Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
J Enzyme Inhib Med Chem ; 38(1): 2212326, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37190931

RESUMEN

Class II histone deacetylases (HDACs) are considered as potential targets to treat Alzheimer's disease (AD). Previously, C-3 substituted phenothiazine-containing compounds with class II HDAC-inhibiting activities was found to promote neurite outgrowth. This study replaced phenothiazine moiety with phenoxazine that contains many C-3 and C-4 substituents. Some resulting compounds bearing the C-4 substituent on a phenoxazine ring displayed potent class II HDAC inhibitory activities. Structure-activity relationship (SAR) of these compounds that inhibited HDAC isoenzymes was disclosed. Molecular modelling analysis demonstrates that the potent activities of C-4 substituted compounds probably arise from π-π stacked interactions between these compounds and class IIa HDAC enzymes. One of these, compound 7d exhibited the most potent class II HDAC inhibition (IC50= 3-870 nM). Notably, it protected neuron cells from H2O2-induced neuron damage at sub-µM concentrations, but with no significant cytotoxicity. These findings show that compound 7d is a lead compound for further development of anti-neurodegenerative agents.


Asunto(s)
Antineoplásicos , Ácidos Hidroxámicos , Ácidos Hidroxámicos/farmacología , Inhibidores de Histona Desacetilasas/farmacología , Peróxido de Hidrógeno/farmacología , Relación Estructura-Actividad , Histona Desacetilasas/metabolismo , Antineoplásicos/farmacología , Histona Desacetilasa 1/farmacología , Proliferación Celular
2.
EMBO J ; 35(2): 176-92, 2016 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-26620551

RESUMEN

During DNA replication, thousands of replication origins are activated across the genome. Chromatin architecture contributes to origin specification and usage, yet it remains unclear which chromatin features impact on DNA replication. Here, we perform a RNAi screen for chromatin regulators implicated in replication control by measuring RPA accumulation upon replication stress. We identify six factors required for normal rates of DNA replication and characterize a function of the bromodomain and PHD finger-containing protein 3 (BRPF3) in replication initiation. BRPF3 forms a complex with HBO1 that specifically acetylates histone H3K14, and genomewide analysis shows high enrichment of BRPF3, HBO1 and H3K14ac at ORC1-binding sites and replication origins found in the vicinity of TSSs. Consistent with this, BRPF3 is necessary for H3K14ac at selected origins and efficient origin activation. CDC45 recruitment, but not MCM2-7 loading, is impaired in BRPF3-depleted cells, identifying a BRPF3-dependent function of HBO1 in origin activation that is complementary to its role in licencing. We thus propose that BRPF3-HBO1 acetylation of histone H3K14 around TSS facilitates efficient activation of nearby replication origins.


Asunto(s)
Ciclo Celular/fisiología , Histona Acetiltransferasas/metabolismo , Histonas/metabolismo , Origen de Réplica/fisiología , Acetilación , Ciclo Celular/genética , Línea Celular , Cromatina/metabolismo , Inmunoprecipitación de Cromatina , Replicación del ADN/genética , Replicación del ADN/fisiología , Histona Acetiltransferasas/genética , Humanos , Inmunohistoquímica , Origen de Réplica/genética
3.
Mol Cell ; 46(1): 1-2, 2012 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-22500733

RESUMEN

In the current issue of Molecular Cell, Yu et al. (2012) establish H3K56 monomethylation (H3K56me1) as a new mammalian chromatin mark, imposed by the G9a methyltransferase and recognized by the replication clamp PCNA.

4.
Expert Opin Drug Discov ; 18(10): 1169-1193, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37525475

RESUMEN

INTRODUCTION: PARP inhibitors block the DNA-repairing mechanism of PARP and represent a promising class of anti-cancer therapy. The last decade has witnessed FDA approvals of several PARP inhibitors, with some undergoing advanced-stage clinical investigation. Medicinal chemists have invested much effort to expand the structure pool of PARP inhibitors. Issues associated with the use of PARP inhibitors that make their standing disconcerting in the pharmaceutical sector have been addressed via the design of new structural assemblages. AREA COVERED: In this review, the authors present a detailed account of the medicinal chemistry campaigns conducted in the recent past for the construction of PARP1/PARP2 inhibitors, PARP1 biased inhibitors, and PARP targeting bifunctional inhibitors as well as PARP targeting degraders (PROTACs). Limitations associated with FDA-approved PARP inhibitors and strategies to outwit the limitations are also discussed. EXPERT OPINION: The PARP inhibitory field has been rejuvenated with numerous tractable entries in the last decade. With numerous magic bullets in hand coupled with unfolded tactics to outwit the notoriety of cancer cells developing resistance toward PARP inhibitors, the dominance of PARP inhibitors as a sagacious option of targeted therapy is highly likely to be witnessed soon.


Asunto(s)
Neoplasias , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Humanos , Reparación del ADN , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico
5.
Eur J Med Chem ; 258: 115505, 2023 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-37302341

RESUMEN

Precise and accurate control of cell cycle progression is required to maintain cell identity and proliferation. Failing to keep it will lead to genome instability and tumorigenesis. Cell Division Cycle 25 (CDC25) phosphatases are the key to regulating the activity of the master cell cycle controller, cyclin-dependent kinases (CDKs). Dysregulation of CDC25 has been shown to associate with several human malignancies. Here, we reported a series of derivatives of the CDC25 inhibitor, NSC663284, bearing quinones as core scaffolds and morpholin alkylamino side chains. Among these derivatives, the cytotoxic activity of the 6-isomer of 5,8-quinolinedione derivatives (6b, 16b, 17b, and 18b) displayed higher potency against colorectal cancer (CRC) cells. Compound 6b possessed the most antiproliferative activity, with IC50 values of 0.59 µM (DLD1) and 0.44 µM (HCT116). The treatment of compound 6b resulted in a remarkable effect on cell cycle progression, blocking S-phase progression in DLD1 cells straight away while slowing S-phase progression and accumulated cells in the G2/M phase in HCT116 cells. Furthermore, we showed that compound 6b inhibited CDK1 dephosphorylation and H4K20 methylation in cells. The treatment with compound 6b induced DNA damage and triggered apoptosis. Our study identifies compound 6b as a potent CDC25 inhibitor that induces genome instability and kills cancer cells through an apoptotic pathway, deserving further investigation to fulfill its candidacy as an anti-CRC agent.


Asunto(s)
Neoplasias Colorrectales , Fosfatasas cdc25 , Humanos , División Celular , Ciclo Celular , Inestabilidad Genómica , Neoplasias Colorrectales/tratamiento farmacológico
6.
Eur J Med Chem ; 227: 113904, 2022 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-34662748

RESUMEN

This study presents the design, synthesis, and characterization of bisindole molecules as anti-cancer agents against Tousled-like kinases (TLKs). We show that compound 2 composed of an indirubin-3'-oxime group linked with a (N-methylpiperidin-2-yl)ethyl moiety possessed inhibitory activity toward both TLK1 and TLK2 in vitro and diminished the phosphorylation level of the downstream substrate anti-silencing function 1 (ASF1) in replicating cells. The treatment of compound 2 impaired DNA replication, slowed S-phase progression, and triggered DNA damage response in replicating cells. Structure optimization further discovered six derivatives exhibiting potent TLK inhibitory activity and revealed the importance of the tertiary amine-containing moiety of the side chain. Moreover, the derivatives 6, 17, 19, and 20 strongly suppressed the growth of triple-negative breast cancer MDA-MB-231 cells, non-small cell lung cancer A549 cells, and colorectal cancer HCT-116 cells, while normal lung fibroblast MRC5 and IMR90 cells showed a lower response to these compounds. Taken together, this study identifies tertiary amine-linked indirubin-3'-oximes as potent anticancer agents that inhibit TLK activity.


Asunto(s)
Antineoplásicos/farmacología , Diseño de Fármacos , Alcaloides Indólicos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Células HEK293 , Humanos , Alcaloides Indólicos/síntesis química , Alcaloides Indólicos/química , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Relación Estructura-Actividad , Células Tumorales Cultivadas
7.
Eur J Med Chem ; 240: 114602, 2022 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-35858522

RESUMEN

A fragment recruitment process was conducted to pinpoint a suitable fragment for installation in the HDAC inhibitory template to furnish agents endowed with the potential to treat lung cancer. Resultantly, Ring C expanded deoxyvasicinone was selected as an appropriate surface recognition part that was accommodated in the HDAC three-component model. Delightfully, fused quinazolinone 6 demonstrating a magnificent anticancer profile against KRAS and EGFR mutant lung cancer cell lines (IC50 = 0.80-0.96 µM) was identified. Results of the mechanistic studies confirmed that the cell growth inhibitory effects of compound 6 stems for HDAC6 (IC50 = 12.9 nM), HDAC1 (IC50 = 49.9 nM) and HDAC3 inhibition (IC50 = 68.5 nM), respectively. Compound 6 also suppressed the colony formation ability of A549 cells, induced apoptosis, and increased autophagic flux. Key interactions of HDAC inhibitor 6 within the active site of HDAC isoforms were figured out through molecular modeling studies. Furthermore, a pH-responsive nanocarrier (Hyaluronic acid - fused quinazolinone 6 nanoparticles) was designed and assessed using a dialysis bag approach under both normal and acidic circumstances that confirmed the pH-sensitive nature of NPs. Delightfully, the nanoparticles demonstrated selective cell viability reduction potential towards the lung cancer cell lines (A549 lung cancer cell lines) and were found to be largely devoid of cell growth inhibitory effects under normal settings (L929, mouse fibroblast cells).


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular , Inhibidores de Histona Desacetilasas/química , Concentración de Iones de Hidrógeno , Neoplasias Pulmonares/metabolismo , Ratones , Sistema de Administración de Fármacos con Nanopartículas , Quinazolinas , Quinazolinonas/administración & dosificación , Quinazolinonas/química , Quinazolinonas/farmacología , Quinazolinonas/uso terapéutico
8.
Biomed Pharmacother ; 138: 111485, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33740521

RESUMEN

Aberrant alteration of epigenetic information disturbs chromatin structure and gene function, thereby facilitating cancer development. Several drugs targeting histone deacetylases (HDACs), a group of epigenetic enzymes, have been approved for treating hematologic malignancies in the clinic. However, patients who suffer from solid tumors often respond poorly to these drugs. In this study, we report a selective entinostat derivative, MPT0L184, with potent cancer-killing activity in both cell-based and mouse xenograft models. A time-course analysis of cell-cycle progression revealed that MPT0L184 treatment elicited an early onset of mitosis but prevented the division of cells with duplicated chromosomes. We show that MPT0L184 possessed potent inhibitory activity toward HDAC1 and 2, and its HDAC-inhibitory activity was required for initiating premature mitotic signaling. HDAC inhibition by MPT0L184 reduced WEE1 expression at the transcription level. In addition, MPT0L184 treatment also downregulated ATR-mediated CHK1 phosphorylation independent of HDAC inhibition. Furthermore, gastric cancer cells resistant to HDAC inhibitors were vulnerable to MPT0L184. Taken together, our study discovers MPT0L184 as a novel HDAC inhibitor that can trigger premature mitosis and potentially counteract drug resistance of cancers.


Asunto(s)
Benzamidas/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Piridinas/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Benzamidas/química , Puntos de Control del Ciclo Celular/fisiología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Femenino , Inhibidores de Histona Desacetilasas/química , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mitosis/efectos de los fármacos , Mitosis/fisiología , Piridinas/química , Transducción de Señal/fisiología , Carga Tumoral/efectos de los fármacos , Carga Tumoral/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
9.
Eur J Med Chem ; 215: 113169, 2021 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-33588178

RESUMEN

The study focuses on the prudent design and synthesis of anilide type class I HDAC inhibitors employing a functionalized pyrrolo[2,3-d]pyrimidine skeleton as the surface recognition part. Utilization of the bicyclic aromatic ring to fabricate the target compounds was envisioned to confer rigidity to the chemical architecture of MS-275 and chidamide. In-vitro enzymatic and cellular assays led to the identification of compound 7 as a potent inhibitor of HDAC1 and 2 isoform that exerted substantial cell growth inhibitory effects against human breast MDA-MB-231, cervical HeLa, breast MDA-MB-468, colorectal DLD1, and colorectal HCT116 cell lines with an IC50 values of 0.05-0.47 µM, better than MS-275 and chidamide. In addition, the anilide 7 was also endowed with a superior antiproliferative profile than MS275 and chidamide towards the human cutaneous T cell lymphoma (HH and HuT78), leukemia (HL60 and KG-1), and HDACi sensitive/resistant gastric cell lines (YCC11 and YCC3/7). Exhaustive exploration of the construct 7 confirmed it to be a microtubule-targeting agent that could trigger the cell-cycle arrest in mitosis. In pursuit of extracting the benefits of evidenced microtubule-destabilizing activity of the anilide 7, it was further evaluated against non-small-cell lung cancer cell lines as well as the multiple-drug resistant uterine cancer cell line (MES-SA/Dx5) and overwhelmingly positive results in context of inhibitory effects were attained. Furthermore, molecular modelling studies were performed and some key interactions of the anilide 7 with the amino acid residues of the active site of HDAC1 isoform and tubulin were figured out.


Asunto(s)
Anilidas/farmacología , Antineoplásicos/farmacología , Inhibidores de Histona Desacetilasas/farmacología , Pirimidinas/farmacología , Pirroles/farmacología , Moduladores de Tubulina/farmacología , Aminopiridinas/química , Anilidas/síntesis química , Anilidas/metabolismo , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Apoptosis/efectos de los fármacos , Benzamidas/química , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Inhibidores de Histona Desacetilasas/síntesis química , Inhibidores de Histona Desacetilasas/metabolismo , Humanos , Simulación del Acoplamiento Molecular , Estructura Molecular , Unión Proteica , Piridinas/química , Pirimidinas/síntesis química , Pirimidinas/metabolismo , Pirroles/síntesis química , Pirroles/metabolismo , Relación Estructura-Actividad , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/síntesis química , Moduladores de Tubulina/metabolismo
10.
Life (Basel) ; 11(12)2021 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-34947945

RESUMEN

Many active substances from marine organisms are produced by symbiotic microorganisms such as bacteria, fungi, and algae. Secondary metabolites from marine actinomycetes exhibited several biological activities and provided interesting drug leads. This study reported the isolation of Lu01-M, a secondary metabolite from the marine actinomycetes Streptomyces sp., with potent anti-proliferative activity against prostate cancers. Lu01-M blocked cell proliferation with IC50 values of 1.03 ± 0.31, 2.12 ± 0.38, 1.27 ± 0.25 µg/mL in human prostate cancer PC3, DU145, and LNCaP cells, respectively. Lu01-M induced cytotoxic activity through multiple mechanisms including cell apoptosis, necroptosis, autophagy, ER stress, and inhibiting colony formation and cell migration. Lu01-M induced cell cycle arrest at the G2/M phase and DNA damage. However, the activity of autophagy induced survival response in cancer cells. Our findings suggested that Lu01-M holds the potential to be developed as an anti-cancer agent against prostate cancers.

11.
Eur J Med Chem ; 196: 112291, 2020 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-32325365

RESUMEN

This study reports the design, synthesis and evaluation of a series of histone deacetylase (HDAC) inhibitors containing purine/purine isoster as a capping group and an N-(2-aminophenyl)-benzamide unit. In vitro cytotoxicity studies reveal that benzamide 14 suppressed the growth of triple-negative breast cancer cells MDA-MB-231 (IC50 = 1.48 µM), MDA-MB-468 (IC50 = 0.65 µM), and liver cancer cells HepG2 (IC50 = 2.44 µM), better than MS-275 (5) and Chidamide (6). Compared to the well-known HDAC inhibitor SAHA, 14 showed a higher toxicity (IC50 = 0.33 µM) in three leukemic cell lines, K-562, KG-1 and THP-1. Moreover, 14 was found to be equally virulent in the HDAC-sensitive and -resistant gastric cell lines, YCC11 and YCC3/7, respectively, indicating the potential of 14 to overcome HDACi resistance. Furthermore, substantial inhibitory effects more pronounced than MS-275 (5) and Chidamide (6) were displayed by 14 towards HDAC1, 2 and 3 isoforms with IC50 values of 0.108, 0.585 and 0.563 µM respectively. Compound 14 also exhibited a potent antitumor efficacy in human MDA-MB-231 breast cancer xenograft mouse model, providing a potential lead for the development of anticancer agents.


Asunto(s)
Antineoplásicos/farmacología , Benzamidas/farmacología , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Purinas/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Benzamidas/síntesis química , Benzamidas/química , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Células Hep G2 , Inhibidores de Histona Desacetilasas/síntesis química , Inhibidores de Histona Desacetilasas/química , Humanos , Masculino , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Simulación del Acoplamiento Molecular , Estructura Molecular , Purinas/química , Relación Estructura-Actividad , Células Tumorales Cultivadas
12.
Eur J Med Chem ; 199: 112490, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32546328

RESUMEN

This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the authors. The authors regret to inform that they would like to withdraw this accepted article, due to serious errors in authorship, affiliations, material sources and supporting grant names/numbers. The authors sincerely apologize for these oversights and miscommunications the study caused.

13.
Eur J Med Chem ; 190: 112074, 2020 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-32045788

RESUMEN

A series of thiochromeno[2,3-c]quinolin-12-one derivatives with various substitutions were synthesized and evaluated as topoisomerase (Topo) inhibitors. Six (8, 10, 12, 14, 19, and 26) of 23 compounds showed strong inhibitory activities against Topo-mediated DNA relaxation and proliferation of five human cell lines including breast (MDA-MB-231, MDA-MB-468 and MCF7), colorectal (HCT116) and non-small cell lung (H1299) cancers. Among these, compounds 14 and 26 exhibited full inhibitory activities against Topo I at 3 µM and Topo IIα at 1 µM. Cancer cells treated with 26 accumulated DNA damage and were arrested at the G2/M phase. With time, cells proceeded to apoptosis, as revealed by increased amounts of cells with fragmented DNA and cleavage of caspase-8 and -9. In contrast, normal breast epithelial cells showed low sensitivity to 26. Taken together, our study identifies 26 as a potent Topo dual-inhibitor with low toxicity to normal cells, and elucidates that the terminal amino group of N-2-aminoethylamino or N-3-aminopropylamino at the 6th position and 8,10-di-halogen substituents on thiochromeno[2,3-c]quinolin-12-one are critical for the Topo-inhibiting and cancer-killing activities.

14.
FEBS Lett ; 593(5): 504-517, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30758841

RESUMEN

Src plays a critical role in regulating cellular responses induced by protease-activated receptor 1 (PAR1). Here, we found that PAR1 activation induces lysosomal degradation of Src. Src is associated and trafficked together with activated PAR1 to early endosomes and then sorted to lysosomes for degradation. Blocking agonist-induced endocytosis of PAR1 by inhibition of dynamin activity suppresses PAR1-induced degradation of Src. However, Src activity is neither required for agonist-induced PAR1 internalization nor required for Src degradation upon PAR1 activation. We show that PAR1 activation triggers endocytosis-dependent lysosomal degradation of Src in both human embryonic kidney 293 and human umbilical vein endothelial cells. Our finding provides a new paradigm for how an irreversibly activated receptor regulates its downstream signalling.


Asunto(s)
Endocitosis , Lisosomas/metabolismo , Receptor PAR-1/metabolismo , Familia-src Quinasas/metabolismo , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Proteolisis , Transducción de Señal
15.
Sci Adv ; 4(8): eaat4985, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30101194

RESUMEN

DNA sequence and epigenetic information embedded in chromatin must be faithfully duplicated and transmitted to daughter cells during cell division. However, how chromatin assembly and DNA replication are integrated remains unclear. We examined the contribution of the Tousled-like kinases 1 and 2 (TLK1/TLK2) to chromatin assembly and maintenance of replication fork integrity. We show that TLK activity is required for DNA replication and replication-coupled nucleosome assembly and that lack of TLK activity leads to replication fork stalling and the accumulation of single-stranded DNA, a phenotype distinct from ASF1 depletion. Consistent with these results, sustained TLK depletion gives rise to replication-dependent DNA damage and p53-dependent cell cycle arrest in G1. We find that deficient replication-coupled de novo nucleosome assembly renders replication forks unstable and highly dependent on the ATR and CHK1 checkpoint kinases, as well as poly(adenosine 5'-diphosphate-ribose) polymerase (PARP) activity, to avoid collapse. Human cancer data revealed frequent up-regulation of TLK genes and an association with poor patient outcome in multiple types of cancer, and depletion of TLK activity leads to increased replication stress and DNA damage in a panel of cancer cells. Our results reveal a critical role for TLKs in chromatin replication and suppression of replication stress and identify a synergistic lethal relationship with checkpoint signaling and PARP that could be exploited in treatment of a broad range of cancers.


Asunto(s)
Replicación del ADN/efectos de los fármacos , Neoplasias/patología , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Mutaciones Letales Sintéticas , Puntos de Control del Ciclo Celular/efectos de los fármacos , Cromatina/genética , Daño del ADN , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Fosforilación , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal , Células Tumorales Cultivadas
16.
Clin Chim Acta ; 320(1-2): 37-42, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11983198

RESUMEN

BACKGROUND: Human herpesvirus 8 (HHV-8), a gamma-herpesvirus, is associated with Kaposi's sarcoma (KS). A variety of assays for the diagnosis HHV-8 infection have been developed. However, the accuracy of current HHV-8 antibody testing is uncertain, mainly in patients with asymptomatic HHV-8 infection. METHODS: We utilized a new immunoblot assay, comprised of three recombinant HHV-8 encoded proteins (ORF16, ORF57, and ORF71) to determine the seroprevalence of HHV-8 in Taiwan. ORF16 encodes a Bcl-2 homologue and is expressed only in the lytic cycle. ORF57 is an immediate-early gene product and can activate gene expression by a post-transcriptional mechanism. ORF71 encodes a viral FLICE inhibitory protein and is a latently expressed protein. We collected 6 KS(+)/HIV(-) patients, 10 KS(-)/HIV(+) individuals, 174 blood donors, and 62 non-KS cancer patients in this study. RESULTS: Among the different populations, ORF57 showed highest reactivities. The frequency of positive results for KS(+)/HIV(-), KS(-)/HIV(+), blood donors, and non-KS cancer patients were 83.3%, 40%, 23%, and 12.9%, respectively. Compared to the widely used lytic immunofluorescence assay, our immunoblotting assay using ORF57 showed higher reactivities among the blood donors. CONCLUSIONS: ORF57 is useful for the detection of HHV-8 infection, especially during the asymptomatic infections.


Asunto(s)
Anticuerpos Antivirales/sangre , Antígenos Virales/inmunología , Herpesvirus Humano 8/inmunología , Adulto , Antígenos Virales/genética , Donantes de Sangre , Western Blotting , Infecciones por Herpesviridae/diagnóstico , Infecciones por Herpesviridae/epidemiología , Infecciones por Herpesviridae/inmunología , Herpesvirus Humano 8/genética , Humanos , Persona de Mediana Edad , Sistemas de Lectura Abierta/genética , Proteínas Recombinantes/inmunología , Sensibilidad y Especificidad , Estudios Seroepidemiológicos , Pruebas Serológicas , Taiwán/epidemiología
17.
Nat Cell Biol ; 16(3): 281-93, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24561620

RESUMEN

To maintain genome function and stability, DNA sequence and its organization into chromatin must be duplicated during cell division. Understanding how entire chromosomes are copied remains a major challenge. Here, we use nascent chromatin capture (NCC) to profile chromatin proteome dynamics during replication in human cells. NCC relies on biotin-dUTP labelling of replicating DNA, affinity purification and quantitative proteomics. Comparing nascent chromatin with mature post-replicative chromatin, we provide association dynamics for 3,995 proteins. The replication machinery and 485 chromatin factors such as CAF-1, DNMT1 and SUV39h1 are enriched in nascent chromatin, whereas 170 factors including histone H1, DNMT3, MBD1-3 and PRC1 show delayed association. This correlates with H4K5K12diAc removal and H3K9me1 accumulation, whereas H3K27me3 and H3K9me3 remain unchanged. Finally, we combine NCC enrichment with experimentally derived chromatin probabilities to predict a function in nascent chromatin for 93 uncharacterized proteins, and identify FAM111A as a replication factor required for PCNA loading. Together, this provides an extensive resource to understand genome and epigenome maintenance.


Asunto(s)
Cromatina/metabolismo , Replicación del ADN , Proteoma/metabolismo , Receptores Virales/metabolismo , Ensamble y Desensamble de Cromatina , Proteínas Cromosómicas no Histona/aislamiento & purificación , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/aislamiento & purificación , Proteínas de Unión al ADN/metabolismo , Células HeLa , Histonas/aislamiento & purificación , Histonas/metabolismo , Humanos , Antígeno Nuclear de Célula en Proliferación/metabolismo , Transporte de Proteínas , Proteoma/aislamiento & purificación , Proteómica , Puntos de Control de la Fase S del Ciclo Celular
18.
Cell Rep ; 2(3): 568-79, 2012 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-22999938

RESUMEN

Tumor suppressor gene silencing through cytosine methylation contributes to cancer formation. Whether DNA demethylation enzymes counteract this oncogenic effect is unknown. Here, we show that TET1, a dioxygenase involved in cytosine demethylation, is downregulated in prostate and breast cancer tissues. TET1 depletion facilitates cell invasion, tumor growth, and cancer metastasis in prostate xenograft models and correlates with poor survival rates in breast cancer patients. Consistently, enforced expression of TET1 reduces cell invasion and breast xenograft tumor formation. Mechanistically, TET1 suppresses cell invasion through its dioxygenase and DNA binding activities. Furthermore, TET1 maintains the expression of tissue inhibitors of metalloproteinase (TIMP) family proteins 2 and 3 by inhibiting their DNA methylation. Concurrent low expression of TET1 and TIMP2 or TIMP3 correlates with advanced node status in clinical samples. Together, these results illustrate a mechanism by which TET1 suppresses tumor development and invasion partly through downregulation of critical gene methylation.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas de Unión al ADN/biosíntesis , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas/biosíntesis , Inhibidor Tisular de Metaloproteinasa-2/biosíntesis , Inhibidor Tisular de Metaloproteinasa-3/biosíntesis , Proteínas Supresoras de Tumor/biosíntesis , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Metilación de ADN/genética , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Proteínas de Unión al ADN/genética , Dioxigenasas/genética , Dioxigenasas/metabolismo , Regulación hacia Abajo/genética , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Oxigenasas de Función Mixta , Invasividad Neoplásica , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas/genética , Inhibidor Tisular de Metaloproteinasa-2/genética , Inhibidor Tisular de Metaloproteinasa-3/genética , Proteínas Supresoras de Tumor/genética
19.
Cell Res ; 21(4): 642-53, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21221131

RESUMEN

Glucose-regulated protein 78 (GRP78), a key regulator of endoplasmic reticulum (ER) stress, facilitates cancer cell growth and viral replication. The mechanism leading to grp78 gene activation during viral infection is largely unknown. In this study, we show that the immediate-early 1 (IE1-72) protein of the human cytomegalovirus (HCMV) is essential for HCMV-mediated GRP78 activation. IE1-72 upregulated grp78 gene expression depending on the ATP-binding site, the zinc-finger domain and the putative leucine-zipper motif of IE1-72, as well as the ER stress response elements (ERSEs) on the grp78 promoter. The purified IE1-72 protein bound to the CCAAT box within ERSE in vitro, whereas deletion mutants of IE1-72 deficient in grp78 promoter stimulation failed to do so. Moreover, IE1-72 binding to the grp78 promoter in infected cells accompanied the recruitment of TATA box-binding protein-associated factor 1 (TAF1), a histone acetyltransferase, and the increased level of acetylated histone H4, an indicator of active-state chromatin. These results provide evidence that HCMV IE1-72 activates grp78 gene expression through direct promoter binding and modulation of the local chromatin structure, indicating an active viral mechanism of cellular chaperone induction for viral growth.


Asunto(s)
Citomegalovirus/metabolismo , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Proteínas de Choque Térmico/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Activación Transcripcional , Western Blotting , Factor de Unión a CCAAT/metabolismo , Línea Celular , Inmunoprecipitación de Cromatina , Citomegalovirus/genética , Chaperón BiP del Retículo Endoplásmico , Expresión Génica , Proteínas de Choque Térmico/genética , Histonas/metabolismo , Humanos , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Factores Asociados con la Proteína de Unión a TATA/metabolismo
20.
Cell Res ; 21(8): 1230-47, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21445097

RESUMEN

Chromatin assembly factor 1 (CAF1) consisting of p150, p60 and p48 is known to assemble histones onto newly synthesized DNA and thus maintain the chromatin structure. Here, we show that CAF1 expression was induced in human cytomegalovirus (HCMV)-infected cells, concomitantly with global chromatin decondensation. This apparent conflict was thought to result, in part, from CAF1 mislocalization to compartments of HCMV DNA synthesis through binding of its largest subunit p150 to viral immediate-early protein 2 (IE2). p150 interaction with p60 and IE2 facilitated HCMV DNA synthesis. The IE2Q548R mutation, previously reported to result in impaired HCMV growth with unknown mechanism, disrupted IE2/p150 and IE2/histones association in our study. Moreover, IE2 interaction with histones partly depends on p150, and the HCMV-induced chromatin decondensation was reduced in cells ectopically expressing the p150 mutant defective in IE2 binding. These results not only indicate that CAF1 was hijacked by IE2 to facilitate the replication of the HCMV genome, suggesting chromatin assembly plays an important role in herpesviral DNA synthesis, but also provide a model of the virus-induced chromatin instability through CAF1.


Asunto(s)
Factor 1 de Ensamblaje de la Cromatina/metabolismo , Citomegalovirus/metabolismo , Interacciones Huésped-Patógeno , Proteínas Inmediatas-Precoces/metabolismo , Transactivadores/metabolismo , Sustitución de Aminoácidos , Línea Celular , Cromatina/metabolismo , Factor 1 de Ensamblaje de la Cromatina/análisis , Ensamble y Desensamble de Cromatina , ADN Viral/metabolismo , Histonas/metabolismo , Humanos , Proteínas Inmediatas-Precoces/análisis , Proteínas Inmediatas-Precoces/genética , Unión Proteica , Subunidades de Proteína/análisis , Subunidades de Proteína/metabolismo , Transactivadores/análisis , Transactivadores/genética , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA