Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 66(4): 503-516.e5, 2017 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-28525742

RESUMO

ADP-ribosylation of proteins is emerging as an important regulatory mechanism. Depending on the family member, ADP-ribosyltransferases either conjugate a single ADP-ribose to a target or generate ADP-ribose chains. Here we characterize Parp9, a mono-ADP-ribosyltransferase reported to be enzymatically inactive. Parp9 undergoes heterodimerization with Dtx3L, a histone E3 ligase involved in DNA damage repair. We show that the Dtx3L/Parp9 heterodimer mediates NAD+-dependent mono-ADP-ribosylation of ubiquitin, exclusively in the context of ubiquitin processing by E1 and E2 enzymes. Dtx3L/Parp9 ADP-ribosylates the carboxyl group of Ub Gly76. Because Gly76 is normally used for Ub conjugation to substrates, ADP-ribosylation of the Ub carboxyl terminus precludes ubiquitylation. Parp9 ADP-ribosylation activity therefore restrains the E3 function of Dtx3L. Mutation of the NAD+ binding site in Parp9 increases the DNA repair activity of the heterodimer. Moreover, poly(ADP-ribose) binding to the Parp9 macrodomains increases E3 activity. Dtx3L heterodimerization with Parp9 enables NAD+ and poly(ADP-ribose) regulation of E3 activity.


Assuntos
Adenosina Difosfato Ribose/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/enzimologia , Poli(ADP-Ribose) Polimerases/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina/metabolismo , Linhagem Celular Tumoral , Reparo do DNA , Células HEK293 , Humanos , Mutação , NAD/metabolismo , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patologia , Poli(ADP-Ribose) Polimerases/genética , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Interferência de RNA , Fatores de Tempo , Transfecção , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
2.
Nucleic Acids Res ; 51(18): 9863-9879, 2023 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-37665026

RESUMO

Repair of DSB induced by IR is primarily carried out by Non-Homologous End Joining (NHEJ), a pathway in which 53BP1 plays a key role. We have discovered that the EMT-inducing transcriptional repressor ZEB1 (i) interacts with 53BP1 and that this interaction occurs rapidly and is significantly amplified following exposure of cells to IR; (ii) is required for the localization of 53BP1 to a subset of double-stranded breaks, and for physiological DSB repair; (iii) co-localizes with 53BP1 at IR-induced foci (IRIF); (iv) promotes NHEJ and inhibits Homologous Recombination (HR); (v) depletion increases resection at DSBs and (vi) confers PARP inhibitor (PARPi) sensitivity on BRCA1-deficient cells. Lastly, ZEB1's effects on repair pathway choice, resection, and PARPi sensitivity all rely on its homeodomain. In contrast to the well-characterized therapeutic resistance of high ZEB1-expressing cancer cells, the novel ZEB1-53BP1-shieldin resection axis described here exposes a therapeutic vulnerability: ZEB1 levels in BRCA1-deficient tumors may serve as a predictive biomarker of response to PARPis.


Assuntos
Reparo do DNA por Junção de Extremidades , Homeobox 1 de Ligação a E-box em Dedo de Zinco , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição/genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , Humanos , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo
3.
Ann Surg ; 278(3): e589-e597, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-36538614

RESUMO

OBJECTIVE: Develop a predictive model to identify patients with 1 pathologic lymph node (pLN) versus >1 pLN using machine learning applied to gene expression profiles and clinical data as input variables. BACKGROUND: Standard management for clinically detected melanoma lymph node metastases is complete therapeutic LN dissection (TLND). However, >40% of patients with a clinically detected melanoma lymph node will only have 1 pLN on final review. Recent data suggest that targeted excision of just the single enlarged LN may provide excellent regional control, with less morbidity than TLND. The selection of patients for less morbid surgery requires accurate identification of those with only 1 pLN. METHODS: The Cancer Genome Atlas database was used to identify patients who underwent TLND for melanoma. Pathology reports in The Cancer Genome Atlas were reviewed to identify the number of pLNs. Patients were included for machine learning analyses if RNA sequencing data were available from a pLN. After feature selection, the top 20 gene expression and clinical input features were used to train a ridge logistic regression model to predict patients with 1 pLN versus >1 pLN using 10-fold cross-validation on 80% of samples. The model was then tested on the remaining holdout samples. RESULTS: A total of 153 patients met inclusion criteria: 64 with one pLN (42%) and 89 with >1 pLNs (58%). Feature selection identified 1 clinical (extranodal extension) and 19 gene expression variables used to predict patients with 1 pLN versus >1 pLN. The ridge logistic regression model identified patient groups with an accuracy of 90% and an area under the receiver operating characteristic curve of 0.97. CONCLUSIONS: Gene expression profiles together with clinical variables can distinguish melanoma metastasis patients with 1 pLN versus >1 pLN. Future models trained using positron emission tomography/computed tomography imaging, gene expression, and relevant clinical variables may further improve accuracy and may predict patients who can be managed with a targeted LN excision rather than a complete TLND.


Assuntos
Melanoma , Neoplasias Cutâneas , Humanos , Metástase Linfática/patologia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/cirurgia , Neoplasias Cutâneas/patologia , Melanoma/genética , Melanoma/cirurgia , Melanoma/patologia , Linfonodos/patologia , Tomada de Decisões , Excisão de Linfonodo , Estudos Retrospectivos
4.
Nucleic Acids Res ; 49(20): 11787-11799, 2021 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-34718766

RESUMO

Extrachromosomal circular DNA (eccDNA) are present within all eukaryotic organisms and actively contribute to gene expression changes. MicroDNA (200-1000bp) are the most abundant type of eccDNA and can amplify tRNA, microRNA, and novel si-like RNA sequences. Due to the heterogeneity of microDNA and the limited technology to directly quantify circular DNA molecules, the specific DNA repair pathways that contribute to microDNA formation have not been fully elucidated. Using a sensitive and quantitative assay that quantifies eight known abundant microDNA, we report that microDNA levels are dependent on resection after double-strand DNA break (DSB) and repair by Microhomology Mediated End Joining (MMEJ). Further, repair of DSB without resection by canonical Non-Homologous End Joining (c-NHEJ) diminishes microDNA formation. MicroDNA levels are induced locally even by a single site-directed DSB, suggesting that excision of genomic DNA by two closely spaced DSB is not necessary for microDNA formation. Consistent with all this, microDNA levels accumulate as cells undergo replication in S-phase, when DNA breaks and repair are elevated, and microDNA levels are decreased if DNA synthesis is prevented. Thus, formation of microDNA occurs during the repair of endogenous or induced DNA breaks by resection-based DNA repair pathways.


Assuntos
Dano ao DNA , Reparo do DNA por Junção de Extremidades , DNA Circular/genética , Replicação do DNA , Células HEK293 , Células HeLa , Humanos
5.
Proc Natl Acad Sci U S A ; 117(10): 5329-5338, 2020 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-32094185

RESUMO

Accumulating evidence suggests participation of RNA-binding proteins with intrinsically disordered domains (IDPs) in the DNA damage response (DDR). These IDPs form liquid compartments at DNA damage sites in a poly(ADP ribose) (PAR)-dependent manner. However, it is greatly unknown how the IDPs are involved in DDR. We have shown previously that one of the IDPs RBM14 is required for the canonical nonhomologous end joining (cNHEJ). Here we show that RBM14 is recruited to DNA damage sites in a PARP- and RNA polymerase II (RNAPII)-dependent manner. Both KU and RBM14 are required for RNAPII-dependent generation of RNA:DNA hybrids at DNA damage sites. In fact, RBM14 binds to RNA:DNA hybrids. Furthermore, RNA:DNA hybrids and RNAPII are detected at gene-coding as well as at intergenic areas when double-strand breaks (DSBs) are induced. We propose that the cNHEJ pathway utilizes damage-induced transcription and intrinsically disordered protein RBM14 for efficient repair of DSBs.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Intrinsicamente Desordenadas/metabolismo , Proteínas de Ligação a RNA/metabolismo , RNA/metabolismo , Quimera , Células HEK293 , Humanos , Autoantígeno Ku/metabolismo , Hibridização de Ácido Nucleico , Domínios Proteicos , RNA/genética , RNA Polimerase II/metabolismo , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/genética
6.
Sensors (Basel) ; 23(21)2023 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-37960642

RESUMO

Self-driving vehicles demand efficient and reliable depth-sensing technologies. Lidar, with its capability for long-distance, high-precision measurement, is a crucial component in this pursuit. However, conventional mechanical scanning implementations suffer from reliability, cost, and frame rate limitations. Solid-state lidar solutions have emerged as a promising alternative, but the vast amount of photon data processed and stored using conventional direct time-of-flight (dToF) prevents long-distance sensing unless power-intensive partial histogram approaches are used. In this paper, we introduce a groundbreaking 'guided' dToF approach, harnessing external guidance from other onboard sensors to narrow down the depth search space for a power and data-efficient solution. This approach centers around a dToF sensor in which the exposed time window of independent pixels can be dynamically adjusted. We utilize a 64-by-32 macropixel dToF sensor and a pair of vision cameras to provide the guiding depth estimates. Our demonstrator captures a dynamic outdoor scene at 3 fps with distances up to 75 m. Compared to a conventional full histogram approach, on-chip data is reduced by over twenty times, while the total laser cycles in each frame are reduced by at least six times compared to any partial histogram approach. The capability of guided dToF to mitigate multipath reflections is also demonstrated. For self-driving vehicles where a wealth of sensor data is already available, guided dToF opens new possibilities for efficient solid-state lidar.

7.
Nucleic Acids Res ; 48(21): e126, 2020 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-33068408

RESUMO

DNA double-strand breaks (DSBs) are highly cytotoxic lesions that can lead to chromosome rearrangements, genomic instability and cell death. Consequently, cells have evolved multiple mechanisms to efficiently repair DSBs to preserve genomic integrity. We have developed a DSB repair assay system, designated CDDR (CRISPR-Cas9-based Dual-fluorescent DSB Repair), that enables the detection and quantification of DSB repair outcomes in mammalian cells with high precision. CDDR is based on the introduction and subsequent resolution of one or two DSB(s) in an intrachromosomal fluorescent reporter following the expression of Cas9 and sgRNAs targeting the reporter. CDDR can discriminate between high-fidelity (HF) and error-prone non-homologous end-joining (NHEJ), as well as between proximal and distal NHEJ repair. Furthermore, CDDR can detect homology-directed repair (HDR) with high sensitivity. Using CDDR, we found HF-NHEJ to be strictly dependent on DNA Ligase IV, XRCC4 and XLF, members of the canonical branch of NHEJ pathway (c-NHEJ). Loss of these genes also stimulated HDR, and promoted error-prone distal end-joining. Deletion of the DNA repair kinase ATM, on the other hand, stimulated HF-NHEJ and suppressed HDR. These findings demonstrate the utility of CDDR in characterizing the effect of repair factors and in elucidating the balance between competing DSB repair pathways.


Assuntos
Bioensaio/métodos , Sistemas CRISPR-Cas/genética , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Corantes Fluorescentes/química , Genes Reporter , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Linhagem Celular Tumoral , DNA Ligase Dependente de ATP/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células HEK293 , Humanos
8.
Curr Issues Mol Biol ; 40: 189-220, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32606249

RESUMO

DNA replication is a tightly regulated conserved process that ensures the faithful transmission of genetic material to define heritable phenotypic traits. Perturbations in this process result in genomic instability, mutagenesis, and diseases, including malignancy. Proteins involved in the initiation, progression, and termination of DNA replication are subject to a plethora of reversible post-translational modifications (PTMs) to provide a proper temporal and spatial control of replication. Among these, modifications involving the covalent attachment of the small protein ubiquitin or the small ubiquitin-like modifier (SUMO) to replication and replication-associated proteins are particularly important for the proper regulation of DNA replication as well as for optimal cellular responses to replication stress. In this article, we describe how the ubiquitination and SUMOylation processes impact DNA replication in eukaryotes and highlight the consequences of deregulated signals emanating from these two versatile regulatory pathways on cellular activities.


Assuntos
Replicação do DNA/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sumoilação/genética , Ubiquitina/metabolismo , Animais , Ciclo Celular/genética , Proliferação de Células/genética , Dano ao DNA/genética , Humanos , Complexo de Endopeptidases do Proteassoma/metabolismo
9.
Mol Cell ; 49(6): 1147-58, 2013 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-23478445

RESUMO

The Cul4-Cdt2 (CRL4(Cdt2)) E3 ubiquitin ligase is a master regulator of cell-cycle progression and genome stability. Despite its central role in the degradation of many cell-cycle regulators, e.g., Cdt1, p21, and Pr-Set7/Set8, little is known about the regulation of its activity. We report that Cdt2 is autoubiquitylated by the CRL4A E3 ubiquitin ligase. Cdt2 is additionally polyubiquitylated and degraded by Cul1-FBXO11 (CRL1(FBXO11)). CRL1(FBXO11)-mediated degradation of Cdt2 stabilizes p21 and Set8, and this is important during the response to TGF-ß, with the Set8 induction being important for turning off the activation of Smad2. The migration of epithelial cells is also stimulated by CRL1(FBXO11)-mediated downregulation of Cdt2 and the consequent stabilization of Set8. This is an interesting example of cross-regulation between specific Cullin 4 and Cullin 1 E3 ubiquitin ligases and highlights the role of ubiquitylation in regulating cellular responses to TGF-ß and the migration of epithelial cells.


Assuntos
Movimento Celular , Proteínas F-Box/fisiologia , Histona-Lisina N-Metiltransferase/metabolismo , Proteínas Nucleares/metabolismo , Proteína-Arginina N-Metiltransferases/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Sequência de Aminoácidos , Linhagem Celular , Sequência Conservada , Proteínas Culina/fisiologia , Cicloeximida/farmacologia , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Leupeptinas/farmacologia , Dados de Sequência Molecular , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Ligação Proteica , Inibidores da Síntese de Proteínas/farmacologia , Proteína-Arginina N-Metiltransferases/genética , Proteína-Arginina N-Metiltransferases/metabolismo , Proteólise , RNA Interferente Pequeno/genética , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta/fisiologia
10.
Ann Surg ; 270(4): 712-722, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31425296

RESUMO

OBJECTIVE: To understand role of barrier molecules in melanomas. BACKGROUND: We have reported poor patient survival and low immune infiltration of melanomas that overexpress a set of genes that include filaggrin (FLG), dystonin (DST), junction plakoglobin (JUP), and plakophilin-3 (PKP3), and are involved in cell-cell adhesions. We hypothesized that these associations are causal, either by interfering with immune cell infiltration or by enhancing melanoma cell growth. METHODS: FLG and DST were knocked out by CRISPR/Cas9 in human DM93 and murine B16-F1 melanoma cells. PKP3 and JUP were overexpressed in murine B16-AAD and human VMM39 melanoma cells by lentiviral transduction. These cell lines were evaluated in vitro for cell proliferation and in vivo for tumor burden, immune composition, cytokine expression, and vascularity. RESULTS: Immune infiltrates were not altered by these genes. FLG/DST knockout reduced proliferation of human DM93 melanoma in vitro, and decreased B16-F1 tumor burden in vivo. Overexpression of JUP, but not PKP3, in B16-AAD significantly increased tumor burden, increased VEGF-A, reduced IL-33, and enhanced vascularity. CONCLUSIONS: FLG and DST support melanoma cell growth in vitro and in vivo. Growth effects of JUP were only evident in vivo, and may be mediated, in part, by enhancing angiogenesis. In addition, growth-promoting effects of FLG and DST in vitro suggest that these genes may also support melanoma cell proliferation through angiogenesis-independent pathways. These findings identify FLG, DST, and JUP as novel therapeutic targets whose down-regulation may provide clinical benefit to patients with melanoma.


Assuntos
Biomarcadores Tumorais/metabolismo , Distonina/metabolismo , Proteínas de Filamentos Intermediários/metabolismo , Melanoma/patologia , Neovascularização Patológica/metabolismo , gama Catenina/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Citocinas/metabolismo , Proteínas Filagrinas , Citometria de Fluxo , Imunofluorescência , Humanos , Melanoma/imunologia , Melanoma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
11.
Mol Cell ; 37(1): 143-9, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20129063

RESUMO

Monoubiquitination of proliferating cell nuclear antigen (PCNA) is a critical posttranslational modification essential for DNA repair by translesion DNA synthesis (TLS). The Rad18 E3 ubiquitin ligase cooperates with the E2 Rad6 to monoubiquitinate PCNA in response to DNA damage. How PCNA is monoubiquitinated in unperturbed cells and whether this plays a role in the repair of DNA associated with replication is not known. We show that the CRL4(Cdt2) E3 ubiquitin ligase complex promotes PCNA monoubiqutination in proliferating cells in the absence of external DNA damage independent of Rad18. PCNA monoubiquitination via CRL4(Cdt2) is constitutively antagonized by the action of the ubiquitin-specific protease 1 (USP1). In vitro, CRL4(Cdt2) monoubiquitinates PCNA at Lys164, the same residue that is monoubiquitinated by Rad18. Significantly, CRL4(Cdt2) is required for TLS in nondamaged cells via a mechanism that is dependent on PCNA monoubiquitination. We propose that CRL4(Cdt2) regulates PCNA-dependent TLS associated with stresses accompanying DNA replication.


Assuntos
Dano ao DNA , Proteínas Nucleares/fisiologia , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ubiquitina-Proteína Ligases/fisiologia , Linhagem Celular , Replicação do DNA , Proteínas de Ligação a DNA/fisiologia , Células HeLa , Humanos , Proteínas Nucleares/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
12.
Mol Cell ; 40(1): 9-21, 2010 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-20932471

RESUMO

PR-Set7/Set8 is a cell-cycle-regulated enzyme that monomethylates lysine 20 of histone H4 (H4K20). Set8 and monomethylated H4K20 are virtually undetectable during G1 and S phases of the cell cycle but increase in late S and in G2. We identify CRL4(Cdt2) as the principal E3 ubiquitin ligase responsible for Set8 proteolytic degradation in the S phase of the cell cycle, which requires Set8-PCNA interaction. Inactivation of the CRL4-Cdt2-PCNA-Set8 degradation axis results in (1) DNA damage and the induction of tumor suppressor p53 and p53-transactivated proapoptotic genes, (2) delayed progression through G2 phase of the cell cycle due to activation of the G2/M checkpoint, (3) specific repression of histone gene transcription and depletion of the histone proteins, and (4) repression of E2F1-dependent gene transcription. These results demonstrate a central role of CRL4(Cdt2)-dependent cell-cycle regulation of Set8 for the maintenance of a stable epigenetic state essential for cell viability.


Assuntos
Ciclo Celular , Proliferação de Células , Montagem e Desmontagem da Cromatina , Proteínas Culina/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Proteínas Nucleares/metabolismo , Processamento de Proteína Pós-Traducional , Sequência de Aminoácidos , Apoptose/genética , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/efeitos da radiação , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Sobrevivência Celular , Montagem e Desmontagem da Cromatina/efeitos dos fármacos , Montagem e Desmontagem da Cromatina/efeitos da radiação , Proteínas Culina/genética , Dano ao DNA , Regulação para Baixo , Fator de Transcrição E2F1/genética , Fator de Transcrição E2F1/metabolismo , Epigênese Genética , Células HeLa , Histona-Lisina N-Metiltransferase/genética , Histonas/genética , Humanos , Metilação , Dados de Sequência Molecular , Proteínas Nucleares/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Isoformas de Proteínas , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos da radiação , Fatores de Tempo , Transcrição Gênica , Ativação Transcricional , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina-Proteína Ligases , Ubiquitinação
13.
Sensors (Basel) ; 18(4)2018 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-29641479

RESUMO

This paper examines methods to best exploit the High Dynamic Range (HDR) of the single photon avalanche diode (SPAD) in a high fill-factor HDR photon counting pixel that is scalable to megapixel arrays. The proposed method combines multi-exposure HDR with temporal oversampling in-pixel. We present a silicon demonstration IC with 96 × 40 array of 8.25 µm pitch 66% fill-factor SPAD-based pixels achieving >100 dB dynamic range with 3 back-to-back exposures (short, mid, long). Each pixel sums 15 bit-planes or binary field images internally to constitute one frame providing 3.75× data compression, hence the 1k frames per second (FPS) output off-chip represents 45,000 individual field images per second on chip. Two future projections of this work are described: scaling SPAD-based image sensors to HDR 1 MPixel formats and shrinking the pixel pitch to 1-3 µm.

14.
Semin Cancer Biol ; 36: 33-51, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26432751

RESUMO

F-box proteins are substrate receptors of the SCF (SKP1-Cullin 1-F-box protein) E3 ubiquitin ligase that play important roles in a number of physiological processes and activities. Through their ability to assemble distinct E3 ubiquitin ligases and target key regulators of cellular activities for ubiquitylation and degradation, this versatile group of proteins is able to regulate the abundance of cellular proteins whose deregulated expression or activity contributes to disease. In this review, we describe the important roles of select F-box proteins in regulating cellular activities, the perturbation of which contributes to the initiation and progression of a number of human malignancies.


Assuntos
Transformação Celular Neoplásica/genética , Proteínas F-Box/genética , Neoplasias/genética , Neoplasias/patologia , Animais , Apoptose/genética , Sobrevivência Celular/genética , Transformação Celular Neoplásica/metabolismo , Progressão da Doença , Proteínas F-Box/metabolismo , Regulação Neoplásica da Expressão Gênica , Instabilidade Genômica , Humanos , Metástase Neoplásica , Neoplasias/metabolismo , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Proteoma/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Ubiquitinas/metabolismo
15.
Adv Exp Med Biol ; 1042: 421-454, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29357069

RESUMO

Proper regulation of DNA replication ensures the faithful transmission of genetic material essential for optimal cellular and organismal physiology. Central to this regulation is the activity of a set of enzymes that induce or reverse posttranslational modifications of various proteins critical for the initiation, progression, and termination of DNA replication. This is particularly important when DNA replication proceeds in cancer cells with elevated rates of genomic instability and increased proliferative capacities. Here, we describe how DNA replication in mammalian cells is regulated via the activity of the ubiquitin-proteasome system as well as the consequence of derailed ubiquitylation signaling involved in this important cellular activity.


Assuntos
Replicação do DNA , Mamíferos/genética , Complexo de Endopeptidases do Proteassoma/fisiologia , Ubiquitina/metabolismo , Ubiquitinação/fisiologia , Animais , Instabilidade Genômica , Humanos , Complexo de Endopeptidases do Proteassoma/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Transdução de Sinais
16.
J Biol Chem ; 288(42): 30509-30514, 2013 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-24022480

RESUMO

After acute DNA damage, the cell arrests S-phase progression by inhibiting origin initiation and fork progression to repair damaged DNA. The intra-S-phase checkpoint kinase Chk1 phosphorylates Cdc25A to target the latter for degradation by CRL1(ß-TrCP) and so inhibit origin firing. The mechanism for inhibiting fork progression, however, has not been identified. Here, we show that degradation of p12, the fourth subunit of DNA polymerase δ, is critical for inhibiting fork progression. CRL4(Cdt2) is an E3 ligase that ubiquitinates and degrades p12 after UV treatment. Cells expressing a stable form of p12 exhibit UV-resistant DNA synthesis. DNA fiber assay and alkaline-sucrose gradient assay demonstrate that the impairment of fork progression after DNA damage requires p12 degradation. These results suggest that ubiquitination of p12 through CRL4(Cdt2) and subsequent degradation form one mechanism by which a cell responds to DNA damage to inhibit fork progression.


Assuntos
Dano ao DNA , DNA Polimerase III/metabolismo , DNA/biossíntese , Proteínas Nucleares/metabolismo , Subunidades Proteicas/metabolismo , Proteólise , Ubiquitina-Proteína Ligases/metabolismo , DNA/genética , DNA Polimerase III/genética , Estabilidade Enzimática/genética , Estabilidade Enzimática/efeitos da radiação , Células HeLa , Humanos , Proteínas Nucleares/genética , Subunidades Proteicas/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitinação/genética , Ubiquitinação/efeitos da radiação , Raios Ultravioleta
17.
Protein Sci ; 33(4): e4945, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38511494

RESUMO

Deltex proteins are a family of E3 ubiquitin ligases that encode C-terminal RING and DTC domains that mediate interactions with E2 ubiquitin-conjugating enzymes and recognize ubiquitination substrates. DTX3L is unique among the Deltex proteins based on its N-terminal domain architecture. The N-terminal D1 and D2 domains of DTX3L mediate homo-oligomerization, and the D3 domain interacts with PARP9, a protein that contains tandem macrodomains with ADP-ribose reader function. While DTX3L and PARP9 are known to heterodimerize, and assemble into a high molecular weight oligomeric complex, the nature of the oligomeric structure, including whether this contributes to the ADP-ribose reader function is unknown. Here, we report a crystal structure of the DTX3L N-terminal D2 domain and show that it forms a tetramer with, conveniently, D2 symmetry. We identified two interfaces in the structure: a major, conserved interface with a surface of 973 Å2 and a smaller one of 415 Å2. Using native mass spectrometry, we observed molecular species that correspond to monomers, dimers and tetramers of the D2 domain. Reconstitution of DTX3L knockout cells with a D1-D2 deletion mutant showed the domain is dispensable for DTX3L-PARP9 heterodimer formation, but necessary to assemble an oligomeric complex with efficient reader function for ADP-ribosylated androgen receptor. Our results suggest that homo-oligomerization of DTX3L is important for the DTX3L-PARP9 complex to read mono-ADP-ribosylation on a ligand-regulated transcription factor.


Assuntos
Leitura , Receptores Androgênicos , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Adenosina Difosfato Ribose/metabolismo
18.
Cancer Res Commun ; 3(4): 592-606, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37077937

RESUMO

The ADP-ribosyltransferase PARP7 modulates protein function by conjugating ADP-ribose to the side chains of acceptor amino acids. PARP7 has been shown to affect gene expression in prostate cancer cells and certain other cell types by mechanisms that include transcription factor ADP-ribosylation. Here, we use a recently developed catalytic inhibitor to PARP7, RBN2397, to study the effects of PARP7 inhibition in androgen receptor (AR)-positive and AR-negative prostate cancer cells. We find that RBN2397 has nanomolar potency for inhibiting androgen-induced ADP-ribosylation of the AR. RBN2397 inhibits the growth of prostate cancer cells in culture when cells are treated with ligands that activate the AR, or the aryl hydrocarbon receptor, and induce PARP7 expression. We show that the growth-inhibitory effects of RBN2397 are distinct from its enhancement of IFN signaling recently shown to promote tumor immunogenicity. RBN2397 treatment also induces trapping of PARP7 in a detergent-resistant fraction within the nucleus, which is reminiscent of how inhibitors such as talazoparib affect PARP1 compartmentalization. Because PARP7 is expressed in AR-negative metastatic tumors and RBN2397 can affect cancer cells through multiple mechanisms, PARP7 may be an actionable target in advanced prostate cancer. Significance: RBN2397 is a potent and selective inhibitor of PARP7 that reduces the growth of prostate cancer cells, including a model for treatment-emergent neuroendocrine prostate cancer. RBN2397 induces PARP7 trapping on chromatin, suggesting its mechanism of action might be similar to clinically used PARP1 inhibitors.


Assuntos
Neoplasias da Próstata , Receptores Androgênicos , Masculino , Humanos , Receptores Androgênicos/genética , Neoplasias da Próstata/tratamento farmacológico , Próstata/metabolismo , ADP Ribose Transferases/genética , Androgênios
19.
bioRxiv ; 2023 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-38076829

RESUMO

Deltex proteins are a family of E3 ubiquitin ligases that encode C-terminal RING and DTC domains that mediate interactions with E2 ubiquitin-conjugating enzymes and recognise ubiquitination substrates. DTX3L is unique among the Deltex proteins based on its N-terminal domain architecture. The N-terminal D1 and D2 domains of DTX3L mediate homo-oligomerisation, and the D3 domain interacts with PARP9, a protein that contains tandem macrodomains with ADP-ribose reader function. While DTX3L and PARP9 are known to heterodimerize, they assemble into a high molecular weight oligomeric complex, but the nature of the oligomeric structure, including whether this contributes to the ADP-ribose reader function is unknown. Here, we report a crystal structure of the DTX3L N-terminal D2 domain and show that it forms a tetramer with, conveniently, D2 symmetry. We identified two interfaces in the structure: a major, conserved interface with a surface of 973 Å2 and a smaller one of 415 Å2. Using native mass spectrometry, we observed molecular species that correspond to monomers, dimers and tetramers of the D2 domain. Reconstitution of DTX3L knockout cells with a D1-D2 deletion mutant showed the domain is dispensable for DTX3L-PARP9 heterodimer formation, but necessary to assemble an oligomeric complex with efficient reader function for ADP-ribosylated androgen receptor. Our results suggest that homo-oligomerisation of DTX3L is important for mono-ADP-ribosylation reading by the DTX3L-PARP9 complex and to a ligand-regulated transcription factor.

20.
J Biol Chem ; 286(35): 30462-30470, 2011 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-21757720

RESUMO

Chromatin-modifying enzymes play a fundamental role in regulating chromatin structure so that DNA replication is spatially and temporally coordinated. For example, the lysine demethylase 4A/Jumonji domain-containing 2A (KDM4A/JMJD2A) is tightly regulated during the cell cycle. Overexpression of JMJD2A leads to altered replication timing and faster S phase progression. In this study, we demonstrate that degradation of JMJD2A is regulated by the proteasome. JMJD2A turnover is coordinated through the SKP1-Cul1-F-box ubiquitin ligase complex that contains cullin 1 and the F-box and leucine-rich repeat protein 4 (FbxL4). This complex interacted with JMJD2A. Ubiquitin overexpression restored turnover and blocked the JMJD2A-dependent faster S phase progression in a cullin 1-dependent manner. Furthermore, increased ubiquitin levels decreased JMJD2A occupancy and BrdU incorporation at target sites. This study highlights a finely tuned mechanism for regulating histone demethylase levels and emphasizes the need to tightly regulate chromatin modifiers so that the cell cycle occurs properly.


Assuntos
Proteínas F-Box/química , Histona Desmetilases com o Domínio Jumonji/química , Proteínas Quinases Associadas a Fase S/química , Ubiquitina-Proteína Ligases/química , Sequência de Bases , Sítios de Ligação , Ciclo Celular , Cromatina/química , Proteínas Culina/química , Replicação do DNA , Histona Desmetilases/química , Humanos , Complexo de Endopeptidases do Proteassoma/química , Ligação Proteica , Estrutura Terciária de Proteína , Ubiquitina/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA