Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 14(1): 5586, 2023 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-37696809

RESUMEN

A large spread in model estimates of the equilibrium climate sensitivity (ECS), defined as the global mean near-surface air-temperature increase following a doubling of atmospheric CO2 concentration, leaves us greatly disadvantaged in guiding policy-making for climate change adaptation and mitigation. In this study, we show that the projected ECS in the latest generation of climate models is highly related to seasonal variations of extratropical low-cloud fraction (LCF) in historical simulations. Marked reduction of extratropical LCF from winter to summer is found in models with ECS > 4.75 K, in accordance with the significant reduction of extratropical LCF under a warming climate in these models. In contrast, a pronounced seasonal cycle of extratropical LCF, as supported by satellite observations, is largely absent in models with ECS < 3.3 K. The distinct seasonality in extratropical LCF in climate models is ascribed to their different prevailing cloud regimes governing the extratropical LCF variability.

2.
Cell Death Differ ; 30(5): 1166-1183, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36813924

RESUMEN

Peptides/small proteins, encoded by noncanonical open reading frames (ORF) of previously claimed non-coding RNAs, have recently been recognized possessing important biological functions, but largely uncharacterized. 1p36 is an important tumor suppressor gene (TSG) locus frequently deleted in multiple cancers, with critical TSGs like TP73, PRDM16, and CHD5 already validated. Our CpG methylome analysis identified a silenced 1p36.3 gene KIAA0495, previously thought coding long non-coding RNA. We found that the open reading frame 2 of KIAA0495 is actually protein-coding and translating, encoding a small protein SP0495. KIAA0495 transcript is broadly expressed in multiple normal tissues, but frequently silenced by promoter CpG methylation in multiple tumor cell lines and primary tumors including colorectal, esophageal and breast cancers. Its downregulation/methylation is associated with poor survival of cancer patients. SP0495 induces tumor cell apoptosis, cell cycle arrest, senescence and autophagy, and inhibits tumor cell growth in vitro and in vivo. Mechanistically, SP0495 binds to phosphoinositides (PtdIns(3)P, PtdIns(3,5)P2) as a lipid-binding protein, inhibits AKT phosphorylation and its downstream signaling, and further represses oncogenic AKT/mTOR, NF-κB, and Wnt/ß-catenin signaling. SP0495 also regulates the stability of autophagy regulators BECN1 and SQSTM1/p62 through modulating phosphoinositides turnover and autophagic/proteasomal degradation. Thus, we discovered and validated a 1p36.3 small protein SP0495, functioning as a novel tumor suppressor regulating AKT signaling activation and autophagy as a phosphoinositide-binding protein, being frequently inactivated by promoter methylation in multiple tumors as a potential biomarker.


Asunto(s)
ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosforilación , Metilación de ADN/genética , Proteínas Portadoras/metabolismo , Genes Supresores de Tumor , Proliferación Celular/genética , Línea Celular Tumoral , Vía de Señalización Wnt , Autofagia/genética , Regulación Neoplásica de la Expresión Génica , ADN Helicasas/metabolismo , Proteínas del Tejido Nervioso/metabolismo
3.
Cell Biosci ; 12(1): 43, 2022 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-35379326

RESUMEN

BACKGROUND: Nuclear factor-κB is a multi-subunit transcription factor that plays a central role in cellular senescence. We previously reported that an increase in the p52 subunit is seen in senescent cells and aged tissue. In the current work, we examined the mechanism by which p52 is activated and whether the increase in p52 promotes senescence. RESULTS: Using both primary mouse embryonic fibroblasts (MEFs) and WI-38 human lung fibroblasts, we examined cells after serial passage and following prolonged culture. An increase in p52 was found in the nucleus relative to pre-senescent cells. The increase in p52 protein was not reflected by an increase in NFKB2 mRNA or by an increase in the abundance of upstream activating kinases, IKKα and NIK. To examine whether p52 promotes senescence, we over-expressed mature p52 in primary MEFs. Significantly more senescence was seen compared to control, a finding not seen with p52 mutated at critical DNA binding residues. In addition, blocking p52 nuclear translocation with the peptide inhibitor, SN52, decreased ß-galactosidase (ß-gal) formation. Subsequent filtration studies demonstrated that proteins in conditioned media (CM) were necessary for the increase in p52 and mass spectrometry identified S100A4 and cyclophilin A (CYPA) as potential factors in CM necessary for induction of p52. The requirement of these proteins in CM for induction of p52 was confirmed using depletion and supplementation studies. In addition, we found that activation of STAT3 signaling was required for the increase in p52. Finally, genome wide ChIP-sequencing analysis confirmed that there is an increase in p52 chromatin enrichment with senescence and identified several downstream factors whose expression is regulated by increased p52 binding. CONCLUSIONS: These results demonstrate that p52 nuclear translocation is increased in senescent cells by factors in conditioned media and that mature p52 induces cellular senescence. The data are consistent with the prior observation that p52 is elevated in aged tissue and support the hypothesis that p52 contributes to organismal aging.

4.
ACS Synth Biol ; 11(1): 92-102, 2022 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-34927418

RESUMEN

Promoters are one of the most critical regulatory elements controlling metabolic pathways. However, the fast and accurate prediction of promoter strength remains challenging, leading to time- and labor-consuming promoter construction and characterization processes. This dilemma is caused by the lack of a big promoter library that has gradient strengths, broad dynamic ranges, and clear sequence profiles that can be used to train an artificial intelligence model of promoter strength prediction. To overcome this challenge, we constructed and characterized a mutant library of Trc promoters (Ptrc) using 83 rounds of mutation-construction-screening-characterization engineering cycles. After excluding invalid mutation sites, we established a synthetic promoter library that consisted of 3665 different variants, displaying an intensity range of more than two orders of magnitude. The strongest variant was ∼69-fold stronger than the original Ptrc and 1.52-fold stronger than a 1 mM isopropyl-ß-d-thiogalactoside-driven PT7 promoter, with an ∼454-fold difference between the strongest and weakest expression levels. Using this synthetic promoter library, different machine learning models were built and optimized to explore the relationships between promoter sequences and transcriptional strength. Finally, our XgBoost model exhibited optimal performance, and we utilized this approach to precisely predict the strength of artificially designed promoter sequences (R2 = 0.88, mean absolute error = 0.15, and Pearson correlation coefficient = 0.94). Our work provides a powerful platform that enables the predictable tuning of promoters to achieve optimal transcriptional strength.


Asunto(s)
Inteligencia Artificial , Redes y Vías Metabólicas , Biblioteca de Genes , Aprendizaje Automático , Regiones Promotoras Genéticas/genética
5.
Front Immunol ; 12: 679482, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34177923

RESUMEN

Infection with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes COVID-19, a disease that involves significant lung tissue damage. How SARS-CoV-2 infection leads to lung injury remains elusive. The open reading frame 8 (ORF8) protein of SARS-CoV-2 (ORF8SARS-CoV-2) is a unique accessory protein, yet little is known about its cellular function. We examined the cellular distribution of ORF8SARS-CoV-2 and its role in the regulation of human lung epithelial cell proliferation and antiviral immunity. Using live imaging and immunofluorescent staining analyses, we found that ectopically expressed ORF8SARS-CoV-2 forms aggregates in the cytosol and nuclear compartments of lung epithelial cells. Using in silico bioinformatic analysis, we found that ORF8SARS-CoV-2 possesses an intrinsic aggregation characteristic at its N-terminal residues 1-18. Cell culture did not reveal any effects of ORF8SARS-CoV-2 expression on lung epithelial cell proliferation and cell cycle progression, suggesting that ORF8SARS-CoV-2 aggregates do not affect these cellular processes. Interestingly, ectopic expression of ORF8SARS-CoV-2 in lung epithelial cells suppressed basal expression of several antiviral molecules, including DHX58, ZBP1, MX1, and MX2. In addition, expression of ORF8SARS-CoV-2 attenuated the induction of antiviral molecules by IFNγ but not by IFNß in lung epithelial cells. Taken together, ORF8SARS-CoV-2 is a unique viral accessory protein that forms aggregates when expressing in lung epithelial cells. It potently inhibits the expression of lung cellular anti-viral proteins at baseline and in response to IFNγ in lung epithelial cells, which may facilitate SARS-CoV-2 escape from the host antiviral innate immune response during early viral infection. In addition, it seems that formation of ORF8SARS-CoV-2 aggregate is independent from the viral infection. Thus, it would be interesting to examine whether any COVID-19 patients exhibit persistent ORF8 SARS-CoV-2 expression after recovering from SARS-CoV-2 infection. If so, the pathogenic effect of prolonged ORF8SARS-CoV-2 expression and its association with post-COVID symptoms warrant investigation in the future.


Asunto(s)
COVID-19/inmunología , Pulmón/patología , Mucosa Respiratoria/fisiología , SARS-CoV-2/fisiología , Proteínas Virales/metabolismo , COVID-19/virología , Regulación de la Expresión Génica , Células HEK293 , Humanos , Inmunidad , Interferón gamma/metabolismo , Espacio Intracelular , Agregación Patológica de Proteínas , Mucosa Respiratoria/virología
6.
Sci Rep ; 11(1): 5665, 2021 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-33707466

RESUMEN

The alkylating agent, temozolomide (TMZ), is the most commonly used chemotherapeutic for the treatment of glioblastoma (GBM). The anti-glioma effect of TMZ involves a complex response that includes G2-M cell cycle arrest and cyclin-dependent kinase 1 (CDK1) activation. While CDK1 phosphorylation is a well-described consequence of TMZ treatment, we find that TMZ also robustly induces CDK1 expression. Analysis of this pathway demonstrates that CDK1 is regulated by NF-κB via a putative κB-site in its proximal promoter. CDK1 was induced in a manner dependent on mature p50 and the atypical inhibitor κB protein, BCL-3. Treatment with TMZ induced binding of NF-κB to the κB-site as assessed by gel shift analysis and chromatin immunoprecipitation. Examination of a CDK1 promoter-reporter demonstrated the functional relevance of the κB-site and underlined the requirement of p50 and BCL-3 for activation. Targeted knockdown of CDK1 or chemical inhibition with the selective CDK1 inhibitor, RO-3306, potentiated the cytotoxic effect of TMZ. These results identify CDK1 as an NF-κB target gene regulated by p50 and BCL-3 and suggest that targeting CDK1 may be a strategy to improve the efficacy of TMZ against GBM.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Proteína Quinasa CDC2/metabolismo , Glioblastoma/metabolismo , FN-kappa B/metabolismo , Temozolomida/farmacología , Proteínas del Linfoma 3 de Células B/metabolismo , Secuencia de Bases , Sitios de Unión , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Proteína Quinasa CDC2/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/genética , Glioblastoma/patología , Humanos , Regiones Promotoras Genéticas/genética
7.
Nat Commun ; 11(1): 5007, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-33024116

RESUMEN

p50, the mature product of NFKB1, is constitutively produced from its precursor, p105. Here, we identify BARD1 as a p50-interacting factor. p50 directly associates with the BARD1 BRCT domains via a C-terminal phospho-serine motif. This interaction is induced by ATR and results in mono-ubiquitination of p50 by the BARD1/BRCA1 complex. During the cell cycle, p50 is mono-ubiquitinated in S phase and loss of this post-translational modification increases S phase progression and chromosomal breakage. Genome-wide studies reveal a substantial decrease in p50 chromatin enrichment in S phase and Cycln E is identified as a factor regulated by p50 during the G1 to S transition. Functionally, interaction with BARD1 promotes p50 protein stability and consistent with this, in human cancer specimens, low nuclear BARD1 protein strongly correlates with low nuclear p50. These data indicate that p50 mono-ubiquitination by BARD1/BRCA1 during the cell cycle regulates S phase progression to maintain genome integrity.


Asunto(s)
Neoplasias de la Mama/metabolismo , Ciclo Celular/fisiología , Inestabilidad Genómica , Subunidad p50 de NF-kappa B/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Sitios de Unión , Neoplasias de la Mama/mortalidad , Línea Celular Tumoral , Femenino , Fibroblastos , Humanos , Lisina/metabolismo , Ratones , Subunidad p50 de NF-kappa B/genética , Neuroblastoma/metabolismo , Dominios Proteicos , Procesamiento Proteico-Postraduccional , Serina/metabolismo , Proteínas Supresoras de Tumor/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
8.
BMC Biol ; 18(1): 32, 2020 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-32209106

RESUMEN

BACKGROUND: Nuclear factor-κB (NF-κB) plays a prominent role in promoting inflammation and resistance to DNA damaging therapy. We searched for proteins that modulate the NF-κB response as a prerequisite to identifying novel factors that affect sensitivity to DNA damaging chemotherapy. RESULTS: Using streptavidin-agarose pull-down, we identified the DExD/H-box RNA helicase, DDX39B, as a factor that differentially interacts with κB DNA probes. Subsequently, using both RNA interference and CRISPR/Cas9 technology, we demonstrated that DDX39B inhibits NF-κB activity by a general mechanism involving inhibition of p65 phosphorylation. Mechanistically, DDX39B mediates this effect by interacting with the pattern recognition receptor (PRR), LGP2, a pathway that required the cellular response to cytoplasmic double-stranded RNA (dsRNA). From a functional standpoint, loss of DDX39B promoted resistance to alkylating chemotherapy in glioblastoma cells. Further examination of DDX39B demonstrated that its protein abundance was regulated by site-specific sumoylation that promoted its poly-ubiquitination and degradation. These post-translational modifications required the presence of the SUMO E3 ligase, PIASx-ß. Finally, genome-wide analysis demonstrated that despite the link to the PRR system, DDX39B did not generally inhibit interferon-stimulated gene expression, but rather acted to attenuate expression of factors associated with the extracellular matrix, cellular migration, and angiogenesis. CONCLUSIONS: These results identify DDX39B, a factor with known functions in mRNA splicing and nuclear export, as an RNA-binding protein that blocks a subset of the inflammatory response. While these findings identify a pathway by which DDX39B promotes sensitization to DNA damaging therapy, the data also reveal a mechanism by which this helicase may act to mitigate autoimmune disease.


Asunto(s)
ARN Helicasas DEAD-box/genética , FN-kappa B/metabolismo , Receptores de Reconocimiento de Patrones/genética , Transducción de Señal , Alquilación , Animales , ARN Helicasas DEAD-box/metabolismo , Sondas de ADN , Quimioterapia , Humanos , Ratones , Receptores de Reconocimiento de Patrones/metabolismo
9.
Cancer Res ; 79(10): 2536-2548, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30940658

RESUMEN

Alkylating chemotherapy is a central component of the management of glioblastoma (GBM). Among the factors that regulate the response to alkylation damage, NF-κB acts to both promote and block cytotoxicity. In this study, we used genome-wide expression analysis in U87 GBM to identify NF-κB-dependent factors altered in response to temozolomide and found the long noncoding RNA (lncRNA) MALAT1 as one of the most significantly upregulated. In addition, we demonstrated that MALAT1 expression was coregulated by p50 (p105) and p53 via novel κB- and p53-binding sites in the proximal MALAT1 coding region. Temozolomide treatment inhibited p50 recruitment to its cognate element as a function of Ser329 phosphorylation while concomitantly increasing p53 recruitment. Moreover, luciferase reporter studies demonstrated that both κB and p53 cis-elements were required for efficient transactivation in response to temozolomide. Depletion of MALAT1 sensitized patient-derived GBM cells to temozolomide cytotoxicity, and in vivo delivery of nanoparticle-encapsulated anti-MALAT1 siRNA increased the efficacy of temozolomide in mice bearing intracranial GBM xenografts. Despite these observations, in situ hybridization of GBM specimens and analysis of publicly available datasets revealed that MALAT1 expression within GBM tissue was not prognostic of overall survival. Together, these findings support MALAT1 as a target for chemosensitization of GBM and identify p50 and p52 as primary regulators of this ncRNA. SIGNIFICANCE: These findings identify NF-κB and p53 as regulators of the lncRNA MALAT1 and suggest MALAT1 as a potential target for the chemosensitization of GBM.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Neoplasias Encefálicas/metabolismo , Glioblastoma/tratamiento farmacológico , FN-kappa B/metabolismo , ARN Largo no Codificante/biosíntesis , Temozolomida/uso terapéutico , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular Tumoral , Daño del ADN/genética , Técnicas de Silenciamiento del Gen , Glioblastoma/metabolismo , Humanos , Masculino , Ratones , Ratones Desnudos , Pronóstico , ARN Largo no Codificante/genética , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Sci Transl Med ; 10(448)2018 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-29973405

RESUMEN

The response of patients with gliomas to alkylating chemotherapy is heterogeneous. However, there are currently no universally accepted predictors of patient response to these agents. We identify the nuclear factor κB (NF-κB) co-regulator B cell CLL/lymphoma 3 (BCL-3) as an independent predictor of response to temozolomide (TMZ) treatment. In glioma patients with tumors that have a methylated O6-methylguanine DNA methyltransferase (MGMT) promoter, high BCL-3 expression was associated with a poor response to TMZ. Mechanistically, BCL-3 promoted a more malignant phenotype by inducing an epithelial-to-mesenchymal transition in glioblastomas through promoter-specific NF-κB dimer exchange. Carbonic anhydrase II (CAII) was identified as a downstream factor promoting BCL-3-mediated resistance to chemotherapy. Experiments in glioma xenograft mouse models demonstrated that the CAII inhibitor acetazolamide enhanced survival of TMZ-treated animals. Our data suggest that BCL-3 might be a useful indicator of glioma response to alkylating chemotherapy and that acetazolamide might be repurposed as a chemosensitizer for treating TMZ-resistant gliomas.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica , Glioma/tratamiento farmacológico , Glioma/genética , Proteínas Proto-Oncogénicas/genética , Factores de Transcripción/genética , Antineoplásicos Alquilantes/farmacología , Proteínas del Linfoma 3 de Células B , Anhidrasa Carbónica II/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Análisis Multivariante , FN-kappa B/metabolismo , Regiones Promotoras Genéticas/genética , Modelos de Riesgos Proporcionales , Multimerización de Proteína , Proteínas Proto-Oncogénicas/metabolismo , Análisis de Supervivencia , Temozolomida/farmacología , Temozolomida/uso terapéutico , Factor de Transcripción ReIA/metabolismo , Factores de Transcripción/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Gastroenterology ; 155(1): 144-155, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29621481

RESUMEN

BACKGROUND & AIMS: Inflammation affects regeneration of the intestinal epithelia; long noncoding RNAs (lncRNAs) regulate cell functions, such as proliferation, differentiation, and migration. We investigated the mechanisms by which the lncRNA H19, imprinted maternally expressed transcript (H19) regulates regeneration of intestinal epithelium using cell cultures and mouse models of inflammation. METHODS: We performed RNA-sequencing transcriptome analyses of intestinal tissues from mice with lipopolysaccharide (LPS)-induced sepsis to identify lncRNAs associated with inflammation; findings were confirmed by quantitative real-time polymerase chain reaction and in situ hybridization analyses of intestinal tissues from mice with sepsis or dextran sulfate sodium (DSS)-induced mucosal wound healing and patients with ulcerative colitis compared to healthy individuals (controls). We screened cytokines for their ability to induce expression of H19 in HT-29 cells and intestinal epithelial cells (IECs), and confirmed findings in crypt epithelial organoids derived from mouse small intestine. IECs were incubated with different signal transduction inhibitors and effects on H19 lncRNA levels were measured. We assessed intestinal epithelial proliferation or regeneration in H19ΔEx1/+ mice given LPS or DSS vs wild-type littermates (control mice). H19 was overexpressed in IECs using lentiviral vectors and cell proliferation was measured. We performed RNA antisense purification, RNA immunoprecipitation, and luciferase reporter assays to study functions of H19 in IECs. RESULTS: In RNA-sequencing transcriptome analysis of lncRNA expression in intestinal tissues from mice, we found that levels of H19 lncRNA changed significantly with LPS exposure. Levels of H19 lncRNA increased in intestinal tissues of patients with ulcerative colitis, mice with LPS-induced and polymicrobial sepsis, or mice with DSS-induced colitis, compared with controls. Increased H19 lncRNA localized to epithelial cells in the intestine, regardless of Lgr5 messenger RNA expression. Exposure of IECs to interleukin 22 (IL22) increased levels of H19 lncRNA with time and dose, which required STAT3 and protein kinase A activity. IL22 induced expression of H19 in mouse intestinal epithelial organoids within 6 hours. Exposure to IL22 increased growth of intestinal epithelial organoids derived from control mice, but not H19ΔEx1/+ mice. Overexpression of H19 in HT-29 cells increased their proliferation. Intestinal mucosa healed more slowly after withdrawal of DSS from H19ΔEx1/+ mice vs control mice. Crypt epithelial cells from H19ΔEx1/+ mice proliferated more slowly than those from control mice after exposure to LPS. H19 lncRNA bound to p53 and microRNAs that inhibit cell proliferation, including microRNA 34a and let-7; H19 lncRNA binding blocked their function, leading to increased expression of genes that promote regeneration of the epithelium. CONCLUSIONS: The level of lncRNA H19 is increased in inflamed intestinal tissues from mice and patients. The inflammatory cytokine IL22 induces expression of H19 in IECs, which is required for intestinal epithelial proliferation and mucosal healing. H19 lncRNA appears to inhibit p53 protein and microRNA 34a and let-7 to promote proliferation of IECs and epithelial regeneration.


Asunto(s)
Colitis Ulcerosa/inmunología , Regulación de la Expresión Génica/inmunología , Interleucinas/inmunología , Mucosa Intestinal/inmunología , ARN Largo no Codificante/genética , Regeneración/fisiología , Sepsis/inmunología , Animales , Estudios de Casos y Controles , Proliferación Celular , Modelos Animales de Enfermedad , Células Epiteliales , Perfilación de la Expresión Génica , Células HT29 , Humanos , Inflamación , Mucosa Intestinal/fisiología , Ratones , ARN Largo no Codificante/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Interleucina-22
12.
Hydrol Process ; 32(22): 3365-3390, 2018 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-31073260

RESUMEN

Accurate and reliable reservoir inflow forecast is instrumental to the efficient operation of the hydroelectric power systems. It has been discovered that natural and anthropogenic aerosols have a great influence on meteorological variables such as temperature, snow water equivalent, and precipitation, which in turn impact the reservoir inflow. Therefore, it is imperative for us to quantify the impact of aerosols on reservoir inflow and to incorporate the aerosol models into future reservoir inflow forecasting models. In this paper, a comprehensive framework was developed to quantify the impact of aerosols on reservoir inflow by integrating the Weather Research and Forecasting model with Chemistry (WRF-Chem) and a dynamic regression model. The statistical dynamic regression model produces forecasts for reservoir inflow based on the meteorological output variables from the WRF-Chem model. The case study was performed on the Florence Lake and Lake Thomas Alva Edison of the Big Creek Hydroelectric Project in the San Joaquin Region. The simulation results show that the presence of aerosols results in a significant reduction of annual reservoir inflow by 4-14%. In the summer, aerosols reduce precipitation, snow water equivalent, and snowmelt that leads to a reduction in inflow by 11-26%. In the spring, aerosols increase temperature and snowmelt which leads to an increase in inflow by 0.6-2%. Aerosols significantly reduce the amount of inflow in the summer when the marginal value of water is extremely high and slightly increase the inflow in the spring when the run-off risk is high. In summary, the presence of aerosols is detrimental to the optimal utilization of hydroelectric power systems.

13.
Earth Space Sci ; 4(8): 574-587, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-29104900

RESUMEN

This paper describes a forward radiative transfer model and retrieval system (FMRS) for the Tropospheric Water and cloud ICE (TWICE) CubeSat instrument. We use the FMRS to simulate radiances for the TWICE's 14 millimeter- and submillimeter-wavelength channels for a tropical atmospheric state produced by a Weather Research and Forecasting model simulation. We also perform simultaneous retrievals of cloud ice particle size, ice water content (IWC), water vapor content (H2O), and temperature from the simulated TWICE radiances using the FMRS. We show that the TWICE instrument is capable of retrieving ice particle size in the range of ~50-1000 µm in mass mean effective diameter with approximately 50% uncertainty. The uncertainties of other retrievals from TWICE are about 1 K for temperature, 50% for IWC, and 20% for H2O.

14.
Cancer Res ; 75(10): 2039-48, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25808868

RESUMEN

Temozolomide is used widely to treat malignant glioma, but the overall response to this agent is generally poor. Resistance to DNA-damaging drugs such as temozolomide has been related to the induction of antiapoptotic proteins. Specifically, the transcription factor NF-κB has been suggested to participate in promoting the survival of cells exposed to chemotherapy. To identify factors that modulate cytotoxicity in the setting of DNA damage, we used an unbiased strategy to examine the NF-κB-dependent expression profile induced by temozolomide. By this route, we defined the decoy receptor DcR1 as a temozolomide response gene induced by a mechanism relying upon p50/NF-κB1. A conserved NF-κB-binding sequence (κB-site) was identified in the proximal promoter and was demonstrated to be required for DcR1 induction by temozolomide. Loss-of-function and gain-of-function studies reveal that the atypical IκB protein, Bcl3, is also required for induction of DcR1 by temozolomide. Mechanistically, DcR1 attenuates temozolomide efficacy by blunting activation of the Fas receptor pathway in p53(+/+) glioma cells. Intracranial xenograft studies show that DcR1 depletion in glioma cells enhances the efficacy of temozolomide. Taken together, our results show how DcR1 upregulation mediates temozolomide resistance and provide a rationale for DcR1 targeting as a strategy to sensitize gliomas to this widely used chemotherapy.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Dacarbazina/análogos & derivados , Subunidad p50 de NF-kappa B/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/metabolismo , Receptores Señuelo del Factor de Necrosis Tumoral/genética , Animales , Proteínas del Linfoma 3 de Células B , Secuencia de Bases , Sitios de Unión , Línea Celular Tumoral , Dacarbazina/farmacología , Resistencia a Antineoplásicos , Proteínas Ligadas a GPI/química , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Glioma/tratamiento farmacológico , Glioma/metabolismo , Humanos , Masculino , Ratones Desnudos , Regiones Promotoras Genéticas , Unión Proteica , Miembro 10c de Receptores del Factor de Necrosis Tumoral , Temozolomida , Activación Transcripcional , Receptores Señuelo del Factor de Necrosis Tumoral/química , Receptores Señuelo del Factor de Necrosis Tumoral/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Nat Commun ; 5: 3972, 2014 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-24875621

RESUMEN

The pioneering factor FOXA1 opens chromatin to facilitate androgen receptor (AR) binding to prostate-specific genes. How FOXA1 controls the AR cistrome, however, is incompletely understood. Here we show that AR directly binds chromatin through the androgen response elements (AREs). FOXA1 is not required for AR-chromatin interaction, but instrumental in recruiting AR to low-affinity half-AREs by opening local chromatin around adjacent FKHD sites. Too much FOXA1 creates excessive open chromatin regions, which serve as reservoirs that retain AR via abundant half-AREs, thereby reducing its availability for specific sites. FOXA1 downregulation, by contrast, relinquishes AR to permissively bind AREs across the genome, resulting in substantial AR-binding events and AR target gene expression even in the absence of androgen. Taken together, our data illustrate the mechanistic details by which cooperativity and equilibrium with FOXA1 define AR cistrome and reveal a previously unknown function of FOXA1 in inhibiting AR signalling and castration-resistant prostate cancer growth.


Asunto(s)
Cromatina/metabolismo , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/genética , Receptores Androgénicos/metabolismo , Línea Celular Tumoral , Humanos , Masculino , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Transducción de Señal
16.
Mol Biol Rep ; 40(11): 6233-44, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24091942

RESUMEN

Retinoid-X-receptor (RXR) plays an essential role in the molting process of decapod crustaceans, by forming a heterodimeric complex with the ecdysteroid receptor. However, its role during female reproduction, especially in the process of ovarian maturation, has not been characterized. To get an insight into the molecular events governing the process of ovarian maturation in shrimps, the full-length cDNA of RXR from Metapenaeus ensis was cloned by extension of truncated cDNA by using the RACE technique. The open reading frame of MeRXR encodes a 410 amino acid protein with a deduced molecular weight of 44.8 kDa, and putative pI of 6.64, which roughly matched our observation from 2DE gel. Phylogenetic analysis showed that MeRXR has high similarity to RXR of Penaeus chinensis and P. japonicus. RT-PCR revealed that MeRXR was universally expressed in all tissues investigated. The variation in MeRXR mRNA expression pattern during ovarian maturation was further analyzed by real-time PCR. In contrast to the decrease in MeRXR at protein level with ovarian maturation, MeRXR mRNA level was low in pre-vitellogenic and mid-vitellogenic ovaries, and increased significantly from mid-vitellogenic to late-vitellogenic stages. This result suggests that MeRXR transcripts in the mature ovary probably act as maternal messages for regulating early molting events during embryonic development.


Asunto(s)
Clonación Molecular , Expresión Génica , Ovario/metabolismo , Penaeidae/genética , Penaeidae/metabolismo , Receptores X Retinoide/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , ADN Complementario/química , ADN Complementario/genética , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Datos de Secuencia Molecular , Especificidad de Órganos/genética , Ovario/efectos de los fármacos , Filogenia , Receptores X Retinoide/metabolismo , Retinoides/farmacología , Alineación de Secuencia , Transcriptoma
17.
Cancer Res ; 73(19): 6068-79, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23913826

RESUMEN

Chromosomal translocations juxtaposing the androgen-responsive TMPRSS2 promoter with the ETS-family transcription factor ERG result in aberrant ERG upregulation in approximately 50% of prostate cancers. Studies to date have shown important roles of ERG in inducing oncogenic properties of prostate cancer. Its molecular mechanisms of action, however, are yet to be fully understood. Here, we report that ERG activates Wnt/LEF1 signaling cascade through multiple mechanisms. ERG bound to the promoters of various Wnt genes to directly increase ligand expression. Consequently, ERG overexpression increased active ß-catenin level in the cells and enhanced TCF/LEF1 luciferase reporter activity, which could be partially blocked by WNT-3A inhibitor IWP-2. Most importantly, our data defined LEF1 as a direct target of ERG and that LEF1 inhibition fully abolished ERG-induced Wnt signaling and target gene expression. Furthermore, functional assays showed that Wnt/LEF1 activation phenocopied that of ERG in inducing cell growth, epithelial-to-mesenchymal transition, and cell invasion, whereas blockade of Wnt signaling attenuated these effects. Concordantly, LEF1 expression is significantly upregulated in ERG-high human prostate cancers. Overall, this study provides an important mechanism of activation of Wnt signaling in prostate cancer and nominates LEF1 as a critical mediator of ERG-induced tumorigenesis. Wnt/LEF1 pathway might provide novel targets for therapeutic management of patients with fusion-positive prostate cancer.


Asunto(s)
Biomarcadores de Tumor/genética , Regulación Neoplásica de la Expresión Génica , Factor de Unión 1 al Potenciador Linfoide/genética , Neoplasias de la Próstata/metabolismo , Transducción de Señal , Transactivadores/metabolismo , Proteína Wnt1/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Inmunoprecipitación de Cromatina , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Humanos , Técnicas para Inmunoenzimas , Factor de Unión 1 al Potenciador Linfoide/antagonistas & inhibidores , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Masculino , Invasividad Neoplásica , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transactivadores/antagonistas & inhibidores , Transactivadores/genética , Activación Transcripcional , Regulador Transcripcional ERG , Proteína Wnt1/antagonistas & inhibidores , Proteína Wnt1/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
18.
Genome Res ; 22(2): 322-31, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22179855

RESUMEN

Androgen receptor (AR) is a hormone-activated transcription factor that plays important roles in prostate development and function, as well as malignant transformation. The downstream pathways of AR, however, are incompletely understood. AR has been primarily known as a transcriptional activator inducing prostate-specific gene expression. Through integrative analysis of genome-wide AR occupancy and androgen-regulated gene expression, here we report AR as a globally acting transcriptional repressor. This repression is mediated by androgen-responsive elements (ARE) and dictated by Polycomb group protein EZH2 and repressive chromatin remodeling. In embryonic stem cells, AR-repressed genes are occupied by EZH2 and harbor bivalent H3K4me3 and H3K27me3 modifications that are characteristic of differentiation regulators, the silencing of which maintains the undifferentiated state. Concordantly, these genes are silenced in castration-resistant prostate cancer rendering a stem cell-like lack of differentiation and tumor progression. Collectively, our data reveal an unexpected role of AR as a transcriptional repressor inhibiting non-prostatic differentiation and, upon excessive signaling, resulting in cancerous dedifferentiation.


Asunto(s)
Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Receptores Androgénicos/metabolismo , Proteínas Represoras/metabolismo , Secuencia de Bases , Línea Celular Tumoral , Análisis por Conglomerados , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/metabolismo , Proteína Potenciadora del Homólogo Zeste 2 , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Motivos de Nucleótidos , Orquiectomía , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Unión Proteica , Elementos de Respuesta , Transducción de Señal , Factores de Transcripción/metabolismo , Activación Transcripcional
19.
Cancer Biol Ther ; 10(6): 617-24, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20603606

RESUMEN

WNT5A is classified as a non-transforming WNT family member whose role in carcinogenesis is still ambiguous. It exhibits tumor suppressor activities in some cancers (thyroid, brain, breast and colorectum), but is aberrantly upregulated in cancers of lung, stomach and prostate. We investigated its epigenetic alterations and functions in esophageal squamous cell carcinoma (ESCC). With semi-quantitative reverse transcription PCR, we found that WNT5A was silenced or downregulated in 5 of 18 ESCC cell lines, but expressed in normal esophagus tissue and immortalized normal esophageal epithelial cell lines. Promoter CpG methylation of WNT5A was detected in 4 of the 5 downregulated ESCC cell lines, while 5-aza-2'-deoxycytidine treatment induced WNT5A expression and demethylated its promoter in silenced cell lines. WNT5A promoter methylation was frequently detected in primary ESCC (24/36, 67%), but less frequently and weakly in paired surgical marginal esophageal tissues (8/36, 22%; p < 0.01), while no methylation was detected in seven normal esophageal epithelial tissues from healthy donors. Ectopic expression of WNT5A resulted in significant inhibition of clonogenicity and motility of ESCC cells, accompanied by a dramatic decrease of intracellular ß-catenin protein level and ß-catenin transcriptional activity. In summary, we show that WNT5A is frequently silenced in ESCC by promoter methylation and exhibits tumor suppressor properties through antagonizing the WNT/ß-catenin pathway. The epigenetic disruption of WNT5A would thus contribute directly to the aberrant activation of WNT/ß-catenin signaling during ESCC pathogenesis.


Asunto(s)
Metilación de ADN , Proteínas Proto-Oncogénicas/genética , Transducción de Señal/genética , Proteínas Wnt/genética , beta Catenina/genética , Western Blotting , Línea Celular Transformada , Línea Celular Tumoral , Hibridación Genómica Comparativa , Islas de CpG/genética , Regulación hacia Abajo/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Wnt/metabolismo , Proteína Wnt-5a , beta Catenina/metabolismo
20.
Cancer Cell ; 17(5): 443-54, 2010 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-20478527

RESUMEN

Chromosomal rearrangements fusing the androgen-regulated gene TMPRSS2 to the oncogenic ETS transcription factor ERG occur in approximately 50% of prostate cancers, but how the fusion products regulate prostate cancer remains unclear. Using chromatin immunoprecipitation coupled with massively parallel sequencing, we found that ERG disrupts androgen receptor (AR) signaling by inhibiting AR expression, binding to and inhibiting AR activity at gene-specific loci, and inducing repressive epigenetic programs via direct activation of the H3K27 methyltransferase EZH2, a Polycomb group protein. These findings provide a working model in which TMPRSS2-ERG plays a critical role in cancer progression by disrupting lineage-specific differentiation of the prostate and potentiating the EZH2-mediated dedifferentiation program.


Asunto(s)
Proteínas de Unión al ADN/genética , Fusión Génica , Proteínas de Fusión Oncogénica/genética , Neoplasias de la Próstata/patología , Receptores Androgénicos/genética , Factores de Transcripción/genética , Inmunoprecipitación de Cromatina , Progresión de la Enfermedad , Proteína Potenciadora del Homólogo Zeste 2 , Humanos , Masculino , Complejo Represivo Polycomb 2 , Neoplasias de la Próstata/genética , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...