Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Biochem Pharmacol ; 194: 114814, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34688635

RESUMEN

Colorectal cancer (CRC) remains the most frequently diagnosed malignancy and also a major contributor to cancer-related death throughout the world. Here, we first revealed the role of histone lysine-specific demethylase 5D (KDM5D) in CRC in males. KDM5D expression in tumor and adjacent tissues of male CRC patients was investigated using immunohistochemistry and RT-qPCR, and the correlation between its expression and patients' prognosis was analyzed. Downregulation of KDM5D in CRC patients was associated with poor prognoses. Overexpression of KDM5D significantly inhibited the growth and metastasis of CRC in vitro and in vivo. The downstream mechanism of KDM5D in CRC was investigated using bioinformatics analysis, and the regulatory relationship was confirmed by ChIP-qPCR and luciferase reporter assays. KDM5D suppressed E2F1 expression by mediating H3K4me3 demethylation. E2F1, highly expressed in CRC, promoted the expression of FKBP4 at the transcriptional level by binding to the FKBP4 promoter. Finally, rescue experiments revealed that overexpression of FKBP4 significantly reversed the inhibitory effect of KDM5D on CRC growth and metastasis. Collectively, KDM5D exerted an anti-tumor and anti-metastatic in CRC through demethylation in E2F1 and suppression of FKBP4 transcription, which might represent a novel target in CRC treatment in male.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Factor de Transcripción E2F1/biosíntesis , Regulación Neoplásica de la Expresión Génica , Histona Demetilasas/biosíntesis , Antígenos de Histocompatibilidad Menor/biosíntesis , Proteínas de Unión a Tacrolimus/biosíntesis , Activación Transcripcional/fisiología , Anciano , Animales , Neoplasias Colorrectales/genética , Bases de Datos Genéticas , Factor de Transcripción E2F1/antagonistas & inhibidores , Células HCT116 , Histona Demetilasas/genética , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Antígenos de Histocompatibilidad Menor/genética , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores , Proteínas de Unión a Tacrolimus/genética , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
2.
Theranostics ; 11(16): 7970-7983, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34335974

RESUMEN

The novel ß-coronavirus, SARS-CoV-2, the causative agent of coronavirus disease 2019 (COVID-19), has infected more than 177 million people and resulted in 3.84 million death worldwide. Recent epidemiological studies suggested that some environmental factors, such as air pollution, might be the important contributors to the mortality of COVID-19. However, how environmental exposure enhances the severity of COVID-19 remains to be fully understood. In the present report, we provided evidence showing that mdig, a previously reported environmentally-induced oncogene that antagonizes repressive trimethylation of histone proteins, is an important regulator for SARS-CoV-2 receptors neuropilin-1 (NRP1) and NRP2, cathepsins, glycan metabolism and inflammation, key determinants for viral infection and cytokine storm of the patients. Depletion of mdig in bronchial epithelial cells by CRISPR-Cas-9 gene editing resulted in a decreased expression of NRP1, NRP2, cathepsins, and genes involved in protein glycosylation and inflammation, largely due to a substantial enrichment of lysine 9 and/or lysine 27 trimethylation of histone H3 (H3K9me3/H3K27me3) on these genes as determined by ChIP-seq. Meanwhile, we also validated that environmental factor arsenic is able to induce mdig, NRP1 and NRP2, and genetic disruption of mdig lowered expression of NRP1 and NRP2. Furthermore, mdig may coordinate with the Neanderthal variants linked to an elevated mortality of COVID-19. These data, thus, suggest that mdig is a key mediator for the severity of COVID-19 in response to environmental exposure and targeting mdig may be the one of the effective strategies in ameliorating the symptom and reducing the mortality of COVID-19.


Asunto(s)
COVID-19/metabolismo , COVID-19/virología , Dioxigenasas/metabolismo , Histona Demetilasas/metabolismo , Neuropilina-1/metabolismo , Proteínas Nucleares/metabolismo , Polisacáridos/metabolismo , SARS-CoV-2/metabolismo , Células Epiteliales Alveolares/metabolismo , Animales , COVID-19/epidemiología , Catepsinas/metabolismo , Línea Celular , Células Cultivadas , Dioxigenasas/biosíntesis , Dioxigenasas/genética , Exposición a Riesgos Ambientales , Histona Demetilasas/biosíntesis , Histona Demetilasas/genética , Histonas/metabolismo , Humanos , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Pandemias , Ratas , SARS-CoV-2/patogenicidad , Glicoproteína de la Espiga del Coronavirus/metabolismo
3.
JCI Insight ; 6(17)2021 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-34314389

RESUMEN

Mitochondrial biogenesis and function are controlled by anterograde regulatory pathways involving more than 1000 nuclear-encoded proteins. Transcriptional networks controlling the nuclear-encoded mitochondrial genes remain to be fully elucidated. Here, we show that histone demethylase LSD1 KO from adult mouse liver (LSD1-LKO) reduces the expression of one-third of all nuclear-encoded mitochondrial genes and decreases mitochondrial biogenesis and function. LSD1-modulated histone methylation epigenetically regulates nuclear-encoded mitochondrial genes. Furthermore, LSD1 regulates gene expression and protein methylation of nicotinamide mononucleotide adenylyltransferase 1 (NMNAT1), which controls the final step of NAD+ synthesis and limits NAD+ availability in the nucleus. Lsd1 KO reduces NAD+-dependent SIRT1 and SIRT7 deacetylase activity, leading to hyperacetylation and hypofunctioning of GABPß and PGC-1α, the major transcriptional factor/cofactor for nuclear-encoded mitochondrial genes. Despite the reduced mitochondrial function in the liver, LSD1-LKO mice are protected from diet-induced hepatic steatosis and glucose intolerance, partially due to induction of hepatokine FGF21. Thus, LSD1 orchestrates a core regulatory network involving epigenetic modifications and NAD+ synthesis to control mitochondrial function and hepatokine production.


Asunto(s)
Hígado Graso/genética , Factores de Crecimiento de Fibroblastos/genética , Regulación de la Expresión Génica , Genes Mitocondriales/genética , Histona Demetilasas/genética , Hígado/metabolismo , ARN/genética , Animales , Células Cultivadas , Epigénesis Genética , Hígado Graso/metabolismo , Hígado Graso/patología , Factores de Crecimiento de Fibroblastos/biosíntesis , Histona Demetilasas/biosíntesis , Hígado/patología , Ratones , Transducción de Señal
4.
Exp Hematol ; 99: 1-11, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34029637

RESUMEN

Relapse is associated with therapy resistance and is a major cause of death in acute myeloid leukemia (AML). It is thought to result from the accretion of therapy-refractory leukemic stem cells. Genetic and transcriptional changes that are recurrently gained at relapse are likely to contribute to the increased stemness and decreased therapy responsiveness at this disease stage. Despite the recent approval of several targeted drugs, chemotherapy with cytosine arabinoside and anthracyclines is still the mainstay of AML therapy. Accordingly, a number of studies have investigated genetic and gene expression changes between diagnosis and relapse of patients subjected to such treatment. Genetic alterations recurrently acquired at relapse were identified, but were restricted to small proportions of patients, and their functional characterization is still largely pending. In contrast, the expression of a substantial number of genes was altered consistently between diagnosis and recurrence of AML. Recent studies corroborated the roles of the upregulation of SOCS2 and CALCRL and of the downregulation of MTSS1 and KDM6A in therapy resistance and/or stemness of AML. These findings spur the assumption that functional investigations of genes consistently altered at recurrence of AML have the potential to promote the development of novel targeted drugs that may help to improve the outcome of this currently often fatal disease.


Asunto(s)
Proteína Similar al Receptor de Calcitonina/biosíntesis , Regulación Leucémica de la Expresión Génica , Histona Demetilasas/biosíntesis , Leucemia Mieloide Aguda , Proteínas de Microfilamentos/biosíntesis , Proteínas de Neoplasias/biosíntesis , Células Madre Neoplásicas/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/biosíntesis , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/terapia , Recurrencia
5.
Cell Cycle ; 20(3): 298-307, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33475455

RESUMEN

Accumulating evidence has reported the role of microRNA (miR) in retinoblastoma (RB). Therefore, the objective was to discuss how miR-362-3p exerted its function in RB cell progression via regulating ubiquitin-specific protease 2 (USP22) and lysine-specific histone demethylase 1 (LSD1). MiR-362-3p, USP22 and LSD1 expression in RB cells and tissues were tested. The biological functions of RB cells were detected via over-expressing miR-362-3p and down-regulating USP22. The target relationship of USP22 and miR-362-3p as well as the interaction of USP22 and LSD1 in RB was verified. Down-regulated miR-362-3p and up-regulated USP22 and LSD1 were demonstrated in RB tissues and cells. Restoring miR-362-3p and depleting USP22 attenuated invasion, proliferation and migration, and facilitated apoptosis of RB cells. USP22 was a target gene of miR-362-3p. USP22 deubiquitinated LSD1 in RB. It is revealed that miR-362-3p targets USP22 and then restrains invasion, proliferation and migration while promotes apoptosis of RB via reducing LSD1 modified by deubiquitination.


Asunto(s)
Histona Demetilasas/biosíntesis , MicroARNs/biosíntesis , Neoplasias de la Retina/metabolismo , Retinoblastoma/metabolismo , Ubiquitina Tiolesterasa/biosíntesis , Línea Celular Tumoral , Proliferación Celular/fisiología , Marcación de Gen , Histona Demetilasas/genética , Humanos , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Neoplasias de la Retina/genética , Retinoblastoma/genética , Ubiquitina Tiolesterasa/genética , Ubiquitinación/fisiología
6.
Biochem J ; 478(4): 721-734, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33410908

RESUMEN

Osteoporosis is a prevalent systemic skeletal disorder entailing bone fragility and increased fracture risk, often emerging in post-menopausal life. Emerging evidence implicates the dysregulation of microRNAs (miRNAs or miRs) in the progression of osteoporosis. This study investigated the effect of miR-199a-3p on osteoporosis and its underlying mechanism. We first examplished an ovariectomized (OVX)-induced rat osteoporosis model, and then isolated mesenchymal stem cells (MSCs) from bone marrow of the model rats. The overexpression and knock down of miR-199a-3p were conducted in OVX rats and MSCs to verify the role of miR-199a-3p on MSC differentiation. Calcium nodules were measured using alizarin red S (ARS) staining. RT-qPCR and Western blot assay were performed to measure the expression of miR-199a-3p, Kdm3a and osteogenic differentiation-related markers in rat tissues and cells. The correlation between miR-199a-3p and Kdm3a was confirmed using dual-luciferase reporter assay. The enrichment of Kdm3a at the Erk2 and Klf2 promoter was assessed using chromatin immunoprecipitation (ChIP) assay. Isolated MSCs were positive for CD29, CD44, CD90, and CD45, suggesting successful isolation of MSCs. There was increased expression of miR-199a-3p and inhibited osteogenic differentiation in OVX rats. Kdm3a was negatively targeted by miR-199a-3p. Our results also demonstrated that Kdm3a elevated the expression of Erk2 and Erk2 by promoting Erk2 and Klf2 demethylation, which further contributed to osteogenic differentiation. Overall, our results revealed a regulatory network of miR-199a-3p in osteogenic differentiation, highlighting miR-199a-3p as a potential target for therapeutic interventions in osteoporosis.


Asunto(s)
Histona Demetilasas/genética , Células Madre Mesenquimatosas/metabolismo , MicroARNs/genética , Osteogénesis/genética , Osteoporosis/genética , Animales , Antígenos CD/biosíntesis , Azepinas/farmacología , Azepinas/uso terapéutico , Huesos/patología , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Femenino , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Redes Reguladoras de Genes , Genes Reporteros , Histona Demetilasas/antagonistas & inhibidores , Histona Demetilasas/biosíntesis , Humanos , Factores de Transcripción de Tipo Kruppel/biosíntesis , Factores de Transcripción de Tipo Kruppel/genética , MicroARNs/biosíntesis , Proteína Quinasa 1 Activada por Mitógenos/biosíntesis , Proteína Quinasa 1 Activada por Mitógenos/genética , Osteoporosis/metabolismo , Osteoporosis/patología , Osteoporosis Posmenopáusica/genética , Ovariectomía , Quinazolinas/farmacología , Quinazolinas/uso terapéutico , Ratas , Ratas Sprague-Dawley
7.
Acta Biochim Biophys Sin (Shanghai) ; 52(10): 1093-1101, 2020 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-33085743

RESUMEN

Previously, Nucleolar protein 66 (NO66) was reported to be closely associated with alcohol exposure-induced injury. However, the role of NO66 in alcohol-induced cytotoxicity remains unclear. In this study, we explored the potential effect and mechanism of NO66 on ethanol-induced apoptosis in human AC16 cardiomyocytes. The AC16 cell lines with NO66 and phosphatase and tensin homolog (PTEN) overexpression were constructed. Cell counting kit-8 (CCK-8), lactate dehydrogenase (LDH) assay, Annexin V-FITC/PI staining, and flow cytometry were used to evaluate the cell viability, membrane damage, and apoptosis, respectively. Quantitative real-time PCR (qRT-PCR) and western blot analysis were applied to measure mRNA and protein expression. The results showed that acute ethanol exposure markedly augmented cytotoxicity and reduced NO66 level in AC16 cardiomyocytes. Overexpression of NO66 partially reversed ethanol-induced apoptosis. NO66 upregulation reversed the decrease in phosphorylation of protein kinase B (Akt) and B-cell lymphoma-2/Bcl-2-associated x (Bcl-2/Bax) ratio and the increase in PTEN, p53, and caspase-3 activity induced by ethanol treatment. Meanwhile, the application of PI3K inhibitor (LY294002) and PTEN overexpression attenuated the inhibition efficiency of NO66 on cell apoptosis. In addition, PTEN overexpression weakened the effect of NO66 on PI3K/Akt activation, without affecting the level of NO66. Our data suggested that NO66 overexpression might play an anti-apoptotic role in ethanol-induced cell injury via reducing PTEN and upregulating the PI3K/Akt pathway.


Asunto(s)
Apoptosis/genética , Dioxigenasas/biosíntesis , Dioxigenasas/genética , Histona Demetilasas/biosíntesis , Histona Demetilasas/genética , Miocitos Cardíacos/metabolismo , Transducción de Señal/efectos de los fármacos , Apoptosis/efectos de los fármacos , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Etanol/toxicidad , Humanos , Miocitos Cardíacos/efectos de los fármacos , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal/genética , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba , Proteína X Asociada a bcl-2/metabolismo
8.
Mediators Inflamm ; 2020: 4092762, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32908449

RESUMEN

MicroRNA-155 (miRNA-155) is abundant in fibroblast-like synoviocytes (FLS) in rheumatoid arthritis (RA). Lysine-specific demethylase 1 (LSD1) has been found that it can ameliorate the severity of RA. Tumor necrosis factor-alpha, interleukin-1 beta, and interleukin-6 are key proinflammatory cytokines implicated in the pathogenesis of RA. In our study, we investigated whether miRNA-155 participates in the expression of LSD1 and proinflammatory cytokines in rheumatoid synovial cells. First of all, flow cytometry and cell counting kit-8 analysis were employed to explore the apoptosis and proliferation of FLS, respectively. Subsequently, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was applied to probe into the level of miRNA-155 in FLS when stimulated by miRNA-155 molecules. Moreover, RT-qPCR was used to explore the relative LSD1 miRNA expression in FLS when stimulated by miRNA-155 molecules, and Western blot and immunofluorescence assay were applied to probe into the expression level of LSD1. Finally, enzyme-linked immunosorbent assay was employed to analyze the secreting level of proinflammatory cytokines in FLS when stimulated by miRNA-155 molecules. RA-FLS showed a higher apoptosis rate than normal FLS. The cell proliferation of both HFLS and MH7A cells was promoted by miRNA-155 upregulation. Meanwhile, the expression of LSD1 and proinflammatory cytokines in the FLS of RA was also changed by miRNA-155 regulation. In conclusion, miRNA-155 participates in the expression of LSD1 and proinflammatory cytokines in rheumatoid synovial cells. These findings imply a potential function and interaction of miRNA-155 and LSD1.


Asunto(s)
Citocinas/metabolismo , Histona Demetilasas/biosíntesis , MicroARNs/biosíntesis , Membrana Sinovial/metabolismo , Sinoviocitos/metabolismo , Apoptosis , Artritis Reumatoide/metabolismo , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Fibroblastos/metabolismo , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Inflamación , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba
9.
J Clin Invest ; 130(11): 5688-5702, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-32701509

RESUMEN

Males and females differ in body composition and fat distribution. Using a mouse model that segregates gonadal sex (ovaries and testes) from chromosomal sex (XX and XY), we showed that XX chromosome complement in combination with a high-fat diet led to enhanced weight gain in the presence of male or female gonads. We identified the genomic dosage of Kdm5c, an X chromosome gene that escapes X chromosome inactivation, as a determinant of the X chromosome effect on adiposity. Modulating Kdm5c gene dosage in XX female mice to levels that are normally present in males resulted in reduced body weight, fat content, and food intake to a degree similar to that seen with altering the entire X chromosome dosage. In cultured preadipocytes, the levels of KDM5C histone demethylase influenced chromatin accessibility (ATAC-Seq), gene expression (RNA-Seq), and adipocyte differentiation. Both in vitro and in vivo, Kdm5c dosage influenced gene expression involved in extracellular matrix remodeling, which is critical for adipocyte differentiation and adipose tissue expansion. In humans, adipose tissue KDM5C mRNA levels and KDM5C genetic variants were associated with body mass. These studies demonstrate that the sex-dependent dosage of Kdm5c contributes to male/female differences in adipocyte biology and highlight X-escape genes as a critical component of female physiology.


Asunto(s)
Adipocitos/enzimología , Adiposidad , Dosificación de Gen , Regulación Enzimológica de la Expresión Génica , Histona Demetilasas , Caracteres Sexuales , Cromosoma X , Animales , Ensamble y Desensamble de Cromatina , Femenino , Histona Demetilasas/biosíntesis , Histona Demetilasas/genética , Humanos , Masculino , Ratones , Ratones Mutantes , Cromosoma X/genética , Cromosoma X/metabolismo
10.
Cell Death Dis ; 10(11): 832, 2019 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-31685800

RESUMEN

GATA3 has emerged as a prominent transcription factor required for maintaining mammary-gland homeostasis. GATA3 loss is associated with aggressive breast cancer development, but the mechanism by which breast cancer is affected by the loss of GATA3 function remains unclear. Here, we report that GATA3 expression is positively correlated with the expression of UTX, a histone H3K27 demethylase contained in the MLL4 methyltransferase complex, and that GATA3 recruits the chromatin-remodeling MLL4 complex and interacts directly with UTX, ASH2L, and RBBP5. Using RNA sequencing and chromatin immunoprecipitation and sequencing, we demonstrate that the GATA3/UTX complex synergistically regulates a cohort of genes including Dicer and UTX, which are critically involved in the epithelial-to-mesenchymal transition (EMT). Our results further show that the GATA3-UTX-Dicer axis inhibits EMT, invasion, and metastasis of breast cancer cells in vitro and the dissemination of breast cancer in vivo. Our study implicates the GATA3-UTX-Dicer axis in breast cancer metastasis and provides new mechanistic insights into the pathophysiological function of GATA3.


Asunto(s)
Neoplasias de la Mama/metabolismo , Factor de Transcripción GATA3/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Histona Demetilasas/biosíntesis , Proteínas de Neoplasias/metabolismo , Activación Transcripcional , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Femenino , Factor de Transcripción GATA3/genética , Histona Demetilasas/genética , Humanos , Células MCF-7 , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética
11.
J Clin Invest ; 129(8): 3121-3133, 2019 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-31232700

RESUMEN

Lumen integrity in vascularization requires fully differentiated endothelial cells (ECs). Here, we report that endothelial-mesenchymal transitions (EndMTs) emerged in ECs of cerebral arteriovenous malformation (AVMs) and caused disruption of the lumen or lumen disorder. We show that excessive Sry-box 2 (Sox2) signaling was responsible for the EndMTs in cerebral AVMs. EC-specific suppression of Sox2 normalized endothelial differentiation and lumen formation and improved the cerebral AVMs. Epigenetic studies showed that induction of Sox2 altered the cerebral-endothelial transcriptional landscape and identified jumonji domain-containing protein 5 (JMJD5) as a direct target of Sox2. Sox2 interacted with JMJD5 to induce EndMTs in cerebral ECs. Furthermore, we utilized a high-throughput system to identify the ß-adrenergic antagonist pronethalol as an inhibitor of Sox2 expression. Treatment with pronethalol stabilized endothelial differentiation and lumen formation, which limited the cerebral AVMs.


Asunto(s)
Diferenciación Celular , Células Endoteliales/metabolismo , Malformaciones Arteriovenosas Intracraneales/metabolismo , Factores de Transcripción SOXB1/biosíntesis , Animales , Células Endoteliales/patología , Etanolaminas/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Histona Demetilasas/biosíntesis , Histona Demetilasas/genética , Humanos , Malformaciones Arteriovenosas Intracraneales/tratamiento farmacológico , Malformaciones Arteriovenosas Intracraneales/genética , Malformaciones Arteriovenosas Intracraneales/patología , Histona Demetilasas con Dominio de Jumonji/biosíntesis , Histona Demetilasas con Dominio de Jumonji/genética , Ratones , Ratones Noqueados , Factores de Transcripción SOXB1/genética , Transcripción Genética/efectos de los fármacos
12.
FASEB J ; 33(3): 4638-4652, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30673507

RESUMEN

Aberrant epigenetic reprogramming is a major factor of developmental failure of cloned embryos. Histone H3 lysine 27 trimethylation (H3K27me3), a histone mark for transcriptional repression, plays important roles in mammalian embryonic development and induced pluripotent stem cell (iPSC) generation. The global loss of H3K27me3 marks may facilitate iPSC generation in mice and humans. However, the H3K27me3 level and its role in bovine somatic cell nuclear transfer (SCNT) reprogramming remain poorly understood. Here, we show that SCNT embryos exhibit global H3K27me3 hypermethylation from the 2- to 8-cell stage and that its removal by ectopically expressed H3K27me3 lysine demethylase (KDM)6A greatly improves nuclear reprogramming efficiency. In contrast, H3K27me3 reduction by H3K27me3 methylase enhancer of zeste 2 polycomb repressive complex knockdown or donor cell treatment with the enhancer of zeste 2 polycomb repressive complex-selective inhibitor GSK343 suppressed blastocyst formation by SCNT embryos. KDM6A overexpression enhanced the transcription of genes involved in cell adhesion and cellular metabolism and X-linked genes. Furthermore, we identified methyl-CpG-binding domain protein 3-like 2, which was reactivated by KDM6A, as a factor that is required for effective reprogramming in bovines. These results show that H3K27me3 functions as an epigenetic barrier and that KDM6A overexpression improves SCNT efficiency by facilitating transcriptional reprogramming.-Zhou, C., Wang, Y., Zhang, J., Su, J., An, Q., Liu, X., Zhang, M., Wang, Y., Liu, J., Zhang, Y. H3K27me3 is an epigenetic barrier while KDM6A overexpression improves nuclear reprogramming efficiency.


Asunto(s)
Bovinos/embriología , Reprogramación Celular/genética , Código de Histonas/genética , Histona Demetilasas/fisiología , Histonas/genética , Técnicas de Transferencia Nuclear , Animales , Blastómeros/metabolismo , Bovinos/genética , Clonación de Organismos , Desarrollo Embrionario/efectos de los fármacos , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Femenino , Fertilización In Vitro , Regulación del Desarrollo de la Expresión Génica , Histona Demetilasas/biosíntesis , Histona Demetilasas/genética , Histonas/metabolismo , Metilación , Microinyecciones , Mórula/citología , Mórula/metabolismo , Procesamiento Proteico-Postraduccional , Interferencia de ARN , ARN Mensajero/administración & dosificación , ARN Mensajero/análisis , ARN Mensajero/genética , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Proteínas Represoras/antagonistas & inhibidores , Análisis de Secuencia de ARN
13.
Oncogene ; 38(1): 17-32, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30072740

RESUMEN

During the evolution into castration or therapy resistance, prostate cancer cells reprogram the androgen responses to cope with the diminishing level of androgens, and undergo metabolic adaption to the nutritionally deprived and hypoxia conditions. AR (androgen receptor) and PKM2 (pyruvate kinase M2) have key roles in these processes. We report in this study, KDM8/JMJD5, a histone lysine demethylase/dioxygnase, exhibits a novel property as a dual coactivator of AR and PKM2 and as such, it is a potent inducer of castration and therapy resistance. Previously, we showed that KDM8 is involved in the regulation of cell cycle and tumor metabolism in breast cancer cells. Its role in prostate cancer has not been explored. Here, we show that KDM8's oncogenic properties in prostate cancer come from its direct interaction (1) with AR to affect androgen response and (2) with PKM2 to regulate tumor metabolism. The interaction with AR leads to the elevated expression of androgen response genes in androgen-deprived conditions. They include ANCCA/ATAD2 and EZH2, which are directly targeted by KDM8 and involved in sustaining the survival of the cells under hormone-deprived conditions. Notably, in enzalutamide-resistant cells, the expressions of both KDM8 and EZH2 are further elevated, so are neuroendocrine markers. Consequently, EZH2 inhibitors or KDM8 knockdown both resensitize the cells toward enzalutamide. In the cytosol, KDM8 associates with PKM2, the gatekeeper of pyruvate flux and translocates PKM2 into the nucleus, where the KDM8/PKM2 complex serves as a coactivator of HIF-1α to upregulate glycolytic genes. Using shRNA knockdown, we validate KDM8's functions as a regulator for both androgen-responsive and metabolic genes. KDM8 thus presents itself as an ideal therapeutic target for metabolic adaptation and castration-resistance of prostate cancer cells.


Asunto(s)
Adenocarcinoma/metabolismo , Proteínas Portadoras/metabolismo , Regulación Neoplásica de la Expresión Génica , Histona Demetilasas/fisiología , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/fisiología , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptores Androgénicos/metabolismo , Hormonas Tiroideas/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas/fisiología , Transporte Activo de Núcleo Celular , Adenocarcinoma/patología , Animales , Benzamidas , Línea Celular Tumoral , Proteínas de Unión al ADN/fisiología , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Proteína Potenciadora del Homólogo Zeste 2/biosíntesis , Proteína Potenciadora del Homólogo Zeste 2/genética , Técnicas de Silenciamiento del Gen , Glucólisis/genética , Xenoinjertos , Histona Demetilasas/biosíntesis , Histona Demetilasas/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Ratones Desnudos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Nitrilos , Feniltiohidantoína/análogos & derivados , Feniltiohidantoína/farmacología , Feniltiohidantoína/uso terapéutico , Neoplasias de la Próstata Resistentes a la Castración/patología , Mapeo de Interacción de Proteínas , ARN Interferente Pequeño/genética , Receptores Androgénicos/genética , Proteínas de Unión a Hormona Tiroide
14.
J Cancer Res Clin Oncol ; 144(11): 2127-2137, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30105513

RESUMEN

PURPOSE: Studies on the performance of epigenetic-based biomarkers in colorectal cancer (CRC) are scarce and have shown contradictory results. Thus, we sought to examine the prognostic value of histone-modifying enzymes (EZH2, SETDB1 and LSD-1) and histone post-translational marks (H3K27me3 and H3K9me3) in CRC. METHODS: A retrospective series of 207 CRC patients primarily submitted to surgery in a cancer center was included in this study. Clinicopathological data were retrieved. One representative paraffin block per case was selected for immunohistochemistry, including normal and CRC tissues whenever possible. The percentage of positive nuclear staining (digital image analysis) was used to classify patients into "low" and "high" expression groups for each biomarker. Correlations between immunoexpression levels, clinicopathological features and clinical outcomes [disease-specific (DSS) and disease-free (DFS) survival] were examined. Statistical significance was set at p < 0.05. RESULTS: CRC tissues showed significantly lower expression of SETDB1 and higher expression of the remainder four biomarkers compared to normal mucosa. High EZH2 expression correlated with disease recurrence/progression, whereas low LSD1 expression and high H3K9me3 and H3K27me3 expression were associated with more advanced stage. In multivariable analysis, cases with high LSD1 expression displayed significantly better DSS and DFS (HR 0.477, 95% confidence interval: 0.247-0.923) adjusted for pathological TNM stage. CONCLUSION: EZH2, SETDB1, LSD1, H3K9me3 and H3K27me3 expression are altered in CRC and may play a role in colorectal carcinogenesis. LSD1 immunoexpression levels independently predicted patient outcome in this cohort. Further investigations, using larger series, are warranted to confirm its potential clinical value and unravel underlying molecular mechanisms.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/biosíntesis , Histona Demetilasas/biosíntesis , Histonas/metabolismo , Proteína Metiltransferasas/biosíntesis , Anciano , Biomarcadores de Tumor/biosíntesis , Neoplasias Colorrectales/patología , Femenino , N-Metiltransferasa de Histona-Lisina , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Lisina/metabolismo , Masculino , Metilación , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos
15.
Exp Hematol ; 58: 44-51.e7, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29111428

RESUMEN

Acute myeloid leukemia (AML) remains an aggressive hematopoietic malignancy that is caused by proliferation of immature myeloid cells and is frequently characterized by perturbations in chromatin-modifying enzymes. Emerging evidence indicates that histone demethylases play a role in tumorigenesis. However, due to the complexity of this enormous family of histone-modifying enzymes, substrate redundancy, and context-specific roles, the contribution of each member remains ambiguous and targeting them remains challenging. Here, we analyzed expression of histone-3-lysine (H3K) demethylases and their cognate substrates in a cohort of de novo AML patients, which demonstrated that the expression of H3K27Me3/2-demethylases and selected members of H3K9Me3/2/1-demethylases are significantly increased in AML. KDM6 upregulation is associated with a global decrease in H3K27Me3 level. Importantly, our data show that pharmacological inhibition of H3K27Me3/2-demethylases or H3K9Me3/2-demethylases, either alone or in combination, could be considered an interesting molecular therapeutic modality in human AML independent of its subtype.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Histona Demetilasas , Histona Demetilasas con Dominio de Jumonji , Leucemia Mieloide Aguda , Proteínas de Neoplasias , Proteínas Nucleares , Línea Celular Tumoral , Histona Demetilasas/antagonistas & inhibidores , Histona Demetilasas/biosíntesis , Humanos , Histona Demetilasas con Dominio de Jumonji/antagonistas & inhibidores , Histona Demetilasas con Dominio de Jumonji/biosíntesis , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/patología , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/biosíntesis , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/biosíntesis
16.
Cancer Res ; 78(4): 938-949, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29259010

RESUMEN

Cancer stem-like cells (CSC) in hepatocellular carcinoma (HCC) are thought to mediate therapeutic resistance and poor survival outcomes, but their intrinsic and extrinsic control is not well understood. In this study, we found that the chromatin modification factor LSD1 is highly expressed in HCC CSC where it decreases during differentiation. LSD1 was responsible for maintaining CSC self-renewal and tumorigenicity in HCC, and its overexpression was sufficient to drive self-renewal of non-CSC. Levels of acetylated LSD1 were low in CSC with high LSD1 activity, and these CSC were capable of self-renewal. Notch signaling activated LSD1 through induction of the sirtuin SIRT1, leading to deacetylation and activation of LSD1 and CSC self-renewal. Notably, we found that LSD1 expression was increased in cancer-associated fibroblasts (CAF) as an upstream driver of Notch3-mediated CSC self-renewal. In clinical specimens of HCC, the presence of CAF, LSD1, and Notch3 strongly associated with poor patient survival. Overall, our results reveal that CAF-induced expression of Notch3 is responsible for LSD1 activation in CSC, driving their self-renewal in HCC.Significance: These seminal findings illuminate a complex pathway in the tissue microenvironment of liver cancer, which is responsible for orchestrating the self-renewal of stem-like cancer cells, with potential implications to improve therapy and limit relapses. Cancer Res; 78(4); 938-49. ©2017 AACR.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Carcinoma Hepatocelular/metabolismo , Histona Demetilasas/metabolismo , Neoplasias Hepáticas/metabolismo , Células Madre Neoplásicas/patología , Receptor Notch3/metabolismo , Animales , Fibroblastos Asociados al Cáncer/patología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular/fisiología , Xenoinjertos , Histona Demetilasas/biosíntesis , Histona Demetilasas/genética , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/metabolismo , Receptor Notch3/genética , Transducción de Señal
17.
Int J Oncol ; 50(3): 1044-1052, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28197626

RESUMEN

Malignant pleural mesothelioma (MPM) is a rare aggressive cancer of the pleura primarily associated with prior exposure to asbestos. The current standard of care for patients suffering from MPM is a combination of cisplatin and pemetrexed (or alternatively cisplatin and raltitrexed). Most patients, however, die within 24 months of diagnosis. New therapies are therefore urgently required for this disease. Inflammation is thought to be a key element in the pathogenesis of MPM, and recently Kdm6 family members (Kdm6a and Kdm6b) have been identified as playing important roles in inflammatory processes. As such these genes could potentially represent novel candidate targets for intervention in MPM. Using RT-PCR we examined the expression of Kdm6aA and Kdm6b in a panel of MPM cell lines and in a cohort of snap-frozen patient samples isolated at surgery comprising benign, epithelial, biphasic and sarcomatoid histologies. Both Kdm6a and Kdm6b were found to be significantly overexpressed in MPM at the mRNA level. However, tests examining if targeting therapeutically Kdm6a/b using a specific small molecule inhibitor (GSK-J4) was potentially useful for treating MPM, revealed that anti-proliferative activity was higher at lower drug concentrations in cell lines derived from normal mesothelial cells compared to those derived from malignant cells. Treatments with GSK-J4 were found to be associated with the induction of apoptosis and increased expression of pro-inflammatory cytokines. As such our results demonstrate that whilst members of the Kdm6 family are overexpressed in MPM they may not be suitable candidates for therapy and may elicit a cytokine storm.


Asunto(s)
Benzazepinas/uso terapéutico , Citocinas/biosíntesis , Histona Demetilasas/biosíntesis , Histona Demetilasas con Dominio de Jumonji/biosíntesis , Neoplasias Pulmonares/tratamiento farmacológico , Mesotelioma/tratamiento farmacológico , Proteínas Nucleares/biosíntesis , Pirimidinas/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Cisplatino/uso terapéutico , Regulación Neoplásica de la Expresión Génica , Histona Demetilasas/genética , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Mesotelioma/genética , Mesotelioma/patología , Mesotelioma Maligno , Proteínas Nucleares/genética , Pemetrexed/uso terapéutico
18.
Int J Oncol ; 50(3): 942-952, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28098854

RESUMEN

A recent study reported that histone lysine specific demethylase 1 (LSD1, KDM1A) is overexpressed in endometrioid endometrial carcinoma (EEC) and associated with tumor progression as well as poor prognosis. However, the physiological function and mechanism of LSD1 in endometrial cancer (EC) remains largely unknown. In this study, we demonstrate that ß-estradiol (E2) treatment increased LSD1 expression via the GPR30/PI3K/AKT pathway in endometrial cancer cells. Both siGPR30 and the PI3K inhibitor LY294002 block this effect. RNAi-mediated silencing of LSD1 abolished estrogen-driven endometrial cancer cell (ECC) proliferation, and induced G1 cell arrest and apoptosis. Mechanistically, we find that LSD1 silencing results in PI3K/AKT signal inactivation, but without the elevation of PTEN expression as expected. This is because the inhibition of LSD1 induces dimethylation of lysine 9 on histone H3 (H3K9m2) accumulation at the promoter region of cyclin D1. Interfering with cyclin D1 leads to PI3K/AKT signal suppression. Re-overexpression of cyclin D1 in LSD1-knockdown ECCs reverses the LSD1 inhibitory action. Our finding connects estrogen signaling with epigenetic regulation in EEC and provides novel experimental support for LSD1 as a potential target for endometrial cancer therapeutics.


Asunto(s)
Ciclina D1/metabolismo , Neoplasias Endometriales/patología , Estrógenos/metabolismo , Histona Demetilasas/metabolismo , Histonas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Cromonas/farmacología , Ciclina D1/genética , Neoplasias Endometriales/mortalidad , Estradiol/farmacología , Femenino , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Histona Demetilasas/biosíntesis , Humanos , Metilación , Persona de Mediana Edad , Morfolinas/farmacología , Fosfohidrolasa PTEN/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Regiones Promotoras Genéticas/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
19.
Mol Reprod Dev ; 84(1): 19-29, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27879032

RESUMEN

Epigenetics is a fundamental regulator underlying many biological functions, such as development and cell differentiation. Epigenetic modifications affect key chromatin regulation, including transcription and DNA repair, which are critical for normal embryo development. In this study, we profiled the expression of epigenetic modifiers and patterns of epigenetic changes in porcine embryos around the period of embryonic genome activation (EGA). We observed that Brahma-related gene 1 (BRG1) and Lysine demethylase 1A (KDM1A), which can alter the methylation status of lysine 4 in histone 3 (H3K4), localize to the nucleus at Day 3-4 of development. We then compared the abundance of epigenetic modifiers between early- and late-cleaving embryos, which were classified based on the time to the first cell cleavage, to investigate if their nuclear localization contributes to developmental competence. The mRNA abundance of BRG1, KDM1A, as well as other lysine demethylases (KDM1B, KDM5A, KDM5B, and KDM5C), were significantly higher in late- compared to early-cleaving embryos near the EGA period, although these difference disappeared at the blastocyst stage. The abundance of H3K4 mono- (H3K4me) and di-methylation (H3K4me2) during the EGA period was reduced in late-cleaving and less developmentally competent embryos. By contrast, BRG1, KDM1A, and H3K4me2 abundance was greater in embryos with more than eight cells at Day 3-4 of development compared to those with fewer than four cells. These findings suggest that altered epigenetic modifications of H3K4 around the EGA period may affect the developmental capacity of porcine embryos to reach the blastocyst stage. Mol. Reprod. Dev. 84: 19-29, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Blastocisto/metabolismo , ADN Helicasas/biosíntesis , Regulación del Desarrollo de la Expresión Génica/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Histona Demetilasas/biosíntesis , Histonas/metabolismo , Animales , Blastocisto/citología , Femenino , Metilación , Porcinos
20.
J Biol Chem ; 292(5): 1798-1807, 2017 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-28031467

RESUMEN

Sall4 (Splat-like 4) plays important roles in maintaining pluripotency of embryonic stem cells and in various developmental processes. Here, we find that Sall4 is highly expressed in oocytes and early embryos. To investigate the roles of SALL4 in oogenesis, we generated Sall4 maternal specific knock-out mice by using CRISPR/Cas9 system, and we find that the maternal deletion of Sall4 causes developmental arrest of oocytes at germinal vesicle stage with non-surrounded nucleus, and the subsequent meiosis resumption is prohibited. We further discover that the loss of maternal Sall4 causes failure in establishment of DNA methylation in oocytes. Furthermore, we find that Sall4 modulates H3K4me3 and H3K27me3 modifications by regulating the expression of key histone demethylases coding genes Kdm5b, Kdm6a, and Kdm6b in oocytes. Moreover, we demonstrate that the aberrant H3K4me3 and H3K27me3 cause mis-expression of genes that are critical for oocytes maturation and meiosis resumption. Taken together, our study explores a pivotal role of Sall4 in regulating epigenetic maturation of mouse oocytes.


Asunto(s)
Metilación de ADN/fisiología , Proteínas de Unión al ADN/metabolismo , Epigénesis Genética/fisiología , Meiosis/fisiología , Oocitos/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Femenino , Histona Demetilasas/biosíntesis , Histona Demetilasas/genética , Histona Demetilasas con Dominio de Jumonji/biosíntesis , Histona Demetilasas con Dominio de Jumonji/genética , Ratones , Ratones Noqueados , Oocitos/citología , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...