Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Electrocardiol ; 83: 41-48, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38306814

RESUMO

Machine learning is poised to revolutionize medicine with algorithms that spot cardiac arrhythmia. An automated diagnostic approach can boost the efficacy of diagnosing life-threatening arrhythmia disorders in routine medical procedures. In this paper, we propose a deep learning network CLINet for ECG signal classification. Our network uses convolution, LSTM and involution layers to bring their unique advantages together. For both convolution and involution layers, we use multiple, large size kernels for multi-scale representation learning. CLINet does not require complicated pre-processing and can handle electrocardiograms of any length. Our network achieves 99.90% accuracy on the ICCAD dataset and 99.94% accuracy on the MIT-BIH dataset. With only 297 K parameters, our model can be easily embedded in smart wearable devices. The source code of CLINet is available at https://github.com/CandleLabAI/CLINet-ECG-Classification-2024.


Assuntos
Aprendizado Profundo , Humanos , Processamento de Sinais Assistido por Computador , Eletrocardiografia/métodos , Algoritmos , Software , Arritmias Cardíacas/diagnóstico
2.
Mol Cancer Res ; 21(9): 947-957, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37314748

RESUMO

Transcription and DNA damage repair act in a coordinated manner. The scaffolding protein SIN3B serves as a transcriptional co-repressor of hundreds of cell cycle-related genes. However, the contribution of SIN3B during the DNA damage response remains unknown. Here, we show that SIN3B inactivation delays the resolution of DNA double-strand breaks and sensitizes cancer cells to DNA-damaging agents, including the chemotherapeutic drugs cisplatin and doxorubicin. Mechanistically, SIN3B is rapidly recruited to DNA damage sites where it directs the accumulation of Mediator of DNA Damage Checkpoint 1 (MDC1). In addition, we show that SIN3B inactivation favors the engagement of the alternative nonhomologous end joining (NHEJ) repair pathway over the canonical NHEJ. Altogether, our findings impute an unexpected function for the transcriptional co-repressor SIN3B as a gatekeeper of genomic integrity and a determining factor in the DNA repair choice pathway, and point to the inhibition of the SIN3B chromatin-modifying complex as a novel therapeutic vulnerability in cancer cells. IMPLICATIONS: Identifying SIN3B as a modulator of DNA damage repair choice provides novel potential therapeutic avenues to sensitize cancer cells to cytotoxic therapies.


Assuntos
Cromatina , Neoplasias , Humanos , Dano ao DNA , Reparo do DNA , DNA/metabolismo , Reparo do DNA por Junção de Extremidades , Proteínas Correpressoras , Apoptose , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo
3.
J Clin Endocrinol Metab ; 106(7): 1929-1955, 2021 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-33755733

RESUMO

CONTEXT: The ability of ovarian steroids to modify ovarian cancer (OC) risk remains controversial. Progesterone is considered to be protective; recent studies indicate no effect or enhanced OC risk. Knowledge of progesterone receptor (PR) signaling during altered physiology that typifies OC development is limited. OBJECTIVE: This study defines PR-driven oncogenic signaling mechanisms in p53-mutant human fallopian tube epithelia (hFTE), a precursor of the most aggressive OC subtype. METHODS: PR expression in clinical samples of serous tubal intraepithelial carcinoma (STIC) lesions and high-grade serous OC (HGSC) tumors was analyzed. Novel PR-A and PR-B isoform-expressing hFTE models were characterized for gene expression and cell cycle progression, emboli formation, and invasion. PR regulation of the DREAM quiescence complex and DYRK1 kinases was established. RESULTS: STICs and HGSC express abundant activated phospho-PR. Progestin promoted reversible hFTE cell cycle arrest, spheroid formation, and invasion. RNAseq/biochemical studies revealed potent ligand-independent/-dependent PR actions, progestin-induced regulation of the DREAM quiescence complex, and cell cycle target genes through enhanced complex formation and chromatin recruitment. Disruption of DREAM/DYRK1s by pharmacological inhibition, HPV E6/E7 expression, or DYRK1A/B depletion blocked progestin-induced cell arrest and attenuated PR-driven gene expression and associated OC phenotypes. CONCLUSION: Activated PRs support quiescence and pro-survival/pro-dissemination cell behaviors that may contribute to early HGSC progression. Our data support an alternative perspective on the tenet that progesterone always confers protection against OC. STICs can reside undetected for decades prior to invasive disease; our studies reveal clinical opportunities to prevent the ultimate development of HGSC by targeting PRs, DREAM, and/or DYRKs.


Assuntos
Processos de Crescimento Celular/genética , Cistadenocarcinoma Seroso/genética , Neoplasias das Tubas Uterinas/genética , Proteínas Interatuantes com Canais de Kv/metabolismo , Receptores de Progesterona/metabolismo , Proteínas Repressoras/metabolismo , Carcinogênese/genética , Linhagem Celular Tumoral , Tubas Uterinas/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Neoplasias Ovarianas/genética , Fenótipo , Proteína Supressora de Tumor p53/metabolismo
4.
Cancers (Basel) ; 13(3)2021 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-33513914

RESUMO

High-risk (HR) human papillomaviruses are known causative agents in 5% of human cancers including cervical, ano-genital and head and neck carcinomas. In part, HR-HPV causes cancer by targeting host-cell tumor suppressors including retinoblastoma protein (pRb) and RB-like proteins p107 and p130. HR-HPV E7 uses a LxCxE motif to bind RB proteins, impairing their ability to control cell-cycle dependent transcription. E7 disrupts DREAM (Dimerization partner, RB-like, E2F and MuvB), a transcriptional repressor complex that can include p130 or p107, but not pRb, which regulates genes required for cell cycle progression. However, it is not known whether disruption of DREAM plays a significant role in HPV-driven tumorigenesis. In the DREAM complex, LIN52 is an adaptor that binds directly to p130 via an E7-like LxSxE motif. Replacement of the LxSxE sequence in LIN52 with LxCxE (LIN52-S20C) increases p130 binding and partially restores DREAM assembly in HPV-positive keratinocytes and human cervical cancer cells, inhibiting proliferation. Our findings demonstrate that disruption of the DREAM complex by E7 is an important process promoting cellular proliferation by HR-HPV. Restoration of the DREAM complex in HR-HPV positive cells may therefore have therapeutic benefits in HR-HPV positive cancers.

5.
Cell Cycle ; 18(5): 531-551, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30773093

RESUMO

Human Dual-specificity tyrosine (Y) Regulated Kinase 1A (DYRK1A) is encoded by a dosage dependent gene whereby either trisomy or haploinsufficiency result in developmental abnormalities. However, the function and regulation of this important protein kinase are not fully understood. Here, we report proteomic analysis of DYRK1A in human cells that revealed a novel role of DYRK1A in DNA double-strand breaks (DSBs) repair, mediated in part by its interaction with the ubiquitin-binding protein RNF169 that accumulates at the DSB sites and promotes homologous recombination repair (HRR) by displacing 53BP1, a key mediator of non-homologous end joining (NHEJ). We found that overexpression of active, but not the kinase inactive DYRK1A in U-2 OS cells inhibits accumulation of 53BP1 at the DSB sites in the RNF169-dependent manner. DYRK1A phosphorylates RNF169 at two sites that influence its ability to displace 53BP1 from the DSBs. Although DYRK1A is not required for the recruitment of RNF169 to the DSB sites and 53BP1 displacement, inhibition of DYRK1A or mutation of the DYRK1A phosphorylation sites in RNF169 decreases its ability to block accumulation of 53BP1 at the DSB sites. Interestingly, CRISPR-Cas9 knockout of DYRK1A in human and mouse cells also diminished the 53BP1 DSB recruitment in a manner that did not require RNF169, suggesting that dosage of DYRK1A can influence the DNA repair processes through both RNF169-dependent and independent mechanisms. Human U-2 OS cells devoid of DYRK1A display an increased HRR efficiency and resistance to DNA damage, therefore our findings implicate DYRK1A in the DNA repair processes.


Assuntos
Dano ao DNA , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Sistemas CRISPR-Cas/genética , Pontos de Checagem do Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Dano ao DNA/efeitos da radiação , Reparo do DNA , Raios gama , Edição de Genes , Humanos , Redes e Vias Metabólicas , Camundongos , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/deficiência , Proteínas Tirosina Quinases/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/genética , Quinases Dyrk
6.
Oncogene ; 38(7): 1080-1092, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30206359

RESUMO

Overexpression of the oncogene MYBL2 (B-Myb) is associated with increased cell proliferation and serves as a marker of poor prognosis in cancer. However, the mechanism by which B-Myb alters the cell cycle is not fully understood. In proliferating cells, B-Myb interacts with the MuvB core complex including LIN9, LIN37, LIN52, RBBP4, and LIN54, forming the MMB (Myb-MuvB) complex, and promotes transcription of genes required for mitosis. Alternatively, the MuvB core interacts with Rb-like protein p130 and E2F4-DP1 to form the DREAM complex that mediates global repression of cell cycle genes in G0/G1, including a subset of MMB target genes. Here, we show that overexpression of B-Myb disrupts the DREAM complex in human cells, and this activity depends on the intact MuvB-binding domain in B-Myb. Furthermore, we found that B-Myb regulates the protein expression levels of the MuvB core subunit LIN52, a key adapter for assembly of both the DREAM and MMB complexes, by a mechanism that requires S28 phosphorylation site in LIN52. Given that high expression of B-Myb correlates with global loss of repression of DREAM target genes in breast and ovarian cancer, our findings offer mechanistic insights for aggressiveness of cancers with MYBL2 amplification, and establish the rationale for targeting B-Myb to restore cell cycle control.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas de Ciclo Celular/biossíntese , Ciclo Celular , Regulação Neoplásica da Expressão Gênica , Proteínas Interatuantes com Canais de Kv/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Ovarianas/metabolismo , Proteínas Repressoras/metabolismo , Transativadores/biossíntese , Neoplasias da Mama/genética , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Feminino , Humanos , Proteínas Interatuantes com Canais de Kv/genética , Complexos Multiproteicos/genética , Proteínas de Neoplasias/genética , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Proteínas Repressoras/genética , Transativadores/genética
7.
Cell Rep ; 25(10): 2797-2807.e8, 2018 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-30517867

RESUMO

The mammalian DREAM complex is responsible for the transcriptional repression of hundreds of cell-cycle-related genes in quiescence. How the DREAM complex recruits chromatin-modifying entities to aid in its repression remains unknown. Using unbiased proteomics analysis, we have uncovered a robust association between the chromatin-associated Sin3B protein and the DREAM complex. We have determined that genetic inactivation of Sin3B results in the de-repression of DREAM target genes during quiescence but is insufficient to allow quiescent cells to resume proliferation. However, inactivation of APC/CCDH1 was sufficient for Sin3B-/- cells, but not parental cells, to re-enter the cell cycle. These studies identify Sin3B as a transcriptional corepressor associated with the DREAM complex in quiescence and reveals a functional cooperation between E2F target repression and APC/CCDH1 in the negative regulation of cell-cycle progression.


Assuntos
Ciclossomo-Complexo Promotor de Anáfase/metabolismo , Ciclo Celular , Histona Desacetilases/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas Repressoras/metabolismo , Humanos , Ligação Proteica , Transcrição Gênica
8.
Proc Natl Acad Sci U S A ; 115(40): 10016-10021, 2018 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-30224471

RESUMO

The MuvB transcriptional regulatory complex, which controls cell-cycle-dependent gene expression, cooperates with B-Myb to activate genes required for the G2 and M phases of the cell cycle. We have identified the domain in B-Myb that is essential for the assembly of the Myb-MuvB (MMB) complex. We determined a crystal structure that reveals how this B-Myb domain binds MuvB through the adaptor protein LIN52 and the scaffold protein LIN9. The structure and biochemical analysis provide an understanding of how oncogenic B-Myb is recruited to regulate genes required for cell-cycle progression, and the MMB interface presents a potential therapeutic target to inhibit cancer cell proliferation.


Assuntos
Proteínas de Ciclo Celular , Ciclo Celular , Complexos Multiproteicos , Proteínas de Neoplasias , Neoplasias , Proteínas Nucleares , Transativadores , Proteínas Supressoras de Tumor , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Cristalografia por Raios X , Humanos , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Neoplasias/química , Neoplasias/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Domínios Proteicos , Transativadores/química , Transativadores/metabolismo , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/metabolismo
9.
Cell Cycle ; 17(14): 1812-1831, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30010460

RESUMO

Vesicle trafficking regulates epithelial cell migration by remodeling matrix adhesions and delivering signaling molecules to the migrating leading edge. Membrane fusion, which is driven by soluble N-ethylmaleimide-sensitive factor associated receptor (SNARE) proteins, is an essential step of vesicle trafficking. Mammalian SNAREs represent a large group of proteins, but few have been implicated in the regulation of cell migration. Ykt6 is a unique SNARE existing in equilibrium between active membrane-bound and inactive cytoplasmic pools, and mediating vesicle trafficking between different intracellular compartments. The biological functions of this protein remain poorly understood. In the present study, we found that Ykt6 acts as a negative regulator of migration and invasion of human prostate epithelial cells. Furthermore, Ykt6 regulates the integrity of epithelial adherens and tight junctions. The observed anti-migratory activity of Ykt6 is mediated by a unique mechanism involving the expressional upregulation of microRNA 145, which selectively decreases the cellular level of Junctional Adhesion Molecule (JAM) A. This decreased JAM-A expression limits the activity of Rap1 and Rac1 small GTPases, thereby attenuating cell spreading and motility. The described novel functions of Ykt6 could be essential for the regulation of epithelial barriers, epithelial repair, and metastatic dissemination of cancer cells.


Assuntos
Movimento Celular , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Molécula A de Adesão Juncional/metabolismo , Fusão de Membrana , MicroRNAs/metabolismo , Proteínas R-SNARE/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Regulação para Baixo/genética , Humanos , Junções Intercelulares/metabolismo , Masculino , MicroRNAs/genética , Neoplasias da Próstata/patologia , Proteínas R-SNARE/genética , Complexo Shelterina , Proteínas de Ligação a Telômeros/metabolismo , Regulação para Cima/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
10.
PLoS Genet ; 12(11): e1006429, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27828963

RESUMO

E-type cyclins (cyclins E1 and E2) are components of the cell cycle machinery that has been conserved from yeast to humans. The major function of E-type cyclins is to drive cell division. It is unknown whether in addition to their 'core' cell cycle functions, E-type cyclins also perform unique tissue-specific roles. Here, we applied high-throughput mass spectrometric analyses of mouse organs to define the repertoire of cyclin E protein partners in vivo. We found that cyclin E interacts with distinct sets of proteins in different compartments. These cyclin E interactors are highly enriched for phosphorylation targets of cyclin E and its catalytic partner, the cyclin-dependent kinase 2 (Cdk2). Among cyclin E interactors we identified several novel tissue-specific substrates of cyclin E-Cdk2 kinase. In proliferating compartments, cyclin E-Cdk2 phosphorylates Lin proteins within the DREAM complex. In the testes, cyclin E-Cdk2 phosphorylates Mybl1 and Dmrtc2, two meiotic transcription factors that represent key regulators of spermatogenesis. In embryonic and adult brains cyclin E interacts with proteins involved in neurogenesis, while in adult brains also with proteins regulating microtubule-based processes and microtubule cytoskeleton. We also used quantitative proteomics to demonstrate re-wiring of the cyclin E interactome upon ablation of Cdk2. This approach can be used to study how protein interactome changes during development or in any pathological state such as aging or cancer.


Assuntos
Ciclina E/genética , Quinase 2 Dependente de Ciclina/genética , Ciclinas/genética , Proteínas Oncogênicas/genética , Proteômica , Animais , Ciclo Celular/genética , Proliferação de Células/genética , Ciclina E/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Ciclinas/metabolismo , Humanos , Masculino , Camundongos , Proteínas Oncogênicas/metabolismo , Fosforilação , Mapas de Interação de Proteínas/genética , Fase S/genética , Espermatogênese/genética , Testículo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA