Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
1.
BMC Med Genomics ; 17(1): 18, 2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-38212800

RESUMEN

BACKGROUND: This study aimed to screen and validate noise-induced hearing loss (NIHL) associated single nucleotide polymorphisms (SNPs), construct genetic risk prediction models, and evaluate higher-order gene-gene, gene-environment interactions for NIHL in Chinese population. METHODS: First, 83 cases and 83 controls were recruited and 60 candidate SNPs were genotyped. Then SNPs with promising results were validated in another case-control study (153 cases and 252 controls). NIHL-associated SNPs were identified by logistic regression analysis, and a genetic risk model was constructed based on the genetic risk score (GRS), and classification and regression tree (CART) analysis was used to evaluate interactions among gene-gene and gene-environment. RESULTS: Six SNPs in five genes were significantly associated with NIHL risk (p < 0.05). A positive dose-response relationship was found between GRS values and NIHL risk. CART analysis indicated that strongest interaction was among subjects with age ≥ 45 years and cumulative noise exposure ≥ 95 [dB(A)·years], without personal protective equipment, and carried GJB2 rs3751385 (AA/AB) and FAS rs1468063 (AA/AB) (OR = 10.038, 95% CI = 2.770, 47.792), compared with the referent group. CDH23, FAS, GJB2, PTPRN2 and SIK3 may be NIHL susceptibility genes. CONCLUSION: GRS values may be utilized in the evaluation of the cumulative effect of genetic risk for NIHL based on NIHL-associated SNPs. Gene-gene, gene-environment interaction patterns play an important role in the incidence of NIHL.


Asunto(s)
Pérdida Auditiva Provocada por Ruido , Ruido en el Ambiente de Trabajo , Humanos , Persona de Mediana Edad , Estudios de Casos y Controles , China/epidemiología , Predisposición Genética a la Enfermedad , Puntuación de Riesgo Genético , Genotipo , Pérdida Auditiva Provocada por Ruido/genética , Pérdida Auditiva Provocada por Ruido/epidemiología , Polimorfismo de Nucleótido Simple , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/genética
2.
Cancer Med ; 10(16): 5524-5533, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34272834

RESUMEN

PURPOSE: The homeobox (HOX) family plays an important role in multi-biological processes, such as morphogenesis and tumors. However, the function of HOXD13 in colon cancer remains unclear. MATERIALS AND METHODS: The Cancer Genome Atlas database was used to analyze the expression of HOXD13 and its effect on the survival rate of colon cancer patients. Wound healing, Transwell, and clone formation were used to evaluate the effects of changes in HOXD13 expression on the function of colon cancer cells. A nude mouse xenograft tumor model was used to test the effects of HOXD13 on tumor growth in vivo. RESULTS: Our results showed that HOXD13 was highly expressed in colon cancer and predicted a poor prognosis for patients. In in vitro experiments, the knockdown of HOXD13 can inhibit the proliferation and invasion of colon cancer cells. In vivo experiments showed the inhibited tumor growth after the knockdown of HODX13. In addition, HOXD13 bound to the protein tyrosine phosphatase receptor type N2 (PTPRN2) promoter and promoted the transcription of PTPRN2. CONCLUSION: We revealed the function and mechanism of HOXD13 in colon cancer and suggest that HOXD13 may be a candidate marker for the diagnosis and treatment of colon cancer.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias del Colon/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/genética , Factores de Transcripción/metabolismo , Animales , Línea Celular Tumoral , Neoplasias del Colon/diagnóstico , Neoplasias del Colon/mortalidad , Neoplasias del Colon/terapia , Progresión de la Enfermedad , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Pronóstico , Regiones Promotoras Genéticas/genética , Tasa de Supervivencia , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Environ Health ; 20(1): 43, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33849548

RESUMEN

BACKGROUND: Between 1962 and 1971, the US Air Force sprayed Agent Orange across Vietnam, exposing many soldiers to this dioxin-containing herbicide. Several negative health outcomes have been linked to Agent Orange exposure, but data is lacking on the effects this chemical has on the genome. Therefore, we sought to characterize the impact of Agent Orange exposure on DNA methylation in the whole blood and adipose tissue of veterans enrolled in the Air Force Health Study (AFHS). METHODS: We received adipose tissue (n = 37) and whole blood (n = 42) from veterans in the AFHS. Study participants were grouped as having low, moderate, or high TCDD body burden based on their previously measured serum levels of dioxin. DNA methylation was assessed using the Illumina 450 K platform. RESULTS: Epigenome-wide analysis indicated that there were no FDR-significantly methylated CpGs in either tissue with TCDD burden. However, 3 CpGs in the adipose tissue (contained within SLC9A3, LYNX1, and TNRC18) were marginally significantly (q < 0.1) hypomethylated, and 1 CpG in whole blood (contained within PTPRN2) was marginally significantly (q < 0.1) hypermethylated with high TCDD burden. Analysis for differentially methylated DNA regions yielded SLC9A3, among other regions in adipose tissue, to be significantly differentially methylated with higher TCDD burden. Comparing whole blood data to a study of dioxin exposed adults from Alabama identified a CpG within the gene SMO that was hypomethylated with dioxin exposure in both studies. CONCLUSION: We found limited evidence of dioxin associated DNA methylation in adipose tissue and whole blood in this pilot study of Vietnam War veterans. Nevertheless, loci in the genes of SLC9A3 in adipose tissue, and PTPRN2 and SMO in whole blood, should be included in future exposure analyses.


Asunto(s)
Tejido Adiposo/metabolismo , Agente Naranja , Sustancias para la Guerra Química , Metilación de ADN , Defoliantes Químicos , Veteranos , Guerra de Vietnam , Proteínas Adaptadoras Transductoras de Señales/genética , Anciano , Anciano de 80 o más Años , Islas de CpG , Exposición a Riesgos Ambientales , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Persona de Mediana Edad , Proyectos Piloto , Dibenzodioxinas Policloradas/sangre , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/genética , Intercambiador 3 de Sodio-Hidrógeno/genética
4.
Sci Rep ; 11(1): 5022, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33658578

RESUMEN

We hypothesized that epigenetics is a link between smoking/allergen exposures and the development of Asthma and chronic obstructive pulmonary disease (ACO). A total of 75 of 228 COPD patients were identified as ACO, which was independently associated with increased exacerbations. Microarray analysis identified 404 differentially methylated loci (DML) in ACO patients, and 6575 DML in those with rapid lung function decline in a discovery cohort. In the validation cohort, ACO patients had hypermethylated PDE9A (+ 30,088)/ZNF323 (- 296), and hypomethylated SEPT8 (- 47) genes as compared with either pure COPD patients or healthy non-smokers. Hypermethylated TIGIT (- 173) gene and hypomethylated CYSLTR1 (+ 348)/CCDC88C (+ 125,722)/ADORA2B (+ 1339) were associated with severe airflow limitation, while hypomethylated IFRD1 (- 515) gene with frequent exacerbation in all the COPD patients. Hypermethylated ZNF323 (- 296) / MPV17L (+ 194) and hypomethylated PTPRN2 (+ 10,000) genes were associated with rapid lung function decline. In vitro cigarette smoke extract and ovalbumin concurrent exposure resulted in specific DNA methylation changes of the MPV17L / ZNF323 genes, while 5-aza-2'-deoxycytidine treatment reversed promoter hypermethylation-mediated MPV17L under-expression accompanied with reduced apoptosis and decreased generation of reactive oxygen species. Aberrant DNA methylations may constitute a determinant for ACO, and provide a biomarker of airflow limitation, exacerbation, and lung function decline.


Asunto(s)
Asma/genética , Metilación de ADN , Epigénesis Genética , Enfermedad Pulmonar Obstructiva Crónica/genética , Fumar/efectos adversos , 3',5'-AMP Cíclico Fosfodiesterasas/genética , 3',5'-AMP Cíclico Fosfodiesterasas/metabolismo , Anciano , Anciano de 80 o más Años , Alérgenos/efectos adversos , Asma/complicaciones , Asma/etiología , Asma/metabolismo , Estudios de Cohortes , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Análisis por Micromatrices , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Persona de Mediana Edad , Fenotipo , Enfermedad Pulmonar Obstructiva Crónica/complicaciones , Enfermedad Pulmonar Obstructiva Crónica/etiología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor de Adenosina A2B/genética , Receptor de Adenosina A2B/metabolismo , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/genética , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Receptores de Leucotrienos/genética , Receptores de Leucotrienos/metabolismo , Pruebas de Función Respiratoria , Septinas/genética , Septinas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
5.
Diabetes ; 70(4): 932-943, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33419759

RESUMEN

We and others previously demonstrated that a type 1 diabetes genetic risk score (GRS) improves the ability to predict disease progression and onset in at-risk subjects with islet autoantibodies. Here, we hypothesized that GRS and islet autoantibodies, combined with age at onset and disease duration, could serve as markers of residual ß-cell function following type 1 diabetes diagnosis. Generalized estimating equations were used to investigate whether GRS along with insulinoma-associated protein-2 autoantibody (IA-2A), zinc transporter 8 autoantibody (ZnT8A), and GAD autoantibody (GADA) titers were predictive of C-peptide detection in a largely cross-sectional cohort of 401 subjects with type 1 diabetes (median duration 4.5 years [range 0-60]). Indeed, a combined model with incorporation of disease duration, age at onset, GRS, and titers of IA-2A, ZnT8A, and GADA provided superior capacity to predict C-peptide detection (quasi-likelihood information criterion [QIC] = 334.6) compared with the capacity of disease duration, age at onset, and GRS as the sole parameters (QIC = 359.2). These findings support the need for longitudinal validation of our combinatorial model. The ability to project the rate and extent of decline in residual C-peptide production for individuals with type 1 diabetes could critically inform enrollment and benchmarking for clinical trials where investigators are seeking to preserve or restore endogenous ß-cell function.


Asunto(s)
Autoanticuerpos/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/metabolismo , Autoanticuerpos/genética , Péptido C/genética , Péptido C/metabolismo , Estudios Transversales , Diabetes Mellitus Tipo 1/genética , Humanos , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/genética , Transportador 8 de Zinc/genética , Transportador 8 de Zinc/metabolismo
6.
J Cell Mol Med ; 25(1): 411-420, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33242228

RESUMEN

Noise-induced hearing loss (NIHL) seriously affects the life quality of humans and causes huge economic losses to society. To identify novel genetic loci involved in NIHL, we conducted a genome-wide association study (GWAS) for this symptom in Chinese populations. GWAS scan was performed in 89 NIHL subjects (cases) and 209 subjects with normal hearing who have been exposed to a similar noise environment (controls), followed by a replication study consisting of 53 cases and 360 controls. We identified that four candidate pathways were nominally significantly associated with NIHL, including the Erbb, Wnt, hedgehog and intraflagellar transport pathways. In addition, two novel index single-nucleotide polymorphisms, rs35075890 in the intron of AUTS2 gene at 7q11.22 (combined P = 1.3 × 10-6 ) and rs10081191 in the intron of PTPRN2 gene at 7q36.3 (combined P = 2.1 × 10-6 ), were significantly associated with NIHL. Furthermore, the expression quantitative trait loci analyses revealed that in brain tissues, the genotypes of rs35075890 are significantly associated with the expression levels of AUTS2, and the genotypes of rs10081191 are significantly associated with the expressions of PTPRN2 and WDR60. In conclusion, our findings highlight two novel loci at 7q11.22 and 7q36.3 conferring susceptibility to NIHL.


Asunto(s)
Estudio de Asociación del Genoma Completo/métodos , Pérdida Auditiva Provocada por Ruido/genética , Polimorfismo de Nucleótido Simple/genética , Proteínas Adaptadoras Transductoras de Señales/genética , China , Proteínas del Citoesqueleto/genética , Predisposición Genética a la Enfermedad/genética , Humanos , Masculino , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/genética , Factores de Transcripción/genética
7.
J Parkinsons Dis ; 9(2): 389-400, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30958317

RESUMEN

BACKGROUND: DNA methylation studies in Parkinson's disease (PD) thus far have focused on disease susceptibility but not progression. OBJECTIVE: In this epigenome-wide association study (EWAS), we aim to identify methylation markers associated with faster cognitive decline or motor progression in PD. METHODS: We included 232 PD patients from the Parkinson's Environment and Gene follow-up study who provided blood samples at enrolment. Information on cognitive and motor function was collected using the Mini-Mental State Examination (MMSE) and Unified Parkinson's Disease Rating Scale (UPDRS). For EWAS analyses, we used a robust measure of correlation: biweight midcorrelations, t-tests, and Cox proportional hazard models. We also conducted weighted correlation network analysis (WGCNA) to identify CpG modules associated with cognitive decline or motor progression in PD. RESULTS: Among 197 individuals of European ancestry, with our EWAS approach we identified 7 genome-wide significant CpGs associated with a MMSE 4-point decline and 8 CpGs associated with faster motor progression (i.e., rate of UPDRS increase ≥5-point/year). The most interesting CpGs for cognitive decline include cg17445913 in KCNB1 (cor = 0.36, p = 6.85×10-7) and cg02920897 in DLEU2 (cor = 0.34, p = 3.23×10-6), while for motor progression it was cg01754178 in PTPRN2 (cor = - 0.34, p = 2.07×10-6). In WGCNA, motor progression related modules were enriched for genes related to neuronal synaptic functions, Wnt signaling pathway, and mitochondrial apoptosis. CONCLUSIONS: Our study provides the first epigenetic evidence that differential methylation in genes previously identified as being associated with cognitive impairment, neuronal synaptic function, Wnt signaling pathway, and mitochondrial apoptosis is associated with cognitive and motor progression in PD.


Asunto(s)
Disfunción Cognitiva/genética , Metilación de ADN/genética , Epigenoma , Enfermedad de Parkinson/genética , Anciano , Apoptosis/genética , Progresión de la Enfermedad , Femenino , Humanos , Estudios Longitudinales , Masculino , Mitocondrias , Pronóstico , Modelos de Riesgos Proporcionales , ARN Largo no Codificante , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/genética , Canales de Potasio Shab/genética , Sinapsis , Transferasas/genética , Vía de Señalización Wnt/genética
8.
J Biol Chem ; 294(21): 8564-8576, 2019 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-30979722

RESUMEN

Type 1 diabetes islet cell autoantigen 512 (ICA512/IA-2) is a tyrosine phosphatase-like intrinsic membrane protein involved in the biogenesis and turnover of insulin secretory granules (SGs) in pancreatic islet ß-cells. Whereas its membrane-proximal and cytoplasmic domains have been functionally and structurally characterized, the role of the ICA512 N-terminal segment named "regulated endocrine-specific protein 18 homology domain" (RESP18HD), which encompasses residues 35-131, remains largely unknown. Here, we show that ICA512 RESP18HD residues 91-131 encode for an intrinsically disordered region (IDR), which in vitro acts as a condensing factor for the reversible aggregation of insulin and other ß-cell proteins in a pH and Zn2+-regulated fashion. At variance with what has been shown for other granule cargoes with aggregating properties, the condensing activity of ICA512 RESP18HD is displayed at a pH close to neutral, i.e. in the pH range found in the early secretory pathway, whereas it is resolved at acidic pH and Zn2+ concentrations resembling those present in mature SGs. Moreover, we show that ICA512 RESP18HD residues 35-90, preceding the IDR, inhibit insulin fibrillation in vitro Finally, we found that glucose-stimulated secretion of RESP18HD upon exocytosis of SGs from insulinoma INS-1 cells is associated with cleavage of its IDR, conceivably to prevent its aggregation upon exposure to neutral pH in the extracellular milieu. Taken together, these findings point to ICA512 RESP18HD being a condensing factor for protein sorting and granulogenesis early in the secretory pathway and for prevention of amyloidogenesis.


Asunto(s)
Amiloide/metabolismo , Insulina/metabolismo , Proteínas Intrínsecamente Desordenadas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/metabolismo , Amiloide/genética , Animales , Línea Celular Tumoral , Humanos , Concentración de Iones de Hidrógeno , Insulina/genética , Proteínas Intrínsecamente Desordenadas/genética , Proteínas del Tejido Nervioso/genética , Ratas , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/genética , Zinc/metabolismo
9.
Sci Rep ; 9(1): 4855, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30890718

RESUMEN

Protein tyrosine phosphatase, receptor type N2 (PTPRN2) encodes a major islet autoantigen in type-1 diabetes. Previous genetic studies have shown its significant association with obesity. PTPRN2 plays an important role in epigenetic regulation of metabolic diseases and cancers. We investigated CpG methylations (cg17429772 and cg158269415) in PTPRN2 by pyrosequencing from blood samples of childhood obesity (n = 638). cg158269415 had significant positive correlations with body mass index (BMI) and waist-hip ratio (WHR). Case-control analysis showed that cg158269415 methylation in blood sample was significantly more hypermethylated in obese cases (n = 252), an average of 2.93% more than that that in controls (n = 386). The cg158269415 methylation has a trimodal distribution pattern with strong dependency on nearby located rs1670344 G > A genotype. Correlations of cg158269415 with BMI and WHR were significant and strong in major G allele carriers (GG + GA). Our study showed that an epigenetic association of PTPRN2 gene with childhood obesity was under certain genetic background. The genetic and epigenetic interplay of PTPRN2 gene may implicate a mechanism of childhood obesity. Whether these small changes in DNA methylation from whole blood are causally or consequently related to childhood obesity outcome and their clinical relevance remains to be determined. However, this study presents a promising obesity risk marker that warrants further investigation.


Asunto(s)
Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Obesidad Infantil/genética , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/genética , Adolescente , Niño , Islas de CpG/genética , Metilación de ADN/genética , Femenino , Humanos , Masculino , Obesidad Infantil/patología
10.
Sci Rep ; 9(1): 2890, 2019 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-30814595

RESUMEN

The intracellular life of insulin secretory granules (ISGs) from biogenesis to secretion depends on their structural (e.g. size) and dynamic (e.g. diffusivity, mode of motion) properties. Thus, it would be useful to have rapid and robust measurements of such parameters in living ß-cells. To provide such measurements, we have developed a fast spatiotemporal fluctuation spectroscopy. We calculate an imaging-derived Mean Squared Displacement (iMSD), which simultaneously provides the size, average diffusivity, and anomalous coefficient of ISGs, without the need to extract individual trajectories. Clustering of structural and dynamic quantities in a multidimensional parametric space defines the ISGs' properties for different conditions. First, we create a reference using INS-1E cells expressing proinsulin fused to a fluorescent protein (FP) under basal culture conditions and validate our analysis by testing well-established stimuli, such as glucose intake, cytoskeleton disruption, or cholesterol overload. After, we investigate the effect of FP-tagged ISG protein markers on the structural and dynamic properties of the granule. While iMSD analysis produces similar results for most of the lumenal markers, the transmembrane marker phogrin-FP shows a clearly altered result. Phogrin overexpression induces a substantial granule enlargement and higher mobility, together with a partial de-polymerization of the actin cytoskeleton, and reduced cell responsiveness to glucose stimulation. Our data suggest a more careful interpretation of many previous ISG-based reports in living ß-cells. The presented data pave the way to high-throughput cell-based screening of ISG structure and dynamics under various physiological and pathological conditions.


Asunto(s)
Glucosa/farmacología , Proteínas Fluorescentes Verdes/metabolismo , Secreción de Insulina/fisiología , Células Secretoras de Insulina/fisiología , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/metabolismo , Vesículas Secretoras/fisiología , Animales , Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Ratas , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/genética , Vesículas Secretoras/efectos de los fármacos , Edulcorantes/farmacología
11.
Scand J Clin Lab Invest ; 79(1-2): 123-125, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30727763

RESUMEN

The GAD65 and IA-2 antibodies (Abs) are biomarkers of the development of type 1 diabetes mellitus (T1DM) in both children and adults. The upper reference limit for the autoantibodies made by the manufacture was established on an adult Chinese population. Here, we established upper reference limits for Northern European adults and children in accordance with the Clinical and Laboratory Standards Institute (CLSI) guidelines. Serum samples from healthy Danish children (0-18 years) and adults (18-70 years) were analysed for GAD65Ab and IA-2Ab using MAGLUMI 800 Chemiluminescence Immunoassay (CLIA). The Kruskal-Wallis test was used for evaluating differences between gender and age groups. No gender or age differences were found for neither GAD65Ab nor IA-2Ab, and a combined upper reference limit for both children and adults could be established. An upper reference limit of 5.1 IU/mL was defined for GAD65Ab and 11.5 U/mL for IA-2Ab. Our results showed a substantial discrepancy with the reference limits established by the manufacturer.


Asunto(s)
Autoanticuerpos/sangre , Glutamato Descarboxilasa/antagonistas & inhibidores , Inmunoensayo/normas , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/antagonistas & inhibidores , Adolescente , Adulto , Anciano , Biomarcadores/sangre , Niño , Preescolar , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/diagnóstico , Diabetes Mellitus Tipo 1/etnología , Diabetes Mellitus Tipo 1/inmunología , Europa (Continente) , Femenino , Expresión Génica , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/inmunología , Voluntarios Sanos , Humanos , Lactante , Recién Nacido , Luminiscencia , Masculino , Persona de Mediana Edad , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/genética , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/inmunología , Valores de Referencia , Población Blanca
12.
Sci Rep ; 9(1): 2382, 2019 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-30787400

RESUMEN

The genetics underlying autism spectrum disorder (ASD) are complex. Approximately 3-5% of ASD cases arise from maternally inherited duplications of 15q11.2-q13.1, termed Duplication 15q syndrome (Dup15q). 15q11.2-q13.1 includes the gene UBE3A which is believed to underlie ASD observed in Dup15q syndrome. UBE3A is an E3 ubiquitin ligase that targets proteins for degradation and trafficking, so finding UBE3A substrates and interacting partners is critical to understanding Dup15q ASD. In this study, we take an unbiased genetics approach to identify genes that genetically interact with Dube3a, the Drosophila melanogaster homolog of UBE3A. We conducted an enhancer/suppressor screen using a rough eye phenotype produced by Dube3a overexpression with GMR-GAL4. Using the DrosDel deficiency kit, we identified 3 out of 346 deficiency lines that enhanced rough eyes when crossed to two separate Dube3a overexpression lines, and subsequently identified IA2, GABA-B-R3, and lola as single genes responsible for rough eye enhancement. Using the FlyLight GAL4 lines to express uas-Dube3a + uas-GFP in the endogenous lola pattern, we observed an increase in the GFP signal compared to uas-GFP alone, suggesting a transcriptional co-activation effect of Dube3a on the lola promoter region. These findings extend the role of Dube3a/UBE3A as a transcriptional co-activator, and reveal new Dube3a interacting genes.


Asunto(s)
Trastorno del Espectro Autista/genética , Proteínas de Drosophila , Drosophila melanogaster/genética , Elementos de Facilitación Genéticos , Genes Supresores , Discapacidad Intelectual/genética , Receptores de GABA-B , Ubiquitina-Proteína Ligasas/fisiología , Animales , Aberraciones Cromosómicas , Cromosomas Humanos Par 15/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología , Epistasis Genética/genética , Estudio de Asociación del Genoma Completo/métodos , Humanos , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/genética , Receptores de GABA-B/genética , Receptores de GABA-B/fisiología , Factores de Transcripción/genética , Activación Transcripcional/genética , Ubiquitina-Proteína Ligasas/genética
13.
BMC Med Genomics ; 11(1): 96, 2018 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-30382873

RESUMEN

BACKGROUND: Glioblastoma multiforme, the most prevalent and aggressive brain tumour, has a poor prognosis. The molecular mechanisms underlying gliomagenesis remain poorly understood. Therefore, molecular research, including various markers, is necessary to understand the occurrence and development of glioma. METHOD: Weighted gene co-expression network analysis (WGCNA) was performed to construct a gene co-expression network in TCGA glioblastoma samples. Gene ontology (GO) and pathway-enrichment analysis were used to identify significance of gene modules. Cox proportional hazards regression model was used to predict outcome of glioblastoma patients. RESULTS: We performed weighted gene co-expression network analysis (WGCNA) and identified a gene module (yellow module) related to the survival time of TCGA glioblastoma samples. Then, 228 hub genes were calculated based on gene significance (GS) and module significance (MS). Four genes (OSMR + SOX21 + MED10 + PTPRN) were selected to construct a Cox proportional hazards regression model with high accuracy (AUC = 0.905). The prognostic value of the Cox proportional hazards regression model was also confirmed in GSE16011 dataset (GBM: n = 156). CONCLUSION: We developed a promising mRNA signature for estimating overall survival in glioblastoma patients.


Asunto(s)
Neoplasias Encefálicas/genética , Redes Reguladoras de Genes/genética , Glioblastoma/genética , Área Bajo la Curva , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Glioblastoma/mortalidad , Glioblastoma/patología , Humanos , Estimación de Kaplan-Meier , Masculino , Complejo Mediador/genética , Persona de Mediana Edad , Subunidad beta del Receptor de Oncostatina M/genética , Pronóstico , Modelos de Riesgos Proporcionales , Curva ROC , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/genética , Factores de Transcripción SOXB2/genética
14.
J Transl Med ; 16(1): 297, 2018 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-30359270

RESUMEN

BACKGROUND: Neurogenin3 (Ngn3) and neurogenic differentiation 1 (NeuroD1), two crucial transcriptional factors involved in human diabetes (OMIM: 601724) and islet development, have been previously found to directly target to the E-boxes of the insulinoma-associated 2 (Insm2) gene promoter, thereby activating the expression of Insm2 in insulin-secretion cells. However, little is known about the function of Insm2 in pancreatic islets and glucose metabolisms. METHODS: Homozygous Insm2-/- mice were generated by using the CRISPR-Cas9 method. Glucose-stimulated insulin secretion and islet morphology were analyzed by ELISA and immunostainings. Expression levels of Insm2-associated molecules were measured using quantitative RT-PCR and Western blots. RESULTS: Fasting blood glucose levels of Insm2-/- mice were higher than wild-type counterparts. Insm2-/- mice also showed reduction in glucose tolerance and insulin/C-peptide levels when compared to the wild-type mice. RT-PCR and Western blot analysis revealed that expression of Insm1 was significantly increased in Insm2-/- mice, suggesting a compensatory response of the homolog gene Insm1. Similarly, transcriptional levels of Ngn3 and NeuroD1 were also increased in Insm2-/- mice. Moreover, Insm2-/- female mice showed a significantly decreased reproductive capacity. CONCLUSIONS: Our findings suggest that Insm2 is important in glucose-stimulated insulin secretion and is involved in the development pathway of neuroendocrine tissues which are regulated by the transcription factors Ngn3, NeuroD1 and Insm1.


Asunto(s)
Eliminación de Gen , Intolerancia a la Glucosa/genética , Secreción de Insulina , Factores de Transcripción/genética , Animales , Secuencia de Bases , Femenino , Genotipo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Masculino , Ratones Noqueados , Modelos Biológicos , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/genética , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/metabolismo
15.
Eur J Hum Genet ; 26(11): 1588-1596, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29976977

RESUMEN

Obesity is a multifactorial condition that is highly heritable. There have been ~60 susceptibility loci identified, but they only account for a fraction of cases. As copy number variations (CNVs) have been implicated in the etiology of a multitude of human disorders including obesity, here, we investigated the contribution of rare (<1% population frequency) CNVs in pediatric cases of obesity. We genotyped 67 such individuals, including 22 with co-morbid developmental delay and prioritized rare CNVs at known obesity-associated loci, as well as, those impacting genes involved in energy homeostasis or related processes. We identified clinically relevant or potentially clinically relevant CNVs in 15% (10/67) of individuals. Of these, 4% (3/67) had 16p11.2 microdeletions encompassing the known obesity risk gene SH2B1. Notably, we identified two unrelated probands harboring different 6p22.2 microduplications encompassing SCGN, a potential novel candidate gene for obesity. Further, we identified other biologically relevant candidate genes for pediatric obesity including ARID5B, GPR39, PTPRN2, and HNF4G. We found previously reported candidate loci for obesity, and new ones, suggesting CNV analysis may assist in the diagnosis of pediatric obesity.


Asunto(s)
Variaciones en el Número de Copia de ADN , Sitios Genéticos , Obesidad/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Adolescente , Preescolar , Proteínas de Unión al ADN/genética , Femenino , Estudio de Asociación del Genoma Completo , Factor Nuclear 4 del Hepatocito/genética , Humanos , Masculino , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/genética , Receptores Acoplados a Proteínas G/genética , Secretagoginas/genética , Factores de Transcripción/genética
16.
Sci Rep ; 8(1): 10074, 2018 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-29973665

RESUMEN

To investigate whether CTLA-4 +49 G/A (rs231775), a tagSNP in Asian, is a functional T1D SNP, we genotyped this SNP with 1035 T1D patients and 2575 controls in Chinese Han population. And 1280 controls measured insulin release and sensitivity based on an oral glucose tolerance test; 283 newly diagnosed T1D patients assayed C-peptide level based on a mixed-meal tolerance test. 31 controls were analyzed for different T cell subsets by multi-color flow cytometry. Under additive model, we found that CTLA-4 +49 G/A was significantly associated with T1D (P = 2.82E-04, OR = 1.25, 95% CI: 1.12-1.41), which was further confirmed by meta-analysis (P = 1.19E-08, OR = 1.65, 95% CI: 1.38-1.96) in Chinese Han population. Although we did not find any association between this SNP and beta-cell function in either healthy individuals or newly diagnosed T1D patients, healthy individuals carrying GG/GA genotypes had lower CTLA-4 expression in naïve or activated CD4 Treg subsets (P = 0.0046 and 0.0317 respectively). A higher positive rate of IA-2A was observed among T1D patients with GG genotype compared with AA (OR = 0.51, 95% CI: 0.30-0.84, p = 0.008). Collectively, CTLA-4 +49 G/A reached a GWAS significant association with T1D risk in Chinese Han population, affects CTLA-4 expression in Treg subsets and subsequently humoral immunity in T1D patients.


Asunto(s)
Antígeno CTLA-4/genética , Diabetes Mellitus Tipo 1/genética , Insulina/genética , Linfocitos T Reguladores/metabolismo , Adulto , Pueblo Asiatico , Péptido C/sangre , Antígeno CTLA-4/sangre , Antígeno CTLA-4/inmunología , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Femenino , Regulación de la Expresión Génica/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Genotipo , Prueba de Tolerancia a la Glucosa , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple/genética , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/genética , Factores de Riesgo , Linfocitos T Reguladores/inmunología , Adulto Joven
17.
Cell Physiol Biochem ; 46(6): 2335-2346, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29742497

RESUMEN

BACKGROUND/AIMS: During the occurrence and progression of hepatocellular carcinoma (HCC), phosphotyrosine phosphatases (PTPs) are usually described as tumor suppressors or proto-oncogenes, and to some degree are correlated with the prognosis of HCC. METHODS: A total of 321 patients from the Cancer Genome Atlas (TCGA) database and 180 patients from our validated cohort with hepatocellular carcinoma were recruited in this study. Kaplan-Meier, univariate and multivariate Cox proportional hazards model were used to evaluate the risk factors for survival. Quantitative real-time PCR (qRT-PCR) and immunohistochemistry (IHC) were applied to detect the expression levels of PTP genes. RESULTS: After screening the data of TCGA, we identified five PTPs as HCC overall survival related PTP genes, among which only three (PTPN12, PTPRN, PTPN18) exhibited differential expression levels in our 180 paired HCC and adjacent tissues (P< 0.001). Further analysis revealed that expression of PTPN18 was positively, but PTPRN was negatively associated with prognosis of HCC both in TCGA cohort and our own cohort. As to PTPN12, results turned out to be opposite according to HBV status. In detail, higher expression of PTPN12 was associated with better outcome in HBV group but worse prognosis in Non-HBV group. CONCLUSION: Our results suggested that PTPN12, PTPRN and PTPN18 were independent prognostic factors in HCC.


Asunto(s)
Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 12/genética , Proteínas Tirosina Fosfatasas no Receptoras/genética , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/genética , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Hepatocelular/diagnóstico , Estudios de Cohortes , Femenino , Regulación de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Neoplasias Hepáticas/diagnóstico , Masculino , Persona de Mediana Edad , Pronóstico , Proteína Tirosina Fosfatasa no Receptora Tipo 12/análisis , Proteínas Tirosina Fosfatasas no Receptoras/análisis , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/análisis
18.
J Biol Chem ; 293(16): 5920-5933, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29483197

RESUMEN

Autocrine insulin signaling is critical for pancreatic ß-cell growth and activity and is at least partially controlled by protein-tyrosine phosphatases (PTPs) that act on insulin receptors (IRs). The receptor-type PTP phogrin primarily localizes on insulin secretory granules in pancreatic ß cells. We recently reported that phogrin knockdown decreases the protein levels of insulin receptor substrate 2 (IRS2), whereas high-glucose stimulation promotes formation of a phogrin-IR complex that stabilizes IRS2. However, the underlying molecular mechanisms by which phogrin affects IRS2 levels are unclear. Here, we found that relative to wildtype mice, IRS2 levels in phogrin-knockout mice islets decreased by 44%. When phogrin was silenced by shRNA in pancreatic ß-cell lines, glucose-induced insulin signaling led to proteasomal degradation of IRS2 via a negative feedback mechanism. Phogrin overexpression in a murine hepatocyte cell line consistently prevented chronic insulin treatment-induced IRS2 degradation. In vitro, phogrin directly bound the IR without the assistance of other proteins and protected recombinant PTP1B from oxidation to potentiate its activity toward the IR. Furthermore, phogrin expression suppressed insulin-induced local generation of hydrogen peroxide and subsequent PTP1B oxidation, which allowed progression of IR dephosphorylation. Together, these results suggest that a transient interaction of phogrin with the IR enables glucose-stimulated autocrine insulin signaling through the regulation of PTP1B activity, which is essential for suppressing feedback-mediated IRS2 degradation in pancreatic ß cells.


Asunto(s)
Glucosa/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/metabolismo , Transducción de Señal , Animales , Línea Celular , Femenino , Silenciador del Gen , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteolisis , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/genética
19.
Vaccine ; 35(51): 7187-7197, 2017 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-29169891

RESUMEN

Type 1 diabetes is a chronic organ-specific autoimmune disease in which selective destruction of insulin-producing ß cells leads to impaired glucose metabolism and its attendant complications. IA2(5)P2-1, a potent immunogenic carrier which designed by our laboratory, can induce high titer specific antibodies when carry a B cell epitope, such as B cell epitopes of DPP4, xanthine oxidase, and Urate transporter protein. In this report, we describe a novel multi-epitope vaccine composing a peptide of DPP4, an anti-diabetic B epitope of Insulinoma antigen-2(IA-2) and a Th2 epitope (P2:IPALDSLTPANED) of P277 peptide in human heat shock protein 60 (HSP60). Immunization with the multi-epitope vaccine in non-obese diabetic (NOD) mice successfully induced specific anti-DPP4 antibody, inhibited plasma DPP4 activity, and increased serum GLP-1 level. Moreover, this antibody titer was correlated with the dose of immunization (20µg, 100µg). Inoculation of this vaccine in NOD mice significantly control blood glucose level, improved glucose excursion and increased insulin level in vivo. Consistent with a lower diabetic and insulitis incidence, a induced splenic T cells proliferation and tolerance were observed. IFN-γ secretion reduced and IL-10 increased significantly in the D41-IA2(5)-P2-1 treated mice compared to P277 and control group due to the potential immunomodulatory effect of the epitope in the vaccine. Immunohistochemical analysis and cytometry showed a rebalance of Th1/Th2 in NOD mice. Our results demonstrate that this multi-epitope vaccine may serve as a promising therapeutic approach for type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/terapia , Dipeptidil Peptidasa 4/inmunología , Epítopos de Linfocito B/inmunología , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/inmunología , Vacunas de Subunidad/inmunología , Vacunas/uso terapéutico , Animales , Formación de Anticuerpos , Glucemia/análisis , Chaperonina 60/inmunología , Citocinas/biosíntesis , Citocinas/inmunología , Diabetes Mellitus Tipo 1/inmunología , Dipeptidil Peptidasa 4/sangre , Dipeptidil Peptidasa 4/química , Epítopos de Linfocito B/química , Epítopos de Linfocito B/genética , Péptido 1 Similar al Glucagón/sangre , Humanos , Insulina/sangre , Ratones , Ratones Endogámicos NOD , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/genética , Linfocitos T/inmunología , Vacunas/administración & dosificación , Vacunas/inmunología , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/química
20.
Nat Genet ; 49(6): 825-833, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28481342

RESUMEN

The contributions of coding mutations to tumorigenesis are relatively well known; however, little is known about somatic alterations in noncoding DNA. Here we describe GECCO (Genomic Enrichment Computational Clustering Operation) to analyze somatic noncoding alterations in 308 pancreatic ductal adenocarcinomas (PDAs) and identify commonly mutated regulatory regions. We find recurrent noncoding mutations to be enriched in PDA pathways, including axon guidance and cell adhesion, and newly identified processes, including transcription and homeobox genes. We identified mutations in protein binding sites correlating with differential expression of proximal genes and experimentally validated effects of mutations on expression. We developed an expression modulation score that quantifies the strength of gene regulation imposed by each class of regulatory elements, and found the strongest elements were most frequently mutated, suggesting a selective advantage. Our detailed single-cancer analysis of noncoding alterations identifies regulatory mutations as candidates for diagnostic and prognostic markers, and suggests new mechanisms for tumor evolution.


Asunto(s)
Adenocarcinoma/genética , Carcinoma Ductal Pancreático/genética , Regulación Neoplásica de la Expresión Génica , Mutación , Neoplasias Pancreáticas/genética , Adenocarcinoma/mortalidad , Carcinoma Ductal Pancreático/mortalidad , Humanos , Neoplasias Pancreáticas/mortalidad , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/genética , Simportadores de Cloruro de Sodio-Potasio/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA