Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 13(1): 14405, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37658135

RESUMO

The ubiquitin‒proteasome system (UPS) and autophagy are the two primary cellular pathways of misfolded or damaged protein degradation that maintain cellular proteostasis. When the proteasome is dysfunctional, cells compensate for impaired protein clearance by activating aggrephagy, a type of selective autophagy, to eliminate ubiquitinated protein aggregates; however, the molecular mechanisms by which impaired proteasome function activates aggrephagy remain poorly understood. Here, we demonstrate that activation of aggrephagy is transcriptionally induced by the transcription factor NRF1 (NFE2L1) in response to proteasome dysfunction. Although NRF1 has been previously shown to induce the expression of proteasome genes after proteasome inhibition (i.e., the proteasome bounce-back response), our genome-wide transcriptome analyses identified autophagy-related p62/SQSTM1 and GABARAPL1 as genes directly targeted by NRF1. Intriguingly, NRF1 was also found to be indispensable for the formation of p62-positive puncta and their colocalization with ULK1 and TBK1, which play roles in p62 activation via phosphorylation. Consistently, NRF1 knockdown substantially reduced the phosphorylation rate of Ser403 in p62. Finally, NRF1 selectively upregulated the expression of GABARAPL1, an ATG8 family gene, to induce the clearance of ubiquitinated proteins. Our findings highlight the discovery of an activation mechanism underlying NRF1-mediated aggrephagy through gene regulation when proteasome activity is impaired.


Assuntos
Proteostase , Fatores de Transcrição , Complexo de Endopeptidases do Proteassoma , Macroautofagia , Regulação da Expressão Gênica , Ubiquitina
2.
iScience ; 26(2): 106045, 2023 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-36818298

RESUMO

Cancer cells coordinate the mTORC1 signals and the related metabolic pathways to robustly and rapidly grow in response to nutrient conditions. Although a CNC-family transcription factor NRF3 promotes cancer development, the biological relevance between NRF3 function and mTORC1 signals in cancer cells remains unknown. Hence, we showed that NRF3 contributes to cancer cell viability through mTORC1 activation in response to amino acids, particularly arginine. NRF3 induced SLC38A9 and RagC expression for the arginine-dependent mTORC1 recruitment onto lysosomes, and it also enhanced RAB5-mediated bulk macropinocytosis and SLC7A1-mediated selective transport for arginine loading into lysosomes. Besides, the inhibition of the NRF3-mTORC1 axis impaired mitochondrial function, leading to cancer cell apoptosis. Consistently, the aberrant upregulation of the axis caused tumor growth and poor prognosis. In conclusion, this study sheds light on the unique function of NRF3 in arginine-dependent mTORC1 activation and the pathophysiological aspects of the NRF3-mTORC1 axis in cancer development.

3.
Mol Cell Biol ; 40(14)2020 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-32366381

RESUMO

Proteasomes are protease complexes essential for cellular homeostasis, and their activity is crucial for cancer cell growth. However, the mechanism of how proteasome activity is maintained in cancer cells has remained unclear. The CNC family transcription factor NFE2L1 induces the expression of almost all proteasome-related genes under proteasome inhibition. Both NFE2L1 and its phylogenetically closest homolog, NFE2L3, are highly expressed in several types of cancer, such as colorectal cancer. Here, we demonstrate that NFE2L1 and NFE2L3 complementarily maintain basal proteasome activity in cancer cells. Double knockdown of NFE2L1 and NFE2L3 impaired basal proteasome activity in cancer cells and cancer cell resistance to a proteasome inhibitor anticancer drug, bortezomib, by significantly reducing the basal expression of seven proteasome-related genes: PSMB3, PSMB7, PSMC2, PSMD3, PSMG2, PSMG3, and POMP Interestingly, the molecular basis behind these cellular consequences was that NFE2L3 repressed NFE2L1 translation by the induction of the gene encoding the translational regulator CPEB3, which binds to the NFE2L1 3' untranslated region and decreases polysome formation on NFE2L1 mRNA. Consistent results were obtained from clinical analysis, wherein patients with cancer having tumors expressing higher levels of CPEB3/NFE2L3 exhibit poor prognosis. These results provide the novel regulatory mechanism of basal proteasome activity in cancer cells through an NFE2L3-CPEB3-NFE2L1 translational repression axis.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Fator 1 Relacionado a NF-E2/metabolismo , Neoplasias/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas de Ligação a RNA/metabolismo , Linhagem Celular Tumoral , Humanos , Biossíntese de Proteínas
4.
Mol Cell Biol ; 40(10)2020 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-32123008

RESUMO

Proteasomes are essential protease complexes that maintain cellular homeostasis, and aberrant proteasomal activity supports cancer development. The regulatory mechanisms and biological function of the ubiquitin-26S proteasome have been studied extensively, while those of the ubiquitin-independent 20S proteasome system remain obscure. Here, we show that the cap 'n' collar (CNC) family transcription factor NRF3 specifically enhances 20S proteasome assembly in cancer cells and that 20S proteasomes contribute to colorectal cancer development through ubiquitin-independent proteolysis of the tumor suppressor p53 and retinoblastoma (Rb) proteins. The NRF3 gene is highly expressed in many cancer tissues and cell lines and is important for cancer cell growth. In cancer cells, NRF3 upregulates the assembly of the 20S proteasome by directly inducing the gene expression of the 20S proteasome maturation protein POMP. Interestingly, NRF3 knockdown not only increases p53 and Rb protein levels but also increases p53 activities for tumor suppression, including cell cycle arrest and induction of apoptosis. Furthermore, protein stability and cell viability assays using two distinct proteasome inhibitor anticancer drugs, the 20S proteasome inhibitor bortezomib and the ubiquitin-activating enzyme E1 inhibitor TAK-243, show that the upregulation of the NRF3-POMP axis leads to ubiquitin-independent proteolysis of p53 and Rb and to impaired sensitivity to bortezomib but not TAK-243. More importantly, the NRF3-POMP axis supports tumorigenesis and metastasis, with higher NRF3/POMP expression levels correlating with poor prognoses in patients with colorectal or rectal adenocarcinoma. These results suggest that the NRF3-POMP-20S proteasome assembly axis is significant for cancer development via ubiquitin-independent proteolysis of tumor suppressor proteins.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Chaperonas Moleculares/metabolismo , Neoplasias/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteína do Retinoblastoma/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Células HCT116 , Células HeLa , Humanos , Proteólise , Ubiquitina/metabolismo
5.
Int J Mol Sci ; 20(13)2019 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-31288376

RESUMO

Remarkable upregulation of the NRF2 (NFE2L2)-related transcription factor NRF3 (NFE2L3) in several cancer tissues and its correlation with poor prognosis strongly suggest the physiological function of NRF3 in tumors. Indeed, we had recently uncovered the function of NRF3, which promotes cancer cell proliferation by p53 degradation via the 20S proteasome. Nevertheless, the molecular mechanism underlying the induction of NRF3 gene expression in cancer cells is highly elusive. We herein describe that NRF3 upregulation is induced by the ß-catenin/TCF4 complex in colon cancer cells. We first confirmed high NRF3 mRNA expression in human colon cancer specimens. The genome database indicated that the human NRF3 gene possesses a species-conserved WRE sequence (TCF/LEF consensus element), implying that the ß-catenin/TCF complex activates NRF3 expression in colon cancer. Consistently, we observed that the ß-catenin/TCF4 complex mediates NRF3 expression by binding directly to the WRE site. Furthermore, inducing NRF3 activates cell proliferation and the expression of the glucose transporter GLUT1. The existence of the ß-catenin/TCF4-NRF3 axis was also validated in the intestine and organoids of Apc-deficient mice. Finally, the positive correlation between NRF3 and ß-catenin target gene expression strongly supports our conclusion. Our findings clearly demonstrate that NRF3 induction in cancer cells is controlled by the Wnt/ß-catenin pathway.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/genética , Neoplasias/genética , Neoplasias/metabolismo , Fator de Transcrição 4/metabolismo , Ativação Transcricional , beta Catenina/metabolismo , Animais , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Proliferação de Células , Biologia Computacional/métodos , Sequência Conservada , Bases de Dados Genéticas , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Modelos Biológicos , Ligação Proteica , RNA Mensageiro/genética , Via de Sinalização Wnt
6.
Bioorg Med Chem ; 27(6): 1119-1129, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30745098

RESUMO

Histone lysine demethylases (KDMs) have drawn much attention as targets of therapeutic agents. KDM5 proteins, which are Fe(II)/α-ketoglutarate-dependent demethylases, are associated with oncogenesis and drug resistance in cancer cells, and KDM5-selective inhibitors are expected to be anticancer drugs. However, few cell-active KDM5 inhibitors have been reported and there is an obvious need to discover more. In this study, we pursued the identification of highly potent and cell-active KDM5-selective inhibitors. Based on the reported KDM5 inhibitors, we designed several compounds by strategically merging two fragments for competitive inhibition with α-ketoglutarate and for KDM5-selective inhibition. Among them, compounds 10 and 13, which have a 3-cyano pyrazolo[1,5-a]pyrimidin-7-one scaffold, exhibited strong KDM5-inhibitory activity and significant KDM5 selectivity. In cellular assays using human lung cancer cell line A549, 10 and 13 increased the levels of trimethylated lysine 4 on histone H3, which is a specific substrate of KDM5s, and induced growth inhibition of A549 cells. These results should provide a basis for the development of cell-active KDM5 inhibitors to highlight the validity of our inhibitor-based fragment merging strategy.


Assuntos
Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Pirimidinas/química , Pirimidinas/farmacologia , Proteína 2 de Ligação ao Retinoblastoma/antagonistas & inibidores , Antineoplásicos/química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Desenho de Fármacos , Epigênese Genética/efeitos dos fármacos , Humanos , Modelos Moleculares , Simulação de Acoplamento Molecular , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Pirazóis/química , Pirazóis/farmacologia , Proteína 2 de Ligação ao Retinoblastoma/metabolismo
7.
Sci Rep ; 7(1): 12494, 2017 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-28970512

RESUMO

Accumulated evidence suggests a physiological relationship between the transcription factor NRF3 (NFE2L3) and cancers. Under physiological conditions, NRF3 is repressed by its endoplasmic reticulum (ER) sequestration. In response to unidentified signals, NRF3 enters the nucleus and modulates gene expression. However, molecular mechanisms underlying the nuclear translocation of NRF3 and its target gene in cancer cells remain poorly understood. We herein report that multiple regulation of NRF3 activities controls cell proliferation. Our analyses reveal that under physiological conditions, NRF3 is rapidly degraded by the ER-associated degradation (ERAD) ubiquitin ligase HRD1 and valosin-containing protein (VCP) in the cytoplasm. Furthermore, NRF3 is also degraded by ß-TRCP, an adaptor for the Skp1-Cul1-F-box protein (SCF) ubiquitin ligase in the nucleus. The nuclear translocation of NRF3 from the ER requires the aspartic protease DNA-damage inducible 1 homolog 2 (DDI2) but does not require inhibition of its HRD1-VCP-mediated degradation. Finally, NRF3 mediates gene expression of the cell cycle regulator U2AF homology motif kinase 1 (UHMK1) for cell proliferation. Collectively, our study provides us many insights into the molecular regulation and biological function of NRF3 in cancer cells.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/genética , Ciclo Celular/genética , Células Epiteliais/metabolismo , Regulação Neoplásica da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas Serina-Treonina Quinases/genética , Animais , Ácido Aspártico Proteases/genética , Ácido Aspártico Proteases/metabolismo , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Células COS , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proliferação de Células , Chlorocebus aethiops , Proteínas Culina/genética , Proteínas Culina/metabolismo , Citosol/metabolismo , Retículo Endoplasmático/metabolismo , Degradação Associada com o Retículo Endoplasmático , Células Epiteliais/patologia , Células HCT116 , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases Associadas a Fase S/genética , Proteínas Quinases Associadas a Fase S/metabolismo , Transdução de Sinais , Fator de Células-Tronco/genética , Fator de Células-Tronco/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Proteína com Valosina/genética , Proteína com Valosina/metabolismo , Proteínas Contendo Repetições de beta-Transducina/genética , Proteínas Contendo Repetições de beta-Transducina/metabolismo
8.
Biochem Biophys Res Commun ; 484(1): 176-183, 2017 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-28088524

RESUMO

The transcription factor Nrf1 (NFE2L1) maintains protein homeostasis (proteostasis) by regulating the gene expression of proteasome subunits in response to proteasome inhibition. The deletion of the Nrf1 gene in neural stem/progenitor cells causes severe neurodegeneration due to the accumulation of ubiquitinated proteins in Purkinje cells and motor neurons (Nrf1 NKO mice). However, the molecular mechanisms governing this neurodegenerative process remain unclear. We demonstrate herein that the loss of Nrf1 leads to the reduced gene expression of the deubiquitinating enzymes (DUBs) but not proteasome subunits in Nrf1 NKO mice between P7 and P18. First, we show that K48-linked polyubiquitinated proteins accumulate in Nrf1-deficient Purkinje cells and cerebral cortex neurons. Nevertheless, loss of Nrf1 does not alter the expression and proteolytic activity of proteasome. A significantly reduced expression of deubiquitinating enzymes was also demonstrated in Nrf1-deficient cerebellar tissue using microarray analysis. The genome database further reveals species-conserved ARE, a Nrf1 recognition element, in the regulatory region of certain DUB genes. Furthermore, we show that Nrf1 can activate Usp9x gene expression related to neurodegeneration. Altogether these findings suggest that neurodegeneration in Nrf1 NKO mice may stem from the dysfunction of the ubiquitin-mediated regulation of neuronal proteins.


Assuntos
Cerebelo/enzimologia , Enzimas Desubiquitinantes/genética , Homeostase/fisiologia , Células-Tronco Neurais/metabolismo , Fator 1 Nuclear Respiratório/fisiologia , Animais , Cerebelo/patologia , Enzimas Desubiquitinantes/metabolismo , Regulação Enzimológica da Expressão Gênica , Camundongos , Camundongos Knockout , Células-Tronco Neurais/citologia , Células-Tronco Neurais/enzimologia , Fator 1 Nuclear Respiratório/genética
9.
Leg Med (Tokyo) ; 13(1): 12-5, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20952240

RESUMO

Unprecedented fidelity and specificity have afforded DNA testing its long reigning status as the gold standard for establishing personal identification. While the method itself is flawless, forensic experts have undoubtedly stumbled across challenging cases in which no reference samples for an unknown person (UP) are available for comparison. In such cases, experts often must resort to an assortment of kinship analyses-primarily those involving alleged parents or children of a UP-to establish personal identification. The present study derives likelihood ratio (LR) distributions from an extensive series of kinship simulations and places actual data, obtained from 120 cases in which personal identification of a UP was established via kinship analyses, to a comprehensive comparison in order to evaluate the efficacy of kinship assessments in establishing personal identification. A commercially available AmpFlSTR Identifiler kit was used to obtain DNA profiles. UP DNAs were extracted and isolated from fingernail (n=87), cardiac blood (24), carpal bone (7) and tooth (2). Buccal cells were procured from alleged kin (AK) for subsequent kinship analyses. In 72 cases 1-3 alleged children were available for comparison; in 46 cases, one or both alleged parents were available; and in the final 2 cases (involving a pair of bodies discovered together in a dwelling), their alleged children were typed for comparison. For each case a LR was calculated based on the DNA typing results. Interestingly, we found that the median LR observed in the actual cases virtually mirrored those of the simulations. With exception to 2 cases in which a silent allele was observed at D19S433, biological relatives showed a LR greater than 100 and in these cases, kinship between the UP and AK were further supported by additional forms of evidence. We show here that in the vast majority of identification cases where direct reference samples are unavailable for a UP, kinship analyses referring to alleged parents/children and using 15 standard loci is more than capable of establishing the identification of a UP. However, discretion should be advised for silent alleles which-albeit rare-are known to occur at loci such as D19S433, along with other mutations which could render a deceivingly reduced LR.


Assuntos
Impressões Digitais de DNA/instrumentação , Família , Antropologia Forense/métodos , Impressões Digitais de DNA/métodos , Humanos , Funções Verossimilhança , Sensibilidade e Especificidade
10.
Mol Cell Biol ; 27(8): 2812-20, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17283053

RESUMO

Fbh1 (F-box DNA helicase 1) orthologues are conserved from Schizosaccharomyces pombe to chickens and humans. Here, we report the disruption of the FBH1 gene in DT40 cells. Although the yeast fbh1 mutant shows an increase in sensitivity to DNA damaging agents, FBH1(-)(/)(-) DT40 clones show no prominent sensitivity, suggesting that the loss of FBH1 might be compensated by other genes. However, FBH1(-)(/)(-) cells exhibit increases in both sister chromatid exchange and the formation of radial structures between homologous chromosomes without showing a defect in homologous recombination. This phenotype is reminiscent of BLM(-)(/)(-) cells and suggests that Fbh1 may be involved in preventing extensive strand exchange during homologous recombination. In addition, disruption of RAD54, a major homologous recombination factor in FBH1(-)(/)(-) cells, results in a marked increase in chromosome-type breaks (breaks on both sister chromatids at the same place) following replication fork arrest. Further, FBH1BLM cells showed additive increases in both sister chromatid exchange and the formation of radial chromosomes. These data suggest that Fbh1 acts in parallel with Bloom helicase to control recombination-mediated double-strand-break repair at replication blocks and to reduce the frequency of crossover.


Assuntos
Adenosina Trifosfatases/metabolismo , Troca Genética , DNA Helicases/metabolismo , Replicação do DNA , Vertebrados/metabolismo , Animais , Camptotecina/farmacologia , Galinhas , Cisplatino/farmacologia , Troca Genética/efeitos dos fármacos , Troca Genética/efeitos da radiação , Dano ao DNA , DNA Helicases/deficiência , DNA Helicases/genética , Replicação do DNA/efeitos dos fármacos , Replicação do DNA/efeitos da radiação , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/metabolismo , Proteína do Grupo de Complementação C da Anemia de Fanconi/deficiência , Proteína do Grupo de Complementação C da Anemia de Fanconi/metabolismo , Raios gama , Deleção de Genes , Marcação de Genes , Instabilidade Genômica/efeitos dos fármacos , Instabilidade Genômica/efeitos da radiação , Metanossulfonato de Metila/farmacologia , Modelos Genéticos , Dados de Sequência Molecular , Mutação/genética , RecQ Helicases , Raios Ultravioleta
11.
Mol Cell Biol ; 25(16): 6948-55, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16055708

RESUMO

Homologous recombination (HR) requires nuclease activities at multiple steps, but the contribution of individual nucleases to the processing of double-strand DNA ends at different stages of HR has not been clearly defined. We used chicken DT40 cells to investigate the role of flap endonuclease 1 (Fen-1) in HR. FEN-1-deficient cells exhibited a significant decrease in the efficiency of immunoglobulin gene conversion while being proficient in recombination between sister chromatids, suggesting that Fen-1 may play a role in HR between sequences of considerable divergence. To clarify whether sequence divergence at DNA ends is truly the reason for the observed HR defect in FEN-1(-/-) cells we inserted a unique I-SceI restriction site in the genome and tested various donor and recipient HR substrates. We found that the efficiency of HR-mediated DNA repair was indeed greatly diminished when divergent sequences were present at the DNA break site. We conclude that Fen-1 eliminates heterologous sequences at DNA damage site and facilitates DNA repair by HR.


Assuntos
Dano ao DNA , Endonucleases Flap/fisiologia , Recombinação Genética , Animais , Sequência de Bases , Ciclo Celular , Galinhas , DNA/química , Reparo do DNA , DNA Complementar/metabolismo , Desoxirribonucleases de Sítio Específico do Tipo II/farmacologia , Endonucleases Flap/metabolismo , Citometria de Fluxo , Raios gama , Cinética , Modelos Genéticos , Dados de Sequência Molecular , Mutação , Plasmídeos/metabolismo , Proteínas de Saccharomyces cerevisiae , Homologia de Sequência do Ácido Nucleico , Troca de Cromátide Irmã , Fatores de Tempo , Transfecção
12.
Mol Cell Biol ; 25(3): 1124-34, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15657438

RESUMO

BRCA2 is a tumor suppressor gene that is linked to hereditary breast and ovarian cancer. Although the Brca2 protein participates in homologous DNA recombination (HR), its precise role remains unclear. From chicken DT40 cells, we generated BRCA2 gene-deficient cells which harbor a truncation at the 3' end of the BRC3 repeat (brca2tr). Comparison of the characteristics of brca2tr cells with those of other HR-deficient DT40 clones revealed marked similarities with rad51 paralog mutants (rad51b, rad51c, rad51d, xrcc2, or xrcc3 cells). The phenotypic similarities include a shift from HR-mediated diversification to single-nucleotide substitutions in the immunoglobulin variable gene segment and the partial reversion of this shift by overexpression of Rad51. Although recent evidence supports at least Xrcc3 and Rad51C playing a role late in HR, our data suggest that Brca2 and the Rad51 paralogs may also contribute to HR at the same early step, with their loss resulting in the stimulation of an alternative, error-prone repair pathway.


Assuntos
Proteína BRCA2/metabolismo , Proteínas de Ligação a DNA/metabolismo , Região Variável de Imunoglobulina/genética , Mutação/genética , Recombinação Genética/genética , Animais , Proteínas Aviárias , Proteína BRCA2/genética , Células Cultivadas , Galinhas , Proteínas de Ligação a DNA/genética , Conversão Gênica/genética , Região Variável de Imunoglobulina/imunologia , Rad51 Recombinase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA