Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.665
Filtrar
1.
JCI Insight ; 7(4)2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35050902

RESUMEN

BACKGROUNDProstate cancer is multifocal with distinct molecular subtypes. The utility of genomic subtyping has been challenged due to inter- and intrafocal heterogeneity. We sought to characterize the subtype-defining molecular alterations of primary prostate cancer across all tumor foci within radical prostatectomy (RP) specimens and determine the prevalence of collision tumors.METHODSFrom the Early Detection Research Network cohort, we identified 333 prospectively collected RPs from 2010 to 2014 and assessed ETS-related gene (ERG), serine peptidase inhibitor Kazal type 1 (SPINK1), phosphatase and tensin homolog (PTEN), and speckle type BTB/POZ protein (SPOP) molecular status. We utilized dual ERG/SPINK1 immunohistochemistry and fluorescence in situ hybridization to confirm ERG rearrangements and characterize PTEN deletion, as well as high-resolution melting curve analysis and Sanger sequencing to determine SPOP mutation status.RESULTSBased on index focus alone, ERG, SPINK1, PTEN, and SPOP alterations were identified in 47.5%, 10.8%, 14.3%, and 5.1% of RP specimens, respectively. In 233 multifocal RPs with ERG/SPINK1 status in all foci, 139 (59.7%) had discordant molecular alterations between foci. Collision tumors, as defined by discrepant ERG/SPINK1 status within a single focus, were identified in 29 (9.4%) RP specimens.CONCLUSIONInterfocal molecular heterogeneity was identified in about 60% of multifocal RP specimens, and collision tumors were present in about 10%. We present this phenomenon as a model for the intrafocal heterogeneity observed in previous studies and propose that future genomic studies screen for collision tumors to better characterize molecular heterogeneity.FUNDINGEarly Detection Research Network US National Cancer Institute (NCI) 5U01 CA111275-09, Center for Translational Pathology at Weill Cornell Medicine (WCM) Department of Pathology and Laboratory Medicine, US NCI (WCM SPORE in Prostate Cancer, P50CA211024-01), R37CA215040, Damon Runyon Cancer Research Foundation, US MetLife Foundation Family Clinical Investigator Award, Norwegian Cancer Society (grant 208197), and South-Eastern Norway Regional Health Authority (grant 2019016 and 2020063).


Asunto(s)
Mutación , Proteínas Nucleares/genética , Fosfohidrolasa PTEN/genética , Neoplasias de la Próstata/genética , ARN Neoplásico/genética , Proteínas Represoras/genética , Inhibidor de Tripsina Pancreática de Kazal/genética , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Análisis Mutacional de ADN , Reordenamiento Génico , Humanos , Inmunohistoquímica , Masculino , Proteínas Nucleares/biosíntesis , Fosfohidrolasa PTEN/biosíntesis , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas Represoras/biosíntesis , Estudios Retrospectivos , Inhibidor de Tripsina Pancreática de Kazal/biosíntesis , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor
2.
Eur J Histochem ; 65(4)2021 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-34783233

RESUMEN

The long non-coding FGD5-AS1 (LncFGD5-AS1) has been reported to be a novel carcinogenic gene and participant in regulating tumor progression by sponging microRNAs (miRNAs). However, the pattern of expression and the biological role of FGD5-AS1 in hepatocellular carcinoma (HCC) remains largely unknown. The expression level of FGD5-AS1 in tumor tissues and cell lines was measured by RT-qPCR. CCK-8, EdU, flow cytometry, wound healing, and transwell chamber assays were performed to investigate the role of FGD5-AS1 in cell proliferation, apoptosis, migration, and invasion in HCC. Dual luciferase reporter, and RNA pull-down assays were performed to identify the regulatory interactions among FGD5-AS1, miR-873-5p and GTP-binding protein 4 (GTPBP4). We found that the expression of FGD5-AS1 was upregulated in HCC tissues and cell lines. Moreover, the knockdown of FGD5-AS1 suppressed cell proliferation, migration and invasion, and induced apoptosis in HCC cells. Further studies demonstrated that FGD5-AS1 could function as a competitive RNA by sponging miR-873-5p in HCC cells. Moreover, GTPBP4 was identified as direct downstream target of miR-873-5p in HCC cells and FGD5-AS1mediated the effects of GTPBP4 by competitively binding with miR-873-5p. Taken together, this study demonstrated the regulatory role of FGD5-AS1 in the progression of HCC and identified the miR-873-5p/GTPBP4 axis as the direct downstream pathway. It represents a promising novel therapeutic strategy for HCC patients.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Proteínas de Unión al GTP/biosíntesis , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/metabolismo , MicroARNs/biosíntesis , Proteínas de Neoplasias/biosíntesis , Proteínas Nucleares/biosíntesis , Oncogenes , ARN Largo no Codificante/metabolismo , ARN Neoplásico/metabolismo , Regulación hacia Arriba , Carcinoma Hepatocelular/genética , Proteínas de Unión al GTP/genética , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , MicroARNs/genética , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , ARN Largo no Codificante/genética , ARN Neoplásico/genética
3.
Genome Biol ; 22(1): 284, 2021 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-34615539

RESUMEN

BACKGROUND: Regulation of protein output at the level of translation allows for a rapid adaptation to dynamic changes to the cell's requirements. This precise control of gene expression is achieved by complex and interlinked biochemical processes that modulate both the protein synthesis rate and stability of each individual mRNA. A major factor coordinating this regulation is the Ccr4-Not complex. Despite playing a role in most stages of the mRNA life cycle, no attempt has been made to take a global integrated view of how the Ccr4-Not complex affects gene expression. RESULTS: This study has taken a comprehensive approach to investigate post-transcriptional regulation mediated by the Ccr4-Not complex assessing steady-state mRNA levels, ribosome position, mRNA stability, and protein production transcriptome-wide. Depletion of the scaffold protein CNOT1 results in a global upregulation of mRNA stability and the preferential stabilization of mRNAs enriched for G/C-ending codons. We also uncover that mRNAs targeted to the ER for their translation have reduced translational efficiency when CNOT1 is depleted, specifically downstream of the signal sequence cleavage site. In contrast, translationally upregulated mRNAs are normally localized in p-bodies, contain disorder-promoting amino acids, and encode nuclear localized proteins. Finally, we identify ribosome pause sites that are resolved or induced by the depletion of CNOT1. CONCLUSIONS: We define the key mRNA features that determine how the human Ccr4-Not complex differentially regulates mRNA fate and protein synthesis through a mechanism linked to codon composition, amino acid usage, and mRNA localization.


Asunto(s)
Regulación de la Expresión Génica , Biosíntesis de Proteínas , Estabilidad del ARN , ARN Mensajero/metabolismo , Factores de Transcripción/fisiología , Codón , Técnicas de Silenciamiento del Gen , Humanos , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Ribosomas/metabolismo , Factores de Transcripción/genética
4.
Bioengineered ; 12(2): 9452-9462, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34696677

RESUMEN

LYAR (Ly-1 antibody reactive) is a transcription factor with a specific DNA-binding domain, which plays a key role in the regulation of embryonic stem cell self-renewal and differentiation. However, the role of LYAR in human cancers remains unclear. This study aimed to analyze the prognostic value of LYAR in cancer. In this study, we evaluated the prognostic value of LYAR in various tumors. We research found that, compared with normal tissues, LYAR levels werehigher in a variety of tumors. LYAR expression level was associated with poor overall survival, progression-free interval, and disease-specific survival. LYAR expression was also related to tumor grade, stage, age, and tumor status. Cell counting kit-8, Transwell, and wound healing assay showed that knocking out LYAR significantly inhibited the proliferation, migration, and invasion of hepatocellular carcinoma cells. In addition, this study found that LYARexpression was significantly positively correlated with MKI67IP, BZW2, and CCT2. Gene set enrichment analysis results showed that samples with high LYAR expression levels were rich in spliceosomes, RNA degradation, pyrimidine metabolism, cell cycle, nucleotide excision repair, and base excision repair.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Movimiento Celular , Proliferación Celular , Proteínas de Unión al ADN/biosíntesis , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/metabolismo , Proteínas de Neoplasias/biosíntesis , Proteínas Nucleares/biosíntesis , Carcinoma Hepatocelular/genética , Proteínas de Unión al ADN/genética , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética
5.
Theranostics ; 11(16): 7970-7983, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34335974

RESUMEN

The novel ß-coronavirus, SARS-CoV-2, the causative agent of coronavirus disease 2019 (COVID-19), has infected more than 177 million people and resulted in 3.84 million death worldwide. Recent epidemiological studies suggested that some environmental factors, such as air pollution, might be the important contributors to the mortality of COVID-19. However, how environmental exposure enhances the severity of COVID-19 remains to be fully understood. In the present report, we provided evidence showing that mdig, a previously reported environmentally-induced oncogene that antagonizes repressive trimethylation of histone proteins, is an important regulator for SARS-CoV-2 receptors neuropilin-1 (NRP1) and NRP2, cathepsins, glycan metabolism and inflammation, key determinants for viral infection and cytokine storm of the patients. Depletion of mdig in bronchial epithelial cells by CRISPR-Cas-9 gene editing resulted in a decreased expression of NRP1, NRP2, cathepsins, and genes involved in protein glycosylation and inflammation, largely due to a substantial enrichment of lysine 9 and/or lysine 27 trimethylation of histone H3 (H3K9me3/H3K27me3) on these genes as determined by ChIP-seq. Meanwhile, we also validated that environmental factor arsenic is able to induce mdig, NRP1 and NRP2, and genetic disruption of mdig lowered expression of NRP1 and NRP2. Furthermore, mdig may coordinate with the Neanderthal variants linked to an elevated mortality of COVID-19. These data, thus, suggest that mdig is a key mediator for the severity of COVID-19 in response to environmental exposure and targeting mdig may be the one of the effective strategies in ameliorating the symptom and reducing the mortality of COVID-19.


Asunto(s)
COVID-19/metabolismo , COVID-19/virología , Dioxigenasas/metabolismo , Histona Demetilasas/metabolismo , Neuropilina-1/metabolismo , Proteínas Nucleares/metabolismo , Polisacáridos/metabolismo , SARS-CoV-2/metabolismo , Células Epiteliales Alveolares/metabolismo , Animales , COVID-19/epidemiología , Catepsinas/metabolismo , Línea Celular , Células Cultivadas , Dioxigenasas/biosíntesis , Dioxigenasas/genética , Exposición a Riesgos Ambientales , Histona Demetilasas/biosíntesis , Histona Demetilasas/genética , Histonas/metabolismo , Humanos , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Pandemias , Ratas , SARS-CoV-2/patogenicidad , Glicoproteína de la Espiga del Coronavirus/metabolismo
6.
Mol Neurobiol ; 58(11): 6049-6061, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34435332

RESUMEN

Diabetic peripheral neuropathy (DPN) is a chronic complication of diabetes, and its neural mechanisms underlying the pathogenesis remain unclear. Autophagy plays an important role in neurodegenerative diseases and nerve tissue injury. Lipin1 is a phosphatidic acid phosphatase enzyme that converts phosphatidic acid (PA) into diacylglycerol (DAG), a precursor of triacylglycerol and phospholipids which plays an important role in maintaining normal peripheral nerve conduction function. However, whether Lipin1 involved in the pathogenesis of DPN via regulation of autophagy is not elucidated. Here, we show that the Lipin1 expression was downregulated in streptozotocin (STZ)-induced DPN rat model. Interestingly, STZ prevented DAG synthesis, and resulted in autophagic hyperactivity, effects which may increase the apoptosis of Schwann cells and lead to demyelination in sciatic nerve in DPN rats. More importantly, upregulation of lipin1 in the DPN rats ameliorated autophagy disorders and pathological changes of the sciatic nerve, which associated with the increase of the motor nerve conductive velocity (MNCV) in DPN rats. In contrast, knockdown of lipin1 exacerbates neuronal abnormalities and facilitates the genesis of DPN phenotypes in rats. In addition, overexpression of lipin1 in RSC96 cells also significantly decreased the autophagic hyperactivity and apoptosis induced by hyperglycemia. These results suggest that lipin1 may exert neuroprotection within the sciatic nerve anomalies and may serve as a potential therapeutic target for the treatment of DPN.


Asunto(s)
Autofagia/fisiología , Enfermedades Desmielinizantes/fisiopatología , Diabetes Mellitus Experimental/complicaciones , Neuropatías Diabéticas/fisiopatología , Degeneración Nerviosa/fisiopatología , Proteínas Nucleares/fisiología , Nervio Ciático/fisiopatología , Animales , Apoptosis , Células Cultivadas , Enfermedades Desmielinizantes/etiología , Enfermedades Desmielinizantes/terapia , Diabetes Mellitus Experimental/sangre , Diglicéridos/biosíntesis , Regulación hacia Abajo , Técnicas de Silenciamiento del Gen , Vectores Genéticos/uso terapéutico , Hiperalgesia/etiología , Hiperalgesia/terapia , Hiperglucemia/etiología , Hiperglucemia/metabolismo , Masculino , Degeneración Nerviosa/etiología , Conducción Nerviosa , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Proteínas Nucleares/uso terapéutico , Ratas , Ratas Wistar , Proteínas Recombinantes/metabolismo , Células de Schwann/metabolismo
7.
Hum Pathol ; 116: 12-21, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34271067

RESUMEN

SMARCA4-deficient uterine sarcoma (SMARCA4-DUS) was recently proposed as a new entity of uterine sarcoma. Reported cases of SMARCA4-DUS showed the loss of SMARCA4 and SMARCA2 expression. However, the prevalence of their deficiency in uterine mesenchymal tumors remains unclear. This study immunohistochemically examined the expression of SMARCA4, SMARCA2, and SMARCB1 in 206 uterine mesenchymal tumors and detected a round cell tumor with the loss of SMARCA4 and SMARCA2 and a low-grade endometrial stromal sarcoma with SMARCA4 deficiency. The remaining 204 cases, including 170 smooth muscle tumors, 22 endometrial stomal nodule/sarcomas, seven undifferentiated uterine sarcomas, two adenosarcomas, one uterine tumor resembling ovarian sex cord tumor, and two perivascular epithelioid cell tumors, retained the expression of both SMARCA4 and SMARCA2. All tumors retained SMARCB1 expression. The round cell tumor with the loss of SMARCA4 and SMARCA2 was composed of diffuse small round cell growth with follicle-like spaces, which resembled small cell carcinoma of the ovary, hypercalcemic type. Immunohistochemically, the tumor showed the proficient expression of mismatch repair proteins and wild-type p53 expression, which favored SMARCA4-DUS; however, the tumor harbored the PIK3CA mutation, and thus, was reclassified as undifferentiated endometrial carcinoma. In conclusion, SMARCA4, SMARCA2, and SMARCB1 were rarely deficient in uterine mesenchymal tumors. SMARCA4 immunohistochemistry has potential in the diagnosis of SMARCA4-DUS with the exclusion of some tumors showing its deficiency, such as endometrial stromal sarcoma and undifferentiated carcinoma. Undifferentiated carcinoma may show an indistinguishable morphology and immunophenotype from SMARCA4-DUS, and thus, molecular analysis is required for their distinction in diagnostic practice.


Asunto(s)
ADN Helicasas/biosíntesis , Proteínas Nucleares/biosíntesis , Proteína SMARCB1/biosíntesis , Sarcoma/diagnóstico , Factores de Transcripción/biosíntesis , Neoplasias Uterinas/diagnóstico , Biomarcadores de Tumor/análisis , Carcinoma/diagnóstico , Diagnóstico Diferencial , Femenino , Humanos , Persona de Mediana Edad , Sarcoma/patología , Neoplasias Uterinas/patología
8.
Biomolecules ; 11(6)2021 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-34063883

RESUMEN

Obesity and type 2 diabetes mellitus (T2DM) are often combined and pathologically affect many tissues due to changes in circulating bioactive molecules. In this work, we evaluated the effect of blood plasma from obese (OB) patients or from obese patients comorbid with diabetes (OBD) on skeletal muscle function and metabolic state. We employed the mouse myoblasts C2C12 differentiation model to test the regulatory effect of plasma exposure at several levels: (1) cell morphology; (2) functional activity of mitochondria; (3) expression levels of several mitochondria regulators, i.e., Atgl, Pgc1b, and miR-378a-3p. Existing databases were used to computationally predict and analyze mir-378a-3p potential targets. We show that short-term exposure to OB or OBD patients' plasma is sufficient to affect C2C12 properties. In fact, the expression of genes that regulate skeletal muscle differentiation and growth was downregulated in both OB- and OBD-treated cells, maximal mitochondrial respiration rate was downregulated in the OBD group, while in the OB group, a metabolic switch to glycolysis was detected. These alterations correlated with a decrease in ATGL and Pgc1b expression in the OB group and with an increase of miR-378a-3p levels in the OBD group.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Diabetes Mellitus/sangre , Metabolismo Energético/efectos de los fármacos , MicroARNs/biosíntesis , Mitocondrias Musculares/metabolismo , Mioblastos Esqueléticos/metabolismo , Obesidad/sangre , Plasma , Adulto , Anciano , Animales , Línea Celular , Femenino , Humanos , Lipasa/biosíntesis , Masculino , Ratones , Persona de Mediana Edad , Proteínas Nucleares/biosíntesis , Factores de Transcripción/biosíntesis
9.
Theranostics ; 11(13): 6592-6606, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33995678

RESUMEN

Purpose: Clinical success of cancer therapy is severely limited by drug resistance, attributed in large part to the loss of function of tumor suppressor genes (TSGs). Developing effective strategies to treat those tumors is challenging, but urgently needed in clinic. Experimental Design: MYOCD is a clinically relevant TSG in lung cancer patients. Our in vitro and in vivo data confirm its tumor suppressive function. Further analysis reveals that MYOCD potently inhibits stemness of lung cancer stem cells. Mechanistically, MYOCD localizes to TGFBR2 promoter region and thereby recruits PRMT5/MEP50 complex to epigenetically silence its transcription. Conclusions: NSCLC cells deficient of MYOCD are particularly sensitive to TGFBR kinase inhibitor (TGFBRi). TGFBRi and stemness inhibitor synergize with existing drugs to treat MYOCD deficient lung cancers. Our current work shows that loss of function of MYOCD creates Achilles' heels in lung cancer cells, which might be exploited in clinic.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas Nucleares/deficiencia , Receptor Tipo II de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Transactivadores/deficiencia , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/genética , Regulación hacia Abajo , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Código de Histonas , Humanos , Neoplasias Pulmonares/genética , Metilación , Ratones Transgénicos , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Células Madre Neoplásicas/patología , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Regiones Promotoras Genéticas , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Procesamiento Proteico-Postraduccional , Proteína-Arginina N-Metiltransferasas/fisiología , Receptor Tipo II de Factor de Crecimiento Transformador beta/genética , Transducción de Señal , Transactivadores/biosíntesis , Transactivadores/genética , Transactivadores/fisiología , Carga Tumoral
10.
Inflammation ; 44(5): 2044-2053, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34041646

RESUMEN

Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is an independent risk factor for mortality in patients with sepsis. In this study, we attempt to investigate the molecular mechanism of circANKRD36 underlying sepsis-induced ALI/ARDS in vitro. We first detected the altered circRNAs in serums of patients with sepsis-induced ARDS using circRNAs microarray. CircANKRD36 expression in serums and LPS-stimulated RAW264.7 cells was measured using qRT-PCR. CCK-8, cell migration, ELISA, and qRT-PCR were applied to the evaluation of cell biological behavior and inflammation reaction. The results showed that circANKRD36 expression was significantly elevated in serum of patients with sepsis-induced ARDS. Knockdown of circANKRD36 inhibited cell viability and migration and alleviated inflammation of lipopolysaccharide-stimulated (LPS-stimulated) RAW264.7 cells. Bioinformatic analysis demonstrated that circANKRD36 serves as a sponge for miR-330 and ROCK1 was directly targeted by miR-330. Furthermore, knockdown of circANKRD36 repressed ROCK1 expression by targeting miR-330. In short, circANKRD36 knockdown suppressed cell viability and migration of LPS-stimulated RAW264.7 cells in vitro via sponging miR-330, which may provide new ideas for the treatment of sepsis-induced ARDS.


Asunto(s)
Movimiento Celular/fisiología , Técnicas de Silenciamiento del Gen/métodos , Lipopolisacáridos/toxicidad , MicroARNs/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Animales , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , MicroARNs/genética , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Células RAW 264.7 , Síndrome de Dificultad Respiratoria/genética , Síndrome de Dificultad Respiratoria/metabolismo
11.
Biochem Biophys Res Commun ; 557: 85-89, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-33862464

RESUMEN

N6-methyladenosine (m6A) mRNA modification has been defined as a crucial regulator in various biological processes. Recent studies indicated an essential role of YTHDF1, an m6A reader, in the maintenance of intestinal stem cells (ISCs), while the detailed mechanism remains to be explored. By searching our m6A sequencing, RNA sequencing, and ribosome profiling data, we identified the transcriptional enhanced associate domain 1 (TEAD1) as a direct target of YTHDF1. We confirmed the presence of m6A modifications in TEAD1 mRNA and its binding with YTHDF1. Knockdown of either m6A methyltransferase METTL3 or YTHDF1 reduced the translation of TEAD1. TEAD1 was highly expressed in ISCs, while depletion of TEAD1 inhibited proliferation and induced differentiation of organoids. Overexpression of TEAD1 reversed the impaired stemness elicited by YTHDF1 depletion. These findings identify TEAD1 as a functional target of m6A-YTHDF1 in sustaining intestinal stemness.


Asunto(s)
Proteínas de Unión al ADN/biosíntesis , Intestinos/citología , Proteínas Nucleares/biosíntesis , Proteínas de Unión al ARN/metabolismo , Células Madre/citología , Células Madre/metabolismo , Factores de Transcripción/biosíntesis , Adenosina/análogos & derivados , Adenosina/genética , Adenosina/metabolismo , Animales , Línea Celular , Proliferación Celular/fisiología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células HCT116 , Humanos , Intestinos/fisiología , Metilación , Ratones , Ratones Noqueados , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Organoides , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Factores de Transcripción de Dominio TEA , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
12.
Cell Calcium ; 96: 102369, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33677175

RESUMEN

Vascular smooth muscle cells are unusual in that differentiated, contractile cells possess the capacity to "de-differentiate" into a synthetic phenotype that is characterized by being replicative, secretory, and migratory. One aspect of this phenotypic modulation is a shift from voltage-gated Ca2+ signalling in electrically coupled, differentiated cells to increased dependence on store-operated Ca2+ entry and sarcoplasmic reticulum Ca2+ release in synthetic cells. Conversely, an increased voltage-gated Ca2+ entry is seen when proliferating A7r5 smooth muscle cells quiesce. We asked whether this change in Ca2+ signalling was linked to changes in the expression of the phenotype-regulating transcriptional co-activator myocardin or α-smooth muscle actin, using correlative epifluorescence Ca2+ imaging and immunocytochemistry. Cells were cultured in growth media (DMEM, 10% serum, 25 mM glucose) or differentiation media (DMEM, 1% serum, 5 mM glucose). Coinciding with growth arrest, A7r5 cells became electrically coupled, and spontaneous Ca2+ signalling showed increasing dependence on L-type voltage-gated Ca2+ channels that were blocked with nifedipine (5 µM). These synchronized oscillations were modulated by ryanodine receptors, based on their sensitivity to dantrolene (5 µM). Actively growing cultures had spontaneous Ca2+ transients that were insensitive to nifedipine and dantrolene but were blocked by inhibition of the sarco-endoplasmic reticulum ATPase with cyclopiazonic acid (10 µM). In cells treated with differentiation media, myocardin and αSMA immunoreactivity increased prior to changes in the Ca2+ signalling phenotype, while chronic inhibition of voltage-gated Ca2+ entry modestly increased immunoreactivity of myocardin. Stepwise regression analyses suggested that changes in myocardin expression had a weak relationship with Ca2+ signalling synchronicity, but not frequency or amplitude. In conclusion, we report a 96-well assay and analytical pipeline to study the link between Ca2+ signalling and smooth muscle differentiation. This assay showed that changes in the expression of two molecular differentiation markers (myocardin and αSMA) tended to precede changes in the Ca2+ signalling phenotype.


Asunto(s)
Aorta/metabolismo , Señalización del Calcio/fisiología , Diferenciación Celular/fisiología , Proteínas Nucleares/biosíntesis , Fenotipo , Transactivadores/biosíntesis , Animales , Aorta/efectos de los fármacos , Bloqueadores de los Canales de Calcio/farmacología , Señalización del Calcio/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular , Dantroleno/farmacología , Expresión Génica , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Proteínas Nucleares/genética , Ratas , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Transactivadores/genética
13.
EMBO J ; 40(6): e106336, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33595114

RESUMEN

About 10% of cancer cells employ the "alternative lengthening of telomeres" (ALT) pathway instead of re-activating the hTERT subunit of human telomerase. The hTR RNA subunit is also abnormally silenced in some ALT+ cells not expressing hTERT, suggesting a possible negative non-canonical impact of hTR on ALT. Indeed, we show that ectopically expressed hTR reduces phosphorylation of ssDNA-binding protein RPA (p-RPAS33 ) at ALT telomeres by promoting the hnRNPA1- and DNA-PK-dependent depletion of RPA. The resulting defective ATR checkpoint signaling at telomeres impairs recruitment of the homologous recombination protein, RAD51. This induces ALT telomere fragility, increases POLD3-dependent C-circle production, and promotes the recruitment of the DNA damage marker 53BP1. In ALT+ cells that naturally retain hTR expression, NHP2 H/ACA ribonucleoprotein levels are downregulated, likely in order to restrain DNA damage response (DDR) activation at telomeres through reduced 53BP1 recruitment. This unexpected role of NHP2 is independent from hTR's non-canonical function in modulating telomeric p-RPAS33 . Collectively, our study shines new light on the interference between telomerase- and ALT-dependent pathways and unravels a crucial role for hTR and NHP2 in DDR regulation at ALT telomeres.


Asunto(s)
Proteínas Nucleares/biosíntesis , ARN/genética , Ribonucleoproteínas Nucleares Pequeñas/biosíntesis , Telomerasa/genética , Homeostasis del Telómero/fisiología , Telómero/genética , Daño del ADN/genética , Reparación del ADN/genética , Proteína Quinasa Activada por ADN/metabolismo , Regulación hacia Abajo , Ribonucleoproteína Nuclear Heterogénea A1/metabolismo , Humanos , Neoplasias/genética , Recombinasa Rad51/metabolismo
14.
J Biochem Mol Toxicol ; 35(4): e22706, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33443779

RESUMEN

Fumonisin B1 (FB1 ) is a common environmental mycotoxin produced by molds such as Fusarium verticillioides. The toxin poses health risks to domestic animals, including pigs, through FB1 -contaminanted feed. However, the cytotoxicity of FB1 to porcine intestines has not been fully analyzed. In the present study, the effects of FB1 on oxidative stress and nutrient transporter-associated genes of the porcine intestinal IPEC-J2 cells were explored. FB1 decreased IPEC-J2 proliferation but did not trigger reactive oxygen species (ROS) overproduction. Meanwhile, FB1 reduced the expression levels of the transporters l-type amino acid transporter-1 (y+ LAT1), solute carrier family 7 member 1 (SLC7A1), solute carrier family 1 member 5 (ASCT2), and excitatory amino acid carrier 1 (EAAC1); in addition, FB1 reduced the levels of the fatty acid transporters long-chain fatty acid transport protein 1 (FATP1) and long-chain fatty acid transport protein 4 (FATP4) as well as glucose transporters Na+ /glucose cotransporter 1 (SGLT1) and glucose transporter 2 (GLUT2). FB1 stimulation increased the expression levels of peptide transporter peptide transporter 1 (PepT1) and metal ion transport-related gene zinc transporter 1 (ZNT1). Moreover, metal ion transporter divalent metal transporter 1 (DMT1) expression was depressed by a higher dosage of FB1 . The data indicate that FB1 results in aberrant expression of nutrient transporters in IPEC-J2 cells, thereby exerting its toxicity even though it fails to exert ROS-dependent oxidative stress.


Asunto(s)
Proteínas Portadoras/biosíntesis , Fumonisinas/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Mucosa Intestinal/metabolismo , Proteínas Nucleares/biosíntesis , Estrés Oxidativo/efectos de los fármacos , Animales , Línea Celular , Fumonisinas/química , Fusarium/química , Mucosa Intestinal/patología , Porcinos
15.
Arch Virol ; 166(3): 755-766, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33420627

RESUMEN

MicroRNAs (miRNAs) are important host molecules involved in human immunodeficiency virus type 1 (HIV-1) infection. Antiretroviral therapy (ART) can affect the miRNA expression profile, but differentially expressed miRNAs still remain to be identified. In this study, we used gene chips to analyze miRNA expression profiles in peripheral blood mononuclear cells from ART-naive HIV-1 patients and those receiving ART, as well as from uninfected individuals. We measured differences in miRNA expression by quantitative polymerase chain reaction (qPCR) in an expanded sample. We found significant differences in the expression of has-miR-191-5p among the three groups (P < 0.05). Furthermore, we showed that hsa-miR-191-5p has an inhibitory effect on HIV-1 replication in cell models in vitro. We identified CCR1 and NUP50 as target molecules of hsa-miR-191-5p and found that hsa-miR-191-5p inhibits the expression of CCR1 and NUP50. Knockdown of NUP50 resulted in significant inhibition of HIV-1 replication. In summary, our research shows that hsa-miR-191-5p expression is reduced in HIV-1-infected patients and acts an inhibitor of HIV-1 infection via a mechanism that may involve targeted repression of NUP50 expression.


Asunto(s)
Regulación de la Expresión Génica/genética , VIH-1/metabolismo , MicroARNs/genética , Proteínas de Complejo Poro Nuclear/biosíntesis , Proteínas Nucleares/biosíntesis , Receptores CCR1/biosíntesis , Adulto , Línea Celular , Regulación hacia Abajo , Femenino , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Células Jurkat , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Reacción en Cadena en Tiempo Real de la Polimerasa , Replicación Viral/genética , Adulto Joven
16.
Angiogenesis ; 24(3): 533-548, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33496909

RESUMEN

Myxofibrosarcoma is genetically complex and lacks effective nonsurgical treatment strategies; thus, elucidation of novel molecular drivers is urgently needed. Reanalyzing public myxofibrosarcoma datasets, we identified mRNA upregulation and recurrent gain of RSF1 and characterized this chromatin remodeling gene. Myxofibrosarcoma cell lines were employed to elucidate the oncogenic mechanisms of RSF1 by genetic manipulation and two IL-1ß-neutralizing antibodies (RD24, P2D7KK), highlighting the regulatory basis and targetability of downstream IL-1ß-mediated angiogenesis. Tumor samples were assessed for RSF1, IL-1ß, and microvascular density (MVD) by immunohistochemistry and for RSF1 gene status by FISH. In vivo, RSF1-silenced and P2D7KK-treated xenografts were analyzed for tumor-promoting effects and the IL-1ß-linked therapeutic relevance of RSF1, respectively. In vitro, RSF1 overexpression promoted invasive and angiogenic phenotypes with a stronger proangiogenic effect. RT-PCR profiling identified IL1B as a top-ranking candidate upregulated by RSF1. RSF1 required hSNF2H and CEBP/ß to cotransactivate the IL1B promoter, which increased the IL1B mRNA level, IL-1ß secretion and angiogenic capacity. Angiogenesis induced by RSF1-upregulated IL-1ß was counteracted by IL1B knockdown and both IL-1ß-neutralizing antibodies. Clinically, RSF1 overexpression was highly associated with RSF1 amplification, IL-1ß overexpression, increased MVD and higher grades (all P ≤ 0.01) and independently predicted shorter disease-specific survival (P = 0.019, hazard ratio: 4.556). In vivo, both RSF1 knockdown and anti-IL-1ß P2D7KK (200 µg twice weekly) enabled significant growth inhibition and devascularization in xenografts. In conclusion, RSF1 overexpression, partly attributable to RSF1 amplification, contributes a novel proangiogenic function by partnering with CEBP/ß to cotransactivate IL1B, highlighting its prognostic, pathogenetic, and therapeutic relevance in myxofibrosarcomas.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Fibrosarcoma/metabolismo , Amplificación de Genes , Regulación Neoplásica de la Expresión Génica , Interleucina-1beta/metabolismo , Proteínas de Neoplasias/metabolismo , Neovascularización Patológica/metabolismo , Proteínas Nucleares/biosíntesis , Transactivadores/biosíntesis , Adenosina Trifosfatasas/genética , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteínas Cromosómicas no Histona/genética , Fibrosarcoma/irrigación sanguínea , Fibrosarcoma/genética , Fibrosarcoma/patología , Humanos , Interleucina-1beta/genética , Proteínas de Neoplasias/genética , Neovascularización Patológica/genética , Proteínas Nucleares/genética , Transactivadores/genética
17.
Inflammation ; 44(1): 194-205, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32812145

RESUMEN

Sepsis is recognized as the acute systemic inflammatory response to severe infection. It is main cause of multiple organ system dysfunction and even organ failure. Long non-coding RNA X inactivate-specific transcript (XIST) is implicated in multiple inflammatory diseases. The aim of present work was to investigate the precise mechanism of XIST underlying sepsis-induced acute liver injury. Rats were underwent cecal ligation and puncture (CLP) to establish sepsis-induced the animal models of acute liver injury. Hematoxylin and eosin (H&E) staining was performed to observe pathological alterations. Corresponding commercial assay kits were employed to analyze the levels of inflammatory cytokines and oxidative stress. Western blot and reverse transcriptional quantitative PCR (RT-qPCR) were performed to determine the expression of proteins and target genes. Finally, TUNEL and CCK-8 assays were performed to test apoptosis rate and cell viability, respectively. In our study, XIST and BRD4 were highly expressed in serum of patients with sepsis-induced acute liver injury. XIST knockdown ameliorated sepsis-induced acute liver injury and inhibited inflammation, oxidative stress, and cell apoptosis in sepsis-induced acute liver injury rats. Interestingly, XIST knockdown downregulated the expression of BRD4, and BRD4 overexpression abolished the impacts of XIST knockdown on inflammation, oxidative stress, and apoptosis of that LPS-induced Kupffer cells. We conclude that lncRNA XIST silencing protects against sepsis-induced acute liver injury via inhibition of the BRD4 expression. Therefore, XIST may be a biomarker for sepsis diagnosis and treatment.


Asunto(s)
Proteínas de Ciclo Celular/biosíntesis , Silenciador del Gen/fisiología , Fallo Hepático Agudo/metabolismo , Proteínas Nucleares/biosíntesis , ARN Largo no Codificante/biosíntesis , Sepsis/metabolismo , Factores de Transcripción/biosíntesis , Adulto , Anciano , Animales , Biomarcadores/metabolismo , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen/métodos , Humanos , Macrófagos del Hígado , Fallo Hepático Agudo/genética , Fallo Hepático Agudo/prevención & control , Masculino , Persona de Mediana Edad , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , ARN Largo no Codificante/antagonistas & inhibidores , ARN Largo no Codificante/genética , Ratas , Ratas Sprague-Dawley , Sepsis/genética , Sepsis/prevención & control , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética
18.
Cell Tissue Res ; 383(3): 979-986, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33242174

RESUMEN

Taste substances are detected by taste receptor cells in the taste buds in the oral epithelium. Individual taste receptor cells contribute to evoking one of the five taste qualities: sweet, umami, bitter, sour, and salty (sodium). They are continuously replaced every few weeks by new ones generated from local epithelial stem cells. A POU transcription factor, Pou2f3 (also known as Skn-1a), regulates the generation and differentiation of sweet, umami, and bitter cells. However, the molecular mechanisms underlying terminal differentiation into these Pou2f3-dependent taste receptor cells remain unknown. To identify the candidate molecules that regulate the differentiation of these taste receptor cells, we searched for taste receptor type-specific transcription factors using RNA-sequence data of sweet and bitter cells. No transcription factor gene showing higher expression in sweet cells than in bitter cells was found. Eyes absent 1 (Eya1) was identified as the only transcription factor gene showing higher expression in bitter cells than in sweet cells. In situ hybridization revealed that Eya1 was predominantly expressed in bitter cells and also in the putative immature/differentiating taste bud cells in circumvallate and fungiform papillae and soft palate. Eya1 is a candidate molecule that regulates the generation and differentiation of bitter cells.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Proteínas Nucleares/biosíntesis , Proteínas Tirosina Fosfatasas/biosíntesis , Papilas Gustativas , Animales , Diferenciación Celular , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Gusto , Papilas Gustativas/citología , Papilas Gustativas/metabolismo
19.
Mol Med Rep ; 23(2)2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33313950

RESUMEN

Gli proteins are key transcription factors of the Hedgehog (HH) signaling pathway, which is associated with tumorigenesis and drug resistance. However, the role of the HH signaling pathway in epithelial ovarian cancer (EOC) remains unclear. Studies have demonstrated that in some tumors, homeobox protein NANOG (NANOG), a known stem cell marker, is a downstream effector of Gli. However, limited research has been conducted on the association between Gli and NANOG in EOC, particularly regarding their roles in the tumor stemness, such as tumor development, drug resistance and patient prognosis. Thus, the aim of the present study was to explore the aforementioned issues. In this study, Gli1, Gli2 and NANOG expression in EOC tissues was assessed using immunohistochemistry. Gene expression was also assessed using western blotting and reverse transcription­quantitative PCR in SKOV3 cells treated with a Gli inhibitor and an HH agonist. Furthermore, cell proliferation, colony­forming ability and cisplatin sensitivity were assessed using Cell Counting Kit­8 and colony formation assays. The results showed that both Gli1 and NANOG were associated with cisplatin resistance and EOC disease stage, while the nuclear expression of Gli2 was significantly associated with cisplatin resistance. Together, the expression of Gli and NANOG predicted poor patient prognosis. Targeting Gli with GANT61 impeded tumor proliferation, reversed cisplatin resistance and colony formation, and reduced NANOG expression. To conclude, Gli and NANOG may be effective indicators of platinum resistance and prognosis in EOC. Targeting Gli may reduce the stemness of ovarian cancer cell, which may be achieved via indirect targeting of NANOG.


Asunto(s)
Carcinoma Epitelial de Ovario/metabolismo , Proteína Homeótica Nanog/metabolismo , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Neoplasias Ováricas/metabolismo , Proteína con Dedos de Zinc GLI1/biosíntesis , Proteína con Dedos de Zinc GLI1/genética , Proteína Gli2 con Dedos de Zinc/biosíntesis , Proteína Gli2 con Dedos de Zinc/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Epitelial de Ovario/genética , Carcinoma Epitelial de Ovario/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Femenino , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/farmacología , Humanos , Estimación de Kaplan-Meier , Persona de Mediana Edad , Proteína Homeótica Nanog/genética , Proteínas Nucleares/antagonistas & inhibidores , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Pronóstico , Piridinas/farmacología , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Células Madre/efectos de los fármacos , Ensayo de Tumor de Célula Madre , Adulto Joven , Proteína con Dedos de Zinc GLI1/antagonistas & inhibidores , Proteína Gli2 con Dedos de Zinc/antagonistas & inhibidores
20.
Biochem Genet ; 59(2): 516-530, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33170398

RESUMEN

Cervical cancer (CC) is a common gynecological malignancy, accounting for 10% of all gynecological cancers. Recently, targeted therapy for CC has shown unprecedented advantages. To improve CC patients' prognosis, there are still urgent needs to develop more promising therapeutic targets. Aldo-keto reductase 1 family member C1 (AKR1C1) is a type of aldosterone reductase and plays a regulatory role in a variety of key metabolic pathways. Several studies indicated that AKR1C1 was highly expressed in a series of tumors, and participated in the progression of these tumors. However, the possible effects of AKR1C1 on CC progression remain unclear. Herein, we revealed AKR1C1 was highly expressed in human CC tissues and correlated with the clinical characteristics of patients with CC. AKR1C1 could regulate the proliferation and invasion of cervical cancer cells in vitro. Further experiments showed that AKR1C1 could regulate TWIST1 expression and AKT pathway. In summary, we confirmed the involvement of AKR1C1 in CC progression, and therefore AKR1C1 may have the potential to be a molecular target for CC treatment.


Asunto(s)
20-Hidroxiesteroide Deshidrogenasas/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/biosíntesis , Proteína 1 Relacionada con Twist/biosíntesis , Neoplasias del Cuello Uterino/metabolismo , 20-Hidroxiesteroide Deshidrogenasas/genética , Femenino , Células HeLa , Humanos , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Proteína 1 Relacionada con Twist/genética , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...