Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Cell Commun Signal ; 22(1): 234, 2024 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-38643181

RESUMEN

BACKGROUND: p66Shc, as a redox enzyme, regulates reactive oxygen species (ROS) production in mitochondria and autophagy. However, the mechanisms by which p66Shc affects autophagosome formation are not fully understood. METHODS: p66Shc expression and its location in the trophoblast cells were detected in vivo and in vitro. Small hairpin RNAs or CRISPR/Cas9, RNA sequencing, and confocal laser scanning microscope were used to clarify p66Shc's role in regulating autophagic flux and STING activation. In addition, p66Shc affects mitochondrial-associated endoplasmic reticulum membranes (MAMs) formation were observed by transmission electron microscopy (TEM). Mitochondrial function was evaluated by detected cytoplastic mitochondrial DNA (mtDNA) and mitochondrial membrane potential (MMP). RESULTS: High glucose induces the expression and mitochondrial translocation of p66Shc, which promotes MAMs formation and stimulates PINK1-PRKN-mediated mitophagy. Moreover, mitochondrial localized p66Shc reduces MMP and triggers cytosolic mtDNA release, thus activates cGAS/STING signaling and ultimately leads to enhanced autophagy and cellular senescence. Specially, we found p66Shc is required for the interaction between STING and LC3II, as well as between STING and ATG5, thereby regulates cGAS/STING-mediated autophagy. We also identified hundreds of genes associated several biological processes including aging are co-regulated by p66Shc and ATG5, deletion either of which results in diminished cellular senescence. CONCLUSION: p66Shc is not only implicated in the initiation of autophagy by promoting MAMs formation, but also helps stabilizing active autophagic flux by activating cGAS/STING pathway in trophoblast.


Asunto(s)
Autofagosomas , Trofoblastos Extravellosos , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo , Autofagosomas/metabolismo , Autofagia , ADN Mitocondrial/metabolismo , Trofoblastos/metabolismo , Glucosa/metabolismo , Nucleotidiltransferasas/metabolismo
2.
Comput Biol Med ; 159: 106858, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37087778

RESUMEN

Foot ulcers are a common complication of diabetes mellitus, which is associated with high morbidity and mortality among diabetic patients. The present study aims to investigate novel wound healing pathways in diabetic foot ulcers (DFU) through proteomics and a network pharmacology analysis. Tandem mass tag (TMT) labeled quantitative proteomics method was performed to evaluate the protein expression profile in wound tissues from healthy controls (HC) and DFU. Kyoto Encyclopedia of Genes (KEGG) and Genomes enrichment analysis (GO) was conducted based on differentially expressed proteins (DEPs) to discover the potential pathways associated with DFU. Western blot analysis was used to confirm the probable DFU-related targets. Proteomics analysis discovered 509 DEPs (248 upregulated and 261 downregulated proteins). Go and KEGG further evaluated the DEPs to discover the DFU-related pathways. According to network pharmacology study, three main targets (metalloproteinase 9 (MMP9), Fatty acid-binding protein 5 (FABP5), and integrin subunit alpha M (ITGAM)) play crucial roles in signaling pathways. Staphylococcus aureus infection and leukocyte transendothelial migration pathways significantly enriched in DFU. In addition, it was confirmed that three critical targets were elevated in diabetes mouse wound tissues. The study confirmed the presence of protein alterations in the wound-healing process of DFU mice and may provide fresh insights into the molecular mechanisms driving DFU.


Asunto(s)
Diabetes Mellitus , Pie Diabético , Ratones , Animales , Pie Diabético/genética , Proteómica , Cicatrización de Heridas
3.
Mol Ther Nucleic Acids ; 28: 293-306, 2022 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-35474733

RESUMEN

Impressive achievements in clinical trials to treat hemophilia establish a milestone in the development of gene therapy. It highlights the significance of AAV-mediated gene delivery to liver. AAV5 is a unique serotype featured by low neutralizing antibody prevalence. Nevertheless, its liver infectivity is relatively weak. Consequently, it is vital to exploit novel AAV5 capsid mutants with robust liver tropism. To this aim, we performed AAV5-NNK library and barcode screening in mice, from which we identified one capsid variant, called AAVzk2. AAVzk2 displayed a similar yield but divergent post-translational modification sites compared with wild-type serotypes. Mice intravenously injected with AAVzk2 demonstrated a stronger liver transduction than AAV5, roughly comparable with AAV8 and AAV9, with undetectable transduction of other tissues or organs such as heart, lung, spleen, kidney, brain, and skeletal muscle, indicating a liver-specific tropism. Further studies showed a superior human hepatocellular transduction of AAVzk2 to AAV5, AAV8 and AAV9, whereas the seroreactivity of AAVzk2 was as low as AAV5. Overall, we provide a novel AAV serotype that facilitates a robust and specific liver gene delivery to a large population, especially those unable to be treated by AAV8 and AAV9.

4.
Mol Cell Endocrinol ; 547: 111598, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35157929

RESUMEN

Maternal pregnancy hyperglycemia is often accompanied by placental dysfunction. During placental development, epithelial-mesenchymal transition (EMT) contributes to the transformation of relatively noninvasive trophoblasts into highly invasive extravillous trophoblasts (EVTs). However, the specific role of EMT in placentas under hyperglycemia environments remains relatively unexplored. Stanniocalcin2 (STC2) regulates EMT in many cancers. In this study, we first demonstrated that STC2 expression was upregulated in GDM placenta. We found that STC2 activated autophagy and suppressed EMT in high-glucose-treated EVTs and was associated with a lack of invasiveness. Specifically, STC2 inhibited the interactions between p62/SQSTM1 (p62) and EMT transcription factors to promote the degradation of Twist1 and Snail via a proteasome-dependent pathway. Furthermore, the PI3K/AKT/AMPK signaling pathway was involved in the regulation of autophagy and EMT by STC2. Taken together, our results reveal that STC2 may serve as a potential prognostic biomarker in GDM and sheds light on the regulatory mechanisms of trophoblast invasion.


Asunto(s)
Transición Epitelial-Mesenquimal , Glicoproteínas , Péptidos y Proteínas de Señalización Intercelular , Trofoblastos , Autofagia , Movimiento Celular , Femenino , Glucosa/genética , Glucosa/farmacología , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Placenta/metabolismo , Embarazo , Trofoblastos/metabolismo
5.
Artículo en Inglés | MEDLINE | ID: mdl-33042863

RESUMEN

Studies have shown that exposure to environmental tobacco smoke can increase the risk of bacterial meningitis, and nicotine is the core component of environmental tobacco smoke. Autophagy is an important way for host cells to eliminate invasive pathogens and resist infection. Escherichia coli K1 strain (E. coli K1) is the most common Gram-negative bacterial pathogen that causes neonatal meningitis. The mechanism of nicotine promoting E. coli K1 to invade human brain microvascular endothelial cells (HBMECs), the main component of the blood-brain barrier, is not clear yet. Our study found that the increase of HBMEC autophagy level during E. coli K1 infection could decrease the survival of intracellular bacteria, while nicotine exposure could inhibit the HBMEC autophagic response of E. coli K1 infection by activating the NF-kappa B and PI3K/Akt/mTOR pathway. We concluded that nicotine could inhibit HBMEC autophagy upon E. coli K1 infection and decrease the scavenging effect on E. coli K1, thus promoting the occurrence and development of neonatal meningitis.


Asunto(s)
Escherichia coli , Nicotina , Autofagia , Encéfalo/metabolismo , Células Cultivadas , Células Endoteliales/metabolismo , Escherichia coli/metabolismo , Humanos , Recién Nacido , Nicotina/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo
6.
Cancer Lett ; 492: 162-173, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32858102

RESUMEN

Estrogen receptor α (ER) acts as an oncogenic signal in endometrial endometrioid carcinoma. ER binding activity largely depends on chromatin remodeling and recruitment of transcription factors to estrogen response elements. A deeper understanding of these regulatory mechanisms may uncover therapeutic targets for ER-dependent endometrial cancers. We show that estrogen induces accessible chromatin and ER binding at a subset of enhancers, which form higher-order super enhancers that are vital for ER signaling. ER positively correlates with active enhancers in primary tumors, and tumors were effectively classified into molecular subtypes with chromatin accessibility dynamics and ER-dependent gene signature. ARID1A binds within ER-bound enhancers and regulates ER-dependent transcription. Knockdown of ARID1A or fulvestrant treatment profoundly affects the gene-expression program, and inhibits cell growth phenotype by affecting the chromatin environment. Importantly, we found dysregulated expression of circadian rhythms genes by estrogen in cancer cells and in primary tumors. Knockdown of ARID1A reduces the chromatin accessibility and ER binding at enhancers of the circadian gene ARNTL and BHLHE41, leading to a decreased expression of these genes. Altogether, we uncover a critical role for ARID1A in ER signaling and therapeutic target in ER-positive endometrial cancer.


Asunto(s)
Factores de Transcripción ARNTL/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Cromatina/metabolismo , Proteínas de Unión al ADN/fisiología , Neoplasias Endometriales/metabolismo , Regulación Neoplásica de la Expresión Génica , Receptores de Estrógenos/fisiología , Factores de Transcripción/fisiología , Femenino , Humanos , Proteínas Musculares/fisiología , Transducción de Señal/fisiología , Factores de Transcripción de Dominio TEA
7.
Mol Cell Endocrinol ; 502: 110674, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31811899

RESUMEN

Autophagy plays an essential role in gestational diabetes mellitus (GDM). Death-associated protein kinase-3 (DAPK3) regulates a variety of cellular functions; however, the relationship between DAPK3 and autophagy is unknown. In this study, we aim to investigate whether DAPK3 is associated with autophagy in GDM, and we found that DAPK3 was upregulated in the placenta of GDM patients and extravillous trophoblast cells under high-glucose conditions. Silencing DAPK3 decreased the assembly of the STX17-SNAP29-VAMP8 complex, leading to the blockade of autophagosome-lysosome fusion by mediating synaptosomal-associated protein 29 (SNAP29). Moreover, knockdown of DAPK3 ameliorates cell invasion and mediates autophagy in high glucose, and does not alter the expression of autophagy-related genes in normal glucose. Our study demonstrates the significance of DAPK3 in autophagy and GDM, which may provide new insights into the molecular mechanisms regulating trophoblast invasion.


Asunto(s)
Autofagosomas/metabolismo , Proteínas Quinasas Asociadas a Muerte Celular/genética , Diabetes Gestacional/genética , Lisosomas/metabolismo , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Línea Celular , Movimiento Celular , Proteínas Quinasas Asociadas a Muerte Celular/metabolismo , Diabetes Gestacional/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Glucosa/efectos adversos , Humanos , Embarazo , Trofoblastos/citología , Trofoblastos/efectos de los fármacos , Trofoblastos/metabolismo , Regulación hacia Arriba
8.
J Infect Dis ; 219(3): 470-479, 2019 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-30202861

RESUMEN

FimH-mediated bacterial invasion and polymorphonuclear neutrophil (PMN) transmigration across human brain microvascular endothelial cells (HBMECs) are required for the pathogenesis of Escherichia coli meningitis. However, the underlying mechanism remains unclear. This study demonstrated that the TnphoA mutant (22A33) and FimH-knockout mutant (ΔFimH) of E coli strain E44, which resulted in inactivation of FimH, were less invasive and less effective in promoting PMN transmigration than their wild-type strain. FimH protein induced PMN transmigration, whereas calmodulin inhibitor significantly blocked this effect. Moreover, immunofluorescence and co-immunoprecipitation analysis indicated that colocalized CD48 and α7 nAChR formed a complex on the surface of HBMECs that is associated with increased cofilin dephosphorylation, which could be remarkably enhanced by FimH+ E44. Our study concluded that FimH-induced E coli K1 invasion and PMN migration across HBMECs may be mediated by the CD48-α7nAChR complex in lipid rafts of HBMEC via Ca2+ signaling and cofilin dephosphorylation.


Asunto(s)
Adhesinas de Escherichia coli/metabolismo , Encéfalo/microbiología , Antígeno CD48/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/microbiología , Escherichia coli/metabolismo , Proteínas Fimbrias/metabolismo , Neutrófilos/microbiología , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Adhesinas de Escherichia coli/genética , Calcio/metabolismo , Movimiento Celular , Escherichia coli/patogenicidad , Proteínas Fimbrias/genética , Técnicas de Silenciamiento del Gen , Humanos , Microdominios de Membrana , Meningitis por Escherichia coli/microbiología , Meningitis por Escherichia coli/patología , Transducción de Señal , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
9.
Free Radic Biol Med ; 121: 26-37, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29684410

RESUMEN

Engineered fluorescent indicators for visualizing mercury ion (Hg2+) are powerful tools to illustrate the intracellular distribution and serious toxicity of the ion. However, the sensitive and specific detection of Hg2+ in living cells and in vivo is challenging. This paper reported the development of fluorescent indicators for Hg2+ in green or red color by inserting a circularly permuted fluorescent protein into a highly mercury-specific repressor. These sensors provided a rapid, sensitive, specific, and real-time read-out of Hg2+ dynamics in solutions, bacteria, subcellular organelles of mammalian cells, and zebrafish, thereby providing a useful new method for Hg2+ detection and bioimaging. In conjunction with the hydrogen peroxide sensor HyPer, we found mercury uptake would trigger subcellular oxidative events at the single-cell level, and provided visual evidence of the causality of mercury and oxidative damage. These sensors would paint the landscape of mercury toxicity to cell functions.


Asunto(s)
Técnicas Biosensibles , Colorantes Fluorescentes/química , Procesamiento de Imagen Asistido por Computador/métodos , Mercurio/análisis , Mercurio/toxicidad , Mitocondrias/patología , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas de Unión al ADN/genética , Escherichia coli/metabolismo , Colorantes Fluorescentes/metabolismo , Células HeLa , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Microscopía Fluorescente , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Oxidación-Reducción , Consumo de Oxígeno , Pez Cebra/metabolismo
10.
Free Radic Biol Med ; 120: 255-265, 2018 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-29580984

RESUMEN

Genetically encoded fluorescent sensors are widely used to visualize secondary messengers, metabolites and dynamic events in living cells. However, almost all of these sensors are based on Aequorea GFPs or GFP-like proteins, which do not correctly maturate and fluoresce under hypoxia or anoxic conditions, greatly limiting their application in biomedical research. Herein, we provide a novel strategy for design of sensors and report a series of thiol redox-sensitive sensor based on a recently discovered oxygen-independent fluorescent protein UnaG from Japanese eel. These redox sensors have large dynamic range, rapid responsiveness, a flexible "switch", and pH-independence, are particularly compatible with hypoxia conditions, and therefore represent a substantial improvement for live-cell redox measurement. We further demonstrated the versatility of these redox sensors, by simultaneously monitoring redox changes and hypoxia state in living cells, thereby proving its capability as a powerful and flexible tool for indexing multidimensional metabolism data in the context of physiological stressors and pathological states. These redox sensors are not only the first case of UnaG-based functional sensors, but also the first case of functional sensors based on non GFP-like proteins. Based on this strategy, more oxygen-independent biosensors could be developed, hence, provide new opportunities for bioimaging.


Asunto(s)
Técnicas Biosensibles/métodos , Anguilas , Proteínas de Peces , Proteínas Luminiscentes , Compuestos de Sulfhidrilo , Animales , Hipoxia de la Célula/fisiología , Proteínas de Peces/química , Proteínas Luminiscentes/química , Oxidación-Reducción
11.
RSC Adv ; 8(5): 2485-2489, 2018 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-35541484

RESUMEN

Glucose is the main source of energy and carbon in organisms and plays a central role in metabolism and cellular homeostasis. However, the sensitive fluctuation of glucose in living cells is difficult to monitor. Thus, we developed a series of ratiometric, highly responsive, single fluorescent protein-based glucose sensors of wide dynamic range by combining a circularly permuted yellow fluorescent protein with a bacterial periplasmic glucose/galactose-binding protein. We used these sensors to monitor glucose transport in living Escherichia coli cells, and found that the cells take up glucose within 10 min to maintain physiological glucose levels, and observed the differences in glucose uptake and glucose metabolism between wild-type and Mlc knockout cells. These sensors can be specific and simple tools for glucose detection in vitro and non-invasive tools for real-time monitoring of glucose metabolism in vivo.

12.
Brief Bioinform ; 19(4): 627-635, 2018 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-28203711

RESUMEN

Long noncoding RNAs (lncRNAs) are a large family of noncoding RNAs that play a critical role in various normal bioprocesses as well as tumorigenesis. However, the expression patterns and biological functions of lncRNAs in acute leukemia have not been well studied. Here, we performed transcriptome-wide lncRNA expression profiling of acute myeloid leukemia (AML) patient samples, along with non-leukemia control hematopoietic samples. We found that lncRNAs were differentially expressed in AML samples relative to control samples. Notably, we identified that lncRNAs upregulated in AML (relative to the control samples) are associated with a lower degree of DNA methylation and a higher ratio of being bound by transcription factors such as SP1, STAT4, ATF-2 and ELK-1 compared with those downregulated in AML. Moreover, an enrichment of H3K4me3 and a depletion of H3K27me3 were observed in upregulated lncRNAs in AML. Expression patterns of three types of lncRNAs (antisense, enhancer and intergenic lncRNAs) have previously been characterized. Of the identified lncRNAs, we found that high expression level lncRNA LOC285758 is associated with the poor prognosis in AML patients. Furthermore, we found that LOC285758 regulates proliferation of AML cell lines by enhancing the expression of HDAC2, a key factor in carcinogenesis. Collectively, our study depicts a landscape of important lncRNAs in AML and provides novel potential therapeutic targets and prognostic markers for AML treatment.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Histona Desacetilasa 2/metabolismo , Leucemia Mieloide Aguda/genética , ARN Largo no Codificante/genética , Transcriptoma , Estudios de Casos y Controles , Histona Desacetilasa 2/genética , Humanos , Células Tumorales Cultivadas
13.
Endocrinology ; 158(8): 2522-2532, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28838138

RESUMEN

Autophagy is a dynamic process that degrades and recycles cellular organelles and proteins to maintain cell homeostasis. Alterations in autophagy occur in various diseases; however, the role of autophagy in gestational diabetes mellitus (GDM) is unknown. In the present study, we characterized the roles and functions of autophagy in GDM patient samples and extravillous trophoblasts cultured with glucose. We found significantly enhanced autophagy in GDM patients. Moreover, high glucose levels enhanced autophagy and cell apoptosis, reducing proliferation and invasion, and these effects were ameliorated through knockdown of ATG5. Genome-wide 5-hydroxymethylcytosine data analysis further revealed the epigenomic regulatory circuitry underlying the induced autophagy and apoptosis in GDM and preeclampsia. Finally, RNA sequencing was performed to identify gene expression changes and critical signaling pathways after silencing of ATG5. Our study has demonstrated the substantial functions of autophagy in GDM and provides potential therapeutic targets for the treatment of GDM patients.


Asunto(s)
Autofagia/fisiología , Diabetes Gestacional/fisiopatología , Placenta/citología , Adulto , Apoptosis/fisiología , Glucemia , Línea Celular , Epigenómica , Femenino , Regulación de la Expresión Génica , Humanos , Hiperglucemia , Embarazo
14.
Sci Rep ; 7(1): 4209, 2017 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-28646144

RESUMEN

In this paper, we propose a novel and sensitive ratiometric analysis method that uses the fractional intensities of time-resolved fluorescence of genetically encoded fluorescent NADH/NAD+ biosensors, Peredox, SoNar, and Frex. When the conformations of the biosensors change upon NADH/NAD+ binding, the fractional intensities (α i τ i ) have opposite changing trends. Their ratios could be exploited to quantify NADH/NAD+ levels with a larger dynamic range and higher resolution versus commonly used fluorescence intensity and lifetime methods. Moreover, only one excitation and one emission wavelength are required for this ratiometric measurement. This eliminates problems of traditional excitation-ratiometric and emission-ratiometric methods. This method could be used to simplify the design and achieve highly sensitive analyte quantification of genetically encoded fluorescent biosensors. Wide potential applications could be developed for imaging live cell metabolism based on this new method.


Asunto(s)
Técnicas Biosensibles/métodos , NAD/análisis , Fluorescencia , Factores de Tiempo
15.
Sci Rep ; 7: 43479, 2017 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-28252043

RESUMEN

High-resolution spatiotemporal imaging of histidine in single living mammalian cells faces technical challenges. Here, we developed a series of ratiometric, highly responsive, and single fluorescent protein-based histidine sensors of wide dynamic range. We used these sensors to quantify subcellular free-histidine concentrations in glucose-deprived cells and glucose-fed cells. Results showed that cytosolic free-histidine concentration was higher and more sensitive to the environment than free histidine in the mitochondria. Moreover, histidine was readily transported across the plasma membrane and mitochondrial inner membrane, which had almost similar transport rates and transport constants, and histidine transport was not influenced by cellular metabolic state. These sensors are potential tools for tracking histidine dynamics inside subcellular organelles, and they will open an avenue to explore complex histidine signaling.


Asunto(s)
Proteínas Bacterianas/química , Técnicas Biosensibles , Membrana Celular/metabolismo , Histidina/análisis , Proteínas Luminiscentes/química , Membranas Mitocondriales/metabolismo , Proteínas de Unión Periplasmáticas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Transporte Biológico , Membrana Celular/efectos de los fármacos , Rastreo Celular , Clonación Molecular , Citosol/efectos de los fármacos , Citosol/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Expresión Génica , Glucosa/metabolismo , Glucosa/farmacología , Células HeLa , Histidina/metabolismo , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Membranas Mitocondriales/efectos de los fármacos , Proteínas de Unión Periplasmáticas/genética , Proteínas de Unión Periplasmáticas/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal
16.
Br J Cancer ; 116(5): 658-668, 2017 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-28141796

RESUMEN

BACKGROUND: DNA methylation at the 5 position of cytosine (5mC) can be converted to 5-hydroxymethylcytosine (5hmC) by the ten-eleven translocation family. The loss of global levels of 5hmC has been regarded as a hallmark in various cancers. 5-hydroxymethylcytosine is distributed at protein-coding gene bodies and promoters; however, the role and distribution of 5hmC at long non-coding RNAs (lncRNAs) is not clear. We investigated the distribution and regulatory roles of 5hmC for lncRNAs in colorectal cancer (CRC). METHODS: We integrated genome-wide profiles of 5hmC, 5mC, transcriptome and histone marks in CRC patients and examined the 5hmC-based clinical outcomes in patients. RESULTS: 5-hydroxymethylcytosine was distributed at lncRNA loci and positively correlated with lncRNA transcription. Dysreulated CRC lncRNAs were regulated by 5hmC directly or through abnormal activities of typical and super-enhancers and promoters modified by 5hmC. In addition, 5hmC was involved in long-range chromatin interactions at lncRNA loci. Finally, lncRNAs regulated by differential 5hmC marks were correlated with different clinical outcomes and tumour status in patients. CONCLUSIONS: 5-hydroxymethylcytosine is critical in regulating the transcription of lncRNA and serve as novel biomarkers for clinical prognosis in CRC.


Asunto(s)
5-Metilcitosina/análogos & derivados , Neoplasias Colorrectales/patología , Epigenómica/métodos , ARN Largo no Codificante/genética , 5-Metilcitosina/metabolismo , Neoplasias Colorrectales/genética , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Humanos , Pronóstico , Análisis de Supervivencia
17.
Breast Cancer Res Treat ; 157(2): 267-279, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27178334

RESUMEN

Breast cancer has been classified into several intrinsic molecular subtypes on the basis of genetic and epigenetic factors. However, knowledge about histone modifications that contribute to the classification and development of biologically distinct breast cancer subtypes remains limited. Here we compared the genome-wide binding patterns of H3K4me3 and H3K27me3 between human mammary epithelial cells and three breast cancer cell lines representing the luminal, HER2, and basal subtypes. We characterized thousands of unique binding events as well as bivalent chromatin signatures unique to each cancer subtype, which were involved in different epigenetic regulation programs and signaling pathways in breast cancer progression. Genes linked to the unique histone mark features exhibited subtype-specific expression patterns, both in cancer cell lines and primary tumors, some of which were confirmed by qPCR in our primary cancer samples. Finally, histone mark-based gene classifiers were significantly correlated with relapse-free survival outcomes in patients. In summary, we have provided a valuable resource for the identification of novel biomarkers of subtype classification and clinical prognosis evaluation in breast cancers.


Asunto(s)
Neoplasias de la Mama/clasificación , Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes , Histonas/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Supervivencia sin Enfermedad , Epigénesis Genética , Femenino , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Código de Histonas , Histonas/metabolismo , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Análisis de Secuencia de ARN , Análisis de Supervivencia
18.
Oncotarget ; 7(17): 23897-908, 2016 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-27004403

RESUMEN

Accumulating evidence highlights the role of long non-coding RNAs (lncRNAs) in tumors. However, the genome-wide expression and roles of lncRNAs in colorectal cancer (CRC) remain unknown. Here, we systematically examined the global gene expressions in primary and synchronous liver metastases CRC tissue, in which thousands of aberrantly expressed lncRNAs were characterized. Co-expression analysis revealed that some lncRNAs correlated to their neighboring mRNAs in expression levels, whereas others formed networks with protein-coding genes in trans. We observed H3K4me3 was enriched at expressed lncRNA transcription start sites (TSSs) and correlated to dysregulated lncRNAs. Furthermore, we identified primary and metastasis tumor linked lncRNA signatures positively correlated with poor-prognosis gene set. Finally, functional experiments demonstrated two candidate lncRNAs were required for proliferation and migration of CRC cells. In summary, we provided a new framework for lncRNA associated clinical prognosis evaluation and target selection of gene therapy in CRC.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Largo no Codificante/genética , Humanos , Pronóstico
19.
Circ Cardiovasc Genet ; 9(2): 110-8, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26896382

RESUMEN

BACKGROUND: The molecular regulation of heart development is regulated by cis- and trans-factors acting on the genome and epigenome. As a class of important regulatory RNAs, the role of long noncoding RNAs (lncRNAs) in human heart development is still poorly understood. Furthermore, factors that interact with lncRNAs in this process are not well characterized. METHODS AND RESULTS: Using RNA sequencing, we systematically define the contrasting lncRNA expression patterns between fetal and adult hearts. We report that lncRNAs upregulated in adult versus fetal heart have different sequence features and distributions. For example, the adult heart expresses more sense lncRNAs compared with fetal heart. We also report the coexpression of lncRNAs and neighboring coding genes that have important functions in heart development. Importantly, the regulation of lncRNA expression during fetal to adult heart development seems to be due, in part, to the coordination of specific developmental epigenetic modifications, such as H3K4me1 and H3k4me3. The expression of promoter-associated lncRNAs in adult and fetal hearts also seems to be related to these epigenetic states. Finally, transcription factor-binding analysis suggests that lncRNAs are directly regulating cardiac gene expression during development. CONCLUSIONS: We provide a systematic analysis of lncRNA control of heart development that gives clues to the roles that specific lncRNAs play in fetal and adult hearts.


Asunto(s)
Feto/metabolismo , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , ARN Largo no Codificante/genética , Adulto , Epigénesis Genética , Redes Reguladoras de Genes , Humanos , Sistemas de Lectura Abierta/genética , Polimorfismo de Nucleótido Simple/genética , Unión Proteica , ARN Largo no Codificante/metabolismo , Factores de Transcripción/metabolismo
20.
Cell Metab ; 21(5): 777-89, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25955212

RESUMEN

The altered metabolism of tumor cells confers a selective advantage for survival and proliferation, and studies have shown that targeting such metabolic shifts may be a useful therapeutic strategy. We developed an intensely fluorescent, rapidly responsive, pH-resistant, genetically encoded sensor of wide dynamic range, denoted SoNar, for tracking cytosolic NAD(+) and NADH redox states in living cells and in vivo. SoNar responds to subtle perturbations of various pathways of energy metabolism in real time, and allowed high-throughput screening for new agents targeting tumor metabolism. Among > 5,500 unique compounds, we identified KP372-1 as a potent NQO1-mediated redox cycling agent that produced extreme oxidative stress, selectively induced cancer cell apoptosis, and effectively decreased tumor growth in vivo. This study demonstrates that genetically encoded sensor-based metabolic screening could serve as a valuable approach for drug discovery.


Asunto(s)
Antineoplásicos/farmacología , Ensayos de Selección de Medicamentos Antitumorales/métodos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , NAD/metabolismo , Neoplasias/tratamiento farmacológico , Tetrazoles/farmacología , Animales , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Compuestos Heterocíclicos de 4 o más Anillos/uso terapéutico , Humanos , Ratones Desnudos , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Vía de Pentosa Fosfato/efectos de los fármacos , Tetrazoles/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...