qRT)-polymerase chain reaction (PCR). IRF5, PPP6C, METTL3, B-cell lymphoma 2 and Bax protein levels were detected using western blot. Cell proliferation, migration, invasion, angiogenesis and apoptosis were determined by using colony formation, 5-ethynyl-2'-deoxyuridine (EdU), transwell, tube formation assay and flow cytometry assay, respectively. Glucose uptake and lactate production were measured using commercial kits. Xenograft tumour assay in vivo was used to explore the role of IRF5. After JASPAR predication, binding between IRF5 and PPP6C promoter was verified using chromatin immunoprecipitation and dual-luciferase reporter assays. Moreover, the interaction between METTL3 and IRF5 was verified using methylated RNA immunoprecipitation (MeRIP). IRF5, PPP6C and METTL3 were highly expressed in CC tissues and cells. IRF5 silencing significantly inhibited cell proliferation, migration, invasion, angiogenesis and glycolytic metabolism in CC cells, while induced cell apoptosis. Furthermore, the absence of IRF5 hindered tumour growth in vivo. At the molecular level, IRF5 might bind with PPP6C to positively regulate the expression of PPP6C mRNA. Meanwhile, IRF5 was identified as a downstream target of METTL3-mediated m6A modification. METTL3-mediated m6A modification of mRNA might promote CC malignant progression by regulating PPP6C, which might provide a promising therapeutic target for CC treatment.


Assuntos
Proliferação de Células , Progressão da Doença , Fatores Reguladores de Interferon , Metiltransferases , Regulação para Cima , Neoplasias do Colo do Útero , Humanos , Feminino , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/patologia , Neoplasias do Colo do Útero/metabolismo , Metiltransferases/genética , Metiltransferases/metabolismo , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo , Linhagem Celular Tumoral , Animais , Proliferação de Células/genética , Camundongos , Regulação Neoplásica da Expressão Gênica , Apoptose/genética , Movimento Celular/genética , Camundongos Nus , Invasividade Neoplásica , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Neovascularização Patológica/metabolismo
10.
Front Cell Infect Microbiol ; 14: 1395716, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38716195

RESUMO

Objective: The relationship between macrophages and the gut microbiota in patients with atherosclerosis remains poorly defined, and effective biological markers are lacking. This study aims to elucidate the interplay between gut microbial communities and macrophages, and to identify biomarkers associated with the destabilization of atherosclerotic plaques. The goal is to enhance our understanding of the underlying molecular pathways and to pave new avenues for diagnostic approaches and therapeutic strategies in the disease. Methods: This study employed Weighted Gene Co-expression Network Analysis (WGCNA) and differential expression analysis on atherosclerosis datasets to identify macrophage-associated genes and quantify the correlation between these genes and gut microbiota gene sets. The Random Forest algorithm was utilized to pinpoint PLEK, IRF8, BTK, CCR1, and CD68 as gut microbiota-related macrophage genes, and a nomogram was constructed. Based on the top five genes, a Non-negative Matrix Factorization (NMF) algorithm was applied to construct gut microbiota-related macrophage clusters and analyze their potential biological alterations. Subsequent single-cell analyses were conducted to observe the expression patterns of the top five genes and the interactions between immune cells. Finally, the expression profiles of key molecules were validated using clinical samples from atherosclerosis patients. Results: Utilizing the Random Forest algorithm, we ultimately identified PLEK, IRF8, CD68, CCR1, and BTK as gut microbiota-associated macrophage genes that are upregulated in atherosclerotic plaques. A nomogram based on the expression of these five genes was constructed for use as an auxiliary tool in clinical diagnosis. Single-cell analysis confirmed the specific expression of gut microbiota-associated macrophage genes in macrophages. Clinical samples substantiated the high expression of PLEK in unstable atherosclerotic plaques. Conclusion: Gut microbiota-associated macrophage genes (PLEK, IRF8, CD68, CCR1, and BTK) may be implicated in the pathogenesis of atherosclerotic plaques and could serve as diagnostic markers to aid patients with atherosclerosis.


Assuntos
Algoritmos , Aterosclerose , Biomarcadores , Microbioma Gastrointestinal , Aprendizado de Máquina , Macrófagos , Placa Aterosclerótica , Receptores CCR1 , Análise de Célula Única , Humanos , Macrófagos/metabolismo , Macrófagos/microbiologia , Placa Aterosclerótica/microbiologia , Biomarcadores/metabolismo , Análise de Célula Única/métodos , Receptores CCR1/metabolismo , Receptores CCR1/genética , Aterosclerose/microbiologia , Aterosclerose/genética , Antígenos de Diferenciação Mielomonocítica/metabolismo , Tirosina Quinase da Agamaglobulinemia/genética , Tirosina Quinase da Agamaglobulinemia/metabolismo , Antígenos CD/metabolismo , Antígenos CD/genética , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Molécula CD68 , Fatores Reguladores de Interferon
11.
J Chem Neuroanat ; 138: 102424, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38670441

RESUMO

Neuroinflammation associated with microglial activation plays a role in the development of Parkinson's disease (PD). The upregulation of interferon regulatory factor 8 (IRF8) in microglia following peripheral nerve injury has been observed to induce microglial activation. This suggests the potential therapeutic significance of IRF8 in PD. This research aims to explore the effects of IRF8 on the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse model and lipopolysaccharide (LPS)-induced neuroinflammation, along with its underlying mechanisms. The study examines the differential expression of IRF8 and its effects on neuropathological changes using a PD mouse model and a PD model established from BV2 cells in vitro. IRF8 was found to be prominently expressed in the substantia nigra pars compacta (SNpc) region of PD mice and LPS-stimulated BV2 cells, while the expression of tyrosine hydroxylase (TH) and dopamine (DA) content in the SNpc region of PD mice was notably reduced. MPTP treatment and LPS stimulation intensified microglial activation, inflammation, and activation of the AMPK/mTOR signaling pathway in vivo and in vitro, respectively. Upon IRF8 silencing in the PD mouse and cell models, the knockdown of IRF8 ameliorated MPTP-induced behavioral deficits, increased the counts of TH and Nissl-positive neurons and DA content, reduced the number of Iba-1-positive microglia, and reduced the content of inflammatory factors, possibly by inhibiting the AMPK/mTOR signaling pathway. Similar outcomes were observed in the PD cell model. In conclusion, the suppression of IRF8 alleviates neuroinflammation through regulating microglial activation in PD models in vivo and in vitro by the AMPK/mTOR signaling pathway.


Assuntos
Fatores Reguladores de Interferon , Camundongos Endogâmicos C57BL , Microglia , Doenças Neuroinflamatórias , Doença de Parkinson , Animais , Microglia/metabolismo , Camundongos , Fatores Reguladores de Interferon/metabolismo , Fatores Reguladores de Interferon/genética , Masculino , Doenças Neuroinflamatórias/metabolismo , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Lipopolissacarídeos , Serina-Treonina Quinases TOR/metabolismo , Técnicas de Silenciamento de Genes , Transdução de Sinais/fisiologia , Parte Compacta da Substância Negra/metabolismo , Parte Compacta da Substância Negra/patologia , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia
12.
J Cell Physiol ; 239(6): e31268, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38577903

RESUMO

Several members of the transforming growth factor beta (TGF-ß) superfamily regulate the proliferation, differentiation, and function of bone-forming osteoblasts and bone-resorbing osteoclasts. However, it is still unknown whether Nodal, a member of the TGF-ß superfamily, serves a function in bone cells. In this study, we found that Nodal did not have any function in osteoblasts but instead negatively regulated osteoclast differentiation. Nodal inhibited RANKL-induced osteoclast differentiation by downregulating the expression of pro-osteoclastogenic genes, including c-fos, Nfatc1, and Blimp1, and upregulating the expression of antiosteoclastogenic genes, including Bcl6 and Irf8. Nodal activated STAT1 in osteoclast precursor cells, and STAT1 downregulation significantly reduced the inhibitory effect of Nodal on osteoclast differentiation. These findings indicate that Nodal activates STAT1 to downregulate or upregulate the expression of pro-osteoclastogenic or antiosteoclastogenic genes, respectively, leading to the inhibition of osteoclast differentiation. Moreover, the inhibitory effect of Nodal on osteoclast differentiation contributed to the reduction of RANKL-induced bone loss in vivo.


Assuntos
Diferenciação Celular , Fatores Reguladores de Interferon , Osteoclastos , Proteínas Proto-Oncogênicas c-bcl-6 , Ligante RANK , Fator de Transcrição STAT1 , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT1/genética , Animais , Osteoclastos/metabolismo , Fosforilação , Ligante RANK/metabolismo , Camundongos , Fatores Reguladores de Interferon/metabolismo , Fatores Reguladores de Interferon/genética , Proteínas Proto-Oncogênicas c-bcl-6/metabolismo , Proteínas Proto-Oncogênicas c-bcl-6/genética , Fator 1 de Ligação ao Domínio I Regulador Positivo/metabolismo , Fator 1 de Ligação ao Domínio I Regulador Positivo/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , Fatores de Transcrição NFATC/metabolismo , Fatores de Transcrição NFATC/genética , Camundongos Endogâmicos C57BL , Transdução de Sinais , Reabsorção Óssea/metabolismo , Reabsorção Óssea/genética , Reabsorção Óssea/patologia , Osteoblastos/metabolismo , Osteogênese/genética , Células RAW 264.7
13.
Cell Rep ; 43(4): 114107, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38613785

RESUMO

The production of type 1 conventional dendritic cells (cDC1s) requires high expression of the transcription factor IRF8. Three enhancers at the Irf8 3' region function in a differentiation stage-specific manner. However, whether and how these enhancers interact physically and functionally remains unclear. Here, we show that the Irf8 3' enhancers directly interact with each other and contact the Irf8 gene body during cDC1 differentiation. The +56 kb enhancer, which functions from multipotent progenitor stages, activates the other 3' enhancers through an IRF8-dependent transcription factor program, that is, in trans. Then, the +32 kb enhancer, which operates in cDC1-committed cells, reversely acts in cis on the other 3' enhancers to maintain the high expression of Irf8. Indeed, mice with compound heterozygous deletion of the +56 and +32 kb enhancers are unable to generate cDC1s. These results illustrate how multiple enhancers cooperate to induce a lineage-determining transcription factor gene during cell differentiation.


Assuntos
Diferenciação Celular , Células Dendríticas , Elementos Facilitadores Genéticos , Fatores Reguladores de Interferon , Fatores Reguladores de Interferon/metabolismo , Fatores Reguladores de Interferon/genética , Animais , Células Dendríticas/metabolismo , Células Dendríticas/citologia , Elementos Facilitadores Genéticos/genética , Camundongos , Camundongos Endogâmicos C57BL
14.
Int J Mol Sci ; 25(8)2024 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-38674095

RESUMO

During periodontitis, the extracellular capsule of Porphyromonas gingivalis favors alveolar bone loss by inducing Th1 and Th17 patterns of lymphocyte response in the infected periodontium. Dendritic cells recognize bacterial antigens and present them to T lymphocytes, defining their activation and polarization. Thus, dendritic cells could be involved in the Th1 and Th17 response induced against the P. gingivalis capsule. Herein, monocyte-derived dendritic cells were obtained from healthy individuals and then stimulated with different encapsulated strains of P. gingivalis or two non-encapsulated isogenic mutants. Dendritic cell differentiation and maturation were analyzed by flow cytometry. The mRNA expression levels for distinct Th1-, Th17-, or T-regulatory-related cytokines and transcription factors, as well as TLR2 and TLR4, were assessed by qPCR. In addition, the production of IL-1ß, IL-6, IL-23, and TNF-α was analyzed by ELISA. The encapsulated strains and non-encapsulated mutants of P. gingivalis induced dendritic cell maturation to a similar extent; however, the pattern of dendritic cell response was different. In particular, the encapsulated strains of P. gingivalis induced higher expression of IRF4 and NOTCH2 and production of IL-1ß, IL-6, IL-23, and TNF-α compared with the non-encapsulated mutants, and thus, they showed an increased capacity to trigger Th1 and Th17-type responses in human dendritic cells.


Assuntos
Citocinas , Células Dendríticas , Porphyromonas gingivalis , Células Th17 , Receptor 2 Toll-Like , Receptor 4 Toll-Like , Porphyromonas gingivalis/imunologia , Humanos , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/microbiologia , Células Th17/imunologia , Células Th17/metabolismo , Receptor 2 Toll-Like/metabolismo , Receptor 2 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Receptor 4 Toll-Like/genética , Citocinas/metabolismo , Diferenciação Celular , Células Th1/imunologia , Fatores Reguladores de Interferon/metabolismo , Fatores Reguladores de Interferon/genética , Receptor Notch2/genética , Receptor Notch2/metabolismo , Células Cultivadas , Cápsulas Bacterianas/imunologia , Cápsulas Bacterianas/metabolismo , Infecções por Bacteroidaceae/imunologia , Infecções por Bacteroidaceae/microbiologia , Fator de Necrose Tumoral alfa/metabolismo
15.
Br J Haematol ; 204(5): 1862-1871, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38613165

RESUMO

Peripheral T-cell lymphomas (PTCL) are morphologically and biologically heterogeneous and a subset expresses CD30, including anaplastic large cell lymphomas (ALCL) and a minority of PTCL, not otherwise specified (PTCL, NOS). ALCL with ALK translocations (ALCL, ALK+) are readily identified by routine diagnostic methods, but differentiating ALCL without ALK translocation (ALCL, ALK-) and PTCL, NOS expressing CD30 (PTCL CD30+) can be challenging. Furthermore, rare PTCL co-express CD30 and CD15 (PTCL CD30+CD15+); some resemble ALCL, ALK- while others resemble classic Hodgkin lymphoma. To explore the relationship between PTCL CD30+CD15+ and ALCL, ALK-, we analysed 19 cases of PTCL with CD30 expression, previously diagnosed as ALCL, ALK- (nine cases) and PTCL CD30+CD15+ (10 cases) for DUSP22/IRF4 rearrangements, coding RNA expression and selected transcriptome analysis using the NanoString nCounter gene expression analysis platform. Unsupervised clustering showed no clear segregation between ALCL, ALK- and PTCL CD30+CD15+. Three cases previously classified as PTCL CD30+CD15+ showed DUSP22/IRF4 rearrangements, favouring a diagnosis of ALCL, ALK-. Our results suggest that cases previously designated PTCL CD30+CD15+, likely fall within the spectrum of ALCL, ALK-; additionally, a subset of ALCL, ALK- with DUSP22/IRF4 rearrangement expresses CD15, consistent with previous reports and expands the immunophenotypic spectrum of this lymphoma subgroup.


Assuntos
Quinase do Linfoma Anaplásico , Antígeno Ki-1 , Antígenos CD15 , Linfoma Anaplásico de Células Grandes , Linfoma de Células T Periférico , Humanos , Linfoma Anaplásico de Células Grandes/genética , Linfoma Anaplásico de Células Grandes/patologia , Linfoma Anaplásico de Células Grandes/diagnóstico , Antígeno Ki-1/metabolismo , Antígeno Ki-1/genética , Antígeno Ki-1/análise , Linfoma de Células T Periférico/genética , Linfoma de Células T Periférico/metabolismo , Linfoma de Células T Periférico/patologia , Linfoma de Células T Periférico/diagnóstico , Masculino , Feminino , Pessoa de Meia-Idade , Adulto , Quinase do Linfoma Anaplásico/genética , Quinase do Linfoma Anaplásico/metabolismo , Antígenos CD15/análise , Antígenos CD15/metabolismo , Idoso , Fosfatases de Especificidade Dupla/genética , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Adulto Jovem , Rearranjo Gênico
16.
EMBO J ; 43(10): 1947-1964, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38605225

RESUMO

Transcription factors BACH2 and IRF4 are both essential for antibody class-switch recombination (CSR) in activated B lymphocytes, while they oppositely regulate the differentiation of plasma cells (PCs). Here, we investigated how BACH2 and IRF4 interact during CSR and plasma-cell differentiation. We found that BACH2 organizes heterochromatin formation of target gene loci in mouse splenic B cells, including targets of IRF4 activation such as Aicda, an inducer of CSR, and Prdm1, a master plasma-cell regulator. Release of these gene loci from heterochromatin in response to B-cell receptor stimulation was coupled to AKT-mTOR pathway activation. In Bach2-deficient B cells, PC genes' activation depended on IRF4 protein accumulation, without an increase in Irf4 mRNA. Mechanistically, a PU.1-IRF4 heterodimer in activated B cells promoted BACH2 function by inducing gene expression of Bach2 and Pten, a negative regulator of AKT signaling. Elevated AKT activity in Bach2-deficient B cells resulted in IRF4 protein accumulation. Thus, BACH2 and IRF4 mutually modulate the activity of each other, and BACH2 inhibits PC differentiation by both the repression of PC genes and the restriction of IRF4 protein accumulation.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica , Diferenciação Celular , Fatores Reguladores de Interferon , Plasmócitos , Animais , Fatores Reguladores de Interferon/metabolismo , Fatores Reguladores de Interferon/genética , Camundongos , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Fatores de Transcrição de Zíper de Leucina Básica/genética , Plasmócitos/metabolismo , Plasmócitos/imunologia , Plasmócitos/citologia , Switching de Imunoglobulina/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Linfócitos B/metabolismo , Linfócitos B/imunologia , Linfócitos B/citologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/genética , Camundongos Knockout , PTEN Fosfo-Hidrolase/metabolismo , PTEN Fosfo-Hidrolase/genética , Camundongos Endogâmicos C57BL , Transativadores/metabolismo , Transativadores/genética , Heterocromatina/metabolismo , Heterocromatina/genética , Serina-Treonina Quinases TOR/metabolismo , Serina-Treonina Quinases TOR/genética
17.
Immunity ; 57(5): 1124-1140.e9, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38636522

RESUMO

Signaling through Notch receptors intrinsically regulates tumor cell development and growth. Here, we studied the role of the Notch ligand Jagged2 on immune evasion in non-small cell lung cancer (NSCLC). Higher expression of JAG2 in NSCLC negatively correlated with survival. In NSCLC pre-clinical models, deletion of Jag2, but not Jag1, in cancer cells attenuated tumor growth and activated protective anti-tumor T cell responses. Jag2-/- lung tumors exhibited higher frequencies of macrophages that expressed immunostimulatory mediators and triggered T cell-dependent anti-tumor immunity. Mechanistically, Jag2 ablation promoted Nr4a-mediated induction of Notch ligands DLL1/4 on cancer cells. DLL1/4-initiated Notch1/2 signaling in macrophages induced the expression of transcription factor IRF4 and macrophage immunostimulatory functionality. IRF4 expression was required for the anti-tumor effects of Jag2 deletion in lung tumors. Antibody targeting of Jagged2 inhibited tumor growth and activated IRF4-driven macrophage-mediated anti-tumor immunity. Thus, Jagged2 orchestrates immunosuppressive systems in NSCLC that can be overcome to incite macrophage-mediated anti-tumor immunity.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Fatores Reguladores de Interferon , Proteína Jagged-2 , Neoplasias Pulmonares , Camundongos Knockout , Macrófagos Associados a Tumor , Proteína Jagged-2/metabolismo , Proteína Jagged-2/genética , Proteína Jagged-2/imunologia , Animais , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/genética , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Camundongos , Humanos , Fatores Reguladores de Interferon/metabolismo , Fatores Reguladores de Interferon/genética , Macrófagos Associados a Tumor/imunologia , Macrófagos Associados a Tumor/metabolismo , Transdução de Sinais , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/genética , Linhagem Celular Tumoral , Camundongos Endogâmicos C57BL , Receptores Notch/metabolismo , Receptor Notch1/metabolismo , Receptor Notch1/genética , Macrófagos/imunologia , Macrófagos/metabolismo , Proteína Jagged-1/metabolismo , Proteína Jagged-1/genética , Evasão Tumoral/imunologia
18.
Int Immunopharmacol ; 133: 112077, 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38615379

RESUMO

Interferon regulatory factor 4 (IRF4) is a member of IRF family of transcription factors which mainly regulates the transcription of IFN. IRF4 is restrictively expressed in immune cells such as T and B cells, macrophages, as well as DC. It is essential for the development and function of these cells. Since these cells take part in the homeostasis of the immune system and dysfunction of them contributes to the initiation and progress of systemic lupus erythematosus (SLE), the roles of IRF4 in the SLE development becomes an important topic. Here we systemically discuss the biological characteristics of IRF4 in various immune cells and analyze the pathologic effects of IRF4 alteration in SLE and the potential targeting therapeutics of SLE.


Assuntos
Fatores Reguladores de Interferon , Lúpus Eritematoso Sistêmico , Lúpus Eritematoso Sistêmico/imunologia , Humanos , Fatores Reguladores de Interferon/metabolismo , Fatores Reguladores de Interferon/genética , Animais , Macrófagos/imunologia , Linfócitos T/imunologia , Linfócitos B/imunologia , Células Dendríticas/imunologia
19.
Dev Comp Immunol ; 156: 105181, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38636698

RESUMO

Interferon regulatory factor 7 (IRF7) is considered the master regulator of virus-induced interferon (IFN) production. However, to avoid an autoimmune response, the expression of IRF7 must be tightly controlled. In this study, we report that zebrafish ubiquitin-specific protease 8 (USP8) promotes IRF7 degradation through an autophagy-lysosome-dependent pathway to inhibit IFN production. First, zebrafish usp8 is induced upon spring viremia of carp virus (SVCV) infection and polyinosinic/polycytidylic acid (poly I:C) stimulation. Second, overexpression of USP8 suppresses SVCV or poly I:C-mediated IFN expression. Mechanistically, USP8 interacts with IRF7 and promotes its degradation via an autophagy-lysosome-dependent pathway. Finally, USP8 significantly suppresses cellular antiviral responses and enhances SVCV proliferation. In summary, our discoveries offer a perspective on the role of zebrafish USP8 and provide additional understanding of the regulation of IRF7 in host antiviral immune response.


Assuntos
Autofagia , Fator Regulador 7 de Interferon , Fatores Reguladores de Interferon , Lisossomos , Rhabdoviridae , Proteínas de Peixe-Zebra , Peixe-Zebra , Animais , Peixe-Zebra/imunologia , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Autofagia/imunologia , Lisossomos/metabolismo , Fator Regulador 7 de Interferon/metabolismo , Fator Regulador 7 de Interferon/genética , Rhabdoviridae/fisiologia , Rhabdoviridae/imunologia , Interferons/metabolismo , Poli I-C/imunologia , Infecções por Rhabdoviridae/imunologia , Proteólise , Doenças dos Peixes/imunologia , Doenças dos Peixes/virologia , Ubiquitina Tiolesterase/metabolismo , Ubiquitina Tiolesterase/genética , Humanos , Imunidade Inata
20.
Blood Adv ; 8(9): 2217-2234, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38457926

RESUMO

ABSTRACT: Multiple myeloma (MM) cells are addicted to MYC and its direct transactivation targets IRF4 for proliferation and survival. MYC and IRF4 are still considered "undruggable," as most small-molecule inhibitors suffer from low potency, suboptimal pharmacokinetic properties, and undesirable off-target effects. Indirect inhibition of MYC/IRF4 emerges as a therapeutic vulnerability in MM. Here, we uncovered an unappreciated tumor-suppressive role of C-terminal binding protein 2 (CTBP2) in MM via strong inhibition of the MYC-IRF4 axis. In contrast to epithelial cancers, CTBP2 is frequently downregulated in MM, in association with shortened survival, hyperproliferative features, and adverse clinical outcomes. Restoration of CTBP2 exhibited potent antitumor effects against MM in vitro and in vivo, with marked repression of the MYC-IRF4 network genes. Mechanistically, CTBP2 impeded the transcription of MYC and IRF4 by histone H3 lysine 27 deacetylation (H3K27ac) and indirectly via activation of the MYC repressor IFIT3. In addition, activation of the interferon gene signature by CTBP2 suggested its concomitant immunomodulatory role in MM. Epigenetic studies have revealed the contribution of polycomb-mediated silencing and DNA methylation to CTBP2 inactivation in MM. Notably, inhibitors of Enhance of zeste homolog 2, histone deacetylase, and DNA methyltransferase, currently under evaluation in clinical trials, were effective in restoring CTBP2 expression in MM. Our findings indicated that the loss of CTBP2 plays an essential role in myelomagenesis and deciphers an additional mechanistic link to MYC-IRF4 dysregulation in MM. We envision that the identification of novel critical regulators will facilitate the development of selective and effective approaches for treating this MYC/IRF4-addicted malignancy.


Assuntos
Oxirredutases do Álcool , Proteínas Correpressoras , Fatores Reguladores de Interferon , Mieloma Múltiplo , Proteínas Proto-Oncogênicas c-myc , Animais , Humanos , Camundongos , Oxirredutases do Álcool/metabolismo , Oxirredutases do Álcool/antagonistas & inibidores , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Fatores Reguladores de Interferon/metabolismo , Fatores Reguladores de Interferon/genética , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/patologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Supressoras de Tumor/metabolismo , Proteínas Correpressoras/antagonistas & inibidores , Proteínas Correpressoras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA