Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Breast Cancer Res ; 24(1): 52, 2022 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-35850772

RESUMEN

BACKGROUND: Targeting vulnerabilities of cancer cells by inhibiting key regulators of cell proliferation or survival represents a promising way to overcome resistance to current therapies. In breast cancer (BC), resistance to endocrine therapy results from constitutively active or aberrant estrogen receptor alpha (ERα) signaling to the genome. Targeting components of the ERα pathway in these tumors represents, therefore, a rational way toward effective new treatments. Interaction proteomics identified several proteins associated with ERα in BC cells, including epigenetic complexes controlling gene transcription comprising the scaffold protein menin and the histone methyltransferase Dot1L. METHODS: We combined chromatin immunoprecipitation, transcriptome sequencing, siRNA-mediated gene knockdown (kd), pharmacological inhibition coupled to cellular and functional assays and interaction proteomics in antiestrogen (AE)-sensitive and AE-resistant human BC cell models to: map menin and Dot1L chromatin localization, search for their common and specific target genes, measure the effects of single or combinatorial knockdown or pharmacological inhibition of these proteins on cell proliferation and survival, and characterize their nuclear interactomes. RESULTS: Dot1L and menin associate in MCF-7 cells chromatin, where they co-localize in a significant fraction of sites, resulting in co-regulation of genes involved, among others, in estrogen, p53, HIF1α and death receptor signaling, regulation of cell cycle and epithelial-to-mesenchymal transition. Specific inhibitors of the two factors synergize with each other for inhibition of cell proliferation of AE (tamoxifen or fulvestrant)-sensitive and AE-resistant BC cells. Menin and Dot1L interactomes share a sizeable fraction of their nuclear partners, the majority being known BC fitness genes. Interestingly, these include B-WICH and WINAC complexes that share BAZ1B, a bromodomain protein comprising a tyrosine-protein kinase domain playing a central role in chromatin remodeling and transcriptional regulation. BAZ1B kd caused significant inhibition of ERα expression, proliferation and transcriptome changes resulting in inhibition of estrogen, myc, mTOR, PI3K and AKT signaling and metabolic pathways in AE-sensitive and AE-resistant BC cells. CONCLUSIONS: Identification of a functional interplay between ERα, Dot1L, menin and BAZ1B and the significant effects of their co-inhibition on cell proliferation and survival in cell models of endocrine therapy-resistant BC reveal a new therapeutic vulnerability of these aggressive diseases.


Asunto(s)
Neoplasias de la Mama , Receptor alfa de Estrógeno , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular , Cromatina/genética , Resistencia a Antineoplásicos/genética , Antagonistas de Estrógenos/uso terapéutico , Moduladores de los Receptores de Estrógeno/farmacología , Receptor alfa de Estrógeno/metabolismo , Estrógenos , Femenino , Regulación Neoplásica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/farmacología , Humanos , Células MCF-7 , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Proteínas Tirosina Quinasas/farmacología , Factores de Transcripción
2.
Antimicrob Agents Chemother ; 66(5): e0225021, 2022 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-35471041

RESUMEN

Fungal infections are a major health concern because of limited antifungal drugs and development of drug resistance. Candida can develop azole drug resistance by overexpression of drug efflux pumps or mutating ERG11, the target of azoles. However, the role of epigenetic histone modifications in azole-induced gene expression and drug resistance is poorly understood in Candida glabrata. In this study, we show that Set1 mediates histone H3K4 methylation in C. glabrata. In addition, loss of SET1 and histone H3K4 methylation increases azole susceptibility in both C. glabrata and S. cerevisiae. This increase in azole susceptibility in S. cerevisiae and C. glabrata strains lacking SET1 is due to distinct mechanisms. For S. cerevisiae, loss of SET1 decreased the expression and function of the efflux pump Pdr5, but not ERG11 expression under azole treatment. In contrast, loss of SET1 in C. glabrata does not alter expression or function of efflux pumps. However, RNA sequencing revealed that C. glabrata Set1 is necessary for azole-induced expression of all 12 genes in the late ergosterol biosynthesis pathway, including ERG11 and ERG3. Furthermore, chromatin immunoprecipitation analysis shows histone H3K4 trimethylation increases upon azole-induced ERG gene expression. In addition, high performance liquid chromatography analysis indicated Set1 is necessary for maintaining proper ergosterol levels under azole treatment. Clinical isolates lacking SET1 were also hypersusceptible to azoles which is attributed to reduced ERG11 expression but not defects in drug efflux. Overall, Set1 contributes to azole susceptibility in a species-specific manner by altering the expression and consequently disrupting pathways known for mediating drug resistance.


Asunto(s)
Azoles , Proteínas de Saccharomyces cerevisiae , Antifúngicos/metabolismo , Antifúngicos/farmacología , Azoles/metabolismo , Azoles/farmacología , Candida glabrata/genética , Candida glabrata/metabolismo , Farmacorresistencia Fúngica/genética , Ergosterol/metabolismo , Regulación Fúngica de la Expresión Génica , Histona Metiltransferasas/genética , Histona Metiltransferasas/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/farmacología , Histonas/genética , Histonas/metabolismo , Pruebas de Sensibilidad Microbiana , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
3.
Breast Cancer Res ; 24(1): 26, 2022 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-35395812

RESUMEN

BACKGROUND: Methyltransferase SETDB1 is highly expressed in breast cancer (BC), however, the mechanisms by which SETDB1 promotes BC progression to endocrine therapy resistance remains elusive. In this study, we examined the mechanisms by which SETDB1 contribute to BC endocrine therapy resistance. METHODS: We utilized therapy sensitive (MCF7 and ZR75), therapy resistant (MCF7-TamR, MCF7-FR, MCF7-PELP1cyto, MCF7-SETDB1) estrogen receptor alpha positive (ER+)BC models and conducted in vitro cell viability, colony formation, 3-dimensional cell growth assays to investigate the role of SETDB1 in endocrine resistance. RNA-seq of parental and SETDB1 knock down ER+ BC cells was used to identify unique pathways. SETDB1 interaction with PELP1 was identified by yeast-two hybrid screen and confirmed by immunoprecipitation and GST-pull down assays. Mechanistic studies were conducted using Western blotting, reporter gene assays, RT-qPCR, and in vitro methylation assays. Xenograft assays were used to establish the role of PELP1 in SETDB1 mediated BC progression. RESULTS: RNA-seq analyses showed that SETDB1 regulates expression of a subset of estrogen receptor (ER) and Akt target genes that contribute to endocrine therapy resistance. Importantly, using yeast-two hybrid screen, we identified ER coregulator PELP1 as a novel interacting protein of SETDB1. Biochemical analyses confirmed SETDB1 and PELP1 interactions in multiple BC cells. Mechanistic studies confirmed that PELP1 is necessary for SETDB1 mediated Akt methylation and phosphorylation. Further, SETDB1 overexpression promotes tamoxifen resistance in BC cells, and PELP1 knockdown abolished these effects. Using xenograft model, we provided genetic evidence that PELP1 is essential for SETDB1 mediated BC progression in vivo. Analyses of TCGA datasets revealed SETDB1 expression is positively correlated with PELP1 expression in ER+ BC patients. CONCLUSIONS: This study suggests that the PELP1/SETDB1 axis play an important role in aberrant Akt activation and serves as a novel target for treating endocrine therapy resistance in breast cancer.


Asunto(s)
Neoplasias de la Mama , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Proteínas Co-Represoras/genética , Proteínas Co-Represoras/metabolismo , Proteínas Co-Represoras/farmacología , Resistencia a Antineoplásicos/genética , Femenino , Regulación Neoplásica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/farmacología , Humanos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Saccharomyces cerevisiae/metabolismo , Tamoxifeno/farmacología , Factores de Transcripción/genética
4.
Clin Transl Med ; 12(2): e746, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35220680

RESUMEN

BACKGROUND: Aging-associated osteoporosis is frequently seen in the elderly in clinic, but efficient managements are limited because of unclear nosogenesis. The current study aims to investigate the role of melatonin on senescent bone marrow stromal cells (BMSCs) and the underlying regulating mechanism. METHODS: Melatonin levels were tested by ELISA. Gene expression profiles were performed by RNA-sequencing, enrichment of H3K36me2 on gene promoters was analyzed by Chromatin Immunoprecipitation Sequencing (ChIP-seq), and chromatin accessibility was determined by Assay for Transposase-Accessible Chromatin with high-throughput sequencing (ATAC-seq). Osteogenesis of BMSCs in vitro was measured by Alizarin Red and Alkaline Phosphatase staining, and in vivo effects of melatonin was assessed by histological staining and micro computed tomography (micro-CT) scan. Correlation of NSD2 expression and severity of senile osteoporosis patients were analyzed by Pearson correlation. RESULTS: Melatonin levels were decreased during aging in human bone marrow, accompanied by downregulation of the histone methyltransferase nuclear receptor binding SET domain protein 2 (NSD2) expression in the senescent BMSCs. Melatonin stimulated the expression of NSD2 through MT1/2-mediated signaling pathways, resulting in the rebalancing of H3K36me2 and H3K27me3 modifications to increase chromatin accessibility of the osteogenic genes, runt-related transcription factor 2 (RUNX2) and bone gamma-carboxyglutamate protein (BGLAP). Melatonin promoted osteogenesis of BMSCs in vitro, and alleviates osteoporosis progression in the aging mice. In clinic, severity of senile osteoporosis (SOP) was negatively correlated with melatonin level in bone marrow, as well as NSD2 expression in BMSCs. Similarly, melatonin remarkably enhanced osteogenic differentiation of BMSCs derived from SOP patients in vitro. CONCLUSIONS: Collectively, our study dissects previously unreported mechanistic insights into the epigenetic regulating machinery of melatonin in meliorating osteogenic differentiation of senescent BMSC, and provides evidence for application of melatonin in preventing aging-associated bone loss.


Asunto(s)
Ensamble y Desensamble de Cromatina/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/farmacología , Melatonina/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Proteínas Represoras/farmacología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Diferenciación Celular/efectos de los fármacos , Ensamble y Desensamble de Cromatina/genética , Ensamble y Desensamble de Cromatina/fisiología , Modelos Animales de Enfermedad , Femenino , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Masculino , Melatonina/uso terapéutico , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL/metabolismo , Persona de Mediana Edad , Osteoblastos/fisiología , Proteínas Represoras/metabolismo
5.
Clin Transl Med ; 11(11): e591, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34841684

RESUMEN

BACKGROUND: The methylation of lysine residues has been involved in the multiple biological and diseases processes. Recently, some particular non-histone proteins have been elucidated to be methylated by SMYD2, a SET and MYND domain protein with lysine methyltransferase activity. METHODS: SMYD2 was evaluated in synovial tissue and cells derived from rheumatoid arthritis patients. We confirmed TRAF2 could be methylated by SMYD2 using Mass spectrometry, pull-down, immunoprecipitation, methyltransferase assay, ubiquitination assay, luciferase reporter assays, and western blot analyses. Using loss- and gain-of function studies, we explored the biological functions of SMYD2 in vitro and in vivo. Using acute and chronic inflammation with different mice models to determine the impact of SMYD2. RESULTS: Here, we first time confirmed that the cytoplasmic protein TRAF2 as the kernel node for NF-κB signaling pathway could be methylated by SMYD2. SMYD2-mediated TRAF2 methylation contributed to the durative sensitization of NF-κB signaling transduction through restraining its own proteolysis and enhancing the activity. In addition, we found knocking down of SMYD2 has different degrees of mitigation in acute and chronic inflammation mice models. Furthermore, as the lysine-specific demethylase, LSD1 could resist methylation on TRAF2 induced by SMYD2. CONCLUSIONS: Our data uncovered an unprecedented cytoplasmic protein network that employed methylation of TRAF2 for the maintenance of NF-κB activation during inflammatory diseases.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/farmacología , Inflamación/tratamiento farmacológico , Metilación/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/uso terapéutico , N-Metiltransferasa de Histona-Lisina/uso terapéutico , Humanos , Inflamación/inmunología , Inflamación/fisiopatología , Transducción de Señal/efectos de los fármacos
6.
Clin Epigenetics ; 13(1): 192, 2021 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-34663428

RESUMEN

To define the role of SETD2 in the WNT5a signaling in the context of osteoclastogenesis, we exploited two different models: in vitro osteoclast differentiation, and K/BxN serum-induced arthritis model. We found that SETD2 and WNT5a were upregulated during osteoclast differentiation and after induction of arthritis. Using gain- and loss-of-function approaches in the myeloid cell, we confirmed that SETD2 regulated the osteoclast markers, and WNT5a via modulating active histone marks by enriching H3K36me3, and by reducing repressive H3K27me3 mark. Additionally, during osteoclastic differentiation, the transcription of Wnt5a was also associated with the active histone H3K9 and H4K8 acetylations. Mechanistically, SETD2 directed induction of NF-κß expression facilitated the recruitment of H3K9Ac and H4K8Ac around the TSS region of the Wnt5a gene, thereby, assisting osteoclast differentiation. Together these findings for the first time revealed that SETD2 mediated epigenetic regulation of Wnt5a plays a critical role in osteoclastogenesis and induced arthritis. Model for the Role of SETD2 dependent regulation of osteoclastic differentiation. A In monocyte cells SETD2-dependent H3K36 trimethylation help to create open chromatin region along with active enhancer mark, H3K27Ac. This chromatin state facilitated the loss of a suppressive H3K27me3 mark. B Additionally, SETD2 mediated induction of NF-κß expression leads to the recruitment of histone acetyl transferases, P300/PCAF, to the Wnt5a gene and establish H3K9Ac and H4K8Ac marks. Along with other activation marks, these acetylation marks help in Wnt5a transcription which leads to osteoclastogenesis.


Asunto(s)
Epigénesis Genética/genética , N-Metiltransferasa de Histona-Lisina/farmacología , Osteogénesis/genética , Proteína Wnt-5a/efectos adversos , Animales , Artritis/inmunología , Artritis/fisiopatología , Modelos Animales de Enfermedad , N-Metiltransferasa de Histona-Lisina/genética , Ratones , Ratones Endogámicos C57BL , Osteogénesis/fisiología , Activación Transcripcional/genética , Proteína Wnt-5a/genética
7.
Clin Epigenetics ; 13(1): 174, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34530900

RESUMEN

BACKGROUND: Multiple myeloma (MM) is a malignancy of plasma cells that largely remains incurable. The search for new therapeutic targets is therefore essential. In addition to a wide panel of genetic mutations, epigenetic alterations also appear as important players in the development of this cancer, thereby offering the possibility to reveal novel approaches and targets for effective therapeutic intervention. RESULTS: Here, we show that a higher expression of the lysine methyltransferase SETD8, which is responsible for the mono-methylation of histone H4 at lysine 20, is an adverse prognosis factor associated with a poor outcome in two cohorts of newly diagnosed patients. Primary malignant plasma cells are particularly addicted to the activity of this epigenetic enzyme. Indeed, the inhibition of SETD8 by the chemical compound UNC-0379 and the subsequent decrease in histone H4 methylation at lysine 20 are highly toxic in MM cells compared to normal cells from the bone marrow microenvironment. At the molecular level, RNA sequencing and functional studies revealed that SETD8 inhibition induces a mature non-proliferating plasma cell signature and, as observed in other cancers, triggers an activation of the tumor suppressor p53, which together cause an impairment of myeloma cell proliferation and survival. However, a deadly level of replicative stress was also observed in p53-deficient myeloma cells treated with UNC-0379, indicating that the cytotoxicity associated with SETD8 inhibition is not necessarily dependent on p53 activation. Consistent with this, UNC-0379 triggers a p53-independent nucleolar stress characterized by nucleolin delocalization and reduction of nucleolar RNA synthesis. Finally, we showed that SETD8 inhibition is strongly synergistic with melphalan and may overcome resistance to this alkylating agent widely used in MM treatment. CONCLUSIONS: Altogether, our data indicate that the up-regulation of the epigenetic enzyme SETD8 is associated with a poor outcome and the deregulation of major signaling pathways in MM. Moreover, we provide evidences that myeloma cells are dependent on SETD8 activity and its pharmacological inhibition synergizes with melphalan, which could be beneficial to improve MM treatment in high-risk patients whatever their status for p53.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/administración & dosificación , Metiltransferasas/farmacología , Mieloma Múltiple/tratamiento farmacológico , Resistencia a Medicamentos/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/farmacología , Humanos , Metiltransferasas/administración & dosificación , Mieloma Múltiple/fisiopatología
8.
Clin Epigenetics ; 13(1): 44, 2021 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-33632299

RESUMEN

BACKGROUND: Trimethylation of lysine 27 and dimethylation of lysine 9 of histone-H3 catalyzed by the histone methyltransferases EZH2 and G9a impede gene transcription in cancer. Our human bronchial epithelial (HBEC) pre-malignancy model studied the role of these histone modifications in transformation. Tobacco carcinogen transformed HBEC lines were characterized for cytosine DNA methylation, transcriptome reprogramming, and the effect of inhibiting EZH2 and G9a on the transformed phenotype. The effects of targeting EZH2 and G9a on lung cancer prevention was assessed in the A/J mouse lung tumor model. RESULTS: Carcinogen exposure induced transformation and DNA methylation of 12-96 genes in the four HBEC transformed (T) lines that was perpetuated in malignant tumors. In contrast, 506 unmethylated genes showed reduced expression in one or more HBECTs with many becoming methylated in tumors. ChIP-on-chip for HBEC2T identified 327 and 143 genes enriched for H3K27me3 and H3K9me2. Treatment of HBEC2T and HBEC13T with DZNep, a lysine methyltransferase inhibitor depleted EZH2, reversed transformation, and induced transcriptional reprogramming. The EZH2 small molecule inhibitor EPZ6438 also affected transformation and expression in HBEC2T, while a G9a inhibitor, UNC0642 was ineffective. Genetic knock down of EZH2 dramatically reduced carcinogen-induced transformation of HBEC2. Only DZNep treatment prevented progression of hyperplasia to adenomas in the NNK mouse lung tumor model through reducing EZH2 and affecting the expression of genes regulating cell growth and invasion. CONCLUSION: These studies demonstrate a critical role for EZH2 catalyzed histone modifications for premalignancy and its potential as a target for chemoprevention of lung carcinogenesis.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Proteína Potenciadora del Homólogo Zeste 2/genética , Código de Histonas/efectos de los fármacos , Neoplasias/prevención & control , Adenosina/análogos & derivados , Adenosina/farmacología , Adenosilhomocisteinasa/antagonistas & inhibidores , Animales , Benzamidas/farmacología , Compuestos de Bifenilo/farmacología , Proliferación Celular/efectos de los fármacos , Islas de CpG , Metilación de ADN/efectos de los fármacos , Proteína Potenciadora del Homólogo Zeste 2/farmacología , Inhibidores Enzimáticos/farmacología , Epigénesis Genética/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Femenino , Código de Histonas/genética , Histona Metiltransferasas/antagonistas & inhibidores , Histona Metiltransferasas/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/farmacología , Histonas/metabolismo , Humanos , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/patología , Ratones , Morfolinas/farmacología , Fenotipo , Piridonas/farmacología , Transcriptoma/efectos de los fármacos
9.
Elife ; 72018 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-30526847

RESUMEN

Protein histidine methylation is a rare post-translational modification of unknown biochemical importance. In vertebrates, only a few methylhistidine-containing proteins have been reported, including ß-actin as an essential example. The evolutionary conserved methylation of ß-actin H73 is catalyzed by an as yet unknown histidine N-methyltransferase. We report here that the protein SETD3 is the actin-specific histidine N-methyltransferase. In vitro, recombinant rat and human SETD3 methylated ß-actin at H73. Knocking-out SETD3 in both human HAP1 cells and in Drosophila melanogaster resulted in the absence of methylation at ß-actin H73 in vivo, whereas ß-actin from wildtype cells or flies was > 90% methylated. As a consequence, we show that Setd3-deficient HAP1 cells have less cellular F-actin and an increased glycolytic phenotype. In conclusion, by identifying SETD3 as the actin-specific histidine N-methyltransferase, our work pioneers new research into the possible role of this modification in health and disease and questions the substrate specificity of SET-domain-containing enzymes.


Asunto(s)
Actinas/metabolismo , Fibroblastos/enzimología , N-Metiltransferasa de Histona-Lisina/genética , Músculo Esquelético/enzimología , Procesamiento Proteico-Postraduccional , Actinas/genética , Secuencia de Aminoácidos , Animales , Sitios de Unión , Línea Celular Tumoral , Secuencia Conservada , Drosophila melanogaster/clasificación , Drosophila melanogaster/enzimología , Drosophila melanogaster/genética , Fibroblastos/citología , Glucólisis/genética , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/química , N-Metiltransferasa de Histona-Lisina/deficiencia , N-Metiltransferasa de Histona-Lisina/farmacología , Humanos , Cinética , Metilación , Modelos Moleculares , Músculo Esquelético/química , Fenotipo , Filogenia , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Ratas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Especificidad por Sustrato
10.
Genes Dev ; 31(12): 1272-1287, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28743695

RESUMEN

Hybrid seed lethality as a consequence of interspecies or interploidy hybridizations is a major mechanism of reproductive isolation in plants. This mechanism is manifested in the endosperm, a dosage-sensitive tissue supporting embryo growth. Deregulated expression of imprinted genes such as ADMETOS (ADM) underpin the interploidy hybridization barrier in Arabidopsis thaliana; however, the mechanisms of their action remained unknown. In this study, we show that ADM interacts with the AT hook domain protein AHL10 and the SET domain-containing SU(VAR)3-9 homolog SUVH9 and ectopically recruits the heterochromatic mark H3K9me2 to AT-rich transposable elements (TEs), causing deregulated expression of neighboring genes. Several hybrid incompatibility genes identified in Drosophila encode for dosage-sensitive heterochromatin-interacting proteins, which has led to the suggestion that hybrid incompatibilities evolve as a consequence of interspecies divergence of selfish DNA elements and their regulation. Our data show that imbalance of dosage-sensitive chromatin regulators underpins the barrier to interploidy hybridization in Arabidopsis, suggesting that reproductive isolation as a consequence of epigenetic regulation of TEs is a conserved feature in animals and plants.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/efectos de los fármacos , Arabidopsis/fisiología , Proteínas de Ciclo Celular/metabolismo , Epigénesis Genética , N-Metiltransferasa de Histona-Lisina/farmacología , Aislamiento Reproductivo , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Proteínas de Ciclo Celular/genética , Regulación de la Expresión Génica de las Plantas , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/metabolismo , Hibridación Genética
11.
AIDS Rev ; 16(4): 246, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25373349

RESUMEN

The eradication of HIV is at this moment one of the greatest challenges in the fight against HIV/AIDS. Despite the prolonged effectiveness of current anti-HIV therapies, capable of keeping patients with undetectable viremia for long periods of time, HIV-infected patients cannot be cured due to the establishment of HIV latent reservoirs. Therefore, several therapeutic strategies are being evaluated to eliminate these viral reservoirs. One of these strategies, termed "shock and kill", aims to attack the latent reservoir by simultaneous treatment with HIV-activating agents to stimulate viral replication in latently infected cells and antiretroviral therapy to block new infections. A number of compounds have been suggested for the shock and kill strategy including histone deacetylase inhibitors (HDACI), histone methyltransferases (HMT), DNA methyltransferase inhibitors (DNMTI), and protein kinase C (PKC) activators.


Asunto(s)
Fármacos Anti-VIH/farmacología , Erradicación de la Enfermedad , Diterpenos/farmacología , Infecciones por VIH/prevención & control , Latencia del Virus/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Erradicación de la Enfermedad/tendencias , Reservorios de Enfermedades , Quimioterapia Combinada , Infecciones por VIH/inmunología , Inhibidores de Histona Desacetilasas/farmacología , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/farmacología , Humanos , Proteína Quinasa C/efectos de los fármacos , Latencia del Virus/inmunología , Replicación Viral/efectos de los fármacos
12.
Clin Chim Acta ; 415: 181-90, 2013 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-23123287

RESUMEN

BACKGROUND: No substantial work has been conducted to date in context to cationic proteins with antilithiatic activity. We explored the antilithiatic cationic proteins present in human calcium oxalate (CaOx) stones and also examined their molecular interactions with calcium oxalate crystals in silico. METHODS: Proteins were isolated from the matrix of human CaOx containing kidney stones. Proteins having MW>3 kDa were subjected to cation exchange chromatography followed by molecular-sieve chromatography. The effect of these purified cationic proteins was tested against CaOx nucleation and growth and on oxalate injured MDCK cells for their activity. Proteins were identified by MALDI-TOF MS. Molecular interaction studies with COM crystals in silico were also investigated. RESULTS: Three antilithiatic cationic proteins were identified as histone-lysine N-methyltransferase, inward rectifier K channel and protein Wnt-2 (MW~53, ~44, and ~42 kDa respectively) by MALDI-TOF MS based on database search with MASCOT server. Further molecular modeling calculations revealed the mode of interaction of these proteins with CaOx at the molecular level. CONCLUSION: We identified histone-lysine N-methyltransferase, inward rectifier K channel and protein Wnt-2 as novel antilithiatic proteins which play a vital role in the kidney function and have been associated with various kidney diseases.


Asunto(s)
Oxalato de Calcio/química , N-Metiltransferasa de Histona-Lisina/química , Cálculos Renales/química , Canales de Potasio de Rectificación Interna/química , Proteína wnt2/química , Adulto , Animales , Sitios de Unión , Cationes , Supervivencia Celular/efectos de los fármacos , Cromatografía en Gel , Cromatografía por Intercambio Iónico , Cristalización , Citoprotección , Perros , N-Metiltransferasa de Histona-Lisina/aislamiento & purificación , N-Metiltransferasa de Histona-Lisina/farmacología , Humanos , Células de Riñón Canino Madin Darby , Simulación del Acoplamiento Molecular , Canales de Potasio de Rectificación Interna/aislamiento & purificación , Canales de Potasio de Rectificación Interna/farmacología , Unión Proteica , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Proteína wnt2/aislamiento & purificación , Proteína wnt2/farmacología
13.
Biochem J ; 448(1): 93-102, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22880885

RESUMEN

The discovery of epigenetic processes as possible pivotal regulatory mechanisms in psychiatric diseases raised the question of how psychoactive drugs may impact the epigenetic machinery. In the present study we set out to explore the specificity and the mode of action of the reported inhibitory effect of the TCA (tricyclic antidepressant) amitriptyline on DNMT (DNA methyltransferase) activity in primary astrocytes from the rat cortex. We found that the impact on DNMT was shared by another TCA, imipramine, and by paroxetine, but not by venlafaxine or the mood stabilizers carbamazepine and valproic acid. DNMT activity in subventricular neural stem cells was refractory to the action of ADs (antidepressants). Among the established DNMTs, ADs primarily targeted DNMT1. The reduction of enzymatic DNMT1 activity was neither due to reduced DNMT1 expression nor due to direct drug interference. We tested putative DNMT1-inhibitory mechanisms and discovered that a known stimulator of DNMT1, the histone methyltransferase G9a, exhibited decreased protein levels and interactions with DNMT1 upon AD exposure. Adding recombinant G9a completely reversed the AD repressive effect on DNMT1 function. In conclusion, the present study presents a model where distinct ADs affect DNMT1 activity via G9a with important repercussions for possible novel treatment regimes.


Asunto(s)
Antidepresivos/farmacología , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , Metilación de ADN/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/metabolismo , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Animales , Astrocitos/efectos de los fármacos , Astrocitos/enzimología , Astrocitos/fisiología , Carbamazepina/farmacología , Células Cultivadas/efectos de los fármacos , Células Cultivadas/enzimología , Células Cultivadas/fisiología , Corteza Cerebral/citología , Corteza Cerebral/embriología , Citalopram/farmacología , Ciclohexanoles/farmacología , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Inhibidores Enzimáticos/farmacología , Glucolípidos/farmacología , N-Metiltransferasa de Histona-Lisina/farmacología , Imipramina/farmacología , Indoles/farmacología , Maleimidas/farmacología , Ratones , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/enzimología , Células-Madre Neurales/fisiología , Neuronas/efectos de los fármacos , Neuronas/enzimología , Neuronas/fisiología , Paroxetina/farmacología , Mapeo de Interacción de Proteínas , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/farmacología , Clorhidrato de Venlafaxina
15.
Int J Med Sci ; 8(2): 161-7, 2011 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-21369371

RESUMEN

Human tumor suppressor gene RIZ encodes two protein products, tumor suppressor RIZ1 and proto-oncoprotein RIZ2, which regulate cellular functions in a Yin-Yang fashion. The only structural difference between them is that RIZ2 lacks the N-terminal PR domain. In this study, we showed that RIZ1 mRNA expression level was elevated in stage IV of eight different types of cancer (stage III for prostate cancer), indicating that RIZ1 might play an important role in tumor metastasis, and the PR domain alone possessed anticancer activity.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/farmacología , N-Metiltransferasa de Histona-Lisina/química , N-Metiltransferasa de Histona-Lisina/farmacología , Proteínas Nucleares/química , Proteínas Nucleares/farmacología , Fragmentos de Péptidos/farmacología , Factores de Transcripción/química , Factores de Transcripción/farmacología , Secuencia de Aminoácidos , Antineoplásicos/química , Clonación Molecular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/fisiología , Humanos , Masculino , Datos de Secuencia Molecular , Neoplasias/genética , Neoplasias/patología , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Fragmentos de Péptidos/química , Estructura Terciaria de Proteína/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Homología de Secuencia de Aminoácido , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/farmacología , Proteínas Supresoras de Tumor/fisiología
16.
Cancer Invest ; 28(8): 806-12, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20594067

RESUMEN

The role of retinoblastoma protein-interacting zinc finger 1 (RIZ1) on the cell growth of mouse and human monocytic leukemia cells was examined. RIZ1 expression was induced in response to tumor necrosis factor (TNF)-α. The expression was dependent on the nuclear factor-κB and AKT signaling. Further, RIZ1 expression led to the augmentation of p53 expression and the silencing of RIZ1 prevented it. On the other hand, a p53 inhibitor enhanced the TNF-α-induced RIZ1 expression. Silencing of RIZ1 augmented the proliferative activity of TNF-α-treated cells. Therefore, it is suggested that RIZ1 negatively regulated the cell proliferation of monocytic leukemia cells via activation of p53.


Asunto(s)
Proteínas de Unión al ADN/farmacología , N-Metiltransferasa de Histona-Lisina/farmacología , Leucemia Mieloide/patología , Proteínas Nucleares/farmacología , Factores de Transcripción/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Animales , División Celular/efectos de los fármacos , Metilación de ADN , Cartilla de ADN , Proteínas de Unión al ADN/genética , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , N-Metiltransferasa de Histona-Lisina/genética , Homeostasis , Humanos , Leucemia Mieloide/genética , Ratones , Neoplasias/genética , Neoplasias/patología , Proteínas Nucleares/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética , Transfección , Factor de Necrosis Tumoral alfa/genética , Proteína p53 Supresora de Tumor/efectos de los fármacos
17.
Br J Cancer ; 90(4): 756-60, 2004 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-14970849

RESUMEN

Rearrangements of the ALL-1/MLL1 gene underlie the majority of infant acute leukaemias, as well as of therapy-related leukaemias developing in cancer patients treated with inhibitors of topoisomerase II, such as VP16 and doxorubicin. The rearrangements fuse ALL-1 to any of >50 partner genes or to itself. Here, we describe the unique features of ALL-1-associated leukaemias, and recent progress in understanding molecular mechanisms involved in the activity of the ALL-1 protein and of its Drosophila homologue TRITHORAX.


Asunto(s)
Cromatina/metabolismo , Proteínas de Unión al ADN/farmacología , Proteínas de Drosophila/farmacología , Regulación Neoplásica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/farmacología , Leucemia Mieloide Aguda/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proto-Oncogenes , Factores de Transcripción , Animales , Transformación Celular Neoplásica , Modelos Animales de Enfermedad , Drosophila/genética , Humanos , Leucemia Mieloide Aguda/fisiopatología , Ratones , Proteína de la Leucemia Mieloide-Linfoide , Leucemia-Linfoma Linfoblástico de Células Precursoras/fisiopatología , Dedos de Zinc
18.
Biochim Biophys Acta ; 622(1): 144-50, 1980 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-6244857

RESUMEN

A highly purified protein methylase III from Neurospora crassa or Saccharomyces cerevisiae specifically methylates a single lysine residue of position 72 of horse heart cytochrome c. The enzymatically methylated cytochrome c has been separated from the unmethylated counterpart species by isoelectric focusing. Simultaneously, the pI values of these two species were found to be 9.49 and 10.03, respectively. Since methyl substitution increases the basicity associated with the epsilon-amino group of lysine residues, the observed decrease in pI value is in opposition to the predicted increase. Space-filling models revealed the possibility of a hydrogen bond between the oxygen of amide of residue-70 asparagine and the epsilon-amino nitrogen of residue-72 lysine in unmethylated horse heart cytochrome C. the enzymatic methylation of residue-72 lysine tends to dissociate this hydrogen bond, thereby possibly inducing the shift of 'effective charge' of the protein molecule. This paper also deals with the pI values of cytochromes c from 13 different sources, determined by the isoelectric focusing technique.


Asunto(s)
Grupo Citocromo c , N-Metiltransferasa de Histona-Lisina/farmacología , Lisina , Proteína Metiltransferasas/farmacología , Animales , Asparagina , Fenómenos Químicos , Química , Caballos , Enlace de Hidrógeno , Focalización Isoeléctrica , Punto Isoeléctrico , Metilación , Modelos Moleculares , Neurospora crassa/enzimología , Saccharomyces cerevisiae/enzimología , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA