Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Hum Genomics ; 17(1): 97, 2023 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-37924098

RESUMEN

BACKGROUND: Alternative splicing (AS) plays a crucial role in transcriptomic diversity and is a hallmark of cancer that profoundly influences the development and progression of prostate cancer (PCa), a prevalent and potentially life-limiting cancer among men. Accumulating evidence has highlighted the association between AS dysregulation and the onset and progression of PCa. However, a comprehensive and integrative analysis of AS profiles at the event level, utilising data from multiple high-throughput cohorts and evaluating the prognosis of PCa progression, remains lacking and calls for thorough exploration. RESULTS: We identified a differentially expressed retained intron event in ZWINT across three distinct cohorts, encompassing an original array-based dataset profiled by us previously and two RNA sequencing (RNA-seq) datasets. Subsequent in-depth analyses of these RNA-seq datasets revealed  141 altered events, of which 21 demonstrated a significant association with patients' biochemical recurrence-free survival (BCRFS). We formulated an AS event-based prognostic signature, capturing six pivotal events in genes CYP4F12, NFATC4, PIGO, CYP3A5, ALS2CL, and FXYD3. This signature effectively differentiated  high-risk patients diagnosed with PCa, who experienced shorter BCRFS, from their low-risk counterparts. Notably, the signature's predictive power surpassed traditional clinicopathological markers in forecasting 5-year BCRFS, demonstrating robust performance in both internal and external validation sets. Lastly, we constructed a novel nomogram that integrates patients' Gleason scores with pathological tumour stages, demonstrating improved prognostication of BCRFS. CONCLUSIONS: Prediction of clinical progression remains elusive in PCa. This research uncovers novel splicing events associated with BCRFS, augmenting existing prognostic tools, thus potentially refining clinical decision-making.


Asunto(s)
Neoplasias de la Próstata , Transcriptoma , Masculino , Humanos , Pronóstico , Transcriptoma/genética , Empalme Alternativo/genética , Biomarcadores de Tumor/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/patología , Proteínas de la Membrana/genética , Proteínas de Neoplasias/genética
2.
J Pathol ; 261(1): 71-84, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37550801

RESUMEN

Aberrant glycosylation is a universal feature of cancer cells, and cancer-associated glycans have been detected in virtually every cancer type. A common change in tumour cell glycosylation is an increase in α2,6 sialylation of N-glycans, a modification driven by the sialyltransferase ST6GAL1. ST6GAL1 is overexpressed in numerous cancer types, and sialylated glycans are fundamental for tumour growth, metastasis, immune evasion, and drug resistance, but the role of ST6GAL1 in prostate cancer is poorly understood. Here, we analyse matched cancer and normal tissue samples from 200 patients and verify that ST6GAL1 is upregulated in prostate cancer tissue. Using MALDI imaging mass spectrometry (MALDI-IMS), we identify larger branched α2,6 sialylated N-glycans that show specificity to prostate tumour tissue. We also monitored ST6GAL1 in plasma samples from >400 patients and reveal ST6GAL1 levels are significantly increased in the blood of men with prostate cancer. Using both in vitro and in vivo studies, we demonstrate that ST6GAL1 promotes prostate tumour growth and invasion. Our findings show ST6GAL1 introduces α2,6 sialylated N-glycans on prostate cancer cells and raise the possibility that prostate cancer cells can secrete active ST6GAL1 enzyme capable of remodelling glycans on the surface of other cells. Furthermore, we find α2,6 sialylated N-glycans expressed by prostate cancer cells can be targeted using the sialyltransferase inhibitor P-3FAX -Neu5Ac. Our study identifies an important role for ST6GAL1 and α2,6 sialylated N-glycans in prostate cancer progression and highlights the opportunity to inhibit abnormal sialylation for the development of new prostate cancer therapeutics. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Neoplasias de la Próstata , Sialiltransferasas , Masculino , Humanos , Glicosilación , Polisacáridos/química , Polisacáridos/metabolismo , Reino Unido , beta-D-Galactósido alfa 2-6-Sialiltransferasa , Antígenos CD/metabolismo
3.
PLoS Genet ; 17(9): e1009803, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34570759

