Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Small ; 18(14): e2106046, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35182014

RESUMEN

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.


Asunto(s)
Nanopartículas , Neoplasias de la Próstata , Animales , Línea Celular Tumoral , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Desnudos , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , ARN Interferente Pequeño/genética
2.
Exp Cell Res ; 389(1): 111848, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31954693

RESUMEN

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.


Asunto(s)
Dieta Alta en Grasa , Neuronas/metabolismo , Obesidad/genética , Proopiomelanocortina/metabolismo , Transcriptoma , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Núcleo Arqueado del Hipotálamo/patología , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Obesos , Ratones Transgénicos , Neuronas/patología , Obesidad/etiología , Obesidad/patología , Proopiomelanocortina/genética
3.
J Biol Chem ; 291(19): 10201-9, 2016 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-26966181

RESUMEN

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.


Asunto(s)
Apoptosis , Pérdida del Embrión , Embrión de Mamíferos , Desarrollo Embrionario/genética , Proteína de Retinoblastoma , Proteínas Quinasas Asociadas a Fase-S , Animales , Apoptosis/genética , Encéfalo/embriología , Encéfalo/patología , Factores de Transcripción E2F/genética , Pérdida del Embrión/genética , Pérdida del Embrión/metabolismo , Pérdida del Embrión/patología , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Femenino , Eliminación de Gen , Masculino , Ratones , Ratones Noqueados , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Proteínas Quinasas Asociadas a Fase-S/genética , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
4.
EMBO J ; 32(6): 844-57, 2013 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-23403926

RESUMEN

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.


Asunto(s)
Dieta Alta en Grasa , Grasas de la Dieta/farmacología , Hipotálamo , Obesidad/genética , Proteína de Retinoblastoma/antagonistas & inhibidores , Proteína de Retinoblastoma/fisiología , Adenilato Quinasa/metabolismo , Adenilato Quinasa/fisiología , Animales , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/metabolismo , Núcleo Arqueado del Hipotálamo/fisiología , Dieta Alta en Grasa/efectos adversos , Regulación hacia Abajo/genética , Factor de Transcripción E2F1/metabolismo , Factor de Transcripción E2F1/fisiología , Femenino , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Hipotálamo/patología , Peso Corporal Ideal/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/fisiología , Obesidad/metabolismo , Obesidad/patología , Fosforilación/efectos de los fármacos , Proopiomelanocortina/metabolismo , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo
5.
J Biol Chem ; 290(9): 5797-809, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-25583987

RESUMEN

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.


Asunto(s)
Sustitución de Aminoácidos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inmunidad Humoral/genética , Hipófisis/metabolismo , Neoplasias Hipofisarias/genética , Proteína de Retinoblastoma/genética , Factores de Edad , Alanina/genética , Alanina/metabolismo , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Carcinogénesis/genética , Carcinogénesis/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Eritrocitos/inmunología , Citometría de Flujo , Técnicas de Sustitución del Gen , Centro Germinal/citología , Centro Germinal/inmunología , Centro Germinal/metabolismo , Humanos , Inmunidad Humoral/inmunología , Inmunohistoquímica , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Hipófisis/patología , Neoplasias Hipofisarias/metabolismo , Proteína de Retinoblastoma/metabolismo , Proteínas Ligasas SKP Cullina F-box/metabolismo , Ovinos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Treonina/genética , Treonina/metabolismo
6.
Biochim Biophys Acta Mol Basis Dis ; 1870(6): 167233, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38744342

RESUMEN

Hepatocellular carcinoma (HCC) poses a significant threat due to its highly aggressive and high recurrence characteristics, necessitating urgent advances in diagnostic and therapeutic approaches. Long non-coding RNAs exert vital roles in HCC tumorigenesis, however the mechanisms of their expression regulation and functions are not fully elucidated yet. Herein, we identify that a novel tumor suppressor 'lnc-PIK3R1' was significantly downregulated in HCC tissues, which was correlated with poor prognosis. Functionally, lnc-PIK3R1 played tumor suppressor roles to inhibit the proliferation and mobility of HCC cells, and to impede the distant implantation of xenograft in mice. Mechanistic studies revealed that lnc-PIK3R1 interacted with miR-1286 and alleviated the repression on GSK3B by miR-1286. Notably, pharmacological inhibition of GSK3ß compromised the tumor suppression effect by lnc-PIK3R1, confirming their functional relevance. Moreover, we identified that oncogenic YY1 acts as a specific transcriptional repressor to downregulate the expression of lnc-PIK3R1 in HCC. In summary, this study highlights the tumor-suppressive effect of lnc-PIK3R1, and provides new insights into the regulation of GSK3ß expression in HCC, which would benefit the development of innovative intervention strategies for HCC.


Asunto(s)
Carcinoma Hepatocelular , Fosfatidilinositol 3-Quinasa Clase Ia , Regulación Neoplásica de la Expresión Génica , Glucógeno Sintasa Quinasa 3 beta , Neoplasias Hepáticas , MicroARNs , ARN Largo no Codificante , Factor de Transcripción YY1 , Humanos , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Glucógeno Sintasa Quinasa 3 beta/genética , MicroARNs/genética , MicroARNs/metabolismo , Animales , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Factor de Transcripción YY1/metabolismo , Factor de Transcripción YY1/genética , Ratones , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Fosfatidilinositol 3-Quinasa Clase Ia/genética , Progresión de la Enfermedad , Proliferación Celular/genética , Línea Celular Tumoral , Masculino , Ratones Desnudos , Femenino
7.
Cell Death Dis ; 15(4): 241, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38561375

RESUMEN

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.


Asunto(s)
Sarcoma , Animales , Humanos , Ratones , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Quinasas Asociadas a Fase-S/genética , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Sarcoma/genética , Sarcoma/patología , Ubiquitinación , Regulación hacia Arriba
8.
Medicine (Baltimore) ; 102(32): e34509, 2023 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-37565888

RESUMEN

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.


Asunto(s)
Estimulación Encefálica Profunda , Enfermedad de Parkinson , Núcleo Subtalámico , Humanos , Núcleo Subtalámico/fisiología , Enfermedad de Parkinson/complicaciones , Enfermedad de Parkinson/terapia , Calidad de Vida , Sueño/fisiología , Resultado del Tratamiento , Ensayos Clínicos Controlados Aleatorios como Asunto
9.
J Cancer ; 14(10): 1935-1945, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37476192

RESUMEN

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.

10.
J Exp Clin Cancer Res ; 42(1): 274, 2023 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-37864196

RESUMEN

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.


Asunto(s)
Neoplasias Endometriales , Proteína con Dominio Pirina 3 de la Familia NLR , Humanos , Ratones , Animales , Femenino , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Caspasa 1/genética , Caspasa 1/metabolismo , Caspasa 1/farmacología , Ratones Desnudos , Piroptosis , Cisplatino/farmacología , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/genética , Glucólisis , Microambiente Tumoral , Proteínas de Unión a Fosfato/genética , Proteínas de Unión a Fosfato/metabolismo , Proteínas de Unión a Fosfato/farmacología , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Receptor Relacionado con Estrógeno ERRalfa
11.
Am J Bot ; 98(9): e239-41, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21846791

RESUMEN

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.


Asunto(s)
Araceae/genética , Repeticiones de Microsatélite/genética , Alelos , Secuencia de Bases , Cartilla de ADN , Genes de Plantas
12.
Sheng Wu Gong Cheng Xue Bao ; 35(8): 1433-1440, 2019 Aug 25.
Artículo en Zh | MEDLINE | ID: mdl-31441614

RESUMEN

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.


Asunto(s)
Núcleo Arqueado del Hipotálamo , Obesidad , Homeostasis , Humanos , Leptina , Proopiomelanocortina
13.
JCI Insight ; 3(17)2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30185666

RESUMEN

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.


Asunto(s)
Aminopiridinas/farmacología , Bencimidazoles/farmacología , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/metabolismo , Dieta Alta en Grasa/efectos adversos , Obesidad/metabolismo , Obesidad/prevención & control , Animales , Modelos Animales de Enfermedad , Aprobación de Drogas , Metabolismo Energético , Hipotálamo/metabolismo , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos , Proteína de Retinoblastoma/metabolismo , Estados Unidos , United States Food and Drug Administration
14.
Cancer Cell ; 31(5): 669-684.e7, 2017 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-28486106

RESUMEN

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.


Asunto(s)
Carcinoma Hepatocelular/enzimología , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Neoplasias Hepáticas/enzimología , Ploidias , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Acetilación , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Proteínas de Ciclo Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Citosol/enzimología , Epistasis Genética , Femenino , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Células Hep G2 , Vía de Señalización Hippo , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Ratones , Ratones Transgénicos , Fenotipo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Embarazo , Proteínas Serina-Treonina Quinasas/genética , Estabilidad Proteica , Proteolisis , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Proteínas Quinasas Asociadas a Fase-S/genética , Transducción de Señal , Factores de Tiempo , Factores de Transcripción , Transfección , Proteínas Señalizadoras YAP , Factores de Transcripción p300-CBP/metabolismo
15.
Sci Rep ; 6: 19275, 2016 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-26763940

RESUMEN

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.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Hepatocitos/metabolismo , Proteínas Nucleares/genética , Prealbúmina/genética , Regiones Promotoras Genéticas , Proteínas Recombinantes de Fusión , Factores de Transcripción/genética , Transducción Genética , Animales , Proteínas de Ciclo Celular , Expresión Génica , Genes Reporteros , Vectores Genéticos/genética , Hepatocitos/efectos de los fármacos , Hepatocitos/trasplante , Lentivirus/genética , Regeneración Hepática , Modelos Animales , Transporte de Proteínas , Ratas , Tamoxifeno/farmacología
16.
PLoS One ; 9(3): e91298, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24614118

RESUMEN

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.


Asunto(s)
Adaptación Fisiológica , Agaricus/enzimología , Agaricus/fisiología , Temperatura , Transaminasas/metabolismo , Ácido 4-Aminobenzoico/farmacología , Adaptación Fisiológica/efectos de los fármacos , Agaricus/efectos de los fármacos , Catalasa/metabolismo , Quitinasas/metabolismo , Electroforesis en Gel Bidimensional , Proteínas Fúngicas/metabolismo , Respuesta al Choque Térmico/efectos de los fármacos , Peróxido de Hidrógeno/metabolismo , Modelos Biológicos , Estrés Oxidativo/efectos de los fármacos , Proteómica , Sulfanilamida , Sulfanilamidas/farmacología , Superóxido Dismutasa/metabolismo , Transgenes
17.
Nat Commun ; 5: 3463, 2014 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-24632684

RESUMEN

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.


Asunto(s)
Apoptosis , Regulación hacia Abajo , Factor de Transcripción E2F1/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Proteína de Retinoblastoma/deficiencia , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Animales , Proliferación Celular , Factor de Transcripción E2F1/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones Noqueados , Neoplasias/fisiopatología , Hipófisis/metabolismo , Proteína de Retinoblastoma/genética , Proteínas Quinasas Asociadas a Fase-S/genética
18.
Cancer Cell ; 24(5): 645-59, 2013 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-24229711

RESUMEN

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.


Asunto(s)
Carcinogénesis/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/fisiología , Neoplasia Intraepitelial Prostática/genética , Neoplasias de la Próstata/genética , Proteína de Retinoblastoma/metabolismo , Proteínas Quinasas Asociadas a Fase-S/genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis , Carcinogénesis/metabolismo , Línea Celular Tumoral , Senescencia Celular , Replicación del ADN , Eliminación de Gen , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/patología , Neoplasia Intraepitelial Prostática/patología , Neoplasias de la Próstata/patología , Proteína de Retinoblastoma/genética , Proteína p53 Supresora de Tumor/genética , Ubiquitina-Proteína Ligasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA