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












Base de datos
Intervalo de año de publicación
1.
Oncogene ; 43(25): 1885-1899, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38664501

RESUMEN

Receptor-interacting protein kinase 4 (RIPK4) is increasingly recognized as a pivotal player in ovarian cancer, promoting tumorigenesis and disease progression. Despite its significance, the posttranslational modifications dictating RIPK4 stability in ovarian cancer remain largely uncharted. In this study, we first established that RIPK4 levels are markedly higher in metastatic than in primary ovarian cancer tissues through single-cell sequencing. Subsequently, we identified UCHL3 as a key deubiquitinase that regulates RIPK4. We elucidate the mechanism that UCHL3 interacts with and deubiquitinates RIPK4 at the K469 site, removing the K48-linked ubiquitin chain and thus enhancing RIPK4 stabilization. Intriguingly, inhibition of UCHL3 activity using TCID leads to increased RIPK4 ubiquitination and degradation. Furthermore, we discovered that GSK3ß-mediated phosphorylation of RIPK4 at Ser420 enhances its interaction with UCHL3, facilitating further deubiquitination and stabilization. Functionally, RIPK4 was found to drive the proliferation and metastasis of ovarian cancer in a UCHL3-dependent manner both in vitro and in vivo. Importantly, positive correlations between RIPK4 and UCHL3 protein expression levels were observed, with both serving as indicators of poor prognosis in ovarian cancer patients. Overall, this study uncovers a novel pathway wherein GSK3ß-induced phosphorylation of RIPK4 strengthens its interaction with UCHL3, leading to increased deubiquitination and stabilization of RIPK4, thereby promoting ovarian cancer metastasis. These findings offer new insights into the molecular underpinnings of ovarian cancer and highlight potential therapeutic targets for enhancing antitumor efficacy.


Asunto(s)
Glucógeno Sintasa Quinasa 3 beta , Neoplasias Ováricas , Ubiquitina Tiolesterasa , Ubiquitinación , Neoplasias Ováricas/patología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/genética , Femenino , Humanos , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Ubiquitina Tiolesterasa/genética , Animales , Ratones , Línea Celular Tumoral , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Metástasis de la Neoplasia , Homeostasis , Fosforilación , Proliferación Celular/genética , Ratones Desnudos
2.
Nat Genet ; 56(4): 637-651, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38565644

RESUMEN

Endometrial carcinoma remains a public health concern with a growing incidence, particularly in younger women. Preserving fertility is a crucial consideration in the management of early-onset endometrioid endometrial carcinoma (EEEC), particularly in patients under 40 who maintain both reproductive desire and capacity. To illuminate the molecular characteristics of EEEC, we undertook a large-scale multi-omics study of 215 patients with endometrial carcinoma, including 81 with EEEC. We reveal an unexpected association between exposome-related mutational signature and EEEC, characterized by specific CTNNB1 and SIGLEC10 hotspot mutations and disruption of downstream pathways. Interestingly, SIGLEC10Q144K mutation in EEECs resulted in aberrant SIGLEC-10 protein expression and promoted progestin resistance by interacting with estrogen receptor alpha. We also identified potential protein biomarkers for progestin response in fertility-sparing treatment for EEEC. Collectively, our study establishes a proteogenomic resource of EEECs, uncovering the interactions between exposome and genomic susceptibilities that contribute to the development of primary prevention and early detection strategies for EEECs.


Asunto(s)
Carcinoma Endometrioide , Hiperplasia Endometrial , Neoplasias Endometriales , Preservación de la Fertilidad , Proteogenómica , Humanos , Femenino , Progestinas/uso terapéutico , Antineoplásicos Hormonales , Hiperplasia Endometrial/tratamiento farmacológico , Preservación de la Fertilidad/métodos , Estudios Retrospectivos , Carcinoma Endometrioide/tratamiento farmacológico , Carcinoma Endometrioide/genética , Carcinoma Endometrioide/patología , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/genética , Neoplasias Endometriales/patología
3.
Acta Obstet Gynecol Scand ; 103(5): 884-896, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38217337

RESUMEN

INTRODUCTION: Previous studies have demonstrated that abnormal body mass index (BMI) is associated with adverse pregnancy outcomes in frozen-thawed embryo transfer cycles. However, the relationship between BMI and pregnancy and perinatal outcomes in patients with polycystic ovary syndrome (PCOS) remains unclear. Furthermore, whether a diagnosis of PCOS could result in adverse pregnancy and perinatal outcomes in women with different BMIs remains unknown. MATERIAL AND METHODS: A historical cohort study included 1667 women with PCOS and 12 256 women without PCOS after a freeze-all policy between January 2016 and December 2020. The outcomes encompassed both pregnancy and perinatal outcomes. Multivariate logistic regression analysis and restricted cubic spline models were performed to eliminate confounding factors when investigating the relationship between BMI and different outcomes. RESULTS: After controlling for covariates, pregnancy outcomes were comparable between underweight women with PCOS and normal weight women with PCOS. However, overweight patients had a lower clinical pregnancy rate and an overall live birth rate. Furthermore, patients with obesity had a lower rate of multiple pregnancies but a higher rate of biochemical pregnancy than in the normal BMI group. Additionally, the restricted cubic spline models showed that as maternal BMI increased to 32 kg/m2, the clinical pregnancy rate and live birth rate after blastocyst transfer decreased, but the risks of preterm birth, gestational diabetes mellitus, macrosomia, large-for-gestational age (LGA) and very LGA increased in patients with PCOS after a freeze-all strategy. Moreover, a diagnosis of PCOS resulted in a higher clinical pregnancy rate and live birth rate and a higher risk of small-for-gestational age in the normal weight group. However, women with PCOS in the overweight group exhibited higher risks of very preterm birth and gestational diabetes mellitus compared with women without PCOS. CONCLUSIONS: This study showed that a higher BMI had a detrimental impact on the pregnancy and perinatal outcomes of PCOS patients undergoing a freeze-all strategy. However, it was only statistically significant in the overweight group. A diagnosis of PCOS had a higher clinical pregnancy rate and live birth rate in normal weight women but higher risks of perinatal complications in normal weight and overweight women.


Asunto(s)
Diabetes Gestacional , Síndrome del Ovario Poliquístico , Nacimiento Prematuro , Embarazo , Humanos , Recién Nacido , Femenino , Síndrome del Ovario Poliquístico/complicaciones , Índice de Masa Corporal , Sobrepeso/complicaciones , Nacimiento Prematuro/epidemiología , Nacimiento Prematuro/etiología , Estudios de Cohortes , Resultado del Embarazo , Estudios Retrospectivos
4.
Lancet Oncol ; 25(1): 76-85, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38048802

RESUMEN

BACKGROUND: Locally advanced cervical cancer constitutes around 37% of cervical cancer cases globally and has a poor prognosis due to limited therapeutic options. Immune checkpoint inhibitors in the neoadjuvant setting could address these challenges. We aimed to investigate the efficacy and safety of neoadjuvant chemo-immunotherapy for locally advanced cervical cancer. METHODS: In this single-arm, phase 2 trial, which was done across eight tertiary hospitals in China, we enrolled patients aged 18-70 years with untreated cervical cancer (IB3, IIA2, or IIB/IIIC1r with a tumour diameter ≥4 cm [International Federation of Gynecology and Obstetrics, 2018]) and an Eastern Cooperative Oncology Group performance status of 0 or 1. Eligible patients underwent one cycle of priming doublet chemotherapy (75-80 mg/m2 cisplatin, intravenously, plus 260 mg/m2 nab-paclitaxel, intravenously), followed by two cycles of a combination of chemotherapy (cisplatin plus nab-paclitaxel) on day 1 with camrelizumab (200 mg, intravenously) on day 2, with a 3-week interval between treatment cycles. Patients with stable disease or progressive disease received concurrent chemoradiotherapy, and patients with a complete response or partial response proceeded to radical surgery. The primary endpoint was the objective response rate, by independent central reviewer according to Response Evaluation Criteria in Solid Tumours, version 1.1. Activity and safety were analysed in patients who received at least one dose of camrelizumab. This study is registered with ClinicalTrials.gov, NCT04516616, and is ongoing. FINDINGS: Between Dec 1, 2020, and Feb 10, 2023, 85 patients were enrolled and all received at least one dose of camrelizumab. Median age was 51 years (IQR 46-57) and no data on race or ethnicity were collected. At data cutoff (April 30, 2023), median follow-up was 11·0 months (IQR 6·0-14·5). An objective response was noted in 83 (98% [95% CI 92-100]) patients, including 16 (19%) patients who had a complete response and 67 (79%) who had a partial response. The most common grade 3-4 treatment-related adverse events during neoadjuvant chemo-immunotherapy were lymphopenia (21 [25%] of 85), neutropenia (ten [12%]), and leukopenia (seven [8%]). No serious adverse events or treatment-related deaths occurred. INTERPRETATION: Neoadjuvant chemo-immunotherapy showed promising antitumour activity and a manageable adverse event profile in patients with locally advanced cervical cancer. The combination of neoadjuvant chemo-immunotherapy with radical surgery holds potential as a novel therapeutic approach for locally advanced cervical cancer. FUNDING: National Key Technology Research and Development Program of China and the National Clinical Research Center of Obstetrics and Gynecology.


Asunto(s)
Trombocitopenia , Neoplasias del Cuello Uterino , Femenino , Humanos , Persona de Mediana Edad , Cisplatino/efectos adversos , Terapia Neoadyuvante/efectos adversos , Neoplasias del Cuello Uterino/tratamiento farmacológico , Anticuerpos Monoclonales Humanizados/efectos adversos , Trombocitopenia/inducido químicamente , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos
5.
Nat Genet ; 55(12): 2175-2188, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37985817

RESUMEN

Cervical squamous cell carcinoma (CSCC) exhibits a limited response to immune-checkpoint blockade. Here we conducted a multiomic analysis encompassing single-cell RNA sequencing, spatial transcriptomics and spatial proteomics, combined with genetic and pharmacological perturbations to systematically develop a high-resolution and spatially resolved map of intratumoral expression heterogeneity in CSCC. Three tumor states (epithelial-cytokeratin, epithelial-immune (Epi-Imm) and epithelial senescence), recapitulating different stages of squamous differentiation, showed distinct tumor immune microenvironments. Bidirectional interactions between epithelial-cytokeratin malignant cells and immunosuppressive cancer-associated fibroblasts form an immune exclusionary microenvironment through transforming growth factor ß pathway signaling mediated by FABP5. In Epi-Imm tumors, malignant cells interact with natural killer and T cells through interferon signaling. Preliminary analysis of samples from a cervical cancer clinical trial ( NCT04516616 ) demonstrated neoadjuvant chemotherapy induces a state transition to Epi-Imm, which correlates with pathological complete remission following treatment with immune-checkpoint blockade. These findings deepen the understanding of cellular state diversity in CSCC.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias del Cuello Uterino , Femenino , Humanos , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/genética , Neoplasias del Cuello Uterino/genética , Inhibidores de Puntos de Control Inmunológico , Relevancia Clínica , Ecosistema , Multiómica , Queratinas/metabolismo , Queratinas/uso terapéutico , Microambiente Tumoral/genética , Proteínas de Unión a Ácidos Grasos/uso terapéutico
6.
J Immunother Cancer ; 11(4)2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37072347

RESUMEN

BACKGROUND: Mismatch repair deficiency (dMMR) is a well-recognized biomarker for response to immune checkpoint blockade (ICB). Strategies to convert MMR-proficient (pMMR) to dMMR phenotype with the goal of sensitizing tumors to ICB are highly sought. The combination of bromodomain containing 4 (BRD4) inhibition and ICB provides a promising antitumor effect. However, the mechanisms underlying remain unknown. Here, we identify that BRD4 inhibition induces a persistent dMMR phenotype in cancers. METHODS: We confirmed the correlation between BRD4 and mismatch repair (MMR) by the bioinformatic analysis on The Cancer Genome Atlas and Clinical Proteomic Tumor Analysis Consortium data, and the statistical analysis on immunohistochemistry (IHC) scores of ovarian cancer specimens. The MMR genes (MLH1,MSH2,MSH6,PMS2) were measured by quantitative reverse transcription PCR, western blot, and IHC. The MMR status was confirmed by whole exome sequencing, RNA sequencing, MMR assay and hypoxanthine-guanine phosphoribosyl transferase gene mutation assay. The BRD4i AZD5153 resistant models were induced both in vitro and in vivo. The transcriptional effects of BRD4 on MMR genes were investigated by chromatin immunoprecipitation among cell lines and data from the Cistrome Data Browser. The therapeutic response to ICB was testified in vivo. The tumor immune microenvironment markers, such as CD4, CD8, TIM-3, FOXP3, were measured by flow cytometry. RESULTS: We identified the positive correlation between BRD4 and MMR genes in transcriptional and translational aspects. Also, the inhibition of BRD4 transcriptionally reduced MMR genes expression, resulting in dMMR status and elevated mutation loads. Furthermore, prolonged exposure to AZD5153 promoted a persistent dMMR signature both in vitro and in vivo, enhancing tumor immunogenicity, and increased sensitivity to α-programmed death ligand-1 therapy despite the acquired drug resistance. CONCLUSIONS: We demonstrated that BRD4 inhibition suppressed expression of genes critical to MMR, dampened MMR, and increased dMMR mutation signatures both in vitro and in vivo, sensitizing pMMR tumors to ICB. Importantly, even in BRD4 inhibitors (BRD4i)-resistant tumor models, the effects of BRD4i on MMR function were maintained rendering tumors sensitive to ICB. Together, these data identified a strategy to induce dMMR in pMMR tumors and further, indicated that BRD4i sensitive and resistant tumors could benefit from immunotherapy.


Asunto(s)
Neoplasias Colorrectales , Proteínas Nucleares , Humanos , Proteínas Nucleares/genética , Inhibidores de Puntos de Control Inmunológico , Reparación de la Incompatibilidad de ADN/genética , Factores de Transcripción/genética , Proteómica , Neoplasias Colorrectales/patología , Mutación , Microambiente Tumoral , Proteínas de Ciclo Celular/genética
7.
J Ovarian Res ; 16(1): 77, 2023 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-37061732

RESUMEN

BACKGROUND: Previous studies have discussed the pregnancy outcomes of diminished ovarian reserve (DOR) patients. However, data on embryonic development potential, neonatal outcomes, and maternal complications of DOR patients still remained unknown. This is the first study to investigate the risk of DOR on pregnancy and perinatal outcomes among women < 38 years. METHODS: Retrospective cohort study was conducted. Patients (< 38 years of age) undergoing their first oocyte retrieval cycle were included. Patients were divided into DOR group and non-DOR group. Pregnancy outcomes of fresh cycle and cumulative live birth rate and perinatal outcomes after one oocyte retrieved cycle were compared between DOR and non-DOR group. RESULT(S): From January 2016 to September 2020, there were 8,179 patients involved: 443 patients in the DOR group and 7,736 patients in the non-DOR group. The incidences of live birth and clinical pregnancy did not differ significantly between patients with or without DOR after fresh cycle transfer, but the cumulative live birth rate was significantly lower in DOR group. Among women who had singleton live births, after binary logistic regression, the rates of maternal complications and neonatal outcomes were comparable in the two groups. CONCLUSION(S): DOR patients (< 38 years of age) showed similar pregnancy outcomes in the first fresh embryo transfer cycle but a lower chance of live birth after a whole oocyte retrieval cycle to non-DOR patients and DOR is not associated with adverse perinatal outcomes.


Asunto(s)
Enfermedades del Ovario , Reserva Ovárica , Embarazo , Humanos , Femenino , Estudios Retrospectivos , Resultado del Embarazo , Transferencia de Embrión , Tasa de Natalidad , Nacimiento Vivo , Fertilización In Vitro , Índice de Embarazo
8.
Cell Genom ; 3(1): 100211, 2023 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-36777180

RESUMEN

Cervical cancer (CC) that is caused by high-risk human papillomavirus (HPV) remains a significant public health problem worldwide. HPV integration sites can be silent or actively transcribed, leading to the production of viral-host fusion transcripts. Herein, we demonstrate that only productive HPV integration sites were nonrandomly distributed across both viral and host genomes, suggesting that productive integration sites are under selection and likely to contribute to CC pathophysiology. Furthermore, using large-scale, multi-omics (clinical, genomic, transcriptional, proteomic, phosphoproteomic, and single-cell) data, we demonstrate that tumors with productive HPV integration are associated with higher E6/E7 proteins and enhanced tumor aggressiveness and immunoevasion. Importantly, productive HPV integration increases from carcinoma in situ to advanced disease. This study improves our understanding of the functional consequences of HPV fusion transcripts on the biology and pathophysiology of HPV-driven CCs, suggesting that productive HPV integration should be evaluated as an indicator of high risk for progression to aggressive cancers.

9.
Nat Commun ; 13(1): 5462, 2022 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-36115848

RESUMEN

Generalization is a fundamental cognitive ability of organisms to deal with the uncertainty in real-world situations. Excessive fear generalization and impaired reward generalization are closely related to many psychiatric disorders. However, the neural circuit mechanism for reward generalization and its role in anxiety-like behaviours remain elusive. Here, we found a robust activation of calbindin 1-neurons (Calb 1) in the posterior basolateral amygdala (pBLA), simultaneous with reward generalization to an ambiguous cue after reward conditioning in mice. We identify the infralimbic medial prefrontal cortex (IL) to the pBLACalb1 (Calb 1 neurons in the pBLA) pathway as being involved in reward generalization for the ambiguity. Activating IL-pBLA inputs strengthens reward generalization and reduces chronic unpredictable mild stress-induced anxiety- and depression-like behaviours in a manner dependent on pBLACalb1 neuron activation. These findings suggest that the IL-pBLACalb1 circuit could be a target to promote stress resilience via reward generalization and consequently ameliorate anxiety- and depression-like behaviours.


Asunto(s)
Ansiedad , Complejo Nuclear Basolateral , Calbindina 1 , Depresión , Neuronas , Corteza Prefrontal , Animales , Ansiedad/genética , Ansiedad/metabolismo , Complejo Nuclear Basolateral/metabolismo , Calbindina 1/genética , Calbindina 1/metabolismo , Depresión/genética , Depresión/metabolismo , Ratones , Neuronas/metabolismo , Neuronas/fisiología , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiología
10.
Adv Sci (Weinh) ; 9(21): e2105469, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35619328

RESUMEN

Targeting the G2/M checkpoint mediator WEE1 has been explored as a novel treatment strategy in ovarian cancer, but mechanisms underlying its efficacy and resistance remains to be understood. Here, it is demonstrated that the WEE1 inhibitor AZD1775 induces endoplasmic reticulum stress and activates the protein kinase RNA-like ER kinase (PERK) and inositol-required enzyme 1α (IRE1α) branches of the unfolded protein response (UPR) in TP53 mutant (mtTP53) ovarian cancer models. This is facilitated through NF-κB mediated senescence-associated secretory phenotype. Upon AZD1775 treatment, activated PERK promotes apoptotic signaling via C/EBP-homologous protein (CHOP), while IRE1α-induced splicing of XBP1 (XBP1s) maintains cell survival by repressing apoptosis. This leads to an encouraging synergistic antitumor effect of combining AZD1775 and an IRE1α inhibitor MKC8866 in multiple cell lines and preclinical models of ovarian cancers. Taken together, the data reveal an important dual role of the UPR signaling network in mtTP53 ovarian cancer models in response to AZD1775 and suggest that inhibition of the IRE1α-XBP1s pathway may enhance the efficacy of AZD1775 in the clinics.


Asunto(s)
Endorribonucleasas , Neoplasias Ováricas , Proteínas Serina-Treonina Quinasas , Benzopiranos , Endorribonucleasas/antagonistas & inhibidores , Endorribonucleasas/metabolismo , Femenino , Humanos , Inositol/metabolismo , Morfolinas , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Pirazoles/farmacología , Pirimidinonas/farmacología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Respuesta de Proteína Desplegada/genética , Respuesta de Proteína Desplegada/fisiología
11.
Mol Plant ; 15(2): 258-275, 2022 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-34715392

RESUMEN

As one of the most important crops in the world, rice (Oryza sativa) is a model plant for metabolome research. Although many studies have focused on the analysis of specific tissues, the changes in metabolite abundance across the entire life cycle have not yet been determined. In this study, combining both targeted and nontargeted metabolite profiling methods, a total of 825 annotated metabolites were quantified in rice samples from different tissues covering the entire life cycle. The contents of metabolites in different tissues of rice were significantly different, with various metabolites accumulating in the plumule and radicle during seed germination. Combining these data with transcriptome data obtained from the same time period, we constructed the Rice Metabolic Regulation Network. The metabolites and co-expressed genes were further divided into 12 clusters according to their accumulation patterns, with members within each cluster displaying a uniform and clear pattern of abundance across development. Using this dataset, we established a comprehensive metabolic profile of the rice life cycle and used two independent strategies to identify novel transcription factors-namely the use of known regulatory genes as bait to screen for new networks underlying lignin metabolism and the unbiased identification of new glycerophospholipid metabolism regulators on the basis of tissue specificity. This study thus demonstrates how guilt-by-association analysis of metabolome and transcriptome data spanning the entire life cycle in cereal crops provides novel resources and tools to aid in understanding the mechanisms underlying important agronomic traits.


Asunto(s)
Oryza , Animales , Perfilación de la Expresión Génica , Regulación de la Expresión Génica de las Plantas/genética , Estadios del Ciclo de Vida , Metaboloma/genética , Oryza/metabolismo , Transcriptoma/genética
12.
Cancer Lett ; 528: 31-44, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34942306

RESUMEN

The CDK4/6 inhibitor, palbociclib has recently entered clinic-trial stage for breast cancer treatment. However, translating its efficacy to other solid tumors has been challenging, especially for aggressive solid tumors. We found that the effect of palbociclib as a single agent was limited due to primary and acquired resistance in multiple ovarian cancer (OC) models. Among these, patient-derived organoid and xenograft models are two most representative models of drug responsiveness in patients with OC. In preclinical models, this study demonstrated that activated MAPK/PI3K-AKT pathway and cell cycle-related proteins induced the resistance to palbociclib, which was overcome by the addition of the bromodomain protein 4 (BRD4) inhibitor AZD5153. Moreover, this study revealed that AZD5153 and palbociclib had a synergistic lethal effect on inducing the cell cycle arrest and increasing apoptosis, even in RB-deficient cell lines. Based on these results, it is anticipated that this class of drugs, including AZD5153, which inhibit the cell cycle-related protein and MAPK/PI3K-AKT pathway, will exhibit synergistic effects with palbociclib in OC.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Proteínas de Ciclo Celular/efectos de los fármacos , Compuestos Heterocíclicos con 2 Anillos/uso terapéutico , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Fosfatidilinositol 3-Quinasas/metabolismo , Piperazinas/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirazoles/uso terapéutico , Piridazinas/uso terapéutico , Piridinas/uso terapéutico , Animales , Antineoplásicos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Línea Celular Tumoral , Femenino , Compuestos Heterocíclicos con 2 Anillos/farmacología , Humanos , Ratones , Piperazinas/farmacología , Pirazoles/farmacología , Piridazinas/farmacología , Piridinas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Front Cell Dev Biol ; 9: 752753, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34746146

RESUMEN

Introduction: Type 2 diabetes mellitus (T2DM) is an independent risk factor of Alzheimer's disease (AD), and populations with mild cognitive impairment (MCI) have high incidence to suffer from AD. Therefore, discerning who may be more vulnerable to MCI, among the increasing T2DM populations, is important for early intervention and eventually decreasing the prevalence rate of AD. This study was to explore whether the change of plasma ß-amyloid (Aß) could be a biomarker to distinguish MCI (T2DM-MCI) from non-MCI (T2DM-nMCI) in T2DM patients. Methods: Eight hundred fifty-two T2DM patients collected from five medical centers were assigned randomly to training and validation cohorts. Plasma Aß, platelet glycogen synthase kinase-3ß (GSK-3ß), apolipoprotein E (ApoE) genotypes, and olfactory and cognitive functions were measured by ELISA, dot blot, RT-PCR, Connecticut Chemosensory Clinical Research Center (CCCRC) olfactory test based on the diluted butanol, and Minimum Mental State Examination (MMSE) test, respectively, and multivariate logistic regression analyses were applied. Results: Elevation of plasma Aß1-42/Aß1-40 is an independent risk factor of MCI in T2DM patients. Although using Aß1-42/Aß1-40 alone only reached an AUC of 0.631 for MCI diagnosis, addition of the elevated Aß1-42/Aß1-40 to our previous model (i.e., activated platelet GSK-3ß, ApoE ε4 genotype, olfactory decline, and aging) significantly increased the discriminating efficiency of T2DM-MCI from T2DM-nMCI, with an AUC of 0.846 (95% CI: 0.794-0.897) to 0.869 (95% CI: 0.822-0.916) in the training cohort and an AUC of 0.848 (95% CI: 0.815-0.882) to 0.867 (95% CI: 0.835-0.899) in the validation cohort, respectively. Conclusion: A combination of the elevated plasma Aß1-42/Aß1-40 with activated platelet GSK-3ß, ApoE ε4 genotype, olfactory decline, and aging could efficiently diagnose MCI in T2DM patients. Further longitudinal studies may consummate the model for early prediction of AD.

14.
Cancer Sci ; 112(11): 4444-4456, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34382294

RESUMEN

WEE1 plays an important role in the regulation of cell cycle G2/M checkpoints and DNA damage response (DDR). Inhibition of WEE1 can increase the instability of the genome and have anti-tumor effects in some solid tumors. However, it has certain limitations for multiple cancer cells from different lineages. Therefore, we consider the use of synthetic lethal interactions to enhance the therapeutic effect. Our experiments proved that WEE1 inhibitor (WEE1i) can activate the ataxia telangiectasia and RAD3-related (ATR) pathway and that blockage of ATR dramatically sensitized the WEE1i-induced cell death. The tumor-selective synthetic lethality between bioavailable WEE1 and ATR inhibitors led to tumor remission in vivo. Mechanistically, the combination promoted the accumulation of cytosolic double-strand DNA, which subsequently activated the stimulator of the interferon gene (STING) pathway and induced the production of type I interferon and CD8+ T cells, thereby inducing anti-tumor immunity. Furthermore, our study found that immune checkpoint programmed death-ligand 1 is upregulated by the combination therapy, and blocking PD-L1 further enhances the effect of the combination therapy. In summary, as an immunomodulator, the combination of WEE1i with ATR inhibitor (ATRi) and immune checkpoint blockers provides a potential new approach for cancer treatment.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Antígeno B7-H1/antagonistas & inhibidores , Proteínas de Ciclo Celular/antagonistas & inhibidores , Neoplasias Colorrectales/tratamiento farmacológico , Terapia Molecular Dirigida/métodos , Neoplasias Ováricas/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos , Muerte Celular , Línea Celular Tumoral , ADN/metabolismo , Daño del ADN , ADN de Neoplasias/biosíntesis , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Femenino , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Inestabilidad Genómica , Humanos , Inmunidad , Inmunoterapia/métodos , Indoles/uso terapéutico , Interferón Tipo I/biosíntesis , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Morfolinas/uso terapéutico , Pirazoles/uso terapéutico , Pirimidinas/uso terapéutico , Pirimidinonas/uso terapéutico , Sulfonamidas/uso terapéutico , Microambiente Tumoral/inmunología , Ensayo de Tumor de Célula Madre , Regulación hacia Arriba
15.
Clin Transl Med ; 11(6): e428, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34185417

RESUMEN

Tau accumulation and cholinergic impairment are characteristic pathologies in Alzheimer's disease (AD). However, the causal role of tau accumulation in cholinergic lesion is elusive. Here, we observed an aberrant tau accumulation in the medial septum (MS) of 3xTg and 5xFAD mice, especially in their cholinergic neurons. Overexpressing hTau in mouse MS (MShTau ) for 6 months but not 3 months induced spatial memory impairment without changing object recognition and anxiety-like behavior, indicating a specific and time-dependent effect of MS-hTau accumulation on spatial cognitive functions. With increasing hTau accumulation, the MShTau mice showed a time-dependent cholinergic neuron loss with reduced cholinergic projections to the hippocampus. Intraperitoneal administration of donepezil, a cholinesterase inhibitor, for 1 month ameliorated the MS-hTau-induced spatial memory deficits with preservation of MS-hippocampal cholinergic pathway and removal of tau load; and the beneficial effects of donepezil was more prominent at low dose. Proteomics revealed that MS-hTau accumulation deregulated multiple signaling pathways with numerous differentially expressed proteins (DEPs). Among them, the vacuolar protein sorting-associated protein 37D (VP37D), an autophagy-related protein, was significantly reduced in MShTau mice; the reduction of VP37D was restored by donepezil, and the effect was more significant at low dose than high dose. These novel evidences reveal a causal role of tau accumulation in linking MS cholinergic lesion to hippocampus-dependent spatial cognitive damages as seen in the AD patients, and the new tau-removal and autophagy-promoting effects of donepezil may extend its application beyond simple symptom amelioration to potential disease modification.


Asunto(s)
Colinérgicos/metabolismo , Hipocampo/patología , Trastornos de la Memoria/patología , Proteoma/metabolismo , Núcleos Septales/patología , Memoria Espacial/fisiología , Proteínas tau/metabolismo , Animales , Hipocampo/metabolismo , Masculino , Trastornos de la Memoria/etiología , Trastornos de la Memoria/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteoma/análisis , Núcleos Septales/metabolismo , Proteínas tau/genética
16.
Front Immunol ; 12: 799171, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35095879

RESUMEN

Background: Ovarian cancer (OC) is one of the most lethal gynecologic cancers. Growing evidence has proven that CDK4/6 plays a key role in tumor immunity and the prognosis of many cancers. However, the expression and function of CDK4/6 in OC remain unclear. Therefore, we aimed to explore the influence of CDK4/6 in OC, especially on immunity. Methods: We analyzed CDK4/6 expression and prognosis using The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO) and Genotype Tissue Expression (GTEx) data. Subsequently, we used the cytoHubba plug-in of Cytoscape software and starBase to identify the noncoding RNAs (ncRNAs) regulating CDK4/6. Finally, we verified the effect of CDK4/6 on immunity in OC cell lines and animal models. Results: CDK4/6 expression was higher in OC tissues than in normal ovarian tissues, and the high expression levels of CDK4/6 contributed to the immunosuppressive state of OC and were thus related to the poor prognosis of OC patients. This was also in general agreement with the results of OC cell line and animal experiments. Mechanistically, the CDK4/6 inhibitor palbociclib increased the secretion of interferon (IFN)-γ and the interferon-stimulated gene (ISG) response, thereby upregulating the expression of antigen-presenting molecules; this effect was partly dependent on the STING pathway and thus activated immunity in OC. Additionally, according to public data, the LRRC75A-AS1-hsa-miR-330-5p axis could inhibit the immune response of OC patients by upregulating CDK4/6, leading to a poor prognosis. Conclusion: CDK4/6 affects the immune microenvironment of OC and correlates with the prognosis of OC patients.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/inmunología , Quinasa 6 Dependiente de la Ciclina/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Neoplasias Ováricas/inmunología , Transcriptoma/inmunología , Microambiente Tumoral/inmunología , Animales , Línea Celular Tumoral , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/genética , Quinasa 6 Dependiente de la Ciclina/metabolismo , Femenino , Ontología de Genes , Humanos , Estimación de Kaplan-Meier , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Ratones Endogámicos C57BL , MicroARNs/genética , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/inmunología , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Piperazinas/farmacología , Pronóstico , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Transducción de Señal/genética , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética
17.
World J Gastroenterol ; 24(20): 2173-2180, 2018 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-29853735

RESUMEN

AIM: To assess the effects of hepatitis E virus (HEV) on the production of type I interferons (IFNs) and determine the underlying mechanisms. METHODS: We measured the production of interferon (IFN)-alpha and -beta (-α/ß) in genotype 3 HEV-infected C3A cells at different time points (0, 8, 12, 24, 48, 72 and 120 h) by enzyme-linked immunosorbent assay (ELISA). The expression levels of IFN-stimulated gene (ISG)15 in HEV-infected C3A cells at different time points were tested by western blotting. The plasmid-expressing open reading frame 3 (ORF3) or control plasmids (green fluorescent protein-expressing) were transfected into C3A cells, and the levels of IFN-α/ß and ISG15 were evaluated, respectively. Furthermore, the plasmid-expressing ISG15 or small interfering RNA-inhibiting ISG15 was transfected into infected C3A cells. Then, the production of IFN-α/ß was also measured by ELISA. RESULTS: We showed that genotype 3 HEV could enhance the production of IFN-α/ß and induce elevation of ISG15 in C3A cells. HEV ORF3 protein could enhance the production of IFN-α/ß and the expression of ISG15. Additionally, ISG15 silencing enhanced the production of IFN-α/ß. Overexpression of ISG15 resulted in the reduction of IFN-α/ß. CONCLUSION: HEV may promote production of IFN-α/ß and expression of ISG15 via ORF3 in the early stages, and increased ISG15 subsequently inhibited the production of IFN-α/ß.


Asunto(s)
Citocinas/metabolismo , Virus de la Hepatitis E/inmunología , Hepatitis E/inmunología , Hepatocitos/metabolismo , Interferón Tipo I/metabolismo , Ubiquitinas/metabolismo , Línea Celular Tumoral , Citocinas/genética , Técnicas de Silenciamiento del Gen , Hepatitis E/virología , Virus de la Hepatitis E/patogenicidad , Hepatocitos/inmunología , Hepatocitos/virología , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata , Interferón Tipo I/inmunología , ARN Interferente Pequeño/metabolismo , Transfección , Ubiquitinas/genética , Proteínas Virales/inmunología
18.
Sci Rep ; 6: 27597, 2016 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-27270888

RESUMEN

Hepatitis E virus (HEV) genotype 1 infection is common and can emerge as outbreaks in developing areas, thus posing a threat to public health. However, due to the absence of feasible animal models, the mechanism of HE pathogenesis remains obscure. The HEV pathogenic mechanism has been suggested to be mediated by the immune system and not by direct viral duplication. We firstly discovered that the open reading frame 3 (ORF3) protein of genotype 1 HEV downregulates TLR3-mediated NF-κB signaling in Human A549 Lung Epithelial Cells (A549 cells) which were exposed to different TLR agonists associated with viral nucleic acids. Additionally, we identified the P2 domain of ORF3 as being responsible for this inhibition. Intriguingly, tumor necrosis factor receptor 1-associated death domain protein (TRADD) expression and receptor-interacting protein kinase 1 (RIP1) K63-ubiquitination were reduced in the presence of both ORF3 and Poly(I:C). Furthermore, we found that Lys377 of RIP1 acts as the functional ubiquitination site for ORF3-associated inhibition. Overall, we found that ORF3 protein downregulates TLR3-mediated NF-κB signaling via TRADD and RIP1. Our findings provide a new perspective on the cellular response in HEV infection and expand our understanding of the molecular mechanisms of HEV pathogenesis in innate immunity.


Asunto(s)
Virus de la Hepatitis E/genética , Hepatitis E/genética , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Unión al ARN/genética , Proteína de Dominio de Muerte Asociada a Receptor de TNF/genética , Proteínas Virales/genética , Células A549 , Regulación de la Expresión Génica/genética , Genotipo , Hepatitis E/virología , Virus de la Hepatitis E/patogenicidad , Humanos , Inmunidad Innata/genética , FN-kappa B/genética , Receptor Toll-Like 3/genética , Ubiquitinación/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...