Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nature ; 616(7956): 339-347, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36991126

RESUMO

There is a need to develop effective therapies for pancreatic ductal adenocarcinoma (PDA), a highly lethal malignancy with increasing incidence1 and poor prognosis2. Although targeting tumour metabolism has been the focus of intense investigation for more than a decade, tumour metabolic plasticity and high risk of toxicity have limited this anticancer strategy3,4. Here we use genetic and pharmacological approaches in human and mouse in vitro and in vivo models to show that PDA has a distinct dependence on de novo ornithine synthesis from glutamine. We find that this process, which is mediated through ornithine aminotransferase (OAT), supports polyamine synthesis and is required for tumour growth. This directional OAT activity is usually largely restricted to infancy and contrasts with the reliance of most adult normal tissues and other cancer types on arginine-derived ornithine for polyamine synthesis5,6. This dependency associates with arginine depletion in the PDA tumour microenvironment and is driven by mutant KRAS. Activated KRAS induces the expression of OAT and polyamine synthesis enzymes, leading to alterations in the transcriptome and open chromatin landscape in PDA tumour cells. The distinct dependence of PDA, but not normal tissue, on OAT-mediated de novo ornithine synthesis provides an attractive therapeutic window for treating patients with pancreatic cancer with minimal toxicity.


Assuntos
Ornitina-Oxo-Ácido Transaminase , Neoplasias Pancreáticas , Poliaminas , Animais , Humanos , Camundongos , Arginina/deficiência , Arginina/metabolismo , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Ornitina/biossíntese , Ornitina/metabolismo , Ornitina-Oxo-Ácido Transaminase/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Poliaminas/metabolismo , Microambiente Tumoral
2.
Heliyon ; 8(10): e10991, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36276728

RESUMO

A processing map is required for Ti alloys to find processing parameters securing a high formability. This study adopted the extreme gradient boosting (XGB) approach of machine learning to predict a flow curve and plot a processing map with less experiments for the first time. The optimum XGB model predicted flow curves of Ti-6Al-2Sn-2Zr-2Mo-2Cr-0.15Si at 1073-1273 K and 10 s-1. The predicted data were used to plot a processing map, which showed a higher accuracy in the instability map as compared with the map without XGB. The XGB model also anticipated the power dissipation map at low strain rates. The low accuracy at high strain rates would be improved by alleviating the bias towards a flow hardening. This work has successfully proven the potential usefulness of XGB for plotting an enhanced processing map in light of a higher accuracy with less experiments.

3.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33653947

RESUMO

Pancreatic ductal adenocarcinoma (PDA) is a lethal, therapy-resistant cancer that thrives in a highly desmoplastic, nutrient-deprived microenvironment. Several studies investigated the effects of depriving PDA of either glucose or glutamine alone. However, the consequences on PDA growth and metabolism of limiting both preferred nutrients have remained largely unknown. Here, we report the selection for clonal human PDA cells that survive and adapt to limiting levels of both glucose and glutamine. We find that adapted clones exhibit increased growth in vitro and enhanced tumor-forming capacity in vivo. Mechanistically, adapted clones share common transcriptional and metabolic programs, including amino acid use for de novo glutamine and nucleotide synthesis. They also display enhanced mTORC1 activity that prevents the proteasomal degradation of glutamine synthetase (GS), the rate-limiting enzyme for glutamine synthesis. This phenotype is notably reversible, with PDA cells acquiring alterations in open chromatin upon adaptation. Silencing of GS suppresses the enhanced growth of adapted cells and mitigates tumor growth. These findings identify nongenetic adaptations to nutrient deprivation in PDA and highlight GS as a dependency that could be targeted therapeutically in pancreatic cancer patients.


Assuntos
Carcinoma Ductal Pancreático/metabolismo , Glutamato-Amônia Ligase/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/metabolismo , Carcinoma Ductal Pancreático/genética , Linhagem Celular Tumoral , Estabilidade Enzimática , Glutamato-Amônia Ligase/genética , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Proteínas de Neoplasias/genética , Neoplasias Pancreáticas/genética
4.
Nat Commun ; 11(1): 6236, 2020 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-33262409

RESUMO

A Correction to this paper has been published: https://doi.org/10.1038/s41467-020-20178-0.

6.
Nat Commun ; 9(1): 3404, 2018 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-30143610

RESUMO

AMP-activated protein kinase (AMPK) plays a key role in controlling energy metabolism in response to physiological and nutritional status. Although AMPK activation has been proposed as a promising molecular target for treating obesity and its related comorbidities, the use of pharmacological AMPK activators has been met with contradictory therapeutic challenges. Here we show a regulatory mechanism for AMPK through its ubiquitination and degradation by the E3 ubiquitin ligase makorin ring finger protein 1 (MKRN1). MKRN1 depletion promotes glucose consumption and suppresses lipid accumulation due to AMPK stabilisation and activation. Accordingly, MKRN1-null mice show chronic AMPK activation in both liver and adipose tissue, resulting in significant suppression of diet-induced metabolic syndrome. We demonstrate also its therapeutic effect by administering shRNA targeting MKRN1 into obese mice that reverses non-alcoholic fatty liver disease. We suggest that ubiquitin-dependent AMPK degradation represents a target therapeutic strategy for metabolic disorders.


Assuntos
Síndrome Metabólica/metabolismo , Ribonucleoproteínas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Adipócitos/metabolismo , Adipócitos/patologia , Animais , Dieta Hiperlipídica/efeitos adversos , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Feminino , Fígado/metabolismo , Fígado/patologia , Masculino , Síndrome Metabólica/genética , Camundongos , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/metabolismo , Ribonucleoproteínas/genética , Ubiquitina-Proteína Ligases/genética
7.
Proc Natl Acad Sci U S A ; 115(16): 4228-4233, 2018 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-29610318

RESUMO

Non-small-cell lung cancer (NSCLC) is a leading cause of cancer death worldwide, with 25% of cases harboring oncogenic Kirsten rat sarcoma (KRAS). Although KRAS direct binding to and activation of PI3K is required for KRAS-driven lung tumorigenesis, the contribution of insulin receptor (IR) and insulin-like growth factor 1 receptor (IGF1R) in the context of mutant KRAS remains controversial. Here, we provide genetic evidence that lung-specific dual ablation of insulin receptor substrates 1/2 (Irs1/Irs2), which mediate insulin and IGF1 signaling, strongly suppresses tumor initiation and dramatically extends the survival of a mouse model of lung cancer with Kras activation and p53 loss. Mice with Irs1/Irs2 loss eventually succumb to tumor burden, with tumor cells displaying suppressed Akt activation and strikingly diminished intracellular levels of essential amino acids. Acute loss of IRS1/IRS2 or inhibition of IR/IGF1R in KRAS-mutant human NSCLC cells decreases the uptake and lowers the intracellular levels of amino acids, while enhancing basal autophagy and sensitivity to autophagy and proteasome inhibitors. These findings demonstrate that insulin/IGF1 signaling is required for KRAS-mutant lung cancer initiation, and identify decreased amino acid levels as a metabolic vulnerability in tumor cells with IR/IGF1R inhibition. Consequently, combinatorial targeting of IR/IGF1R with autophagy or proteasome inhibitors may represent an effective therapeutic strategy in KRAS-mutant NSCLC.


Assuntos
Carcinogênese/metabolismo , Carcinoma Pulmonar de Células não Pequenas/prevenção & controle , Genes ras , Proteínas Substratos do Receptor de Insulina/fisiologia , Fator de Crescimento Insulin-Like I/fisiologia , Insulina/farmacologia , Neoplasias Pulmonares/prevenção & controle , Proteínas Proto-Oncogênicas p21(ras)/fisiologia , Células A549 , Aminoácidos/metabolismo , Animais , Autofagia , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/fisiopatologia , Códon de Terminação , Humanos , Proteínas Substratos do Receptor de Insulina/deficiência , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/fisiopatologia , Camundongos , Proteínas de Neoplasias/fisiologia , Proteólise , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais/fisiologia
8.
Cell Death Differ ; 25(6): 1050-1062, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29472714

RESUMO

Oncogene-induced senescence (OIS) is a critical tumor-suppressor mechanism, which prevents hyper-proliferation and transformation of cells. c-Myc promotes OIS through the transcriptional activation of p14ARF followed by p53 activation. Although the oncogene-mediated transcriptional regulation of p14ARF has been well addressed, the post-translational modification of p14ARF regulated by oncogenic stress has yet to be investigated. Here, we found that c-Myc increased p14ARF protein stability by inducing the transcription of ubiquitin-specific protease 10 (USP10). USP10, in turn, mediated the deubiquitination of p14ARF, preventing its proteasome-dependent degradation. USP10-null mouse embryonic fibroblasts and human primary cells depleted of USP10 bypassed c-Myc-induced senescence via the destabilization of p14ARF, and these cells displayed accelerated hyper-proliferation and transformation. Clinically the c-Myc-USP10-p14ARF axis was disrupted in non-small cell lung cancer patients, resulting in significantly worse overall survival. Our studies indicate that USP10 induced by c-Myc has a crucial role in OIS by maintaining the stability of key tumor suppressor p14ARF.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Senescência Celular , Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteína Supressora de Tumor p14ARF/metabolismo , Ubiquitina Tiolesterase/metabolismo , Ubiquitinação , Células A549 , Animais , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Células MCF-7 , Camundongos , Camundongos Knockout , Estabilidade Proteica , Proteínas Proto-Oncogênicas c-myc/genética , Proteína Supressora de Tumor p14ARF/genética , Ubiquitina Tiolesterase/genética
9.
Sci Rep ; 7: 40023, 2017 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-28059128

RESUMO

PPARγ (Peroxisome proliferator-activated receptor γ) is a nuclear receptor involved in lipid homeostasis and related metabolic diseases. Acting as a transcription factor, PPARγ is a master regulator for adipocyte differentiation. Here, we reveal that CHIP (C-terminus of HSC70-interacting protein) suppresses adipocyte differentiation by functioning as an E3 ligase of PPARγ. CHIP directly binds to and induces ubiquitylation of the PPARγ protein, leading to proteasome-dependent degradation. Stable overexpression or knockdown of CHIP inhibited or promoted adipogenesis, respectively, in 3T3-L1 cells. On the other hand, a CHIP mutant defective in E3 ligase could neither regulate PPARγ protein levels nor suppress adipogenesis, indicating the importance of CHIP-mediated ubiquitylation of PPARγ in adipocyte differentiation. Lastly, a CHIP null embryo fibroblast exhibited augmented adipocyte differentiation with increases in PPARγ and its target protein levels. In conclusion, CHIP acts as an E3 ligase of PPARγ, suppressing PPARγ-mediated adipogenesis.


Assuntos
Adipócitos/citologia , PPAR gama/metabolismo , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/metabolismo , Células 3T3-L1 , Adipócitos/metabolismo , Adipogenia , Animais , Sítios de Ligação , Diferenciação Celular , Linhagem Celular , Regulação da Expressão Gênica , Células HEK293 , Humanos , Camundongos , PPAR gama/química , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Proteólise , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/genética
10.
Cancer Res ; 77(2): 343-354, 2017 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-27793846

RESUMO

The tumor suppressor function of p14ARF is regulated at a posttranslational level via mechanisms yet to be fully understood. Here, we report the identification of an unconventional p14ARF degradation pathway induced by the chaperone HSP90 in association with the E3 ubiquitin ligase C-terminus of HSP70-interacting protein (CHIP). The ternary complex of HSP90, CHIP, and p14ARF was required to induce the lysosomal degradation of p14ARF by an ubiquitination-independent but LAMP2A-dependent mechanism. Depletion of HSP90 or CHIP induced p14ARF-dependent senescence in human fibroblasts. Premature senescence observed in cells genetically deficient in CHIP was rescued in cells that were doubly deficient in CHIP and p14ARF. Notably, non-small cell lung cancer cells (NSCLC) positive for p14ARF were sensitive to treatment with the HSP90 inhibitor geldanamycin. Furthermore, overexpression of HSP90 and CHIP with a concomitant loss of p14ARF correlated with poor prognosis in patients with NSCLC. Our findings identify a relationship between p14ARF and its chaperones that suggest new therapeutic strategies in cancers that overexpress HSP90. Cancer Res; 77(2); 343-54. ©2016 AACR.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Senescência Celular/fisiologia , Proteínas de Choque Térmico HSP90/metabolismo , Neoplasias Pulmonares/patologia , Proteína Supressora de Tumor p14ARF/metabolismo , Animais , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Fibroblastos/metabolismo , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/metabolismo , Lisossomos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos de Riscos Proporcionais , Ubiquitina-Proteína Ligases/metabolismo
11.
Nat Commun ; 6: 7769, 2015 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-26183061

RESUMO

The activity of the phosphatase and tensin homologue (PTEN) is known to be suppressed via post-translational modification. However, the mechanism and physiological significance by which post-translational modifications lead to PTEN suppression remain unclear. Here we demonstrate that PTEN destabilization is induced by EGFR- or oncogenic PI3K mutation-mediated AKT activation in cervical cancer. EGFR/PI3K/AKT-mediated ubiquitination and degradation of PTEN are dependent on the MKRN1 E3 ligase. These processes require the stabilization of MKRN1 via AKT-mediated phosphorylation. In cervical cancer patients with high levels of pAKT and MKRN1 expression, PTEN protein levels are low and correlate with a low 5-year survival rate. Taken together, our results demonstrate that PI3K/AKT signals enforce positive-feedback regulation by suppressing PTEN function.


Assuntos
Carcinoma/genética , Regulação Neoplásica da Expressão Gênica , Proteínas do Tecido Nervoso/metabolismo , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ribonucleoproteínas/metabolismo , Displasia do Colo do Útero/genética , Neoplasias do Colo do Útero/genética , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Carcinogênese/genética , Carcinoma/metabolismo , Carcinoma/patologia , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Movimento Celular , Receptores ErbB/metabolismo , Retroalimentação Fisiológica , Feminino , Células HeLa , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Mutação , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas , Fosforilação , Prognóstico , Processamento de Proteína Pós-Traducional , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serina-Treonina Quinases TOR/metabolismo , Ubiquitinação , Neoplasias do Colo do Útero/metabolismo , Displasia do Colo do Útero/metabolismo
12.
J Natl Cancer Inst ; 104(21): 1660-72, 2012 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-23104211

RESUMO

BACKGROUND: We investigated whether Makorin ring finger protein 1 (MKRN1), an E3 ligase, affects p14ARF-associated cellular senescence and tumorigenesis by posttranslational modification in gastric tumorigenesis. METHODS: A link between MKRN1 and ARF was examined in MKRN1 null mouse embryonic fibroblasts (MEFs) and in human fibroblasts and gastric cancer cells by silencing MKRN1 using small interfering RNA (siRNA) and short hairpin RNA (shRNA). Ubiquitination and proteasomal degradation assays were used to assess p14ARF degradation associated with MKRN1. MKRN1 and p14ARF expression levels were analyzed with immunohistochemistry in malignant and normal tissues from gastric cancer patients and with χ(2) tests. The tumor growth of gastric cancer cells stably expressing MKRN1 shRNA, p14ARF shRNA, or both was examined in mouse xenograft models (n = 4-6) and analyzed with unpaired t tests. All statistical tests were two-sided. RESULTS: MKRN1 knockout MEFs exhibited premature senescence and growth retardation with increased p19ARF protein expression. Similar results were obtained for human fibroblasts or gastric cancer cell lines by MKRN1 knockdown. Biochemical analyses confirmed that MKRN1 targets p14ARF for ubiquitination and subsequent proteasome-dependent degradation. A statistically significant association was shown between MKRN1 overexpression and p14ARF underexpression (P = .016). Xenograft analyses using p53-functional AGS or -dysfunctional SNU601 cells displayed statistically significant tumor growth retardation by silencing MKRN1, which was reversed under depletion of p14ARF (AGS cells, MKRN1 knockdown tumors vs MKRN1 and p14ARF knockdown tumors: 164.6 vs 464.8mm(3), difference = 300.2mm(3), 95% CI = 189.1 to 411.3mm(3), P < .001). CONCLUSIONS: We demonstrated that MKRN1 functions as a novel E3 ligase of p14ARF and that it potentially regulates cellular senescence and tumorigenesis in gastric cancer.


Assuntos
Transformação Celular Neoplásica/metabolismo , Senescência Celular , Proteínas do Tecido Nervoso/metabolismo , Ribonucleoproteínas/metabolismo , Neoplasias Gástricas/metabolismo , Proteína Supressora de Tumor p14ARF/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Camundongos Nus , Proteínas do Tecido Nervoso/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Biossíntese de Proteínas , RNA Interferente Pequeno , Ribonucleoproteínas/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Neoplasias Gástricas/fisiopatologia , Análise Serial de Tecidos , Transplante Heterólogo , Proteína Supressora de Tumor p53/metabolismo , Ubiquitinação
13.
Int J Offender Ther Comp Criminol ; 55(2): 268-86, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20142625

RESUMO

Most research on sentencing outcomes reveals that legally relevant factors such as the seriousness of the offense and prior criminal record are primary determinants. There is, however, a substantial body of research that finds that extralegal factors such as a defendant's sex also influence these outcomes. Most of these latter studies conclude that female defendants receive less severe outcomes compared to their male counterparts. Most of this research, however, is limited to Western societies. To extend this body of research, the current study examines sex differences in sentencing practices for a sample of narcotics offenders in South Korea. Results support previous research; female drug offenders in South Korea are generally treated more leniently than their male counterparts. Tests for interaction effects reveal that the defendant's sex also interacts with other constellations of factors to produce lenient treatment for certain female defendants. These tests, however, also reveal that lenient sentence outcomes are not extended to all female defendants; those with prior drug convictions do not fare better than their male counterparts at the incarceration decision.


Assuntos
Crime/estatística & dados numéricos , Aplicação da Lei , Entorpecentes , Transtornos Relacionados ao Uso de Opioides , Preconceito , Adolescente , Adulto , Idoso , Comércio/legislação & jurisprudência , Controle de Medicamentos e Entorpecentes/legislação & jurisprudência , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Paternalismo , República da Coreia , Fatores Sexuais , Fatores Socioeconômicos , Estereotipagem
14.
EMBO J ; 28(14): 2100-13, 2009 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-19536131

RESUMO

Makorin Ring Finger Protein 1 (MKRN1) is a transcriptional co-regulator and an E3 ligase. Here, we show that MKRN1 simultaneously functions as a differentially negative regulator of p53 and p21. In normal conditions, MKRN1 could destabilize both p53 and p21 through ubiquitination and proteasome-dependent degradation. As a result, depletion of MKRN1 induced growth arrest through activation of p53 and p21. Interestingly, MKRN1 used earlier unknown sites, K291 and K292, for p53 ubiquitination and subsequent degradation. Under severe stress conditions, however, MKRN1 primarily induced the efficient degradation of p21. This regulatory process contributed to the acceleration of DNA damage-induced apoptosis by eliminating p21. MKRN1 depletion diminished adriamycin or ultraviolet-induced cell death, whereas ectopic expression of MKRN1 facilitated apoptosis. Furthermore, MKRN1 stable cell lines that constantly produced low levels of p53 and p21 exhibited stabilization of p53, but not p21, with increased cell death on DNA damage. Our results indicate that MKRN1 exhibits dual functions of keeping cells alive by suppressing p53 under normal conditions and stimulating cell death by repressing p21 under stress conditions.


Assuntos
Apoptose , Ciclo Celular , Proteínas do Tecido Nervoso/metabolismo , Ribonucleoproteínas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Dano ao DNA , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Proteínas do Tecido Nervoso/genética , Complexo de Endopeptidases do Proteassoma , Ribonucleoproteínas/genética , Ubiquitinação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...