Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 317
Filtrar
1.
Nat Commun ; 15(1): 3561, 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38670996

RESUMEN

Lysine lactylation (Kla) links metabolism and gene regulation and plays a key role in multiple biological processes. However, the regulatory mechanism and functional consequence of Kla remain to be explored. Here, we report that HBO1 functions as a lysine lactyltransferase to regulate transcription. We show that HBO1 catalyzes the addition of Kla in vitro and intracellularly, and E508 is a key site for the lactyltransferase activity of HBO1. Quantitative proteomic analysis further reveals 95 endogenous Kla sites targeted by HBO1, with the majority located on histones. Using site-specific antibodies, we find that HBO1 may preferentially catalyze histone H3K9la and scaffold proteins including JADE1 and BRPF2 can promote the enzymatic activity for histone Kla. Notably, CUT&Tag assays demonstrate that HBO1 is required for histone H3K9la on transcription start sites (TSSs). Besides, the regulated Kla can promote key signaling pathways and tumorigenesis, which is further supported by evaluating the malignant behaviors of HBO1- knockout (KO) tumor cells, as well as the level of histone H3K9la in clinical tissues. Our study reveals HBO1 serves as a lactyltransferase to mediate a histone Kla-dependent gene transcription.


Asunto(s)
Histonas , Factor C1 de la Célula Huésped , Lisina , Transcripción Genética , Histonas/metabolismo , Humanos , Lisina/metabolismo , Células HEK293 , Animales , Línea Celular Tumoral , Sitio de Iniciación de la Transcripción , Regulación de la Expresión Génica , Ratones , Procesamiento Proteico-Postraduccional
2.
Genet Res (Camb) ; 2022: 9304264, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36299684

RESUMEN

Background: Our study aimed to investigate the relationship between extracellular matrix 1 (ECM1) gene polymorphism and progression of liver fibrosis in the Chinese population. Methods: A total 656 patients with hepatitis B virus (HBV) infection and 298 healthy individuals of the Chinese Han population were recruited for a retrospective case-control study. Of the disease group, 104 cases had chronic hepatitis B (CHB), 266 had LC, and 286 had hepatocellular carcinoma (HCC). Subjects were frequency-matched according to age and gender. Polymorphisms of the ECM1 gene were examined using the MassARRAY SNP genotyping method. Results: There were no associations between genotype and allele frequencies of ECM1 rs3737240 and rs13294 loci with the risk of CHB and CHB-related HCC. After adjustment for age, sex, smoking status, and drinking habits, the GT genotype was dramatically related to a reduced risk of chronic HBV infection in both non-HCC (OR = 0.68, 95% CI: 0.49-0.94) and total chronic HBV infection patients (OR = 0.75, 95% CI: 0.56-1.00). Haplotype analyses revealed twelve protective haplotypes against total chronic HBV infection and four against non-HCC chronic HBV infection. Conclusion: ECM1 gene polymorphism in rs3834087 and rs3754217 loci is associated with a reduced risk of chronic HBV infection but not with liver fibrosis development and the occurrence of HCC.


Asunto(s)
Carcinoma Hepatocelular , Enfermedades del Sistema Digestivo , Hepatitis B Crónica , Neoplasias Hepáticas , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Estudios de Casos y Controles , Estudios Retrospectivos , Factor C1 de la Célula Huésped/genética , Predisposición Genética a la Enfermedad , Polimorfismo de Nucleótido Simple/genética , Carcinoma Hepatocelular/patología , Hepatitis B Crónica/complicaciones , Hepatitis B Crónica/genética , Hepatitis B Crónica/epidemiología , Virus de la Hepatitis B , Cirrosis Hepática/genética , Cirrosis Hepática/complicaciones , Frecuencia de los Genes , Genotipo , Enfermedades del Sistema Digestivo/complicaciones , Proteínas de la Matriz Extracelular/genética , China/epidemiología
3.
Am J Hum Genet ; 109(2): 361-372, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35051358

RESUMEN

Nuclear deubiquitinase BAP1 (BRCA1-associated protein 1) is a core component of multiprotein complexes that promote transcription by reversing the ubiquitination of histone 2A (H2A). BAP1 is a tumor suppressor whose germline loss-of-function variants predispose to cancer. To our knowledge, there are very rare examples of different germline variants in the same gene causing either a neurodevelopmental disorder (NDD) or a tumor predisposition syndrome. Here, we report a series of 11 de novo germline heterozygous missense BAP1 variants associated with a rare syndromic NDD. Functional analysis showed that most of the variants cannot rescue the consequences of BAP1 inactivation, suggesting a loss-of-function mechanism. In T cells isolated from two affected children, H2A deubiquitination was impaired. In matching peripheral blood mononuclear cells, histone H3 K27 acetylation ChIP-seq indicated that these BAP1 variants induced genome-wide chromatin state alterations, with enrichment for regulatory regions surrounding genes of the ubiquitin-proteasome system (UPS). Altogether, these results define a clinical syndrome caused by rare germline missense BAP1 variants that alter chromatin remodeling through abnormal histone ubiquitination and lead to transcriptional dysregulation of developmental genes.


Asunto(s)
Proteína BRCA1/genética , Mutación de Línea Germinal , Mutación con Pérdida de Función , Mutación Missense , Trastornos del Neurodesarrollo/genética , Proteínas Supresoras de Tumor/genética , Ubiquitina Tiolesterasa/genética , Adolescente , Proteína BRCA1/inmunología , Niño , Preescolar , Cromatina/química , Cromatina/inmunología , Ensamble y Desensamble de Cromatina/genética , Ensamble y Desensamble de Cromatina/inmunología , Familia , Femenino , Regulación de la Expresión Génica , Heterocigoto , Histonas/genética , Histonas/inmunología , Factor C1 de la Célula Huésped/genética , Factor C1 de la Célula Huésped/inmunología , Humanos , Lactante , Masculino , Trastornos del Neurodesarrollo/inmunología , Trastornos del Neurodesarrollo/patología , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/inmunología , Linfocitos T/inmunología , Linfocitos T/patología , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/inmunología , Ubiquitina/genética , Ubiquitina/inmunología , Ubiquitina Tiolesterasa/deficiencia , Ubiquitina Tiolesterasa/inmunología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/inmunología , Ubiquitinación
4.
Nat Commun ; 13(1): 134, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013307

RESUMEN

Combined methylmalonic acidemia and homocystinuria (cblC) is the most common inborn error of intracellular cobalamin metabolism and due to mutations in Methylmalonic Aciduria type C and Homocystinuria (MMACHC). Recently, mutations in the transcriptional regulators HCFC1 and RONIN (THAP11) were shown to result in cellular phenocopies of cblC. Since HCFC1/RONIN jointly regulate MMACHC, patients with mutations in these factors suffer from reduced MMACHC expression and exhibit a cblC-like disease. However, additional de-regulated genes and the resulting pathophysiology is unknown. Therefore, we have generated mouse models of this disease. In addition to exhibiting loss of Mmachc, metabolic perturbations, and developmental defects previously observed in cblC, we uncovered reduced expression of target genes that encode ribosome protein subunits. We also identified specific phenotypes that we ascribe to deregulation of ribosome biogenesis impacting normal translation during development. These findings identify HCFC1/RONIN as transcriptional regulators of ribosome biogenesis during development and their mutation results in complex syndromes exhibiting aspects of both cblC and ribosomopathies.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/genética , Homocistinuria/genética , Factor C1 de la Célula Huésped/genética , Oxidorreductasas/genética , Proteínas Represoras/genética , Ribosomas/genética , Deficiencia de Vitamina B 12/genética , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Errores Innatos del Metabolismo de los Aminoácidos/patología , Animales , Modelos Animales de Enfermedad , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica , Homocistinuria/metabolismo , Homocistinuria/patología , Factor C1 de la Célula Huésped/deficiencia , Humanos , Masculino , Ratones , Ratones Noqueados , Mutación , Biogénesis de Organelos , Oxidorreductasas/deficiencia , Biosíntesis de Proteínas , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Proteínas Represoras/deficiencia , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Ribosomas/metabolismo , Ribosomas/patología , Vitamina B 12/metabolismo , Deficiencia de Vitamina B 12/metabolismo , Deficiencia de Vitamina B 12/patología
5.
Mol Cell ; 81(12): 2611-2624.e10, 2021 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-33857404

RESUMEN

The Shieldin complex shields double-strand DNA breaks (DSBs) from nucleolytic resection. Curiously, the penultimate Shieldin component, SHLD1, is one of the least abundant mammalian proteins. Here, we report that the transcription factors THAP1, YY1, and HCF1 bind directly to the SHLD1 promoter, where they cooperatively maintain the low basal expression of SHLD1, thereby ensuring a proper balance between end protection and resection during DSB repair. The loss of THAP1-dependent SHLD1 expression confers cross-resistance to poly (ADP-ribose) polymerase (PARP) inhibitor and cisplatin in BRCA1-deficient cells and shorter progression-free survival in ovarian cancer patients. Moreover, the embryonic lethality and PARPi sensitivity of BRCA1-deficient mice is rescued by ablation of SHLD1. Our study uncovers a transcriptional network that directly controls DSB repair choice and suggests a potential link between DNA damage and pathogenic THAP1 mutations, found in patients with the neurodevelopmental movement disorder adult-onset torsion dystonia type 6.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Animales , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteínas de Ciclo Celular/genética , ADN/metabolismo , Roturas del ADN de Doble Cadena/efectos de los fármacos , Reparación del ADN por Unión de Extremidades/efectos de los fármacos , Reparación del ADN/genética , Distonía/genética , Femenino , Factor C1 de la Célula Huésped/metabolismo , Proteínas Mad2/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Reparación del ADN por Recombinación/efectos de los fármacos , Proteínas de Unión a Telómeros/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Factor de Transcripción YY1/metabolismo
6.
Elife ; 102021 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-33416496

RESUMEN

The oncoprotein transcription factor MYC is a major driver of malignancy and a highly validated but challenging target for the development of anticancer therapies. Novel strategies to inhibit MYC may come from understanding the co-factors it uses to drive pro-tumorigenic gene expression programs, providing their role in MYC activity is understood. Here we interrogate how one MYC co-factor, host cell factor (HCF)-1, contributes to MYC activity in a human Burkitt lymphoma setting. We identify genes connected to mitochondrial function and ribosome biogenesis as direct MYC/HCF-1 targets and demonstrate how modulation of the MYC-HCF-1 interaction influences cell growth, metabolite profiles, global gene expression patterns, and tumor growth in vivo. This work defines HCF-1 as a critical MYC co-factor, places the MYC-HCF-1 interaction in biological context, and highlights HCF-1 as a focal point for development of novel anti-MYC therapies.


Tumours form when cells lose control of their growth. Usually, cells produce signals that control how much and how often they divide. But if these signals become faulty, cells may grow too quickly or multiply too often. For example, a group of proteins known as MYC proteins activate growth genes in a cell, but too much of these proteins causes cells to grow uncontrollably. With one third of all cancer deaths linked to excess MYC proteins, these molecules could be key targets for anti-cancer drugs. However, current treatments fail to target these proteins. One option for treating cancers linked to MYC proteins could be to target proteins that work alongside MYC proteins, such as the protein HCF-1, which can attach to MYC proteins. To test if HCF-1 could be a potential drug target, Popay et al. first studied how HCF-1 and MYC proteins interacted using specific cancer cells grown in the laboratory. This revealed that when the two proteins connected, they activated genes that trigger rapid cell growth. When these cancer cells were then injected into mice, tumours quickly grew. However, when the MYC and HCF-1 attachments in the cancer cells were disrupted, the tumours shrunk. This suggests that if anti-cancer drugs were able to target HCF-1 proteins, they could potentially reduce or even reverse the growth of tumours. While further research is needed to identify drug candidates, these findings reveal a promising target for treating tumours that stem from over-abundant MYC proteins.


Asunto(s)
Expresión Génica , Genes Mitocondriales , Factor C1 de la Célula Huésped/genética , Biogénesis de Organelos , Proteínas Proto-Oncogénicas c-myc/genética , Ribosomas/fisiología , Animales , Linfoma de Burkitt , Femenino , Factor C1 de la Célula Huésped/metabolismo , Humanos , Ratones , Ratones Desnudos , Proteínas Proto-Oncogénicas c-myc/metabolismo
7.
Cell Death Dis ; 11(10): 907, 2020 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-33097698

RESUMEN

The eukaryotic cell cycle involves a highly orchestrated series of events in which the cellular genome is replicated during a synthesis (S) phase and each of the two resulting copies are segregated properly during mitosis (M). Host cell factor-1 (HCF-1) is a transcriptional co-regulator that is essential for and has been implicated in basic cellular processes, such as transcriptional regulation and cell cycle progression. Although a series of HCF-1 transcriptional targets have been identified, few functional clues have been provided, especially for chromosome segregation. Our results showed that HCF-1 activated CDC42 expression by binding to the -881 to -575 region upstream of the CDC42 transcription start site, and the regulation of CDC42 expression by HCF-1 was correlated with cell cycle progression. The overexpression of a spontaneously cycling and constitutively active CDC42 mutant (CDC42F28L) rescued G1 phase delay and multinucleate defects in mitosis upon the loss of HCF-1. Therefore, these results establish that HCF-1 ensures proper cell cycle progression by regulating the expression of CDC42, which indicates a possible mechanism of cell cycle coordination and the regulation mode of typical Rho GTPases.


Asunto(s)
Factor C1 de la Célula Huésped/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Ciclo Celular/fisiología , Segregación Cromosómica , Ciclina A/biosíntesis , Ciclina A/genética , Progresión de la Enfermedad , Puntos de Control de la Fase G1 del Ciclo Celular , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HeLa , Factor C1 de la Célula Huésped/genética , Humanos , Mitosis , Regiones Promotoras Genéticas , Proteína de Unión al GTP cdc42/biosíntesis , Proteína de Unión al GTP cdc42/genética
8.
BMB Rep ; 53(12): 634-639, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33050986

RESUMEN

In prostate cancer, the androgen receptor (AR) transcription factor is a major regulator of cell proliferation and metastasis. To identify new AR regulators, we focused on Mixed lineage leukemia 5 (MLL5), a histone-regulating enzyme, because significantly higher MLL5 expression was detected in prostate cancer tissues than in matching normal tissues. When we expressed shRNAs targeting MLL5 gene in prostate cancer cell line, the growth rate and AR activity were reduced compared to those in control cells, and migration ability of the knockdown cells was reduced significantly. To determine the molecular mechanisms of MLL5 on AR activity, we proved that AR physically interacted with MLL5 and other co-factors, including SET-1 and HCF-1, using an immunoprecipitation method. The chromatin immunoprecipitation analysis showed reduced binding of MLL5, co-factors, and AR enzymes to AR target gene promoters in MLL5 shRNA-expressing cells. Histone H3K4 methylation on the AR target gene promoters was reduced, and H3K9 methylation at the same site was increased in MLL5 knockdown cells. Finally, xenograft tumor formation revealed that reduction of MLL5 in prostate cancer cells retarded tumor growth. Our results thus demonstrate the important role of MLL5 as a new epigenetic regulator of AR in prostate cancer. [BMB Reports 2020; 53(12): 634-639].


Asunto(s)
Proteínas de Unión al ADN/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Factor C1 de la Célula Huésped/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Proteínas de Unión al ADN/genética , Regulación Neoplásica de la Expresión Génica/genética , N-Metiltransferasa de Histona-Lisina/fisiología , Histonas/metabolismo , Factor C1 de la Célula Huésped/fisiología , Humanos , Masculino , Metilación , Regiones Promotoras Genéticas/genética , Neoplasias de la Próstata/metabolismo , Procesamiento Proteico-Postraduccional/genética , Receptores Androgénicos/metabolismo , Receptores Androgénicos/fisiología , Transducción de Señal/genética , Factores de Transcripción/metabolismo
9.
Cell Rep ; 29(6): 1645-1659.e9, 2019 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-31693902

RESUMEN

Molecular chaperones such as heat-shock proteins (HSPs) help in protein folding. Their function in the cytosol has been well studied. Notably, chaperones are also present in the nucleus, a compartment where proteins enter after completing de novo folding in the cytosol, and this raises an important question about chaperone function in the nucleus. We performed a systematic analysis of the nuclear pool of heat-shock protein 90. Three orthogonal and independent analyses led us to the core functional interactome of HSP90. Computational and biochemical analyses identify host cell factor C1 (HCFC1) as a transcriptional regulator that depends on HSP90 for its stability. HSP90 was required to maintain the expression of HCFC1-targeted cell-cycle genes. The regulatory nexus between HSP90 and the HCFC1 module identified in this study sheds light on the relevance of chaperones in the transcription of cell-cycle genes. Our study also suggests a therapeutic avenue of combining chaperone and transcription inhibitors for cancer treatment.


Asunto(s)
Cromatina/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Genes cdc , Proteínas HSP90 de Choque Térmico/metabolismo , Factor C1 de la Célula Huésped/metabolismo , Animales , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular/genética , Secuenciación de Inmunoprecipitación de Cromatina , Quinasa 9 Dependiente de la Ciclina/antagonistas & inhibidores , Citosol/metabolismo , Bases de Datos Genéticas , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/genética , Factor C1 de la Célula Huésped/genética , Humanos , Ratones , Unión Proteica , Mapas de Interacción de Proteínas , RNA-Seq
10.
Cell Oncol (Dordr) ; 42(5): 579-589, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31183804

RESUMEN

BACKGROUND: The p53 and HSF1 transcription factors are key players in cellular responses to stress. They activate important signaling pathways triggering adaptive mechanisms that maintain cellular homeostasis. HSF1 is mainly activated by proteotoxic stress, and its induction leads to the synthesis of chaperones that provide proteome integrity. The p53 protein, which is primarily activated in response to DNA damage, causes cell cycle arrest allowing for DNA repair or directs cells to apoptosis, thereby maintaining genome integrity. Both signaling pathways are also involved in neoplastic transformation and tumor progression. Loss of tumor suppressor abilities of the wild-type p53 protein results in oncogenesis, whereas proper HSF1 action, though non-oncogenic itself, actively supports this process. CONCLUSIONS: Here, we describe in detail the interplay between the p53 and HSF1 signaling pathways, with particular emphasis on the molecular mechanisms involved, as well as their importance for normal cellular behavior, cancer development, the effectiveness of anti-cancer therapies and their toxicity. Detailed knowledge of the complex interplay between HSF1 and p53 may form a basis for the design of new protocols for cancer treatment.


Asunto(s)
Carcinogénesis/genética , Factor C1 de la Célula Huésped/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis , Carcinogénesis/metabolismo , Senescencia Celular/genética , Citoprotección/genética , Citoprotección/fisiología , Reparación del ADN , Progresión de la Enfermedad , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Neovascularización Patológica/metabolismo , Proteínas Nucleares/metabolismo , Dependencia del Oncogén/genética , Transducción de Señal , Estrés Fisiológico/genética , Proteína p53 Supresora de Tumor/genética
11.
Theranostics ; 9(8): 2183-2197, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31149037

RESUMEN

O-GlcNAc transferase (OGT) is overexpressed in aggressive prostate cancer. OGT modifies intra-cellular proteins via single sugar conjugation (O-GlcNAcylation) to alter their activity. We recently discovered the first fast-acting OGT inhibitor OSMI-2. Here, we probe the stability and function of the chromatin O-GlcNAc and identify transcription factors that coordinate with OGT to promote proliferation of prostate cancer cells. Methods: Chromatin immunoprecipitation (ChIP) coupled to sequencing (seq), formaldehyde-assisted isolation of regulatory elements, RNA-seq and reverse-phase protein arrays (RPPA) were used to study the importance of OGT for chromatin structure and transcription. Mass spectrometry, western blot, RT-qPCR, cell cycle analysis and viability assays were used to establish the role of OGT for MYC-related processes. Prostate cancer patient data profiled for both mRNA and protein levels were used to validate findings. Results: We show for the first time that OGT inhibition leads to a rapid loss of O-GlcNAc chromatin mark. O-GlcNAc ChIP-seq regions overlap with super-enhancers (SE) and MYC binding sites. OGT inhibition leads to down-regulation of SE-dependent genes. We establish the first O-GlcNAc chromatin consensus motif, which we use as a bait for mass spectrometry. By combining the proteomic data from oligonucleotide enrichment with O-GlcNAc and MYC ChIP-mass spectrometry, we identify host cell factor 1 (HCF-1) as an interaction partner of MYC. Inhibition of OGT disrupts this interaction and compromises MYC's ability to confer androgen-independent proliferation to prostate cancer cells. We show that OGT is required for MYC-mediated stabilization of mitotic proteins, including Cyclin B1, and/or the increased translation of their coding transcripts. This implies that increased expression of mRNA is not always required to achieve increased protein expression and confer aggressive phenotype. Indeed, high expression of Cyclin B1 protein has strong predictive value in prostate cancer patients (p=0.000014) while mRNA does not. Conclusions: OGT promotes SE-dependent gene expression. OGT activity is required for the interaction between MYC and HCF-1 and expression of MYC-regulated mitotic proteins. These features render OGT essential for the androgen-independent, MYC-driven proliferation of prostate cancer cells. Androgen-independency is the major mechanism of prostate cancer progression, and our study identifies OGT as an essential mediator in this process.


Asunto(s)
Proliferación Celular , N-Acetilglucosaminiltransferasas/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Animales , Línea Celular Tumoral , Células Cultivadas , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Ciclina B1/genética , Ciclina B1/metabolismo , Elementos de Facilitación Genéticos , Factor C1 de la Célula Huésped/genética , Factor C1 de la Célula Huésped/metabolismo , Humanos , Masculino , Ratones , N-Acetilglucosaminiltransferasas/genética , Neoplasias de la Próstata/genética , Activación Transcripcional
12.
Nucleic Acids Res ; 47(11): 5792-5808, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-31049581

RESUMEN

HCF-2 is a member of the host-cell-factor protein family, which arose in early vertebrate evolution as a result of gene duplication. Whereas its paralog, HCF-1, is known to act as a versatile chromatin-associated protein required for cell proliferation and differentiation, much less is known about HCF-2. Here, we show that HCF-2 is broadly present in human and mouse cells, and possesses activities distinct from HCF-1. Unlike HCF-1, which is excluded from nucleoli, HCF-2 is nucleolar-an activity conferred by one and a half C-terminal Fibronectin type 3 repeats and inhibited by the HCF-1 nuclear localization signal. Elevated HCF-2 synthesis in HEK-293 cells results in phenotypes reminiscent of HCF-1-depleted cells, including inhibition of cell proliferation and mitotic defects. Furthermore, increased HCF-2 levels in HEK-293 cells lead to inhibition of cell proliferation and metabolism gene-expression programs with parallel activation of differentiation and morphogenesis gene-expression programs. Thus, the HCF ancestor appears to have evolved into a small two-member protein family possessing contrasting nuclear versus nucleolar localization, and cell proliferation and differentiation functions.


Asunto(s)
Perfilación de la Expresión Génica , Factor C1 de la Célula Huésped/fisiología , Factores de Transcripción/fisiología , Animales , Línea Celular , Línea Celular Tumoral , Nucléolo Celular , Proliferación Celular , Cromatina/química , Fibroblastos/metabolismo , Duplicación de Gen , Células HEK293 , Células HeLa , Factor C1 de la Célula Huésped/metabolismo , Humanos , Células Jurkat , Células MCF-7 , Ratones , Mitosis , Señales de Localización Nuclear/metabolismo , Fenotipo , Plásmidos/metabolismo , ARN Interferente Pequeño/metabolismo , Factores de Transcripción/metabolismo
13.
Cell ; 177(3): 722-736.e22, 2019 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-30955890

RESUMEN

Insulin receptor (IR) signaling is central to normal metabolic control and dysregulated in prevalent chronic diseases. IR binds insulin at the cell surface and transduces rapid signaling via cytoplasmic kinases. However, mechanisms mediating long-term effects of insulin remain unclear. Here, we show that IR associates with RNA polymerase II in the nucleus, with striking enrichment at promoters genome-wide. The target genes were highly enriched for insulin-related functions including lipid metabolism and protein synthesis and diseases including diabetes, neurodegeneration, and cancer. IR chromatin binding was increased by insulin and impaired in an insulin-resistant disease model. Promoter binding by IR was mediated by coregulator host cell factor-1 (HCF-1) and transcription factors, revealing an HCF-1-dependent pathway for gene regulation by insulin. These results show that IR interacts with transcriptional machinery at promoters and identify a pathway regulating genes linked to insulin's effects in physiology and disease.


Asunto(s)
Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Receptor de Insulina/metabolismo , Animales , Línea Celular Tumoral , Cromatina/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Factor C1 de la Célula Huésped/antagonistas & inhibidores , Factor C1 de la Célula Huésped/genética , Factor C1 de la Célula Huésped/metabolismo , Humanos , Insulina/metabolismo , Insulina/farmacología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Unión Proteica , Subunidades de Proteína/metabolismo , Interferencia de ARN , ARN Polimerasa II/metabolismo , ARN Interferente Pequeño/metabolismo , Receptor de Insulina/química , Transducción de Señal/efectos de los fármacos
14.
Cell Death Dis ; 9(11): 1118, 2018 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-30389914

RESUMEN

Although additional sex combs-like 1 (ASXL1) has been extensively described in hematologic malignancies, little is known about the molecular role of ASXL1 in organ development. Here, we show that Asxl1 ablation in mice results in postnatal lethality due to cyanosis, a respiratory failure. This lung defect is likely caused by higher proliferative potential and reduced expression of surfactant proteins, leading to reduced air space and defective lung maturation. By microarray analysis, we identified E2F1-responsive genes, including Nmyc, as targets repressed by Asxl1. Nmyc and Asxl1 are reciprocally expressed during the fetal development of normal mouse lungs, whereas Nmyc downregulation is impaired in Asxl1-deficient lungs. Together with E2F1 and ASXL1, host cell factor 1 (HCF-1), purified as an Asxl1-bound protein, is recruited to the E2F1-binding site of the Nmyc promoter. The interaction occurs between the C-terminal region of Asxl1 and the N-terminal Kelch domain of HCF-1. Trimethylation (me3) of histone H3 lysine 27 (H3K27) is enriched in the Nmyc promoter upon Asxl1 overexpression, whereas it is downregulated in Asxl1-deleted lung and -depleted A549 cells, similar to H3K9me3, another repressive histone marker. Overall, these findings suggest that Asxl1 modulates proliferation of lung epithelial cells via the epigenetic repression of Nmyc expression, deficiency of which may cause hyperplasia, leading to dyspnea.


Asunto(s)
Factor de Transcripción E2F1/genética , Represión Epigenética , Células Epiteliales/metabolismo , Pulmón/metabolismo , Proteína Proto-Oncogénica N-Myc/genética , Proteínas Represoras/genética , Insuficiencia Respiratoria/genética , Células A549 , Animales , Factor de Transcripción E2F1/metabolismo , Embrión de Mamíferos , Células Epiteliales/patología , Feto , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Genes Letales , Células HEK293 , Histonas/genética , Histonas/metabolismo , Factor C1 de la Célula Huésped/genética , Factor C1 de la Célula Huésped/metabolismo , Humanos , Pulmón/crecimiento & desarrollo , Pulmón/patología , Ratones , Ratones Noqueados , Proteína Proto-Oncogénica N-Myc/metabolismo , Organogénesis/genética , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Represoras/deficiencia , Insuficiencia Respiratoria/metabolismo , Insuficiencia Respiratoria/patología , Transducción de Señal
15.
Mol Cell Biol ; 38(17)2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29941490

RESUMEN

Cancer cells often heavily depend on the ubiquitin-proteasome system (UPS) for their growth and survival. Irrespective of their strong dependence on the proteasome activity, cancer cells, except for multiple myeloma, are mostly resistant to proteasome inhibitors. A major cause of this resistance is the proteasome bounce-back response mediated by NRF1, a transcription factor that coordinately activates proteasome subunit genes. To identify new targets for efficient suppression of UPS, we explored, using immunoprecipitation and mass spectrometry, the possible existence of nuclear proteins that cooperate with NRF1 and identified O-linked N-acetylglucosamine transferase (OGT) and host cell factor C1 (HCF-1) as two proteins capable of forming a complex with NRF1. O-GlcNAcylation catalyzed by OGT was essential for NRF1 stabilization and consequent upregulation of proteasome subunit genes. Meta-analysis of breast and colorectal cancers revealed positive correlations in the relative protein abundance of OGT and proteasome subunits. OGT inhibition was effective at sensitizing cancer cells to a proteasome inhibitor both in culture cells and a xenograft mouse model. Since active O-GlcNAcylation is a feature of cancer metabolism, our study has clarified a novel linkage between cancer metabolism and UPS function and added a new regulatory axis to the regulation of the proteasome activity.


Asunto(s)
Factor 1 Relacionado con NF-E2/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Inhibidores de Proteasoma/farmacología , Acetilglucosamina/metabolismo , Animales , Línea Celular Tumoral , Resistencia a Antineoplásicos/fisiología , Femenino , Glicosilación , Células HEK293 , Células HeLa , Factor C1 de la Célula Huésped/química , Factor C1 de la Célula Huésped/genética , Factor C1 de la Célula Huésped/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , N-Acetilglucosaminiltransferasas/antagonistas & inhibidores , N-Acetilglucosaminiltransferasas/genética , N-Acetilglucosaminiltransferasas/metabolismo , Factor 1 Relacionado con NF-E2/química , Factor 1 Relacionado con NF-E2/genética , Neoplasias/genética , Factor Nuclear 1 de Respiración , Regiones Promotoras Genéticas , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , Ubiquitina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas con Repetición de beta-Transducina/química , Proteínas con Repetición de beta-Transducina/metabolismo
16.
J Virol ; 92(17)2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29899098

RESUMEN

Following productive infection, bovine herpesvirus 1 (BoHV-1) establishes latency in sensory neurons. As in other alphaherpesviruses, expression of BoHV-1 immediate early (IE) genes is regulated by an enhancer complex containing the viral IE activator VP16, the cellular transcription factor Oct-1, and transcriptional coactivator HCF-1, which is assembled on an IE enhancer core element (TAATGARAT). Expression of the IE transcription unit that encodes the viral IE activators bICP0 and bICP4 may also be induced by the activated glucocorticoid receptor (GR) via two glucocorticoid response elements (GREs) located upstream of the enhancer core. Strikingly, lytic infection and reactivation from latency are consistently enhanced by glucocorticoid treatment in vivo As the coactivator HCF-1 is essential for IE gene expression of alphaherpesviruses and recruited by multiple transcription factors, we tested whether HCF-1 is required for glucocorticoid-induced IE gene expression. Depletion of HCF-1 reduced GR-mediated activation of the IE promoter in mouse neuroblastoma cells (Neuro-2A). More importantly, HCF-1-mediated GR activation of the promoter was dependent on the presence of GRE sites but independent of the TAATGARAT enhancer core element. HCF-1 was also recruited to the GRE region of a promoter lacking the enhancer core, consistent with a direct role of the coactivator in mediating GR-induced transcription. Similarly, during productive lytic infection, HCF-1 and GR occupied the IE region containing the GREs. These studies indicate HCF-1 is critical for GR activation of the viral IE genes and suggests that glucocorticoid induction of viral reactivation proceeds via an HCF-1-GR mechanism in the absence of the viral IE activator VP16.IMPORTANCE BoHV-1 transcription is rapidly activated during stress-induced reactivation from latency. The immediate early transcription unit 1 (IEtu1) promoter is regulated by the GR via two GREs. The IEtu1 promoter regulates expression of two viral transcriptional regulatory proteins, infected cell proteins 0 and 4 (bICP0 and bICP4), and thus must be stimulated during reactivation. This study demonstrates that activation of the IEtu1 promoter by the synthetic corticosteroid dexamethasone requires HCF-1. Interestingly, the GRE sites, but not the IE enhancer core element (TAATGARAT), were required for HCF-1-mediated GR promoter activation. The GR and HCF-1 were recruited to the IEtu1 promoter in transfected and infected cells. Collectively, these studies indicate that HCF-1 is critical for GR activation of the viral IE genes and suggest that an HCF-1-GR complex can stimulate the IEtu1 promoter in the absence of the viral IE activator VP16.


Asunto(s)
Regulación Viral de la Expresión Génica , Genes Inmediatos-Precoces , Glucocorticoides/metabolismo , Herpesvirus Bovino 1/fisiología , Factor C1 de la Célula Huésped/metabolismo , Receptores de Glucocorticoides/metabolismo , Transcripción Genética , Animales , Línea Celular , Técnicas de Silenciamiento del Gen , Factor C1 de la Célula Huésped/genética , Ratones , Neuronas/virología
17.
Mem. Inst. Oswaldo Cruz ; 113(10): e180311, 2018. graf
Artículo en Inglés | LILACS | ID: biblio-955107

RESUMEN

BACKGROUND Scedosporium apiospermum is a ubiquitous, emerging and multidrug-resistant fungal pathogen with still rather unknown virulence mechanisms. OBJECTIVES/METHODS The cellular basis of the in vitro interaction between fungi and host cells/tissues is the determinant factor for the development of a successful in vivo infection. Herein, we evaluated the interaction of S. apiospermum conidia with lung epithelial (A549), lung fibroblast (MRC-5) and RAW 264.7 macrophages by light and scanning/transmission electron microscopy. FINDINGS After 4 h of fungi-host cell contact, the percentage of infected mammalian cells and the number of fungi per infected cell was measured by light microscopy, and the following association indexes were calculated for A549, MRC-5 and macrophage cells: 73.2 ± 25.9, 69.7 ± 22.5 and 59.7 ± 11.1, respectively. Both conidia and germinated conidia were regularly observed interacting with the evaluated cells, with a higher prevalence of non-germinated conidia. Interestingly, nests of germinated conidia were evidenced at the surface of lung cells by scanning electron microscopy. Some germination projections and hyphae were seen penetrating/evading the mammalian cells. Furthermore, internalised conidia were seen within vacuoles as visualised by transmission electron microscopy. MAIN CONCLUSIONS The present study contributes to a better understanding of S. apiospermum pathogenesis by demonstrating the first steps of the infection process of this opportunistic fungus.


Asunto(s)
Humanos , Scedosporium , Macrófagos , Carcinoma de Pulmón de Células no Pequeñas , Factor C1 de la Célula Huésped
18.
Cell Host Microbe ; 21(4): 507-517.e5, 2017 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-28407486

RESUMEN

The cellular transcriptional coactivator HCF-1 is required for initiation of herpes simplex virus (HSV) lytic infection and for reactivation from latency in sensory neurons. HCF-1 stabilizes the viral Immediate Early (IE) gene enhancer complex and mediates chromatin transitions to promote IE transcription initiation. In infected cells, HCF-1 was also found to be associated with a network of transcription elongation components including the super elongation complex (SEC). IE genes exhibit characteristics of genes controlled by transcriptional elongation, and the SEC-P-TEFb complex is specifically required to drive the levels of productive IE mRNAs. Significantly, compounds that enhance the levels of SEC-P-TEFb also potently stimulated HSV reactivation from latency both in a sensory ganglia model system and in vivo. Thus, transcriptional elongation of HSV IE genes is a key limiting parameter governing both the initiation of HSV infection and reactivation of latent genomes.


Asunto(s)
Regulación Viral de la Expresión Génica , Genes Inmediatos-Precoces , Simplexvirus/fisiología , Elongación de la Transcripción Genética , Activación Viral , Animales , Línea Celular , Células Epiteliales/virología , Ganglios Sensoriales/virología , Factor C1 de la Célula Huésped/metabolismo , Humanos , Ratones , Simplexvirus/genética , Factores de Transcripción/metabolismo
19.
Biochem Biophys Res Commun ; 483(2): 793-802, 2017 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-27845045

RESUMEN

High-risk human papilloma virus (HPV) 16/18 infections are often found in lung cancer. The cellular mechanisms involved in the metastatic spread of HPV-infected cervical cancer cells remain largely elusive. High O-linked-N-acetylglucosamine (O-GlcNAc) modification has also been observed in lung cancer. In the present study, we assessed the relationship between O-GlcNAc transferase (OGT) and HPV 16/18 E6/E7, or C-X-C chemokine receptor type 4 (CXCR4), in HeLa cells and in lungs of xenografted mice. Depleting OGT with an OGT-specific shRNA significantly decreased levels of E6 and E7 oncoproteins in HeLa cells and xenograft tumors, and reduced tumor formation in vivo. Western blotting and immunofluorescence analysis showed significantly decreased expression levels of E6, E7, and HCF-1 in the lungs of xenografted mice treated with an OGT-specific shRNA compared to those treated with non-targeting shRNA. Additionally, levels of E7 or OGT co-localized with Ki-67 were significantly decreased in the lungs of xenografted mice treated with OGT-specific shRNA compared to those treated with non-targeting shRNA. Moreover, levels of CXCR4 were significantly decreased in HeLa cells and in the lungs of xenografted mice treated with OGT-specific shRNA compared to those treated with non-targeting shRNA; this may be related to reduced adhesion or invasion of circulating HPV-positive tumor cells. These findings provide novel evidence that OGT functions in metastatic spread of HPV E6/E7-positive tumor cells to the lungs through E6/E7, HCF-1 and CXCR4, suggesting OGT might be a therapeutic target for HPV-positive lung cancer.


Asunto(s)
Papillomavirus Humano 16/patogenicidad , Papillomavirus Humano 18/patogenicidad , Neoplasias Pulmonares/etiología , N-Acetilglucosaminiltransferasas/metabolismo , Infecciones por Papillomavirus/etiología , Animales , Proteínas de Unión al ADN/metabolismo , Células HeLa , Xenoinjertos , Factor C1 de la Célula Huésped/metabolismo , Humanos , Pulmón/metabolismo , Pulmón/virología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Ratones , Ratones Desnudos , N-Acetilglucosaminiltransferasas/antagonistas & inhibidores , N-Acetilglucosaminiltransferasas/genética , Proteínas Oncogénicas Virales/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Infecciones por Papillomavirus/metabolismo , Infecciones por Papillomavirus/virología , ARN Interferente Pequeño/genética , Receptores CXCR4/metabolismo , Proteínas Represoras/metabolismo
20.
J Am Heart Assoc ; 5(12)2016 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-27919931

RESUMEN

BACKGROUND: Cytochrome oxidase (COX) IV complex regulates energy production in mitochondria. Impaired COX gene expression is related to obesity and type 2 diabetes mellitus, but whether it is directly related to the incidence of cardiovascular events is unknown. We investigated whether COX gene expression in monocytes is predictive for cardiovascular events in coronary artery disease patients. To avoid monocyte isolation from fresh blood, we then aimed to validate our findings in monocyte-derived microvesicles isolated from plasma. METHODS AND RESULTS: We enrolled 142 consecutive patients undergoing diagnostic coronary angiography between June 2010 and January 2011 and followed 67 patients with stable coronary artery disease prospectively for at least 3 years. Twenty-two patients experienced a new cardiovascular event (32.8%). Circulating CD14+ monocytes and microvesicles were isolated with magnetic beads, and COX mRNA levels were measured with quantitative polymerase chain reaction, after normalization with 5 validated house-keeping genes. Patients in the lowest tertile of mitochondrial cytochrome oxidase, subunit I (MT-COI) in monocytes at baseline had a higher risk for developing a new event after adjusting for age, sex, (ex)smoking, body mass index, blood pressure, diabetes mellitus, low-density lipoprotein- and high-density lipoprotein-cholesterol, triglycerides, high-sensitivity C-reactive protein, interleukin-6, and number of diseased vessels (harzard ratio [HR], 3.95; 95% CI, 1.63-9.57). Patients in the lowest tertile of MT-COI in monocyte-specific microvesicles had also a higher risk of developing a new event (adjusted HR, 5.00; 95% CI, 1.77-14). CONCLUSIONS: In the current blinded study, low MT-COI in monocytes of coronary artery disease patients identifies a population at risk for new cardiovascular events. For the first time, we show that signatures in monocyte-specific microvesicles in plasma have similar predictive properties.


Asunto(s)
Micropartículas Derivadas de Células/metabolismo , Enfermedad de la Arteria Coronaria/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Monocitos/metabolismo , Enfermedades Cardiovasculares/mortalidad , Angiografía Coronaria , Enfermedad de la Arteria Coronaria/diagnóstico por imagen , Femenino , Factor C1 de la Célula Huésped , Humanos , Masculino , Persona de Mediana Edad , Mitocondrias/metabolismo , Isquemia Miocárdica/epidemiología , Isquemia Miocárdica/metabolismo , Pronóstico , Modelos de Riesgos Proporcionales , Estudios Prospectivos , ARN Mensajero , Reacción en Cadena en Tiempo Real de la Polimerasa , Accidente Cerebrovascular/epidemiología , Accidente Cerebrovascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA