Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Dis ; 15(4): 241, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38561375

RESUMO

Soft-tissue sarcomas (STS) emerges as formidable challenges in clinics due to the complex genetic heterogeneity, high rates of local recurrence and metastasis. Exploring specific targets and biomarkers would benefit the prognosis and treatment of STS. Here, we identified RCC1, a guanine-nucleotide exchange factor for Ran, as an oncogene and a potential intervention target in STS. Bioinformatics analysis indicated that RCC1 is highly expressed and correlated with poor prognosis in STS. Functional studies showed that RCC1 knockdown significantly inhibited the cell cycle transition, proliferation and migration of STS cells in vitro, and the growth of STS xenografts in mice. Mechanistically, we identified Skp2 as a downstream target of RCC1 in STS. Loss of RCC1 substantially diminished Skp2 abundance by compromising its protein stability, resulting in the upregulation of p27Kip1 and G1/S transition arrest. Specifically, RCC1 might facilitate the nucleo-cytoplasmic trafficking of Skp2 via direct interaction. As a result, the cytoplasmic retention of Skp2 would further protect it from ubiquitination and degradation. Notably, recovery of Skp2 expression largely reversed the phenotypes induced by RCC1 knockdown in STS cells. Collectively, this study unveils a novel RCC1-Skp2-p27Kip1 axis in STS oncogenesis, which holds promise for improving prognosis and treatment of this formidable malignancy.


Assuntos
Sarcoma , Animais , Humanos , Camundongos , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Quinases Associadas a Fase S/genética , Proteínas Quinases Associadas a Fase S/metabolismo , Sarcoma/genética , Sarcoma/patologia , Ubiquitinação , Regulação para Cima
2.
J Exp Clin Cancer Res ; 42(1): 274, 2023 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-37864196

RESUMO

BACKGROUND: Tumor cells can resist chemotherapy-induced pyroptosis through glycolytic reprogramming. Estrogen-related receptor alpha (ERRα) is a central regulator of cellular energy metabolism associated with poor cancer prognosis. Herein, we refine the oncogenic role of ERRα in the pyroptosis pathway and glycolytic metabolism. METHODS: The interaction between ERRα and HIF-1α was verified using co-immunoprecipitation. The transcriptional binding sites of ERRα and NLRP3 were confirmed using dual-luciferase reporter assay and cleavage under targets and tagmentation (CUT&Tag). Flow cytometry, transmission electron microscopy, scanning electron microscopy, cell mito stress test, and extracellular acidification rate analysis were performed to investigate the effects of ERRα on the pyroptosis pathway and glycolytic metabolism. The results of these experiments were further confirmed in endometrial cancer (EC)-derived organoids and nude mice. In addition, the expression of ERRα-related pyroptosis genes was analyzed using The Cancer Genome Atlas and Gene Expression Omnibus database. RESULTS: Triggered by a hypoxic microenvironment, highly expressed ERRα could bind to the promoter of NLRP3 and inhibit caspase-1/GSDMD signaling, which reduced inflammasome activation and increased pyroptosis resistance, thereby resulting in the resistance of cancer cells to cisplatin. Moreover, ERRα activated glycolytic rate-limiting enzyme to bridge glycolytic metabolism and pyroptosis in EC. This phenomenon was further confirmed in EC-derived organoids and nude mice. CUT & Tag sequencing and The Cancer Genome Atlas database analysis showed that ERRα participated in glycolysis and programmed cell death, which resulted in EC progression. CONCLUSIONS: ERRα inhibits pyroptosis in an NLRP3-dependent manner and induces glycolytic metabolism, resulting in cisplatin resistance in EC cells.


Assuntos
Neoplasias do Endométrio , Proteína 3 que Contém Domínio de Pirina da Família NLR , Humanos , Camundongos , Animais , Feminino , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Caspase 1/genética , Caspase 1/metabolismo , Caspase 1/farmacologia , Camundongos Nus , Piroptose , Cisplatino/farmacologia , Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/genética , Glicólise , Microambiente Tumoral , Proteínas de Ligação a Fosfato/genética , Proteínas de Ligação a Fosfato/metabolismo , Proteínas de Ligação a Fosfato/farmacologia , Proteínas Citotóxicas Formadoras de Poros/metabolismo
3.
Medicine (Baltimore) ; 102(32): e34509, 2023 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-37565888

RESUMO

BACKGROUND: Sleep disorders significantly affect the quality of life in Parkinson disease (PD) patients. Deep brain stimulation of the subthalamic nucleus has been reported to improve motor symptoms and decrease medication usage. However, the impact of subthalamic nucleus deep brain stimulation (STN-DBS) on sleep quality in PD patients remains to be definitively determined. This systematic review and meta-analysis, conducted following the preferred reporting items for systematic reviews and meta-analyses guidelines, aimed to clarify the effect of STN-DBS on sleep quality in PD patients. METHODS: A rigorous literature search identified 6 studies, including 1 randomized controlled trial and 5 self-controlled trials, totaling 154 patients who underwent deep brain stimulation, providing 308 pairs of data for analysis. Parkinson disease sleep scale was the primary measure of interest, while the Movement Disorder Society-sponsored revision of the unified Parkinson disease rating scale was documented in all trials. Study quality was assessed using the Newcastle-Ottawa scale. RESULTS: STN-DBS significantly improved Parkinson disease sleep scale scores (mean difference = 20.41, 95% CI: [13.03, 27.79], I² = 60.8%, P < .001), indicating enhanced sleep quality. Furthermore, a significant reduction in movement disorder society unified Parkinson disease rating scale part III scores postoperatively (mean difference = -12.59, 95% CI: [-14.70, -10.49], I² = 89.9%, P < .001) suggested improved motor function. PD medication usage was also significantly reduced postoperatively (mean difference = -314.71, 95% CI: [-468.13, -161.28], I² = 52.9%, P < .001). A sensitivity analysis confirmed the robustness of the main findings. The sample size was adequate, allowing for conclusive inferences. CONCLUSION: The present study, which comprises a comprehensive systematic review and meta-analysis, offers compelling evidence that STN-DBS can ameliorate sleep quality, augment motor function, and curtail medication consumption among individuals afflicted with PD.


Assuntos
Estimulação Encefálica Profunda , Doença de Parkinson , Núcleo Subtalâmico , Humanos , Núcleo Subtalâmico/fisiologia , Doença de Parkinson/complicações , Doença de Parkinson/terapia , Qualidade de Vida , Sono/fisiologia , Resultado do Tratamento , Ensaios Clínicos Controlados Aleatórios como Assunto
4.
J Cancer ; 14(10): 1935-1945, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37476192

RESUMO

Objective: We retrospectively studied cancer mortality and incidence in China from 1990 to 2019, investigated the cancer trends and risk factors, and analyzed the effects of Gross Domestic Product (GDP) on cancer mortality and incidence. Methods: Data was obtained in "Our world in data" in October 2022 to explore mortality rates of different cancers and their trends and the roles of cancer risk factors, including GDP, air pollution, etc. Results: Over the past 30 years, cancer had been China's second leading cause of death. Tracheal, bronchial, and lung cancers, with an annual growth rate of 6.5%, were the most frequently diagnosed cancers. The burden of different cancers changed as the mortality rate of cancer changed. The age-standardized cancer mortality rate had decreased by 19.0%; cancer deaths in all age groups had increased. While the number of cancer deaths in the elderly aged ≥70 did not increase distinctively, its percentage increased by 52.1% and 1.7% annually. The percentage of patients with new-onset cancer increased by 240% and 8.6% annually. For every USD 1,000 increase in GDP, cancer deaths decreased by 2.3/100,000. Tobacco, meat, and alcohol consumption and BMI had increased and were not conducive to the future control of cancer. Conclusions: We summarized the incidence and mortality of major cancers and their trends in China over the past 30 years and analyzed the effects of GDP and the roles of cancer risk factors. Overall GDP growth and effective control of air pollution reduced cancer mortality, while population aging, smoking, alcohol consumption, BMI increasing, and meat consumption brought challenges for cancer control.

5.
Small ; 18(14): e2106046, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35182014

RESUMO

Advanced prostate cancer, harboring multiple mutations of tumor suppressor genes, is refractory to conventional therapies. Knockout of the Skp2 gene blocks pRb/p53 doubly deficient prostate cancer in mice, which inspired the authors to develop an approach for delivering siRNA that would efficiently silence Skp2 (siSkp2) in vivo. Here, a facile strategy is reported to directly assemble siSkp2 with the natural compound quercetin (Que) into supramolecular nanoparticles (NPs). This carrier-free siSkp2 delivery system could effectively protect siSkp2 from degradation in serum and enhance its cellular internalization. Furthermore, the siSkp2/Que NPs exhibit synergistic effects in Skp2 silencing, because they can degrade the mRNA and protein of Skp2 simultaneously. Indeed, siSkp2/Que NPs remarkably diminish the Skp2 abundance and further inhibit the proliferation and migration of TMU cells (RB1/TP53/KRAS triple mutations) in vitro. The in vivo results further show that i.v. administration of siSkp2/Que NPs efficiently accumulates in tumor sites and strongly inhibits the growth of TMU tumors in nude mice. Importantly, the siSkp2/Que NPs do not induce any abnormality in the treated mice, which suggests satisfactory biocompatibility. Collectively, this study describes a tractable siRNA self-assembled strategy for Skp2 silencing, which might be a promising nanodrug to cure multitherapy-resistant advanced prostate cancer.


Assuntos
Nanopartículas , Neoplasias da Próstata , Animais , Linhagem Celular Tumoral , Humanos , Masculino , Camundongos , Camundongos Knockout , Camundongos Nus , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , RNA Interferente Pequeno/genética
6.
Exp Cell Res ; 389(1): 111848, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31954693

RESUMO

Loss of neuron homeostasis in the arcuate nucleus (ARC) is responsible for diet-induced-obesity (DIO). We previously reported that loss of Rb1 gene compromised the homeostasis of anorexigenic POMC neurons in ARC and induced obesity in mice. To evaluate the development of DIO, we propose to analyze the transcriptomic alteration of POMC neurons in mice following high fat diet (HFD) feeding. We isolated these neurons from established DIO mice and performed transcriptomic profiling using RNA-seq. In total, 1066 genes (628 upregulated and 438 downregulated) were identified as differentially expressed genes (DEGs). Pathway enrichment analysis with these DEGs further revealed that "cell cycle," "apoptosis," "chemokine signaling," and "sphingolipid metabolism" pathways were correlated with DIO development. Moreover, we validated that the pRb protein, a key regulator of "cell cycle pathway," was inactivated by phosphorylation in POMC neurons by HFD feeding. Importantly, the reversal of deregulated cell cycle by stereotaxic delivering of the unphosphorylated pRbΔP in ARC significantly meliorated the DIO. Collectively, our study provides insights into the mechanisms related to the loss of homeostasis of POMC neurons in DIO, and suggests pRb phosphorylation as a potential intervention target to treat DIO.


Assuntos
Dieta Hiperlipídica , Neurônios/metabolismo , Obesidade/genética , Pró-Opiomelanocortina/metabolismo , Transcriptoma , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/patologia , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Obesos , Camundongos Transgênicos , Neurônios/patologia , Obesidade/etiologia , Obesidade/patologia , Pró-Opiomelanocortina/genética
7.
Sheng Wu Gong Cheng Xue Bao ; 35(8): 1433-1440, 2019 Aug 25.
Artigo em Chinês | MEDLINE | ID: mdl-31441614

RESUMO

The social problems and medical burdens caused by obesity have become more serious in recent years. Obesity is mainly caused by the imbalance of energy intake and consumption in the body. The central nervous system and related neurons regulate the balance of energy metabolism. The hypothalamic arcuate nucleus (ARC) contains anorexigenic proopiomelanocortin (POMC) neurons and orexigenic neuropeptid Y(NPY)/agouti-related protein (AgRP) neurons that regulate the feeding behavior of body. High-fat diet induces phosphorylation of Rb protein in POMC neurons, and inactivation of Rb phosphorylation leads to re-entry of POMC neurons from the resting-state into the cell cycle, which rapidly shifts to apoptosis. High-fat diet also causes the inhibition of neuronal regeneration, induces inflammation and neuronal damage, loss of neuronal homeostasis, leptin resistance, and ultimately leads to obesity. This review discusses the relationship between loss of neuronal homeostasis and dietary obesity, as well as the underlying mechanisms, which might provide the evidence for prevention and treatment of these diseases.


Assuntos
Núcleo Arqueado do Hipotálamo , Obesidade , Homeostase , Humanos , Leptina , Pró-Opiomelanocortina
8.
JCI Insight ; 3(17)2018 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-30185666

RESUMO

When obesity is caused by consumption of a high-fat diet, the tumor suppressor pRb is phosphoinactivated in the neurons of the mediobasal hypothalamus, a brain area critical for energy-balance regulation. However, the functional relevance of pRb phosphoinactivation in the mediobasal hypothalamus to diet-induced obesity remains unknown. Here, we show that inhibiting pRb phosphorylation in the mediobasal hypothalamus can prevent and treat diet-induced obesity in mice. Expressing an unphosphorylable pRb nonselectively in the mediobasal hypothalamus or conditionally in anorexigenic POMC neurons inhibits diet-induced obesity. Intracerebroventricular delivery of US Food and Drug Administration-approved (FDA-approved) cyclin-dependent kinase 4 (CDK4) inhibitor abemaciclib inhibits pRb phosphorylation in the mediobasal hypothalamus and prevents diet-induced obesity. Oral administration of abemaciclib at doses approved for human use reduces fat mass in diet-induced obese mice by increasing lipid oxidation without significantly reducing lean mass. With analysis of recent literature identifying CDK4 as the most abundantly expressed neuronal CDK in the mediobasal hypothalamus, our work uncovers CDK4 as the major kinase for hypothalamic pRb phosphoinactivation and a highly effective central antiobesity target. As three CDK4/6 inhibitors have recently received FDA approval for life-long breast cancer therapy, our study provides a preclinical basis for their expedient repurposing for obesity management.


Assuntos
Aminopiridinas/farmacologia , Benzimidazóis/farmacologia , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 4 Dependente de Ciclina/metabolismo , Dieta Hiperlipídica/efeitos adversos , Obesidade/metabolismo , Obesidade/prevenção & controle , Animais , Modelos Animais de Doenças , Aprovação de Drogas , Metabolismo Energético , Hipotálamo/metabolismo , Metabolismo dos Lipídeos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação/efeitos dos fármacos , Proteína do Retinoblastoma/metabolismo , Estados Unidos , United States Food and Drug Administration
9.
Cancer Cell ; 31(5): 669-684.e7, 2017 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-28486106

RESUMO

Polyploidy can lead to aneuploidy and tumorigenesis. Here, we report that the Hippo pathway effector Yap promotes the diploid-polyploid conversion and polyploid cell growth through the Akt-Skp2 axis. Yap strongly induces the acetyltransferase p300-mediated acetylation of the E3 ligase Skp2 via Akt signaling. Acetylated Skp2 is exclusively localized to the cytosol, which causes hyper-accumulation of the cyclin-dependent kinase inhibitor p27, leading to mitotic arrest and subsequently cell polyploidy. In addition, the pro-apoptotic factors FoxO1/3 are overly degraded by acetylated Skp2, resulting in polyploid cell division, genomic instability, and oncogenesis. Importantly, the depletion or inactivation of Akt or Skp2 abrogated Hippo signal deficiency-induced liver tumorigenesis, indicating their epistatic interaction. Thus, we conclude that Hippo-Yap signaling suppresses cell polyploidy and oncogenesis through Skp2.


Assuntos
Carcinoma Hepatocelular/enzimologia , Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Neoplasias Hepáticas/enzimologia , Ploidias , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases Associadas a Fase S/metabolismo , Acetilação , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Proteínas de Ciclo Celular , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Citosol/enzimologia , Epistasia Genética , Feminino , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Regulação Neoplásica da Expressão Gênica , Predisposição Genética para Doença , Células Hep G2 , Via de Sinalização Hippo , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Transgênicos , Fenótipo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Gravidez , Proteínas Serina-Treonina Quinases/genética , Estabilidade Proteica , Proteólise , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Proteínas Quinases Associadas a Fase S/genética , Transdução de Sinais , Fatores de Tempo , Fatores de Transcrição , Transfecção , Proteínas de Sinalização YAP , Fatores de Transcrição de p300-CBP/metabolismo
10.
J Biol Chem ; 291(19): 10201-9, 2016 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-26966181

RESUMO

Tumor suppressor pRb represses Skp2, a substrate-recruiting subunit of the SCF(Skp2) ubiquitin ligase. Rb1(+/-) mice incur "two-hit" pituitary tumorigenesis; Skp2(-/-);Rb1(+/-) mice do not. Rb1(-/-) embryos die on embryonic day (E) 14.5-15.5. Here, we report that Skp2(-/-);Rb1(-/-) embryos died on E11.5, establishing an organismal level synthetic lethal relationship between Rb1 and Skp2 On E10.5, Rb1(-/-) placentas showed similarly active proliferation and similarly inactive apoptosis as WT placenta, whereas Rb1(-/-) embryos showed ectopic proliferation without increased apoptosis in the brain. Combining Skp2(-/-) did not reduce proliferation or increase apoptosis in the placentas but induced extensive apoptosis in the brain. We conditionally deleted Rb1 in neuronal lineage with Nes-Cre and reproduced the brain apoptosis in E13.5 Nes-Cre;Rb1(lox/lox);Skp2(-/-) embryos, demonstrating their synthetic lethal relationship at a cell autonomous level. Nes-Cre-mediated Rb1 deletion increased expression of proliferative E2F target genes in the brains of Skp2(+/+) embryos; the increases rose higher with activation of expression of apoptotic E2F target genes in Skp2(-/-) embryos. The brain apoptosis was independent of p53 but coincident with proliferation. The highly activated expression of proliferative and apoptotic E2F target genes subsided with gradually reduced roles of Skp2 in preventing p27 protein accumulation in the brain in late gestation, allowing the embryos to reach full term with normally sized brains. These findings establish that Rb1 and Skp2 deletions are synthetic lethal and suggest how this lethal relationship might be circumvented, which could help design better therapies for pRb-deficient cancer.


Assuntos
Apoptose , Perda do Embrião , Embrião de Mamíferos , Desenvolvimento Embrionário/genética , Proteína do Retinoblastoma , Proteínas Quinases Associadas a Fase S , Animais , Apoptose/genética , Encéfalo/embriologia , Encéfalo/patologia , Fatores de Transcrição E2F/genética , Perda do Embrião/genética , Perda do Embrião/metabolismo , Perda do Embrião/patologia , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Feminino , Deleção de Genes , Masculino , Camundongos , Camundongos Knockout , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Proteínas Quinases Associadas a Fase S/genética , Proteínas Quinases Associadas a Fase S/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
11.
Sci Rep ; 6: 19275, 2016 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-26763940

RESUMO

Liver repopulation by transplanted hepatocytes has not been achieved previously in a normal liver microenvironment. Here we report that adult rat hepatocytes transduced ex vivo with a lentivirus expressing a human YapERT2 fusion protein (hYapERT2) under control of the hepatocyte-specific transthyretin (TTR) promoter repopulate normal rat liver in a tamoxifen-dependent manner. Transplanted hepatocytes expand very slowly but progressively to produce 10% repopulation at 6 months, showing clusters of mature hepatocytes that are fully integrated into hepatic parenchyma, with no evidence for dedifferentiation, dysplasia or malignant transformation. Thus, we have developed the first vector designed to regulate the growth control properties of Yap that renders it capable of producing effective cell therapy. The level of liver repopulation achieved has significant translational implications, as it is 2-3x the level required to cure many monogenic disorders of liver function that have no underlying hepatic pathology and is potentially applicable to diseases of other tissues and organs.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Hepatócitos/metabolismo , Proteínas Nucleares/genética , Pré-Albumina/genética , Regiões Promotoras Genéticas , Proteínas Recombinantes de Fusão , Fatores de Transcrição/genética , Transdução Genética , Animais , Proteínas de Ciclo Celular , Expressão Gênica , Genes Reporter , Vetores Genéticos/genética , Hepatócitos/efeitos dos fármacos , Hepatócitos/transplante , Lentivirus/genética , Regeneração Hepática , Modelos Animais , Transporte Proteico , Ratos , Tamoxifeno/farmacologia
12.
J Biol Chem ; 290(9): 5797-809, 2015 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-25583987

RESUMO

p27Kip1 (p27) is an inhibitor of cyclin-dependent kinases. Inhibiting p27 protein degradation is an actively developing cancer therapy strategy. One focus has been to identify small molecule inhibitors to block recruitment of Thr-187-phosphorylated p27 (p27T187p) to SCF(Skp2/Cks1) ubiquitin ligase. Since phosphorylation of Thr-187 is required for this recruitment, p27T187A knockin (KI) mice were generated to determine the effects of systemically blocking interaction between p27 and Skp2/Cks1 on tumor susceptibility and other proliferation related mouse physiology. Rb1(+/-) mice develop pituitary tumors with full penetrance and the tumors are invariably Rb1(-/-), modeling tumorigenesis by two-hit loss of RB1 in humans. Immunization induced humoral immunity depends on rapid B cell proliferation and clonal selection in germinal centers (GCs) and declines with age in mice and humans. Here, we show that p27T187A KI prevented pituitary tumorigenesis in Rb1(+/-) mice and corrected decline in humoral immunity in older mice following immunization with sheep red blood cells (SRBC). These findings reveal physiological contexts that depend on p27 ubiquitination by SCF(Skp2-Cks1) ubiquitin ligase and therefore help forecast clinical potentials of Skp2/Cks1-p27T187p interaction inhibitors. We further show that GC B cells and T cells use different mechanisms to regulate their p27 protein levels, and propose a T helper cell exhaustion model resembling that of stem cell exhaustion to understand decline in T cell-dependent humoral immunity in older age.


Assuntos
Substituição de Aminoácidos , Inibidor de Quinase Dependente de Ciclina p27/genética , Imunidade Humoral/genética , Hipófise/metabolismo , Neoplasias Hipofisárias/genética , Proteína do Retinoblastoma/genética , Fatores Etários , Alanina/genética , Alanina/metabolismo , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Carcinogênese/genética , Carcinogênese/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Eritrócitos/imunologia , Citometria de Fluxo , Técnicas de Introdução de Genes , Centro Germinativo/citologia , Centro Germinativo/imunologia , Centro Germinativo/metabolismo , Humanos , Imunidade Humoral/imunologia , Imuno-Histoquímica , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Hipófise/patologia , Neoplasias Hipofisárias/metabolismo , Proteína do Retinoblastoma/metabolismo , Proteínas Ligases SKP Culina F-Box/metabolismo , Ovinos , Linfócitos T/imunologia , Linfócitos T/metabolismo , Treonina/genética , Treonina/metabolismo
13.
Nat Commun ; 5: 3463, 2014 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-24632684

RESUMO

One mechanism of tumour suppression by pRb is repressing E2F1. Hence, E2f1 deletion diminishes tumorigenesis following Rb1 loss. However, E2F1 promotes both proliferation and apoptosis. It therefore remains unclear how de-repressed E2F1 promotes tumorigenesis. Another mechanism of pRb function is repressing Skp2 to elevate p27 to arrest proliferation. However, Skp2 deletion induced apoptosis, not proliferation arrest, in Rb1-deficient pituitary tumorigenesis. Here we show that Rb1 deletion induces higher expression of E2F1 target genes in the absence of Skp2. E2F1 binds less cyclin A but more target promoters when Rb1 is deleted with Skp2 knockout or p27T187A knockin, suggesting that stabilized p27 prevents cyclin A from binding and inhibiting E2F1. In Rb1-deficient pituitary tumorigenesis, Skp2 deletion or p27T187A mutation converts E2F1's role from proliferative to apoptotic. These findings delineate a pRb-Skp2-p27-cyclin A-E2F1 pathway that determines whether E2F1 is proliferative or apoptotic in Rb1-deficient tumorigenesis.


Assuntos
Apoptose , Regulação para Baixo , Fator de Transcrição E2F1/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Proteína do Retinoblastoma/deficiência , Proteínas Quinases Associadas a Fase S/metabolismo , Animais , Proliferação de Células , Fator de Transcrição E2F1/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos Knockout , Neoplasias/fisiopatologia , Hipófise/metabolismo , Proteína do Retinoblastoma/genética , Proteínas Quinases Associadas a Fase S/genética
14.
PLoS One ; 9(3): e91298, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24614118

RESUMO

Most mushrooms are thermo-sensitive to temperatures over 23°C, which greatly restricts their agricultural cultivation. Understanding mushroom's innate heat-tolerance mechanisms may facilitate genetic improvements of their thermotolerance. Agaricus bisporus strain 02 is a relatively thermotolerant mushroom strain, while strain 8213 is quite thermo-sensitive. Here, we compared their responses at proteomic level to heat treatment at 33°C. We identified 73 proteins that are differentially expressed between 02 and 8213 or induced upon heat stress in strain 02 itself, 48 of which with a known identity. Among them, 4 proteins are constitutively more highly expressed in 02 than 8213; and they can be further upregulated in response to heat stress in 02, but not in 8213. One protein is encoded by the para-aminobenzoic acid (PABA) synthase gene Pabs, which has been shown to scavenge the reactive oxygen species in vitro. Pabs mRNA and its chemical product PABA show similar heat stress induction pattern as PABA synthase protein and are more abundant in 02, indicating transcriptional level upregulation of Pabs upon heat stress. A specific inhibitor of PABA synthesis impaired thermotolerance of 02, while exogenous PABA or transgenic overexpression of 02 derived PABA synthase enhanced thermotolerance of 8213. Furthermore, compared to 8213, 02 accumulated less H2O2 but more defense-related proteins (e.g., HSPs and Chitinase) under heat stress. Together, these results demonstrate a role of PABA in enhancing mushroom thermotolerance by removing H2O2 and elevating defense-related proteins.


Assuntos
Adaptação Fisiológica , Agaricus/enzimologia , Agaricus/fisiologia , Temperatura , Transaminases/metabolismo , Ácido 4-Aminobenzoico/farmacologia , Adaptação Fisiológica/efeitos dos fármacos , Agaricus/efeitos dos fármacos , Catalase/metabolismo , Quitinases/metabolismo , Eletroforese em Gel Bidimensional , Proteínas Fúngicas/metabolismo , Resposta ao Choque Térmico/efeitos dos fármacos , Peróxido de Hidrogênio/metabolismo , Modelos Biológicos , Estresse Oxidativo/efeitos dos fármacos , Proteômica , Sulfanilamida , Sulfanilamidas/farmacologia , Superóxido Dismutase/metabolismo , Transgenes
15.
Cancer Cell ; 24(5): 645-59, 2013 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-24229711

RESUMO

pRb and p53 are two major tumor suppressors. Here, we found that p53 activates expression of Pirh2 and KPC1, two of the three ubiquitin ligases for p27. Loss of p53 in the absence of Skp2, the third ubiquitin ligase for p27, shrinks the cellular pool of p27 ubiquitin ligases to accumulate p27 protein. In the absence of pRb and p53, p27 was unable to inhibit DNA synthesis in spite of its abundance, but could inhibit division of cells that maintain DNA replication with rereplication. This mechanism blocked pRb/p53 doubly deficient pituitary and prostate tumorigenesis lastingly coexistent with bromodeoxyuridine-labeling neoplastic lesions, revealing an unconventional cancer cell vulnerability when pRb and p53 are inactivated.


Assuntos
Carcinogênese/genética , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Neoplasia Prostática Intraepitelial/genética , Neoplasias da Próstata/genética , Proteína do Retinoblastoma/metabolismo , Proteínas Quinases Associadas a Fase S/genética , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose , Carcinogênese/metabolismo , Linhagem Celular Tumoral , Senescência Celular , Replicação do DNA , Deleção de Genes , Humanos , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Neoplasias Hipofisárias/genética , Neoplasias Hipofisárias/patologia , Neoplasia Prostática Intraepitelial/patologia , Neoplasias da Próstata/patologia , Proteína do Retinoblastoma/genética , Proteína Supressora de Tumor p53/genética , Ubiquitina-Proteína Ligases/metabolismo
16.
EMBO J ; 32(6): 844-57, 2013 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-23403926

RESUMO

pRb is frequently inactivated in tumours by mutations or phosphorylation. Here, we investigated whether pRb plays a role in obesity. The Arcuate nucleus (ARC) in hypothalamus contains antagonizing POMC and AGRP/NPY neurons for negative and positive energy balance, respectively. Various aspects of ARC neurons are affected in high-fat diet (HFD)-induced obesity mouse model. Using this model, we show that HFD, as well as pharmacological activation of AMPK, induces pRb phosphorylation and E2F target gene de-repression in ARC neurons. Some affected neurons express POMC; and deleting Rb1 in POMC neurons induces E2F target gene de-repression, cell-cycle re-entry, apoptosis, and a hyperphagia-obesity-diabetes syndrome. These defects can be corrected by combined deletion of E2f1. In contrast, deleting Rb1 in the antagonizing AGRP/NPY neurons shows no effects. Thus, pRb-E2F1 is an obesity suppression mechanism in ARC POMC neurons and HFD-AMPK inhibits this mechanism by phosphorylating pRb in this location.


Assuntos
Dieta Hiperlipídica , Gorduras na Dieta/farmacologia , Hipotálamo , Obesidade/genética , Proteína do Retinoblastoma/antagonistas & inibidores , Proteína do Retinoblastoma/fisiologia , Adenilato Quinase/metabolismo , Adenilato Quinase/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/fisiologia , Dieta Hiperlipídica/efeitos adversos , Regulação para Baixo/genética , Fator de Transcrição E2F1/metabolismo , Fator de Transcrição E2F1/fisiologia , Feminino , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Hipotálamo/patologia , Peso Corporal Ideal/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/fisiologia , Obesidade/metabolismo , Obesidade/patologia , Fosforilação/efeitos dos fármacos , Pró-Opiomelanocortina/metabolismo , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo
17.
Am J Bot ; 98(9): e239-41, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21846791

RESUMO

PREMISE OF THE STUDY: Microsatellite primers were developed for taro (Colocasia esculenta) to investigate its population genetics and evolutionary history through germplasm improvement. • METHODS AND RESULTS: Nineteen microsatellite loci were identified in three populations. The number of alleles per locus ranged from two to seven, with a mean of 4.68. The observed and expected heterozygosities ranged from 0.231 to 0.820 and from 0.126 to 0.742, respectively. • CONCLUSIONS: These new genetic markers will be useful for the study of taro germplasm management and population evolution in southwestern China.


Assuntos
Araceae/genética , Repetições de Microssatélites/genética , Alelos , Sequência de Bases , Primers do DNA , Genes de Plantas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...