RESUMEN

SNIP1 (Smad nuclear interacting protein 1) is a widely expressed transcriptional suppressor of the TGF-ß signal-transduction pathway which plays a key role in human spliceosome function. Here, we describe extensive genetic studies and clinical findings of a complex inherited neurodevelopmental disorder in 35 individuals associated with a SNIP1 NM_024700.4:c.1097A>G, p.(Glu366Gly) variant, present at high frequency in the Amish community. The cardinal clinical features of the condition include hypotonia, global developmental delay, intellectual disability, seizures, and a characteristic craniofacial appearance. Our gene transcript studies in affected individuals define altered gene expression profiles of a number of molecules with well-defined neurodevelopmental and neuropathological roles, potentially explaining clinical outcomes. Together these data confirm this SNIP1 gene variant as a cause of an autosomal recessive complex neurodevelopmental disorder and provide important insight into the molecular roles of SNIP1, which likely explain the cardinal clinical outcomes in affected individuals, defining potential therapeutic avenues for future research.


Asunto(s)
Alelos , Amish/genética , Trastornos del Neurodesarrollo/genética , Proteínas de Unión al ARN/genética , Expresión Génica/genética , Genes Recesivos , Humanos
4.
Gut ; 72(2): 295-305, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-35902214

RESUMEN

OBJECTIVE: Antitumour necrosis factor (TNF) drugs impair serological responses following SARS-CoV-2 vaccination. We sought to assess if a third dose of a messenger RNA (mRNA)-based vaccine substantially boosted anti-SARS-CoV-2 antibody responses and protective immunity in infliximab-treated patients with IBD. DESIGN: Third dose vaccine induced anti-SARS-CoV-2 spike (anti-S) receptor-binding domain (RBD) antibody responses, breakthrough SARS-CoV-2 infection, reinfection and persistent oropharyngeal carriage in patients with IBD treated with infliximab were compared with a reference cohort treated with vedolizumab from the impaCt of bioLogic therApy on saRs-cov-2 Infection and immuniTY (CLARITY) IBD study. RESULTS: Geometric mean (SD) anti-S RBD antibody concentrations increased in both groups following a third dose of an mRNA-based vaccine. However, concentrations were lower in patients treated with infliximab than vedolizumab, irrespective of whether their first two primary vaccine doses were ChAdOx1 nCoV-19 (1856 U/mL (5.2) vs 10 728 U/mL (3.1), p<0.0001) or BNT162b2 vaccines (2164 U/mL (4.1) vs 15 116 U/mL (3.4), p<0.0001). However, no differences in anti-S RBD antibody concentrations were seen following third and fourth doses of an mRNA-based vaccine, irrespective of the combination of primary vaccinations received. Post-third dose, anti-S RBD antibody half-life estimates were shorter in infliximab-treated than vedolizumab-treated patients (37.0 days (95% CI 35.6 to 38.6) vs 52.0 days (95% CI 49.0 to 55.4), p<0.0001).Compared with vedolizumab-treated, infliximab-treated patients were more likely to experience SARS-CoV-2 breakthrough infection (HR 2.23 (95% CI 1.46 to 3.38), p=0.00018) and reinfection (HR 2.10 (95% CI 1.31 to 3.35), p=0.0019), but this effect was uncoupled from third vaccine dose anti-S RBD antibody concentrations. Reinfection occurred predominantly during the Omicron wave and was predicted by SARS-CoV-2 antinucleocapsid concentrations after the initial infection. We did not observe persistent oropharyngeal carriage of SARS-CoV-2. Hospitalisations and deaths were uncommon in both groups. CONCLUSIONS: Following a third dose of an mRNA-based vaccine, infliximab was associated with attenuated serological responses and more SARS-CoV-2 breakthrough infection and reinfection which were not predicted by the magnitude of anti-S RBD responses, indicative of vaccine escape by the Omicron variant. TRIAL REGISTRATION NUMBER: ISRCTN45176516.


Asunto(s)
COVID-19 , Enfermedades Inflamatorias del Intestino , Vacunas , Humanos , SARS-CoV-2 , COVID-19/epidemiología , COVID-19/prevención & control , Vacunas contra la COVID-19 , Infliximab/uso terapéutico , Pandemias , Reinfección/epidemiología , Reinfección/prevención & control , Vacuna BNT162 , ChAdOx1 nCoV-19 , Anticuerpos Antivirales , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico
5.
Hum Mol Genet ; 28(16): 2763-2774, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31098640

RESUMEN

Changes to islet cell identity in response to type 2 diabetes (T2D) have been reported in rodent models, but are less well characterized in humans. We assessed the effects of aspects of the diabetic microenvironment on hormone staining, total gene expression, splicing regulation and the alternative splicing patterns of key genes in EndoC-ßH1 human beta cells. Genes encoding islet hormones [somatostatin (SST), insulin (INS), Glucagon (GCG)], differentiation markers [Forkhead box O1 (FOXO1), Paired box 6, SRY box 9, NK6 Homeobox 1, NK6 Homeobox 2] and cell stress markers (DNA damage inducible transcript 3, FOXO1) were dysregulated in stressed EndoC-ßH1 cells, as were some serine arginine rich splicing factor splicing activator and heterogeneous ribonucleoprotein particle inhibitor genes. Whole transcriptome analysis of primary T2D islets and matched controls demonstrated dysregulated splicing for ~25% of splicing events, of which genes themselves involved in messenger ribonucleic acid processing and regulation of gene expression comprised the largest group. Approximately 5% of EndoC-ßH1 cells exposed to these factors gained SST positivity in vitro. An increased area of SST staining was also observed ex vivo in pancreas sections recovered at autopsy from donors with type 1 diabetes (T1D) or T2D (9.3% for T1D and 3% for T2D, respectively compared with 1% in controls). Removal of the stressful stimulus or treatment with the AKT Serine/Threonine kinase inhibitor SH-6 restored splicing factor expression and reversed both hormone staining effects and patterns of gene expression. This suggests that reversible changes in hormone expression may occur during exposure to diabetomimetic cellular stressors, which may be mediated by changes in splicing regulation.


Asunto(s)
Empalme Alternativo , Islotes Pancreáticos/metabolismo , Estrés Fisiológico/genética , Línea Celular , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Estrés del Retículo Endoplásmico , Expresión Génica , Perfilación de la Expresión Génica , Glucagón/metabolismo , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Células Neuroendocrinas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología
6.
FASEB J ; 33(1): 1086-1097, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30088951

RESUMEN

Cellular plasticity is a key facet of cellular homeostasis requiring correct temporal and spatial patterns of alternative splicing. Splicing factors, which orchestrate this process, demonstrate age-related dysregulation of expression; they are emerging as potential influences on aging and longevity. The upstream drivers of these alterations are still unclear but may involve aberrant cellular signaling. We compared the phosphorylation status of proteins in multiple signaling pathways in early and late passage human primary fibroblasts. We then assessed the impact of chemical inhibition or targeted knockdown of direct downstream targets of the ERK and AKT pathways on splicing factor expression, cellular senescence, and proliferation kinetics in senescent primary human fibroblasts. Components of the ERK and AKT signaling pathways demonstrated altered activation during cellular aging. Inhibition of AKT and ERK pathways led to up-regulation of splicing factor expression, reduction in senescent cell load, and partial reversal of multiple cellular senescence phenotypes in a dose-dependent manner. Furthermore, targeted knockdown of the genes encoding the downstream targets FOXO1 or ETV6 was sufficient to mimic these observations. Our results suggest that age-associated dysregulation of splicing factor expression and cellular senescence may derive in part from altered activity of ERK and AKT signaling and may act in part through the ETV6 and FOXO1 transcription factors. Targeting the activity of downstream effectors of ERK and AKT may therefore represent promising targets for future therapeutic intervention.-Latorre, E., Ostler, E. L., Faragher, R. G. A., Harries, L. W. FOXO1 and ETV6 genes may represent novel regulators of splicing factor expression in cellular senescence.


Asunto(s)
Senescencia Celular , Proteína Forkhead Box O1/genética , Proteínas Proto-Oncogénicas c-ets/genética , Factores de Empalme de ARN/metabolismo , Proteínas Represoras/genética , Proliferación Celular , Células Cultivadas , Fibroblastos/citología , Fibroblastos/enzimología , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Quinasas/metabolismo , Transducción de Señal , Proteína ETS de Variante de Translocación 6
7.
Exp Cell Res ; 384(1): 111559, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31425691

RESUMEN

Altered expression of miRNAs is evident in the islets of diabetic human donors, but the effects of specific aspects of the diabetic microenvironment and identity of gene ontology pathways demonstrating target gene enrichment in response to each is understudied. We assessed changes in the miRNA milieu in response to high/low glucose, hypoxia, dyslipidaemia and inflammatory factors in a humanised EndoC-ßH1 beta cell culture system and performed miRPath analysis for each treatment individually. The 10 miRNAs demonstrating the greatest dysregulation across treatments were then independently validated and Gene Set Enrichment Analysis to confirm targeted pathways undertaken. 171 of 392 miRNAs displayed altered expression in response to one or more cellular stressors. miRNA changes were treatment specific, but their target genes were enriched in conserved pathways. 5 miRNAs (miR-136-5p, miR299-5p, miR-454-5p, miR-152 and miR-185) were dysregulated in response to multiple stressors and survived validation in independent samples (p = 0.008, 0.002, 0.012, 0.005 and 0.024 respectively). Target genes of dysregulated miRNAs were clustered into FOXO1, HIPPO and Lysine degradation pathways (p = 0.02, p = 5.84 × 10-5 and p = 3.00 × 10-3 respectively). We provide evidence that the diabetic microenvironment may induce changes to the expression of miRNAs targeting genes enriched in pathways involved in cell stress response and cell survival.


Asunto(s)
Microambiente Celular/genética , Diabetes Mellitus/metabolismo , Proteína Forkhead Box O1/metabolismo , Células Secretoras de Insulina/metabolismo , Lisina/metabolismo , MicroARNs/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Línea Celular , Supervivencia Celular/fisiología , Perfilación de la Expresión Génica/métodos , Ontología de Genes , Redes Reguladoras de Genes/fisiología , Vía de Señalización Hippo , Humanos
9.
BMC Cancer ; 19(1): 86, 2019 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-30658617

RESUMEN

BACKGROUND: Obesity increases breast cancer (BC) risk in post-menopausal women by mostly unknown molecular mechanisms which may partly be regulated by microRNAs (miRNAs). METHODS: We isolated RNA from paired benign and malignant biopsies from 83 BC patients and determined miRNA profiles in samples from 12 women at the extremes of the BMI distribution by RNA-seq. Candidates were validated in all samples. Associations between miR-10b expression and validated target transcript levels, and effects of targeted manipulation of miR-10b levels in a primary BC cell line on proliferation and invasion potential, were explored. RESULTS: Of the 148 miRNAs robustly expressed in breast tissues, the levels of miR-21, miR-10b, miR-451a, miR-30c, and miR-378d were significantly associated with presence of cancer. Of these, miR-10b showed a stronger down-regulation in the tumors of the obese subjects, as opposed to the lean. In ductal but not lobular tumors, significant inverse correlations were observed between the tumor levels of miR-10b and miR-30c and the mRNA levels of cancer-relevant target genes SRSF1, PIEZO1, MAPRE1, CDKN2A, TP-53 and TRA2B, as well as tumor grade. Suppression of miR-10b levels in BT-549 primary BC-derived cells increased cell proliferation and invasive capacity, while exogenous miR-10b mimic decreased invasion. Manipulation of miR-10b levels also inversely affected the mRNA levels of miR-10b targets BCL2L11, PIEZO1 and NCOR2. CONCLUSIONS: Our findings suggest that miR-10b may be a mediator between obesity and cancer in post-menopausal women, regulating several known cancer-relevant genes. MiR-10b expression may have diagnostic and therapeutic implications for the incidence and prognosis of BC in obese women.


Asunto(s)
Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Obesidad/genética , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/patología , Regulación hacia Abajo , Femenino , Perfilación de la Expresión Génica , Humanos , Persona de Mediana Edad , Pronóstico , Células Tumorales Cultivadas
10.
Biogerontology ; 20(5): 649-663, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31292793

RESUMEN

Dysregulation of splicing factor expression is emerging as a driver of human ageing; levels of transcripts encoding splicing regulators have previously been implicated in ageing and cellular senescence both in vitro and in vivo. We measured the expression levels of an a priori panel of 20 age- or senescence-associated splicing factors by qRT-PCR in peripheral blood samples from the InCHIANTI Study of Aging, and assessed longitudinal relationships with human ageing phenotypes (cognitive decline and physical ability) using multivariate linear regression. AKAP17A, HNRNPA0 and HNRNPM transcript levels were all predictively associated with severe decline in MMSE score (p = 0.007, 0.001 and 0.008 respectively). Further analyses also found expression of these genes was associated with a performance decline in two other cognitive measures; the Trail Making Test and the Purdue Pegboard Test. AKAP17A was nominally associated with a decline in mean hand-grip strength (p = 0.023), and further analyses found nominal associations with two other physical ability measures; the Epidemiologic Studies of the Elderly-Short Physical Performance Battery and calculated speed (m/s) during a timed 400 m fast walking test. These data add weight to the hypothesis that splicing dyregulation may contribute to the development of some ageing phenotypes in the human population.


Asunto(s)
Envejecimiento/fisiología , Antígenos/genética , Disfunción Cognitiva , Fuerza de la Mano/fisiología , Ribonucleoproteína Heterogénea-Nuclear Grupo M/genética , Ribonucleoproteínas Nucleares Heterogéneas/genética , Glicoproteínas de Membrana/genética , Factores de Empalme de ARN , Velocidad al Caminar/genética , Anciano , Senescencia Celular/fisiología , Disfunción Cognitiva/diagnóstico , Disfunción Cognitiva/genética , Correlación de Datos , Femenino , Humanos , Masculino , Pruebas de Estado Mental y Demencia , Rendimiento Físico Funcional , Valor Predictivo de las Pruebas , Factores de Empalme de ARN/sangre , Factores de Empalme de ARN/genética
11.
Clin Sci (Lond) ; 132(3): 313-325, 2018 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-29330351

RESUMEN

Coronary heart disease (CHD) is a leading cause of morbidity in people over 65 years of age; >40% of all deaths are due to this condition. The association between increasing age and CHD is well documented; the accumulation of senescent cells in cardiac and vascular tissues may represent one factor underpinning this observation. We aimed to identify senescence-related expression changes in primary human senescent cardiomyocytes and endothelial cells and to relate transcript expression in peripheral blood leucocytes to prevalent and incident CHD in the InCHIANTI study of aging. We quantified splicing factor expression and splicing patterns of candidate transcripts in proliferative and senescent later passage endothelial cells and cardiomyocytes using qRTPCR. Senescence-associated isoforms also expressed in peripheral blood leucocytes were then examined for associations with CHD status in 134 pairs of age, sex and BMI-matched CHD cases and controls. Splicing factor expression was dysregulated in senescent cardiomyocytes, as previously reported for endothelial cells, as was the expression of alternatively expressed cardiac and vascular candidate genes in both cell types. We found nominal associations between the expression of VEGFA156b and FNI-EIIIIA isoforms in peripheral blood mRNA and CHD status. Dysregulated splicing factor expression is a key feature of senescent cardiomyocytes and endothelial cells. Altered splicing of key cardiac or endothelial genes may contribute to the risk of CHD in the human population.


Asunto(s)
Enfermedad Coronaria/epidemiología , Enfermedad Coronaria/genética , Células Endoteliales/citología , Factor A de Crecimiento Endotelial Vascular/genética , Empalme Alternativo/genética , Células Cultivadas , Senescencia Celular/genética , Endotelio Vascular , Humanos , Incidencia , Isoformas de Proteínas/genética , ARN Interferente Pequeño/genética
12.
Nucleic Acids Res ; 44(13): 6318-34, 2016 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-27342281

RESUMEN

4E-Transporter binds eIF4E via its consensus sequence YXXXXLΦ, shared with eIF4G, and is a nucleocytoplasmic shuttling protein found enriched in P-(rocessing) bodies. 4E-T inhibits general protein synthesis by reducing available eIF4E levels. Recently, we showed that 4E-T bound to mRNA however represses its translation in an eIF4E-independent manner, and contributes to silencing of mRNAs targeted by miRNAs. Here, we address further the mechanism of translational repression by 4E-T by first identifying and delineating the interacting sites of its major partners by mass spectrometry and western blotting, including DDX6, UNR, unrip, PAT1B, LSM14A and CNOT4. Furthermore, we document novel binding between 4E-T partners including UNR-CNOT4 and unrip-LSM14A, altogether suggesting 4E-T nucleates a complex network of RNA-binding protein interactions. In functional assays, we demonstrate that joint deletion of two short conserved motifs that bind UNR and DDX6 relieves repression of 4E-T-bound mRNA, in part reliant on the 4E-T-DDX6-CNOT1 axis. We also show that the DDX6-4E-T interaction mediates miRNA-dependent translational repression and de novo P-body assembly, implying that translational repression and formation of new P-bodies are coupled processes. Altogether these findings considerably extend our understanding of the role of 4E-T in gene regulation, important in development and neurogenesis.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Proteínas de Unión al ADN/metabolismo , Factor 4E Eucariótico de Iniciación/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Biosíntesis de Proteínas , Proteínas Proto-Oncogénicas/metabolismo , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos/genética , Sitios de Unión , ARN Helicasas DEAD-box/genética , Proteínas de Unión al ADN/genética , Factor 4E Eucariótico de Iniciación/genética , Factor 4G Eucariótico de Iniciación/genética , Factor 4G Eucariótico de Iniciación/metabolismo , Regulación de la Expresión Génica/genética , Células HEK293 , Células HeLa , Humanos , Proteínas de Transporte Nucleocitoplasmático/genética , Unión Proteica , Mapas de Interacción de Proteínas/genética , Proteínas Proto-Oncogénicas/genética , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/genética , Factores de Transcripción/genética
13.
BMC Cell Biol ; 18(1): 31, 2017 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-29041897

RESUMEN

BACKGROUND: Altered expression of mRNA splicing factors occurs with ageing in vivo and is thought to be an ageing mechanism. The accumulation of senescent cells also occurs in vivo with advancing age and causes much degenerative age-related pathology. However, the relationship between these two processes is opaque. Accordingly we developed a novel panel of small molecules based on resveratrol, previously suggested to alter mRNA splicing, to determine whether altered splicing factor expression had potential to influence features of replicative senescence. RESULTS: Treatment with resveralogues was associated with altered splicing factor expression and rescue of multiple features of senescence. This rescue was independent of cell cycle traverse and also independent of SIRT1, SASP modulation or senolysis. Under growth permissive conditions, cells demonstrating restored splicing factor expression also demonstrated increased telomere length, re-entered cell cycle and resumed proliferation. These phenomena were also influenced by ERK antagonists and agonists. CONCLUSIONS: This is the first demonstration that moderation of splicing factor levels is associated with reversal of cellular senescence in human primary fibroblasts. Small molecule modulators of such targets may therefore represent promising novel anti-degenerative therapies.


Asunto(s)
Senescencia Celular/efectos de los fármacos , Senescencia Celular/genética , Factores de Empalme de ARN/genética , Bibliotecas de Moléculas Pequeñas/farmacología , Estilbenos/farmacología , Empalme Alternativo/efectos de los fármacos , Empalme Alternativo/genética , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Fibroblastos , Humanos , Factores de Empalme de ARN/metabolismo , Resveratrol , Estilbenos/química
14.
Diabetologia ; 59(1): 161-169, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26474776

RESUMEN

AIMS/HYPOTHESIS: Ageing can lead to reduced insulin sensitivity and loss of pancreatic beta cell function, predisposing individuals to the development of diabetes. The aim of this study was to assess the contribution of microRNAs (miRNAs) to age-associated beta cell dysfunction. METHODS: The global mRNA and miRNA profiles of 3- and 12-month-old rat islets were collected by microarray. The functional impact of age-associated differences in miRNA expression was investigated by mimicking the observed changes in primary beta cells from young animals. RESULTS: Beta cells from 12-month-old rats retained normal insulin content and secretion, but failed to proliferate in response to mitotic stimuli. The islets of these animals displayed modifications at the level of several miRNAs, including upregulation of miR-34a, miR-124a and miR-383, and downregulation of miR-130b and miR-181a. Computational analysis of the transcriptomic modifications observed in the islets of 12-month-old rats revealed that the differentially expressed genes were enriched for miR-34a and miR-181a targets. Indeed, the induction of miR-34a and reduction of miR-181a in the islets of young animals mimicked the impaired beta cell proliferation observed in old animals. mRNA coding for alpha-type platelet-derived growth factor receptor, which is critical for compensatory beta cell mass expansion, is directly inhibited by miR34a and is likely to be at least partly responsible for the effects of this miRNA. CONCLUSIONS/INTERPRETATION: Changes in the level of specific miRNAs that occur during ageing affect the proliferative capacity of beta cells. This might reduce their ability to expand under conditions of increased insulin demand, favouring the development of type 2 diabetes.


Asunto(s)
Envejecimiento , Regulación de la Expresión Génica , Células Secretoras de Insulina/citología , Islotes Pancreáticos/citología , MicroARNs/metabolismo , Animales , Apoptosis , Proliferación Celular , Diabetes Mellitus Tipo 2/fisiopatología , Modelos Animales de Enfermedad , Humanos , Insulina/metabolismo , Resistencia a la Insulina , Células Secretoras de Insulina/patología , Islotes Pancreáticos/patología , Masculino , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Transcriptoma , Transfección
15.
Diabetes Obes Metab ; 18(12): 1167-1175, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27550203

RESUMEN

Type 2 diabetes (T2D) affects 415 million people worldwide and is characterized by chronic hyperglycaemia and insulin resistance, progressing to insufficient insulin production, as a result of ß-cell failure. Over time, chronic hyperglycaemia can ultimately lead to loss of ß-cell function, leaving patients insulin-dependent. Until recently the loss of ß-cell mass seen in T2D was considered to be the result of increased rates of apoptosis; however, it has been proposed that apoptosis alone cannot account for the extent of ß-cell mass loss seen in the disease, and that a loss of function may also occur as a result of changes in ß-cell differentiation status. In the present review, we consider current knowledge of determinants of ß-cell fate in the context of understanding its relevance to disease process in T2D, and also the impact of a diabetogenic environment (hyperglycaemia, hypoxia, inflammation and dyslipidaemia) on the expression of genes involved in maintenance of ß-cell identity. We describe current knowledge of the impact of the diabetic microenvironment on gene regulatory processes such alternative splicing, the expression of disallowed genes and epigenetic modifications. Elucidating the molecular mechanisms that underpin changes to ß-cell differentiation status and the concomitant ß-cell failure offers potential treatment targets for the future management of patients with T2D.


Asunto(s)
Desdiferenciación Celular , Diabetes Mellitus Tipo 2/metabolismo , Regulación de la Expresión Génica , Hiperglucemia/metabolismo , Células Secretoras de Insulina/citología , Animales , Apoptosis , Diferenciación Celular , Diabetes Mellitus Tipo 2/genética , Epigénesis Genética , Humanos , Hipoxia/metabolismo , Inflamación/metabolismo , Células Secretoras de Insulina/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
16.
Diabetologia ; 58(4): 745-8, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25634229

RESUMEN

AIMS/HYPOTHESIS: Intronic single nucleotide polymorphisms (SNPs) in the CDKAL1 gene are associated with risk of developing type 2 diabetes. A strong correlation between risk alleles and lower levels of the non-coding RNA, CDKAL1-v1, has recently been reported in whole blood extracted from Japanese individuals. We sought to replicate this association in two independent cohorts: one using whole blood from white UK-resident individuals, and one using a collection of human pancreatic islets, a more relevant tissue type to study with respect to the aetiology of diabetes. METHODS: Levels of CDKAL1-v1 were measured by real-time PCR using RNA extracted from human whole blood (n = 70) and human pancreatic islets (n = 48). Expression with respect to genotype was then determined. RESULTS: In a simple linear regression model, expression of CDKAL1-v1 was associated with the lead type 2 diabetes-associated SNP, rs7756992, in whole blood and islets. However, these associations were abolished or substantially reduced in multiple regression models taking into account rs9366357 genotype: a moderately linked SNP explaining a much larger amount of the variation in CDKAL1-v1 levels, but not strongly associated with risk of type 2 diabetes. CONCLUSIONS/INTERPRETATION: Contrary to previous findings, we provide evidence against a role for dysregulated expression of CDKAL1-v1 in mediating the association between intronic SNPs in CDKAL1 and susceptibility to type 2 diabetes. The results of this study illustrate how caution should be exercised when inferring causality from an association between disease-risk genotype and non-coding RNA expression.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/genética , Diabetes Mellitus Tipo 2/genética , Polimorfismo de Nucleótido Simple , ARN no Traducido/genética , Estudios de Casos y Controles , Quinasa 5 Dependiente de la Ciclina/sangre , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/etnología , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Intrones , Islotes Pancreáticos/química , Modelos Lineales , Fenotipo , ARN no Traducido/sangre , Factores de Riesgo , Reino Unido/epidemiología , Población Blanca/genética , ARNt Metiltransferasas
17.
Biogerontology ; 16(4): 423-34, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25700689

RESUMEN

MicroRNAs are non-coding RNAs with roles in many cellular processes. Tissue-specific miRNA profiles associated with senescence have been described for several cell and tissue types. We aimed to characterise miRNAs involved in core, rather than tissue-specific, senescence pathways by assessment of common miRNA expression differences in two different cell types, with follow-up of predicted targets in human peripheral blood. MicroRNAs were profiled in early and late passage primary lung and skin fibroblasts to identify commonly-deregulated miRNAs. Expression changes of their bioinformatically-predicted mRNA targets were then assessed in both cell types and in human peripheral blood from elderly participants in the InCHIANTI study. 57/178 and 26/492 microRNAs were altered in late passage skin and lung cells respectively. Three miRNAs (miR-92a, miR-15b and miR-125a-3p) were altered in both tissues. 14 mRNA targets of the common miRNAs were expressed in lung and skin fibroblasts, of which two demonstrated up-regulation in late passage skin and lung cells (LYST; p = 0.02 [skin] and 0.02 [lung] INMT; p = 0.03 [skin] and 0.04 [lung]). ZMPSTE24 and LHFPL2 demonstrated altered expression in late passage skin cells only (p = 0.01 and 0.05 respectively). LHFPL2 was also positively correlated with age in peripheral blood (p value = 6.6 × 10(-5)). We find that the majority of senescence-associated miRNAs demonstrate tissue-specific effects. However, miRNAs showing common effects across tissue types may represent those associated with core, rather than tissue-specific senescence processes.


Asunto(s)
Envejecimiento/metabolismo , Senescencia Celular , Fibroblastos/metabolismo , Pulmón/metabolismo , MicroARNs/metabolismo , Piel/metabolismo , Factores de Edad , Envejecimiento/sangre , Envejecimiento/genética , Línea Celular , Proliferación Celular , Senescencia Celular/genética , Biología Computacional , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Humanos , Pulmón/citología , MicroARNs/genética , Piel/citología , Factores de Tiempo
19.
Biochem Soc Trans ; 42(4): 1184-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25110023

RESUMEN

miRNAs are small non-coding regulators of gene expression that are estimated to regulate over 60% of all human genes. Each miRNA can target multiple mRNA targets and as such, miRNAs are responsible for some of the 'fine tuning' of gene expression and are implicated in regulation of all cellular processes. miRNAs bind to target genes by sequence complementarity, resulting in target degradation or translational blocking and usually a reduction in target gene expression. Like mRNA, miRNAs are transcribed from genomic DNA and are processed in several steps that are heavily reliant on correct secondary and tertiary structure. Secondary structure is determined by RNA sequence, which is in turn determined by the sequence of the genome. The human genome, however, like most eukaryotes is variable. Large numbers of SNPs (single nucleotide polymorphisms), small insertions and deletions (indels) and CNVs (copy number variants) have been described in our genome. Should this genetic variation occur in regions critical for the correct secondary structure or target binding, it may interfere with normal gene regulation and cause disease. In this review, we outline the consequences of genetic variation involving different aspects of miRNA biosynthesis, processing and regulation, with selected examples of incidences when this has potential to affect human disease.


Asunto(s)
MicroARNs/genética , Variación Genética/genética , Genoma Humano/genética , Humanos , Polimorfismo de Nucleótido Simple/genética , Regiones Promotoras Genéticas/genética
20.
Reproduction ; 147(4): 419-26, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24231368

RESUMEN

Bisphenol A (BPA) is a widely used plastics constituent that has been associated with endocrine, immune and metabolic effects. Evidence for how BPA exerts significant biological effects at chronic low levels of exposure has remained elusive. In adult men, exposure to BPA has been associated with higher expression of two nuclear receptors, oestrogen receptor-ß (ERß) and oestrogen-related-receptor-α (ERRα), in peripheral white blood cells in vivo. In this study, we explore the expression of ESR2 (ERß) and ESRRA (ERRα) in human leukaemic T-cell lymphoblasts (Jurkat cells) exposed to BPA in vitro. We show that exposure to BPA led to enhanced expression of ESRRA within 6 h of exposure (mean±s.e.m.: 1.43±0.08-fold increase compared with the control, P<0.05). After 72 h, expression of ESRRA remained significantly enhanced at concentrations of BPA ≥1 nM. Oxidative metabolism of BPA by rat liver S9 fractions yields the potent oestrogenic metabolite, 4-methyl-2,4-bis(4-hydroxyphenyl)pent-1-ene (MBP). Exposure of cells to 1-100 nM MBP increased the expression of both ESRRA (significantly induced, P<0.05, at 1, 10, 100 nM) and ESR2 (1.32±0.07-fold increase at 100 nM exposure, P<0.01). ERRα is a major control point for oxidative metabolism in many cell types, including T-cells. Following exposure to both BPA and MBP, we found that cells showed a decrease in cell proliferation rate. Taken together, these results confirm the bioactivity of BPA against putative T-cell targets in vitro at concentrations relevant to general human exposure.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Fenoles/farmacología , Receptores de Estrógenos/genética , Linfocitos T/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Células Jurkat , Receptores de Estrógenos/metabolismo , Linfocitos T/fisiología , Factores de Tiempo , Receptor Relacionado con Estrógeno ERRalfa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA