Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 302
Filtrar
1.
Nat Immunol ; 24(2): 225-238, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36624165

RESUMEN

Skin is exposed to various environmental assaults and undergoes morphological changes immediately after birth. Proper localization and function of immune cells in the skin is crucial for protection and establishment of skin tissue homeostasis. Here we report the discovery of a developmentally programmed process that directs preferential localization of invariant natural killer T (iNKT) cells to the skin for early local homeostatic regulation. We show that iNKT cells are programmed predominantly with a CCR10+ skin-homing phenotype during thymic development in infant and young mice. Early skin localization of iNKT cells is critical for proper commensal bacterial colonization and tissue development. Mechanistically, skin iNKT cells provide a local source of transferrin that regulates iron metabolism in hair follicle progenitor cells and helps hair follicle development. These findings provide molecular insights into the establishment and physiological functions of iNKT cells in the skin during early life.


Asunto(s)
Células T Asesinas Naturales , Ratones , Animales , Piel , Homeostasis , Ratones Endogámicos C57BL , Ratones Noqueados
2.
Cell ; 152(5): 1037-50, 2013 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-23452852

RESUMEN

Although somatic cell reprogramming to generate inducible pluripotent stem cells (iPSCs) is associated with profound epigenetic changes, the roles and mechanisms of epigenetic factors in this process remain poorly understood. Here, we identify Jmjd3 as a potent negative regulator of reprogramming. Jmjd3-deficient MEFs produced significantly more iPSC colonies than did wild-type cells, whereas ectopic expression of Jmjd3 markedly inhibited reprogramming. We show that the inhibitory effects of Jmjd3 are produced through both histone demethylase-dependent and -independent pathways. The latter pathway involves Jmjd3 targeting of PHF20 for ubiquitination and degradation via recruitment of an E3 ligase, Trim26. Importantly, PHF20-deficient MEFs could not be converted to fully reprogrammed iPSCs, even with knockdown of Jmjd3, Ink4a, or p21, indicating that PHF20 is required for reprogramming. Our findings demonstrate, to the best of our knowledge, a previously unrecognized role of Jmjd3 in cellular reprogramming and provide molecular insight into the mechanisms by which the Jmjd3-PHF20 axis controls this process.


Asunto(s)
Reprogramación Celular , Proteínas de Homeodominio/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Animales , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Proteínas de Unión al ADN , Embrión de Mamíferos/citología , Fibroblastos/metabolismo , Cinética , Ratones , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Factores de Transcripción , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Regulación hacia Arriba
3.
PLoS Genet ; 19(1): e1010595, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36656901

RESUMEN

Defective ribosome biogenesis (RiBi) underlies a group of clinically diverse human diseases collectively known as the ribosomopathies, core manifestations of which include cytopenias and developmental abnormalities that are believed to stem primarily from an inability to synthesize adequate numbers of ribosomes and concomitant activation of p53. The importance of a correctly functioning RiBi machinery for maintaining tissue homeostasis is illustrated by the observation that, despite having a paucity of certain cell types in early life, ribosomopathy patients have an increased risk for developing cancer later in life. This suggests that hypoproliferative states trigger adaptive responses that can, over time, become maladaptive and inadvertently drive unchecked hyperproliferation and predispose to cancer. Here we describe an experimentally induced ribosomopathy in the mouse and show that a normal level of hepatic ribosomal protein S6 (Rps6) is required for proper bile duct development and preservation of hepatocyte viability and that its insufficiency later promotes overgrowth and predisposes to liver cancer which is accelerated in the absence of the tumor-suppressor PTEN. We also show that the overexpression of c-Myc in the liver ameliorates, while expression of a mutant hyperstable form of p53 partially recapitulates specific aspects of the hepatopathies induced by Rps6 deletion. Surprisingly, co-deletion of p53 in the Rps6-deficient background fails to restore biliary development or significantly improve hepatic function. This study not only reveals a previously unappreciated dependence of the developing liver on adequate levels of Rps6 and exquisitely controlled p53 signaling, but suggests that the increased cancer risk in ribosomopathy patients may, in part, stem from an inability to preserve normal tissue homeostasis in the face of chronic injury and regeneration.


Asunto(s)
Neoplasias , Proteína p53 Supresora de Tumor , Humanos , Animales , Ratones , Proteína S6 Ribosómica/genética , Proteína S6 Ribosómica/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Hepatocitos/metabolismo , Fenotipo , Conductos Biliares/metabolismo , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo
4.
EMBO Rep ; 24(10): e57032, 2023 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-37650863

RESUMEN

Bromodomain-containing protein 4 (BRD4) is overexpressed and functionally implicated in various myeloid malignancies. However, the role of BRD4 in normal hematopoiesis remains largely unknown. Here, utilizing an inducible Brd4 knockout mouse model, we find that deletion of Brd4 (Brd4Δ/Δ ) in the hematopoietic system impairs hematopoietic stem cell (HSC) self-renewal and differentiation, which associates with cell cycle arrest and senescence. ATAC-seq analysis shows increased chromatin accessibility in Brd4Δ/Δ hematopoietic stem/progenitor cells (HSC/HPCs). Genome-wide mapping with cleavage under target and release using nuclease (CUT&RUN) assays demonstrate that increased global enrichment of H3K122ac and H3K4me3 in Brd4Δ/Δ HSC/HPCs is associated with the upregulation of senescence-specific genes. Interestingly, Brd4 deletion increases clipped H3 (cH3) which correlates with the upregulation of senescence-specific genes and results in a higher frequency of senescent HSC/HPCs. Re-expression of BRD4 reduces cH3 levels and rescues the senescence rate in Brd4Δ/Δ HSC/HPCs. This study unveils an important role of BRD4 in HSC/HPC function by preventing H3 clipping and suppressing senescence gene expression.


Asunto(s)
Histonas , Factores de Transcripción , Animales , Ratones , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Histonas/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Senescencia Celular/genética , Células Madre Hematopoyéticas/metabolismo , Diferenciación Celular , Hematopoyesis
5.
Am J Epidemiol ; 193(7): 940-950, 2024 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-38576195

RESUMEN

Avanzando Caminos (Leading Pathways): The Hispanic/Latino Cancer Survivorship Cohort Study aims to examine the influence of sociocultural, medical, stress-related, psychosocial, lifestyle, behavioral, and biological factors on symptom burden, health-related quality of life, and clinical outcomes among Hispanics/Latinos who have been previously treated for cancer. Avanzando Caminos is a prospective, cohort-based study of 3000 Hispanics/Latinos who completed primary cancer treatment within the past 5 years that is representative of the general Hispanic/Latino population in the United States. Participants will complete self-report measures at baseline (time [T] 1), 6 months (T2), 1 year (T3), 2 years (T4), 3 years (T5), 4 years (T6), and 5 years (T7). Blood samples drawn for assessment of leukocyte gene expression, cardiometabolic markers, and genetic admixture will be collected at baseline (T1), 1 year (T3), 3 years (T5), and 5 years (T7). Medical and cancer characteristics and clinical outcomes will be extracted from the electronic medical record and/or state cancer registry at each time point. Data analysis will include general latent variable modeling and latent growth modeling. Avanzando Caminos will fill critical gaps in knowledge in order to guide future secondary and tertiary prevention efforts to mitigate cancer disparities and optimize health-related quality of life among Hispanic/Latino cancer survivors.


Asunto(s)
Supervivientes de Cáncer , Hispánicos o Latinos , Calidad de Vida , Humanos , Hispánicos o Latinos/estadística & datos numéricos , Estudios Prospectivos , Supervivientes de Cáncer/estadística & datos numéricos , Masculino , Femenino , Estados Unidos/epidemiología , Neoplasias/etnología , Adulto , Persona de Mediana Edad , Proyectos de Investigación , Anciano , Factores Socioeconómicos
6.
Brief Bioinform ; 23(1)2022 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-34553223

RESUMEN

With the rapid development of single-cell sequencing techniques, several large-scale cell atlas projects have been launched across the world. However, it is still challenging to integrate single-cell RNA-seq (scRNA-seq) datasets with diverse tissue sources, developmental stages and/or few overlaps, due to the ambiguity in determining the batch information, which is particularly important for current batch-effect correction methods. Here, we present SCORE, a simple network-based integration methodology, which incorporates curated molecular network features to infer cellular states and generate a unified workflow for integrating scRNA-seq datasets. Validating on real single-cell datasets, we showed that regardless of batch information, SCORE outperforms existing methods in accuracy, robustness, scalability and data integration.


Asunto(s)
Análisis de la Célula Individual , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Secuenciación del Exoma
7.
Brief Bioinform ; 23(1)2022 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-34929734

RESUMEN

Since its selection as the method of the year in 2013, single-cell technologies have become mature enough to provide answers to complex research questions. With the growth of single-cell profiling technologies, there has also been a significant increase in data collected from single-cell profilings, resulting in computational challenges to process these massive and complicated datasets. To address these challenges, deep learning (DL) is positioned as a competitive alternative for single-cell analyses besides the traditional machine learning approaches. Here, we survey a total of 25 DL algorithms and their applicability for a specific step in the single cell RNA-seq processing pipeline. Specifically, we establish a unified mathematical representation of variational autoencoder, autoencoder, generative adversarial network and supervised DL models, compare the training strategies and loss functions for these models, and relate the loss functions of these models to specific objectives of the data processing step. Such a presentation will allow readers to choose suitable algorithms for their particular objective at each step in the pipeline. We envision that this survey will serve as an important information portal for learning the application of DL for scRNA-seq analysis and inspire innovative uses of DL to address a broader range of new challenges in emerging multi-omics and spatial single-cell sequencing.


Asunto(s)
Aprendizaje Profundo , RNA-Seq/métodos , Análisis de la Célula Individual/métodos , Algoritmos , Análisis por Conglomerados , Perfilación de la Expresión Génica/métodos , Humanos , Aprendizaje Automático , Análisis de Secuencia de ARN/métodos , Transcriptoma
8.
Nature ; 559(7715): E11, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29950716

RESUMEN

In this Letter, the sentence beginning "This work was funded…." in the Acknowledgements should have read "CPRIT (RP140105) to J.C.R." rather than "CPRIT (RP150445) to J.C.R." This error has been corrected online.

9.
Nature ; 555(7696): 387-391, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29513652

RESUMEN

Ewing sarcoma is an aggressive paediatric cancer of the bone and soft tissue. It results from a chromosomal translocation, predominantly t(11;22)(q24:q12), that fuses the N-terminal transactivation domain of the constitutively expressed EWSR1 protein with the C-terminal DNA binding domain of the rarely expressed FLI1 protein. Ewing sarcoma is highly sensitive to genotoxic agents such as etoposide, but the underlying molecular basis of this sensitivity is unclear. Here we show that Ewing sarcoma cells display alterations in regulation of damage-induced transcription, accumulation of R-loops and increased replication stress. In addition, homologous recombination is impaired in Ewing sarcoma owing to an enriched interaction between BRCA1 and the elongating transcription machinery. Finally, we uncover a role for EWSR1 in the transcriptional response to damage, suppressing R-loops and promoting homologous recombination. Our findings improve the current understanding of EWSR1 function, elucidate the mechanistic basis of the sensitivity of Ewing sarcoma to chemotherapy (including PARP1 inhibitors) and highlight a class of BRCA-deficient-like tumours.


Asunto(s)
Proteína BRCA1/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica , Conformación de Ácido Nucleico , Proteínas de Fusión Oncogénica/metabolismo , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína EWS de Unión a ARN/metabolismo , Reparación del ADN por Recombinación , Sarcoma de Ewing/genética , Transcripción Genética , Proteína BRCA1/metabolismo , Línea Celular Tumoral , Daño del ADN , Humanos , Proteínas de Fusión Oncogénica/genética , Proteína Proto-Oncogénica c-fli-1/genética , Proteína EWS de Unión a ARN/genética , Sarcoma de Ewing/metabolismo
10.
Environ Res ; 244: 117912, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38097058

RESUMEN

Energy transition policies are of great significance in adjusting the structure of energy supply and demand and coping with climate change. The new energy demonstration city pilot (NEDCP) policy, as an important pilot project in China's energy transition process, lacks a scientific assessment of the carbon reduction effect of the NEDCP policy and an in-depth explanation of the mechanism of the NEDCP. Based on panel data of 209 Chinese cities at the prefectural and higher levels from 2007 to 2019, this study takes the NEDCP policy as a quasi-natural experiment, using a difference-in-differences model combined with firm-level data to identify the impact of the NEDCP policy on urban carbon dioxide (CO2) emissions. This study analyzes the impact of heterogeneity of urban characteristics on the policy effect from multiple perspectives, and further investigates its mechanism. The conclusions are shown in the following aspects. (1) The implementation of the NEDCP policy decreases urban CO2 emissions significantly. Meanwhile, a series of robustness tests, including the instrumental variables method, propensity score matching difference-in-differences method, placebo test, exclusion of policy interference test, and machine learning method, support this conclusion. (2) The NEDCP policy achieves carbon reduction effects mainly through scale and structure effects. (3) The results of the heterogeneity test show that the NEDCP policy is more effective in cities with higher administrative levels, energy-demanding cities, cities in the southeast of Hu-line, and cities with a higher degree of nationalization. Therefore, the Chinese government should summarize the implementation experience of the NEDCP policy and expand its scope of application. The evaluation of the NEDCP policy in China has important reference value for the energy transition of other developing countries.


Asunto(s)
Dióxido de Carbono , Políticas , Ciudades , Proyectos Piloto , China , Desarrollo Económico
11.
Breast Cancer Res ; 25(1): 131, 2023 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-37904250

RESUMEN

BACKGROUND: Mammary physiology is distinguished in containing adult stem/progenitor cells that are actively amending the breast tissue throughout the reproductive lifespan of women. Despite their importance in both mammary gland development, physiological maintenance, and reproduction, the exact role of mammary stem/progenitor cells in mammary tumorigenesis has not been fully elucidated in humans or animal models. The implications of modulating adult stem/progenitor cells in women could lead to a better understanding of not only their function, but also toward possible breast cancer prevention led us to evaluate the efficacy of rapamycin in reducing mammary stem/progenitor cell activity and malignant progression markers. METHODS: We analyzed a large number of human breast tissues for their basal and luminal cell composition with flow cytometry and their stem and progenitor cell function with sphere formation assay with respect to age and menopausal status in connection with a clinical study (NCT02642094) involving a low-dose (2 mg/day) and short-term (5-7 days) treatment of the mTOR inhibitor sirolimus. The expression of biomarkers in biopsies and surgical breast samples were measured with quantitative analysis of immunohistochemistry. RESULTS: Sirolimus treatment significantly abrogated mammary stem cell activity, particularly in postmenopausal patients. It did not affect the frequency of luminal progenitors but decreased their self-renewal capacity. While sirolimus had no effect on basal cell population, it decreased luminal cell population, particularly in postmenopausal patients. It also significantly diminished prognostic biomarkers associated with breast cancer progression from ductal carcinoma in situ to invasive breast cancer including p16INK4A, COX-2, and Ki67, as well as markers of the senescence-associated secretary phenotype, thereby possibly functioning in preventing early breast cancer progression. CONCLUSION: Overall, these findings indicate a link from mTOR signaling to mammary stem and progenitor cell activity and cancer progression. Trial registration This study involves a clinical trial registered under the ClinicalTrials.gov identifier NCT02642094 registered December 30, 2015.


Asunto(s)
Neoplasias de la Mama , Animales , Humanos , Femenino , Neoplasias de la Mama/genética , Glándulas Mamarias Animales/metabolismo , Células Madre/metabolismo , Biomarcadores/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Sirolimus/farmacología , Sirolimus/metabolismo , Células Epiteliales/metabolismo
12.
Brief Bioinform ; 22(6)2021 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-34097004

RESUMEN

Transcriptomic and epigenetic alterations during early embryo development have been proven to play essential roles in regulating the cell fate. Nowadays, advances in single-cell transcriptomics and epigenomics profiling techniques provide large volumes of data for understanding the molecular regulatory mechanisms in early embryos and facilitate the investigation of assisted reproductive technology as well as preimplantation genetic testing. However, the lack of integrated data collection and unified analytic procedures greatly limits their usage in scientific research and clinical application. Hence, it is necessary to establish a database integrating the regulatory information of human and mouse early embryos with unified analytic procedures. Here, we introduce DevOmics (http://devomics.cn/), which contains normalized gene expression, DNA methylation, histone modifications (H3K4me3, H3K9me3, H3K27me3, H3K27ac), chromatin accessibility and 3D chromatin architecture profiles of human and mouse early embryos spanning six developmental stages (zygote, 2cell, 4cell, 8cell, morula and blastocyst (ICM, TE)). The current version of DevOmics provides Search and Advanced Search for retrieving genes a researcher is interested in, Analysis Tools including the differentially expressed genes (DEGs) analysis for acquiring DEGs between different types of samples, allelic explorer for displaying allele-specific gene expression as well as epigenetic modifications and correlation analysis for showing the dynamic changes in different layers of data across developmental stages, as well as Genome Browser and Ortholog for visualization. DevOmics offers a user-friendly website for biologists and clinicians to decipher molecular regulatory mechanisms of human and mouse early embryos.


Asunto(s)
Biología Computacional/métodos , Bases de Datos Genéticas , Embrión de Mamíferos/embriología , Embrión de Mamíferos/fisiología , Desarrollo Embrionario , Genómica/métodos , Programas Informáticos , Animales , Mapeo Cromosómico , Metilación de ADN , Epigenómica/métodos , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Ratones , Interfaz Usuario-Computador , Navegador Web
13.
Reprod Biomed Online ; 47(1): 26-34, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37105855

RESUMEN

RESEARCH QUESTION: Are there any differences between conjoined twin fetuses at the molecular level? DESIGN: Skin tissues were collected from thoracopagus conjoined twins at 15+4 weeks of gestation. The skin tissues were collected from the thigh side of conjoined twins after the abortion procedure. All specimens were obtained after written informed patient consent and were fully anonymized. All relevant ethical regulations were followed. Every specimen underwent multiomics sequencing analysis to determine associations among the DNA methylome, transcriptome and mutations in the exon regions in the conjoined twins. RESULTS: The global methylation pattern was similar in the two fetuses of conjoined twins, while significant differences were seen in local regions such as CpG islands (P = 0.026), enhancers (P < 0.001) and various repetitive elements (P < 0.05), which showed significant differences. The conjoined twins also differed in genes related to growth and development, cellular component morphogenesis and cellular stress, both in terms of DNA methylation levels and gene expression levels. Exon data analysis revealed that the common mutations in conjoined twins mainly occurred in neural development, lipid metabolism and microtubule morphogenesis. Specific mutations were associated with cellular component biosynthesis, behaviour and germ cell development. CONCLUSION: Conjoined twins were similar to each other globally, but there were significant differences related to growth and development, cellular component morphogenesis and cellular stress. The current study reveals the molecular features of conjoined twins for the first time, laying the foundation for future exploration of the mechanism of conjoined twins.


Asunto(s)
Aborto Inducido , Gemelos Siameses , Embarazo , Femenino , Humanos , Multiómica , Feto
14.
Nucleic Acids Res ; 49(20): e116, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34417605

RESUMEN

N6-methyladenosine (m6A) is the most abundant form of mRNA modification and controls many aspects of RNA metabolism including gene expression. However, the mechanisms by which m6A regulates cell- and condition-specific gene expression are still poorly understood, partly due to a lack of tools capable of identifying m6A sites that regulate gene expression under different conditions. Here we develop m6A-express, the first algorithm for predicting condition-specific m6A regulation of gene expression (m6A-reg-exp) from limited methylated RNA immunoprecipitation sequencing (MeRIP-seq) data. Comprehensive evaluations of m6A-express using simulated and real data demonstrated its high prediction specificity and sensitivity. When only a few MeRIP-seq samples may be available for the cellular or treatment conditions, m6A-express is particularly more robust than the log-linear model. Using m6A-express, we reported that m6A writers, METTL3 and METTL14, competitively regulate the transcriptional processes by mediating m6A-reg-exp of different genes in Hela cells. In contrast, METTL3 induces different m6A-reg-exp of a distinct group of genes in HepG2 cells to regulate protein functions and stress-related processes. We further uncovered unique m6A-reg-exp patterns in human brain and intestine tissues, which are enriched in organ-specific processes. This study demonstrates the effectiveness of m6A-express in predicting condition-specific m6A-reg-exp and highlights the complex, condition-specific nature of m6A-regulation of gene expression.


Asunto(s)
Adenosina/análogos & derivados , Procesamiento Postranscripcional del ARN , Análisis de Secuencia de ARN/métodos , Adenosina/metabolismo , Encéfalo/metabolismo , Células HeLa , Células Hep G2 , Humanos , Mucosa Intestinal/metabolismo
15.
Proc Natl Acad Sci U S A ; 117(33): 20109-20116, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32747526

RESUMEN

Herpesviruses are ubiquitous human pathogens that cause a wide range of health complications. Currently, there is an incomplete understanding of cellular factors that contribute to herpesvirus infection. Here, we report an antiviral necroptosis-based genetic screen to identify novel host cell factors required for infection with the ß-herpesvirus murine cytomegalovirus (MCMV). Our genome-wide CRISPR-based screen harnessed the capacity of herpesvirus mutants that trigger antiviral necroptotic cell death upon early viral gene expression. Vascular endothelial growth factor (VEGF) and semaphorin-binding receptor Neuropilin-1 (Nrp-1) emerge as crucial determinants of MCMV infection. We find that elimination of Nrp-1 impairs early viral gene expression and reduces infection rates in endothelial cells, fibroblasts, and macrophages. Furthermore, preincubation of virus with soluble Nrp-1 dramatically inhibits infection by reducing virus attachment. Thus, Nrp-1 is a key determinant of the initial phase of MCMV infection.


Asunto(s)
Infecciones por Citomegalovirus/metabolismo , Muromegalovirus/metabolismo , Necroptosis/fisiología , Neuropilina-1/metabolismo , Animales , Línea Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Infecciones por Citomegalovirus/genética , Eliminación de Gen , Regulación Viral de la Expresión Génica , Ratones , Muromegalovirus/genética , Neuropilina-1/genética
16.
Int J Mol Sci ; 24(18)2023 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-37762536

RESUMEN

BACKGROUND: Ulcerative colitis (UC) is a chronic, incurable condition characterized by mucosal inflammation and intestinal epithelial cell (IEC) damage. The circadian clock gene NR1D1, implicated in UC and the critical mitophagy process for epithelial repair, needs further exploration regarding its role in mitophagy regulation in UC. METHODS: We created a jet lag mouse model and induced colitis with dextran sulfate sodium (DSS), investigating NR1D1's role. Intestinal-specific Nr1d1 knockout mice were also generated. RNA sequencing, chromatin immunoprecipitation (ChIP), and dual-luciferase reporter assays helped ascertain NR1D1's regulatory effect on BNIP3 expression. The mitochondrial state in IECs was assessed through transmission electron microscopy, while confocal microscopy evaluated mitophagy-associated protein expression in colon tissue and CCD841 cells. Cell apoptosis and reactive oxygen species (ROS) were measured via flow cytometry. RESULTS: We observed reduced NR1D1 expression in the IECs of UC patients, accentuated under jet lag and DSS exposure in mice. NR1D1 ablation led to disrupted immune homeostasis and declined mitophagy in IECs. NR1D1, usually a transcriptional repressor, was a positive regulator of BNIP3 expression, leading to impaired mitophagy, cellular inflammation, and apoptosis. Administering the NR1D1 agonist SR9009 ameliorated colitis symptoms, primarily by rectifying defective mitophagy. CONCLUSIONS: Our results suggest that NR1D1 bridges the circadian clock and UC, controlling BNIP3-mediated mitophagy and representing a potential therapeutic target. Its agonist, SR9009, shows promise in UC symptom alleviation.


Asunto(s)
Colitis Ulcerosa , Colitis , Animales , Humanos , Ratones , Colitis/inducido químicamente , Colitis/genética , Colitis Ulcerosa/genética , Inflamación , Síndrome Jet Lag , Proteínas de la Membrana/genética , Mitofagia , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/genética , Proteínas Proto-Oncogénicas/genética
17.
J Clean Prod ; 414: 137755, 2023 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-37304130

RESUMEN

The COVID-19 pandemic prompted several nations, including China, to enact unprecedented lockdown measures, leading to significant alterations in environmental conditions. Previous studies have solely analysed the impact of lockdown measures on air pollutants or carbon dioxide (CO2) emissions during the COVID-19 pandemic in China, but few have focused on the spatio-temporal change characteristics and synergistic effects between the two. In this study, we constructed a methodological framework to examine the spatiotemporal characteristics and co-effects of air quality (PM2.5, SO2, and NO2) and CO2 changes in 324 prefecture-level cities in China due to the COVID-19 blockade measures from January 24 to April 30, 2020, using the regression discontinuity in time method and co-effect control coordinate system. The results show that a significant improvement in air quality and CO2 emissions during the lockdown period, with notable north‒south heterogeneity. During the major lockdown period (January 24 to February 29), the measures resulted in respective reductions of 5.6%, 16.6%, and 25.1% in the concentrations of SO2, NO2, and CO2 nationwide. The proportions of cities with negative treatment effects on PM2.5, SO2, NO2, and CO2 were 39.20%, 70.99%, 84.6%, and 99.38%, respectively. Provinces where concentrations of CO2 and NO2 declined by over 30% were primarily concentrated in southern areas of the 'Yangtze River Defense Line'. Starting from March, the improvement effect of air quality and CO2 has weakened, and the concentration of air pollutants has rebounded. This study offers crucial insights into the causal effects of lockdown measures on air quality changes, and reveals the synergy between air quality and CO2, thereby providing a reference for devising effective air quality improvement and energy-saving emission reduction strategies.

18.
Neurosciences (Riyadh) ; 28(1): 42-47, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36617452

RESUMEN

OBJECTIVES: To explore the clinicopathological features and prognosis of multifocal high-grade gliomas (M-HGGs) with H3F3A mutation in adults. METHODS: Four adult patients with H3F3A-mutant M-HGGs who were treated at our institution from August 2020 to December 2021 were reviewed, including clinical, pathological and radiologic data. A series of 16 adult patients with M-HGGs without H3F3A mutation was used as a comparative group. Progression-free survival (PFS) and overall survival (OS) were compared between the groups using the Kaplan-Meier method. RESULTS: All patients were IDH wild-type and TERT wild-type, and P53 was overexpressed. A patient with the H3 G34R mutation and 1 of 3 patients with the H3 K27 M mutation had MGMT promoter methylation. The lesions with the H3 G34R mutation were located in the cerebral hemisphere; the lesions with H3 K27 alterations were mainly in the midline structure, and the cerebral hemisphere could also be involved. One patient underwent subtotal resection (STR), and 3 patients underwent biopsy. All patients received radiotherapy, and the median PFS and OS were 9.5 months and 14.5 months, respectively. The clinical outcomes were similar to those of non-H3F3A-mutated M-HGGs patients (median PFS and OS were 7.0 months and 18.0 months, respectively). CONCLUSION: We describe the clinicopathological features and outcomes of 4 adult M-HGGs patients with H3F3A mutation, and found this mutation doesn't appear to have a negative outcome with the administration of current therapies.


Asunto(s)
Neoplasias Encefálicas , Glioma , Adulto , Humanos , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Glioma/diagnóstico por imagen , Glioma/genética , Glioma/terapia , Pronóstico , Mutación/genética
19.
Breast Cancer Res ; 24(1): 26, 2022 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-35395812

RESUMEN

BACKGROUND: Methyltransferase SETDB1 is highly expressed in breast cancer (BC), however, the mechanisms by which SETDB1 promotes BC progression to endocrine therapy resistance remains elusive. In this study, we examined the mechanisms by which SETDB1 contribute to BC endocrine therapy resistance. METHODS: We utilized therapy sensitive (MCF7 and ZR75), therapy resistant (MCF7-TamR, MCF7-FR, MCF7-PELP1cyto, MCF7-SETDB1) estrogen receptor alpha positive (ER+)BC models and conducted in vitro cell viability, colony formation, 3-dimensional cell growth assays to investigate the role of SETDB1 in endocrine resistance. RNA-seq of parental and SETDB1 knock down ER+ BC cells was used to identify unique pathways. SETDB1 interaction with PELP1 was identified by yeast-two hybrid screen and confirmed by immunoprecipitation and GST-pull down assays. Mechanistic studies were conducted using Western blotting, reporter gene assays, RT-qPCR, and in vitro methylation assays. Xenograft assays were used to establish the role of PELP1 in SETDB1 mediated BC progression. RESULTS: RNA-seq analyses showed that SETDB1 regulates expression of a subset of estrogen receptor (ER) and Akt target genes that contribute to endocrine therapy resistance. Importantly, using yeast-two hybrid screen, we identified ER coregulator PELP1 as a novel interacting protein of SETDB1. Biochemical analyses confirmed SETDB1 and PELP1 interactions in multiple BC cells. Mechanistic studies confirmed that PELP1 is necessary for SETDB1 mediated Akt methylation and phosphorylation. Further, SETDB1 overexpression promotes tamoxifen resistance in BC cells, and PELP1 knockdown abolished these effects. Using xenograft model, we provided genetic evidence that PELP1 is essential for SETDB1 mediated BC progression in vivo. Analyses of TCGA datasets revealed SETDB1 expression is positively correlated with PELP1 expression in ER+ BC patients. CONCLUSIONS: This study suggests that the PELP1/SETDB1 axis play an important role in aberrant Akt activation and serves as a novel target for treating endocrine therapy resistance in breast cancer.


Asunto(s)
Neoplasias de la Mama , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Proteínas Co-Represoras/genética , Proteínas Co-Represoras/metabolismo , Proteínas Co-Represoras/farmacología , Resistencia a Antineoplásicos/genética , Femenino , Regulación Neoplásica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/farmacología , Humanos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Saccharomyces cerevisiae/metabolismo , Tamoxifeno/farmacología , Factores de Transcripción/genética
20.
Int J Cancer ; 151(6): 930-943, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35657344

RESUMEN

Integrin α6 (ITGA6) forms integrin receptors with either integrin ß1 (ITGB1) or integrin ß4 (ITGB4). How it functions to regulate hepatocellular carcinoma (HCC) progression is not well-elucidated. We found that ITGA6 RNA and protein expression levels are significantly elevated in human HCC tissues in comparison with paired adjacent nontumor tissues by RNA sequencing, RT-qPCR, Western blotting and immunofluorescence staining. Stable knockdown of ITGA6 with different ITGA6 shRNA expression lentivectors significantly inhibited proliferation, migration and anchorage-independent growth of HCC cell lines in vitro, and xenograft tumor growth in vivo. The inhibition of anchorage-dependent and -independent growth of HCC cell lines was also confirmed with anti-ITGA6 antibody. ITGA6 knockdown was shown to induce cell-cycle arrest at G0/G1 phase. Immunoprecipitation assay revealed apparent interaction of ITGA6 with ITGB4, but not ITGB1. Expression studies showed that ITGA6 positively regulates the expression of ITGB4 with no or negative regulation of ITGB1 expression. Finally, while high levels of ITGA6 and ITGB4 together were associated with significantly worse survival of HCC patients in TCGA data set, the association was not significant for high levels of ITGA6 and ITGB1. In conclusion, ITGA6 is upregulated in HCC tumors and has a malignant promoting role in HCC cells through integrin α6ß4 complex. Thus, integrin α6ß4 may be a therapeutic target for treating patients with HCC.


Asunto(s)
Carcinoma Hepatocelular , Integrina alfa6 , Integrina alfa6beta4 , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Integrina alfa6/genética , Integrina alfa6/metabolismo , Integrina alfa6beta4/genética , Integrina alfa6beta4/metabolismo , Integrina beta4/genética , Integrina beta4/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda