Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Mol Cell ; 38(5): 662-74, 2010 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-20541999

RESUMO

Expression of the INK4b/ARF/INK4a tumor suppressor locus in normal and cancerous cell growth is controlled by methylation of histone H3 at lysine 27 (H3K27me) as directed by the Polycomb group proteins. The antisense noncoding RNA ANRIL of the INK4b/ARF/INK4a locus is also important for expression of the protein-coding genes in cis, but its mechanism has remained elusive. Here we report that chromobox 7 (CBX7) within the polycomb repressive complex 1 binds to ANRIL, and both CBX7 and ANRIL are found at elevated levels in prostate cancer tissues. In concert with H3K27me recognition, binding to RNA contributes to CBX7 function, and disruption of either interaction impacts the ability of CBX7 to repress the INK4b/ARF/INK4a locus and control senescence. Structure-guided analysis reveals the molecular interplay between noncoding RNA and H3K27me as mediated by the conserved chromodomain. Our study suggests a mechanism by which noncoding RNA participates directly in epigenetic transcriptional repression.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina , Inativação Gênica , Histonas , Lisina/metabolismo , RNA não Traduzido/metabolismo , Proteínas Repressoras/metabolismo , Animais , Linhagem Celular Tumoral , Senescência Celular/fisiologia , Inibidor de Quinase Dependente de Ciclina p15/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Masculino , Metilação , Modelos Moleculares , Dados de Sequência Molecular , Família Multigênica , Ressonância Magnética Nuclear Biomolecular , Complexo Repressor Polycomb 1 , Proteínas do Grupo Polycomb , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Estrutura Terciária de Proteína , RNA não Traduzido/genética , Proteínas Repressoras/química , Proteínas Repressoras/genética , Transcrição Gênica
2.
Molecules ; 23(8)2018 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-30072621

RESUMO

Tumor suppressor p53-directed apoptosis triggers loss of normal cells, which contributes to the side-effects from anticancer therapies. Thus, small molecules with potential to downregulate the activation of p53 could minimize pathology emerging from anticancer therapies. Acetylation of p53 by the histone acetyltransferase (HAT) domain is the hallmark of coactivator CREB-binding protein (CBP) epigenetic function. During genotoxic stress, CBP HAT-mediated acetylation is essential for the activation of p53 to transcriptionally govern target genes, which control cellular responses. Here, we present a small molecule, NiCur, which blocks CBP HAT activity and downregulates p53 activation upon genotoxic stress. Computational modeling reveals that NiCur docks into the active site of CBP HAT. On CDKN1A promoter, the recruitment of p53 as well as RNA Polymerase II and levels of acetylation on histone H3 were diminished by NiCur. Specifically, NiCur reduces the levels of acetylation at lysine 27 on histone H3, which concomitantly increases the levels of trimethylation at lysine 27. Finally, NiCur attenuates p53-directed apoptosis by inhibiting the Caspase 3 activity and cleavage of Poly (ADP-ribose) polymerase (PARP) in normal gastrointestinal epithelial cells. Collectively, NiCur demonstrates the potential to reprogram the chromatin landscape and modulate biological outcomes of CBP-mediated acetylation under normal and disease conditions.


Assuntos
Proteína de Ligação a CREB/antagonistas & inibidores , Regulação para Baixo , Histonas/metabolismo , Lisina/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Acetilação , Animais , Apoptose/efeitos dos fármacos , Proteína de Ligação a CREB/química , Proteína de Ligação a CREB/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cromatina/metabolismo , Curcumina/análogos & derivados , Curcumina/síntese química , Curcumina/química , Curcumina/farmacologia , Dano ao DNA , Regulação para Baixo/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Enterócitos/efeitos dos fármacos , Enterócitos/metabolismo , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Humanos , Concentração Inibidora 50 , Metilação , Domínios Proteicos , Ratos , Relação Estrutura-Atividade
3.
Proc Natl Acad Sci U S A ; 111(50): 17995-8000, 2014 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-25453078

RESUMO

A variety of human cancers, including nonsmall cell lung (NSCLC), breast, and colon cancers, are driven by the human epidermal growth factor receptor (HER) family of receptor tyrosine kinases. Having shown that bisphosphonates, a class of drugs used widely for the therapy of osteoporosis and metastatic bone disease, reduce cancer cell viability by targeting HER1, we explored their potential utility in the prevention and therapy of HER-driven cancers. We show that bisphosphonates inhibit colony formation by HER1(ΔE746-A750)-driven HCC827 NSCLCs and HER1(wt)-expressing MB231 triple negative breast cancers, but not by HER(low)-SW620 colon cancers. In parallel, oral gavage with bisphosphonates of mice xenografted with HCC827 or MB231 cells led to a significant reduction in tumor volume in both treatment and prevention protocols. This result was not seen with mice harboring HER(low) SW620 xenografts. We next explored whether bisphosphonates can serve as adjunctive therapies to tyrosine kinase inhibitors (TKIs), namely gefitinib and erlotinib, and whether the drugs can target TKI-resistant NSCLCs. In silico docking, together with molecular dynamics and anisotropic network modeling, showed that bisphosphonates bind to TKIs within the HER1 kinase domain. As predicted from this combinatorial binding, bisphosphonates enhanced the effects of TKIs in reducing cell viability and driving tumor regression in mice. Impressively, the drugs also overcame erlotinib resistance acquired through the gatekeeper mutation T790M, thus offering an option for TKI-resistant NSCLCs. We suggest that bisphosphonates can potentially be repurposed for the prevention and adjunctive therapy of HER1-driven cancers.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/prevenção & controle , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/prevenção & controle , Difosfonatos/farmacologia , Receptores ErbB/antagonistas & inibidores , Animais , Western Blotting , Difosfonatos/uso terapêutico , Reposicionamento de Medicamentos/métodos , Feminino , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos BALB C , Simulação de Dinâmica Molecular , Ligação Proteica , Transdução de Sinais/efeitos dos fármacos , Sais de Tetrazólio , Tiazóis , Ensaio Tumoral de Célula-Tronco
4.
Proc Natl Acad Sci U S A ; 111(50): 17989-94, 2014 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-25453081

RESUMO

Bisphosphonates are the most commonly prescribed medicines for osteoporosis and skeletal metastases. The drugs have also been shown to reduce cancer progression, but only in certain patient subgroups, suggesting that there is a molecular entity that mediates bisphosphonate action on tumor cells. Using connectivity mapping, we identified human epidermal growth factor receptors (human EGFR or HER) as a potential new molecular entity for bisphosphonate action. Protein thermal shift and cell-free kinase assays, together with computational modeling, demonstrated that N-containing bisphosphonates directly bind to the kinase domain of HER1/2 to cause a global reduction in downstream signaling. By doing so, the drugs kill lung, breast, and colon cancer cells that are driven by activating mutations or overexpression of HER1. Knocking down HER isoforms thus abrogates cell killing by bisphosphonates, establishing complete HER dependence and ruling out a significant role for other receptor tyrosine kinases or the enzyme farnesyl pyrophosphate synthase. Consistent with this finding, colon cancer cells expressing low levels of HER do not respond to bisphosphonates. The results suggest that bisphosphonates can potentially be repurposed for the prevention and therapy of HER family-driven cancers.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Difosfonatos/farmacologia , Receptores ErbB/antagonistas & inibidores , Modelos Moleculares , Anisotropia , Western Blotting , Linhagem Celular Tumoral , Cristalografia , Difosfonatos/metabolismo , Receptores ErbB/química , Receptores ErbB/metabolismo , Fluorescência , Humanos , Simulação de Dinâmica Molecular , Ligação Proteica , Conformação Proteica , Sais de Tetrazólio , Tiazóis
5.
Nat Cell Biol ; 10(9): 1114-22, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19160493

RESUMO

Viruses recruit host proteins to secure viral genome maintenance and replication. However, whether they modify host histones directly to interfere with chromatin-based transcription is unknown. Here we report that Paramecium bursaria chlorella virus 1 (PBCV-1) encodes a functional SET domain histone Lys methyltransferase (HKMTase) termed vSET, which is linked to rapid inhibition of host transcription after viral infection. We show that vSET is packaged in the PBCV-1 virion, and that it contains a nuclear localization signal and probably represses host transcription by methylating histone H3 at Lys 27 (H3K27), a modification known to trigger gene silencing in eukaryotes. We also show that vSET induces cell accumulation at the G2/M phase by recruiting the Polycomb repressive complex CBX8 to the methylated H3K27 site in a heterologous system, vSET-like proteins that have H3K27 methylation activity are conserved in chlorella viruses. Our findings suggest a viral mechanism to repress gene transcription by direct modification of chromatin by PBCV-1 vSET.


Assuntos
Chlorella/genética , Epigênese Genética , Regulação da Expressão Gênica , Histona-Lisina N-Metiltransferase/metabolismo , Phycodnaviridae/enzimologia , Transcrição Gênica , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Núcleo Celular/enzimologia , Núcleo Celular/genética , Chlorella/citologia , Chlorella/virologia , Histona-Lisina N-Metiltransferase/química , Histonas/metabolismo , Metilação , Dados de Sequência Molecular , Transporte Proteico , Proteínas Repressoras/metabolismo , Proteínas Virais/química , Vírion/enzimologia
6.
J Biol Chem ; 288(32): 23458-72, 2013 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-23720780

RESUMO

Toxins play a major role in the pathogenesis of Bacillus anthracis by subverting the host defenses. However, besides toxins, B. anthracis expresses effector proteins, whose role in pathogenesis are yet to be investigated. Here we present that suppressor-of-variegation, enhancer-of-zeste, trithorax protein from B. anthracis (BaSET) methylates human histone H1, resulting in repression of NF-κB functions. Notably, BaSET is secreted and undergoes nuclear translocation to enhance H1 methylation in B. anthracis-infected macrophages. Compared with wild type Sterne, delayed growth kinetics and altered septum formation were observed in the BaSET knock-out (BaΔSET) bacilli. Uncontrolled BaSET expression during complementation of the BaSET gene in BaΔSET partially restored growth during stationary phase but resulted in substantially shorter bacilli throughout the growth cycle. Importantly, in contrast to Sterne, the BaΔSET B. anthracis is avirulent in a lethal murine bacteremia model of infection. Collectively, BaSET is required for repression of host transcription as well as proper B. anthracis growth, making it a potentially unique virulence determinant.


Assuntos
Antraz/enzimologia , Bacillus anthracis , Proteínas de Bactérias/biossíntese , Epigênese Genética , Macrófagos/metabolismo , NF-kappa B/metabolismo , Proteínas Metiltransferases/biossíntese , Transcrição Gênica , Fatores de Virulência/biossíntese , Animais , Antraz/genética , Antraz/patologia , Bacillus anthracis/enzimologia , Bacillus anthracis/genética , Bacillus anthracis/patogenicidade , Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica/fisiologia , Células HeLa , Humanos , Macrófagos/microbiologia , Macrófagos/patologia , Camundongos , NF-kappa B/genética , Proteínas Metiltransferases/genética , Fatores de Virulência/genética
7.
Proteins ; 82(7): 1534-41, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24470253

RESUMO

The fibroblast growth factor receptor (FGFR) substrate 2 (FRS2) family proteins function as scaffolding adapters for receptor tyrosine kinases (RTKs). The FRS2α proteins interact with RTKs through the phosphotyrosine-binding (PTB) domain and transfer signals from the activated receptors to downstream effector proteins. Here, we report the nuclear magnetic resonance structure of the FRS2α PTB domain bound to phosphorylated TrkB. The structure reveals that the FRS2α-PTB domain is comprised of two distinct but adjacent pockets for its mutually exclusive interaction with either nonphosphorylated juxtamembrane region of the FGFR, or tyrosine phosphorylated peptides TrkA and TrkB. The new structural insights suggest rational design of selective small molecules through targeting of the two conjunct pockets in the FRS2α PTB domain.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Proteínas Tirosina Quinases/química , Proteínas Tirosina Quinases/metabolismo , Sítios de Ligação , Humanos , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Receptor trkB
8.
Brain Sci ; 13(2)2023 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-36831729

RESUMO

During oncogenesis, alterations in driver genes called driver alterations (DAs) modulate the transcriptome, methylome and proteome through oncogenic signaling pathways. These modulatory effects of any DA may be analyzed by examining differentially expressed mRNAs (DEMs), differentially methylated genes (DMGs) and differentially expressed proteins (DEPs) between tumor samples with and without that DA. We aimed to analyze these modulations with 12 common driver genes in Isocitrate Dehydrogenase 1 wildtype glioblastomas (IDH1-W-GBs). Using Cbioportal, groups of tumor samples with and without DAs in these 12 genes were generated from the IDH1-W-GBs available from "The Cancer Genomics Atlas Firehose Legacy Study Group" (TCGA-FL-SG) on Glioblastomas (GBs). For all 12 genes, samples with and without DAs were compared for DEMs, DMGs and DEPs. We found that DAs in PTEN were unassociated with any DEM or DMG in contrast to DAs in all other drivers, which were associated with several DEMs and DMGs. This contrasting PTEN-related property of being unassociated with differential gene expression or methylation in IDH1-W-GBs was unaffected by concurrent DAs in other common drivers or by the types of DAs affecting PTEN. From the lists of DEMs and DMGs associated with some common drivers other than PTEN, enriched gene ontology terms and insights into the co-regulatory effects of these drivers on the transcriptome were obtained. The findings from this study can improve our understanding of the molecular mechanisms underlying gliomagenesis with potential therapeutic benefits.

9.
Methods ; 53(1): 97-101, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20828615

RESUMO

Human immunodeficiency virus (HIV) when integrated into a host chromosome exists in a transcriptionally inactive but replication-competent state. Such latent infection represents a major challenge to HIV eradication efforts because a permanent virus reservoir resided in the infected cell is able to spike the viral load on immune suppression or during interruption of highly active anti-retroviral therapy. Understanding the molecular mechanisms that control HIV proviral latency and its reactivation could provide new perspectives on host factors as therapeutic targets for abolishing cellular reservoirs of dormant HIV. Although the control of HIV latency is multifactorial, chromatin structure and the chromatin-associated transcriptional machinery are known to be important factors. For instance, transcription initiation of the HIV provirus involves a complex molecular interplay between chromatin-associated proteins and the virus-encoded trans-activator, Tat. The first part of this review discusses our current understanding of the elements involved in HIV transcriptional activation and viral mRNA elongation, mainly post-translational modifications of HIV Tat and its interactions with host chromatin-modifying enzymes and chromatin-remodeling complexes. The second part highlights new experimental therapeutic approaches aimed at administrating activators of HIV gene expression to reduce or eliminate the pool of latently HIV-infected cells.


Assuntos
Cromatina/metabolismo , Infecções por HIV/tratamento farmacológico , HIV-1/genética , Ativação Transcricional , Acetilação , Fármacos Anti-HIV/farmacologia , Fármacos Anti-HIV/uso terapêutico , Sítios de Ligação , Cromatina/genética , Desenho de Fármacos , Infecções por HIV/virologia , HIV-1/efeitos dos fármacos , HIV-1/fisiologia , Interações Hospedeiro-Patógeno , Humanos , Mapeamento de Interação de Proteínas/métodos , Integração Viral , Latência Viral , Fatores de Transcrição de p300-CBP/antagonistas & inibidores , Fatores de Transcrição de p300-CBP/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana/química , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo
10.
J Am Chem Soc ; 133(7): 2040-3, 2011 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-21271695

RESUMO

The CREB binding protein (CBP) is a human transcriptional coactivator consisting of several conserved functional modules, which interacts with distinct transcription factors including nuclear receptors, CREB, and STAT proteins. Despite the importance of CBP in transcriptional regulation, many questions regarding the role of its particular domains in CBP functions remain unanswered. Therefore, developing small molecules capable of selectively modulating a single domain of CBP is of invaluable aid at unraveling its prominent activities. Here we report the design, synthesis, and biological evaluation of conformationally restricted peptides as novel modulators for the acetyl-lysine binding bromodomain (BRD) of CBP. Utilizing a target structure-guided and computer-aided rational design approach, we developed a series of cyclic peptides with affinity for CBP BRD significantly greater than those of its biological ligands, including lysine-acetylated histones and tumor suppressor p53. The best cyclopeptide of the series exhibited a K(d) of 8.0 µM, representing a 24-fold improvement in affinity over that of the linear lysine 382-acetylated p53 peptide. This lead peptide is highly selective for CBP BRD over BRDs from other transcriptional proteins. Cell-based functional assays carried out in colorectal carcinoma HCT116 cells further demonstrated the efficacy of this compound to modulate p53 stability and function in response to DNA damage. Our results strongly argue that these CBP modulators can effectively inhibit p53 transcriptional activity by blocking p53K382ac binding to CBP BRD and promoting p53 instability by changes of its post-translational modification states, a different mechanism than that of the p53 inhibitors reported to date.


Assuntos
Proteína de Ligação a CREB/efeitos dos fármacos , Desenho de Fármacos , Peptídeos Cíclicos/síntese química , Proteína Supressora de Tumor p53/antagonistas & inibidores , Linhagem Celular Tumoral , Humanos , Concentração Inibidora 50 , Modelos Moleculares , Estrutura Molecular , Peptídeos Cíclicos/química , Peptídeos Cíclicos/farmacologia , Relação Estrutura-Atividade
11.
Biomolecules ; 11(3)2021 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-33803759

RESUMO

The molecular interplay between nucleosomal packaging and the chromatin landscape regulates the transcriptional programming and biological outcomes of downstream genes. An array of epigenetic modifications plays a pivotal role in shaping the chromatin architecture, which controls DNA access to the transcriptional machinery. Acetylation of the amino acid lysine is a widespread epigenetic modification that serves as a marker for gene activation, which intertwines the maintenance of cellular homeostasis and the regulation of signaling during stress. The biochemical horizon of acetylation ranges from orchestrating the stability and cellular localization of proteins that engage in the cell cycle to DNA repair and metabolism. Furthermore, lysine acetyltransferases (KATs) modulate the functions of transcription factors that govern cellular response to microbial infections, genotoxic stress, and inflammation. Due to their central role in many biological processes, mutations in KATs cause developmental and intellectual challenges and metabolic disorders. Despite the availability of tools for detecting acetylation, the mechanistic knowledge of acetylation-mediated cellular processes remains limited. This review aims to integrate molecular and structural bases of KAT functions, which would help design highly selective tools for understanding the biology of KATs toward developing new disease treatments.


Assuntos
Lisina Acetiltransferases/metabolismo , Acetilação , Reparo do DNA/genética , Reparo do DNA/fisiologia , Epigênese Genética/genética , Epigênese Genética/fisiologia , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/fisiologia , Humanos , Lisina Acetiltransferases/genética
12.
J Med Chem ; 50(10): 2285-8, 2007 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-17444627

RESUMO

Human immunodeficiency virus 1 (HIV-1) trans-activator Tat recruits the human transcriptional coactivator PCAF (p300/CREB binding protein-associated factor) to facilitate transcription of the integrated HIV-1 provirus. We report here structure-based lead optimization of small-molecule inhibitors that block selectively Tat and PCAF association in cells. Our lead optimization was guided by grand-canonical ensemble simulation of the receptor/lead complex that leads to definition of chemical modifications with improved lead affinity through displacing weakly bound water molecules at the ligand-receptor interface.


Assuntos
Proteínas de Ciclo Celular/química , Produtos do Gene tat/química , HIV-1/metabolismo , Histona Acetiltransferases/química , Modelos Moleculares , Propilaminas/química , Fatores de Transcrição/química , Proteínas de Ciclo Celular/metabolismo , Produtos do Gene tat/genética , Produtos do Gene tat/metabolismo , Genes Reporter , Repetição Terminal Longa de HIV , HIV-1/genética , Histona Acetiltransferases/metabolismo , Humanos , Ligantes , Luciferases/genética , Luciferases/metabolismo , Método de Monte Carlo , Regiões Promotoras Genéticas , Propilaminas/farmacologia , Estrutura Terciária de Proteína , Fatores de Transcrição/metabolismo , Transcrição Gênica , Água/química , Fatores de Transcrição de p300-CBP , Produtos do Gene tat do Vírus da Imunodeficiência Humana
13.
Ann N Y Acad Sci ; 1376(1): 18-28, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26919042

RESUMO

Su(var)3-9, Enhancer-of-zeste, Trithorax (SET) domain-mediated lysine methylation, one of the major epigenetic marks, has been found to regulate chromatin-mediated gene transcription. Published studies have established further that methylation is not restricted to nuclear proteins but is involved in many cellular processes, including growth, differentiation, immune regulation, and cancer progression. The biological complexity of lysine methylation emerges from its capacity to cause gene activation or gene repression owing to the specific position of methylated-lysine moieties on the chromatin. Accumulating evidence suggests that despite the absence of chromatin, viruses and prokaryotes also express SET proteins, although their functional roles remain relatively less investigated. One possibility could be that SET proteins in lower organisms have more than one biological function, for example, in regulating growth or in manipulating host transcription machinery in order to establish infection. Thus, elucidating the role of an SET protein in host-pathogen interactions requires a thorough understanding of their functions. This review discusses the biological role of lysine methylation in prokaryotes and lower eukaryotes, as well as the underlying structural complexity and functional diversity of SET proteins.


Assuntos
Interações Hospedeiro-Patógeno/genética , Lisina/metabolismo , Células Procarióticas/metabolismo , Transcrição Gênica , Sequência de Aminoácidos , Animais , Humanos , Metilação , Domínios Proteicos
14.
Cancers (Basel) ; 7(3): 1622-42, 2015 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-26295410

RESUMO

The dynamics of lysine acetylation serve as a major epigenetic mark, which regulates cellular response to inflammation, DNA damage and hormonal changes. Microarray assays reveal changes in gene expression, but cannot predict regulation of a protein function by epigenetic modifications. The present study employs computational tools to inclusively analyze microarray data to understand the potential role of acetylation during development of androgen-independent PCa. The data revealed that the androgen receptor interacts with 333 proteins, out of which at least 92 proteins were acetylated. Notably, the number of cellular proteins undergoing acetylation in the androgen-dependent PCa was more as compared to the androgen-independent PCa. Specifically, the 32 lysine-acetylated proteins in the cellular models of androgen-dependent PCa were mainly involved in regulating stability as well as pre- and post-processing of mRNA. Collectively, the data demonstrate that protein lysine acetylation plays a crucial role during the transition of androgen-dependent to -independent PCa, which importantly, could also serve as a functional axis to unravel new therapeutic targets.

15.
Mol Endocrinol ; 28(6): 872-85, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24702180

RESUMO

In prostate cancer (PCa), the functional synergy between androgen receptor (AR) and nuclear factor-κ B (NF-κB) escalates the resistance to therapeutic regimens and promotes aggressive tumor growth. Although the underlying mechanisms are less clear, gene regulatory abilities of coactivators can bridge the transcription functions of AR and NF-κB. The present study shows that MYST1 (MOZ, YBF2 and SAS2, and TIP60 protein 1) costimulates AR and NF-κB functions in PCa cells. We demonstrate that activation of NF-κB promotes deacetylation of MYST1 by sirtuin 1. Further, the mutually exclusive interactions of MYST1 with sirtuin 1 vs AR regulate the acetylation of lysine 16 on histone H4. Notably, in AR-lacking PC3 cells and in AR-depleted LNCaP cells, diminution of MYST1 activates the cleavage of poly(ADP-ribose) polymerase and caspase 3 that leads to apoptosis. In contrast, in AR-transformed PC3 cells (PC3-AR), depletion of MYST1 induces cyclin-dependent kinase (CDK) N1A/p21, which results in G2M arrest. Concomitantly, the levels of phospho-retinoblastoma, E2F1, CDK4, and CDK6 are reduced. Finally, the expression of tumor protein D52 (TPD52) was unequivocally affected in PC3, PC3-AR, and LNCaP cells. Taken together, the results of this study reveal that the functional interactions of MYST1 with AR and NF-κB are critical for PCa progression.


Assuntos
Proliferação de Células , Histona Acetiltransferases/fisiologia , NF-kappa B/fisiologia , Receptores Androgênicos/fisiologia , Acetilação , Apoptose , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Pontos de Checagem da Fase G2 do Ciclo Celular , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Neoplasias da Próstata , Processamento de Proteína Pós-Traducional , Elementos de Resposta , Sirtuína 1/metabolismo , Ativação Transcricional , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
16.
J Med Chem ; 56(22): 9251-64, 2013 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-24144283

RESUMO

BRD4, characterized by two acetyl-lysine binding bromodomains and an extra-terminal (ET) domain, is a key chromatin organizer that directs gene activation in chromatin through transcription factor recruitment, enhancer assembly, and pause release of the RNA polymerase II complex for transcription elongation. BRD4 has been recently validated as a new epigenetic drug target for cancer and inflammation. Our current knowledge of the functional differences of the two bromodomains of BRD4, however, is limited and is hindered by the lack of selective inhibitors. Here, we report our structure-guided development of diazobenzene-based small-molecule inhibitors for the BRD4 bromodomains that have over 90% sequence identity at the acetyl-lysine binding site. Our lead compound, MS436, through a set of water-mediated interactions, exhibits low nanomolar affinity (estimated Ki of 30-50 nM), with preference for the first bromodomain over the second. We demonstrated that MS436 effectively inhibits BRD4 activity in NF-κB-directed production of nitric oxide and proinflammatory cytokine interleukin-6 in murine macrophages. MS436 represents a new class of bromodomain inhibitors and will facilitate further investigation of the biological functions of the two bromodomains of BRD4 in gene expression.


Assuntos
Benzeno/química , Benzeno/farmacologia , Desenho de Fármacos , Animais , Linhagem Celular , Fenômenos Químicos , Ligantes , Camundongos , Modelos Moleculares , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Estrutura Terciária de Proteína/efeitos dos fármacos , Relação Estrutura-Atividade , Especificidade por Substrato , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo
17.
Nutr Metab (Lond) ; 8: 12, 2011 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-21371315

RESUMO

The biochemical landscape of lysine acetylation has expanded from a small number of proteins in the nucleus to a multitude of proteins in the cytoplasm. Since the first report confirming acetylation of the tumor suppressor protein p53 by a lysine acetyltransferase (KAT), there has been a surge in the identification of new, non-histone targets of KATs. Added to the known substrates of KATs are metabolic enzymes, cytoskeletal proteins, molecular chaperones, ribosomal proteins and nuclear import factors. Emerging studies demonstrate that no fewer than 2000 proteins in any particular cell type may undergo lysine acetylation. As described in this review, our analyses of cellular acetylated proteins using DAVID 6.7 bioinformatics resources have facilitated organization of acetylated proteins into functional clusters integral to cell signaling, the stress response, proteolysis, apoptosis, metabolism, and neuronal development. In addition, these clusters also depict association of acetylated proteins with human diseases. These findings not only support lysine acetylation as a widespread cellular phenomenon, but also impel questions to clarify the underlying molecular and cellular mechanisms governing target selectivity by KATs. Present challenges are to understand the molecular basis for the overlapping roles of KAT-containing co-activators, to differentiate between global versus dynamic acetylation marks, and to elucidate the physiological roles of acetylated proteins in biochemical pathways. In addition to discussing the cellular 'acetylome', a focus of this work is to present the widespread and dynamic nature of lysine acetylation and highlight the nexus that exists between epigenetic-directed transcriptional regulation and metabolism.

18.
Chem Biol ; 18(4): 531-41, 2011 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-21513889

RESUMO

As a master transcription factor in cellular responses to external stress, tumor suppressor p53 is tightly regulated. Excessive p53 activity during myocardial ischemia causes irreversible cellular injury and cardiomyocyte death. p53 activation is dependent on lysine acetylation by the lysine acetyltransferase and transcriptional coactivator CREB-binding protein (CBP) and on acetylation-directed CBP recruitment for p53 target gene expression. Here, we report a small molecule ischemin, developed with a structure-guided approach to inhibit the acetyl-lysine binding activity of the bromodomain of CBP. We show that ischemin alters post-translational modifications on p53 and histones, inhibits p53 interaction with CBP and transcriptional activity in cells, and prevents apoptosis in ischemic cardiomyocytes. Our study suggests small molecule modulation of acetylation-mediated interactions in gene transcription as a new approach to therapeutic interventions of human disorders such as myocardial ischemia.


Assuntos
Apoptose/efeitos dos fármacos , Proteína de Ligação a CREB/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Compostos Azo/química , Compostos Azo/metabolismo , Compostos Azo/farmacologia , Proteína de Ligação a CREB/química , Linhagem Celular Tumoral , Citoproteção/efeitos dos fármacos , Dano ao DNA , Descoberta de Drogas , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Modelos Moleculares , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Miócitos Cardíacos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Transdução de Sinais/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/química , Relação Estrutura-Atividade , Transcrição Gênica/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo
19.
PLoS One ; 5(5): e10486, 2010 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-20463968

RESUMO

A hallmark of p53 function is to regulate a transcriptional program in response to extracellular and intracellular stress that directs cell cycle arrest, apoptosis, and cellular senescence. Independent of the role of p53 in the nucleus, some of the anti-proliferative functions of p53 reside within the mitochondria [1]. p53 can arrest cell growth in response to mitochondrial p53 in an EJ bladder carcinoma cell environment that is naïve of p53 function until induced to express p53 [2]. TP53 can independently partition with endogenous nuclear and mitochondrial proteins consistent with the ability of p53 to enact senescence. In order to address the role of p53 in navigating cellular senescence through the mitochondria, we identified SirT3 to rescue EJ/p53 cells from induced p53-mediated growth arrest. Human SirT3 function appears coupled with p53 early during the initiation of p53 expression in the mitochondria by biochemical and cellular localization analysis. Our evidence suggests that SirT3 partially abrogates p53 activity to enact growth arrest and senescence. Additionally, we identified the chaperone protein BAG-2 in averting SirT3 targeting of p53 -mediated senescence. These studies identify a complex relationship between p53, SirT3, and chaperoning factor BAG-2 that may link the salvaging and quality assurance of the p53 protein for control of cellular fate independent of transcriptional activity.


Assuntos
Mitocôndrias/enzimologia , Sirtuína 3/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Acetilação , Linhagem Celular Tumoral , Proliferação de Células , Senescência Celular , Deleção de Genes , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Chaperonas Moleculares , NAD/metabolismo , Fenótipo , Ligação Proteica , Estabilidade Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Frações Subcelulares/enzimologia , Proteína Supressora de Tumor p53/química , Neoplasias da Bexiga Urinária/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA