Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Oral Dis ; 2022 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-36398465

RESUMEN

OBJECTIVES: To provide a comprehensive characterization of DNA methylome of oral tongue squamous cell carcinoma (OTSCC) and identify novel tumor-specific DNA methylation markers for early detection using saliva. MATERIAL AND METHODS: Genome-wide DNA methylation analysis including six OTSCC matched adjacent non-tumoral tissue and saliva was performed using Infinium MethylationEPIC array. Differentially methylated levels of selected genes in our OTSCC cohort were further validated using OTSCC methylation data from The Cancer Genome Atlas database (TCGA). The methylation levels of a set of tumor-specific hypermethylated genes associated with a downregulated expression were evaluated in saliva. Receiver operating characteristic (ROC) curves were performed to assess the diagnostic value of DNA methylation markers. RESULTS: A total of 25,890 CpGs (20,505 hypomethylated and 5385 hypermethylated) were differentially methylated (DMCpGs) between OTSCC and adjacent non-tumoral tissue. Hypermethylation of 11 tumor-specific genes was validated in OTSCC TCGA cohort. Of these 11 genes, A2BP1, ANK1, ALDH1A2, GFRA1, TTYH1, and PDE4B were also hypermethylated in saliva. These six salivary methylated genes showed high diagnostic accuracy (≥0.800) for discriminating patients from controls. CONCLUSIONS: This is the first largest genome-wide DNA methylation study on OTSCC that identifies a group of novel tumor-specific DNA methylation markers with diagnostic potential in saliva.

2.
J Cell Mol Med ; 15(9): 1927-40, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21029370

RESUMEN

The role of obestatin, a 23-amino-acid peptide encoded by the ghrelin gene, on the control of the metabolism of pre-adipocyte and adipocytes as well as on adipogenesis was determined. For in vitro assays, pre-adipocyte and adipocyte 3T3-L1 cells were used to assess the obestatin effect on cell metabolism and adipogenesis based on the regulation of the key enzymatic nodes, Akt and AMPK and their downstream targets. For in vivo assays, white adipose tissue (WAT) was obtained from male rats under continuous subcutaneous infusion of obestatin. Obestatin activated Akt and its downstream targets, GSK3α/ß, mTOR and S6K1, in 3T3-L1 adipocyte cells. Simultaneously, obestatin inactivated AMPK in this cell model. In keeping with this, ACC phosphorylation was also decreased. This fact was confirmed in vivo in white adipose tissue (omental, subcutaneous and gonadal) obtained from male rats under continuous sc infusion of obestatin (24 and 72 hrs). The relevance of obestatin as regulator of adipocyte metabolism was supported by AS160 phosphorylation, GLUT4 translocation and augment of glucose uptake in 3T3-L1 adipocyte cells. In contrast, obestatin failed to modify translocation of fatty acid transporters, FATP1, FATP4 and FAT/CD36, to plasma membrane. Obestatin treatment in combination with IBMX and DEX showed to regulate the expression of C/EBPα, C/EBPß, C/EBPδ and PPARγ promoting adipogenesis. Remarkable, preproghrelin expression, and thus obestatin expression, increased during adipogenesis being sustained throughout terminal differentiation. Neutralization of endogenous obestatin secreted by 3T3-L1 cells by anti-obestatin antibody decreased adipocyte differentiation. Furthermore, knockdown experiments by preproghrelin siRNA supported that obestatin contributes to adipogenesis. In summary, obestatin promotes adipogenesis in an autocrine/paracrine manner, being a regulator of adipocyte metabolism. These data point to a putative role in the pathogenesis of metabolic syndrome.


Asunto(s)
Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adipogénesis/efectos de los fármacos , Ghrelina/farmacología , Células 3T3-L1 , Adenilato Quinasa/metabolismo , Adipocitos/enzimología , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/enzimología , Animales , Comunicación Autocrina/efectos de los fármacos , Western Blotting , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Activación Enzimática/efectos de los fármacos , Proteínas de Transporte de Ácidos Grasos/metabolismo , Ghrelina/metabolismo , Glucosa/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Masculino , Ratones , Comunicación Paracrina/efectos de los fármacos , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
3.
Clin Epigenetics ; 10: 60, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29719581

RESUMEN

Background: Lack of vitamin D (VD) has been associated with colorectal cancer (CRC). VD has anti-inflammatory effects and regulates several cellular pathways by means of its receptor, including epigenetic modifications. Adipose tissue dysfunction has been related to low-grade inflammation, which is related to diseases like cancer. The aim of this study was to explore the relationship between serum 25-hydroxyvitamin D (25(OH)D), adipose tissue gene expression of VD receptor (VDR), pro-inflammatory markers, and the epigenetic factor DNA methyltransferase 3a (DNMT3A) as well as VDR promoter methylation in CRC. Methods: Blood and visceral adipose tissue from 57 CRC and 50 healthy control subjects were collected. CRC subjects had lower serum 25(OH)D levels and higher VDR gene expression, and these were negatively correlated in the CRC group. Results: Adipose tissue NFκB1, IL6, and IL1B gene expression were higher in the CRC subjects than in the control subjects. 25(OH)D correlated negatively with NFκB1 and CRP. In turn, CRP correlated positively with NFκB1, IL6, IL1B, and VDR gene expression as well as NFκB1 that correlated positively with IL6 and IL1B. DNMT3A mRNA was negatively correlated with serum 25(OH)D and positively correlated with VDR DNA methylation. VDR DNA methylation at position 4 had lower levels in the CRC group. Global NFκB1 methylation at dinucleotide 3 was lower in the CRC group. Conclusion: Our results suggest that adipose tissue may be a key factor in CRC development. The low 25(OH)D levels and high adipose tissue VDR expression in CRC may, at least in part, mediate this relationship by modifying adipose tissue DNA methylation and promoting inflammation.


Asunto(s)
Neoplasias Colorrectales/genética , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN , Subunidad p50 de NF-kappa B/genética , Receptores de Calcitriol/genética , Regulación hacia Arriba , Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Anciano , Proteína C-Reactiva/metabolismo , Estudios de Casos y Controles , Neoplasias Colorrectales/sangre , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/metabolismo , ADN Metiltransferasa 3A , Epigénesis Genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Interleucina-1beta/genética , Interleucina-6/genética , Masculino , Persona de Mediana Edad , Subunidad p50 de NF-kappa B/metabolismo , Regiones Promotoras Genéticas , Receptores de Calcitriol/metabolismo , Vitamina D/análogos & derivados , Vitamina D/sangre
4.
Endocrine ; 55(2): 435-446, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27738888

RESUMEN

The secretion of the hepatokine alpha-2-Heremans-Schmid glycoprotein/Fetuin A, implicated in pathological processes including systemic insulin resistance, by adipose tissue has been recently described. Thus, we have recently identified its presence in white adipose tissue secretomes by mass spectrometry. However, the secretion pattern and function of adipose-derived alpha-2-Heremans-Schmid glycoprotein are poorly understood. The aim of this study is to evaluate the expression and secretion of total and active phosphorylated alpha-2-Heremans-Schmid glycoprotein by adipose tissue from visceral and subcutaneous localizations in animals at different physiological and nutritional status including anorexia and obesity. Alpha-2-Heremans-Schmid glycoprotein expression and secretion in visceral adipose tissue and subcutaneous adipose tissue explants from animals under fasting and exercise training, at pathological situations such as anorexia and obesity, and from human obese individuals were assayed by immunoblotting, quantitative real-time polymerase chain reaction and enzyme-linked immunosorbent assay. We reveal that visceral adipose tissue expresses and secretes more alpha-2-Heremans-Schmid glycoprotein than subcutaneous adipose tissue, and that this secretion is diminished after fasting and exercise training. Visceral adipose tissue from anorectic animals showed reduced alpha-2-Heremans-Schmid glycoprotein secretion; on the contrary, alpha-2-Heremans-Schmid glycoprotein is over-secreted by visceral adipose tissue in the occurrence of obesity. While secretion of active-PhophoSer321α2HSG by visceral adipose tissue is independent of body mass index, we found that the fraction of active-alpha-2-Heremans-Schmid glycoprotein secreted by subcutaneous adipose tissue increments significantly in situations of obesity. Functional studies show that the inhibition of adipose-derived alpha-2-Heremans-Schmid glycoprotein increases insulin sensitivity in differentiated adipocytes. In conclusion, visceral adipose tissue secretes more alpha-2-Heremans-Schmid glycoprotein than subcutaneous adipose tissue and this secretion is more sensitive to nutritional and physiological changes. The over-secretion of alpha-2-Heremans-Schmid glycoprotein by visceral adipose tissue, the increased secretion of the active phosphorylated form by subcutaneous adipose tissuein obese animals, and the adipose-derived alpha-2-Heremans-Schmid glycoprotein capacity to inhibit the insulin pathway suggest the participation of adipose-derived alpha-2-Heremans-Schmid glycoprotein in the deleterious effects of obesity.


Asunto(s)
Grasa Intraabdominal/metabolismo , Obesidad/metabolismo , Grasa Subcutánea/metabolismo , alfa-2-Glicoproteína-HS/metabolismo , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Animales , Células Cultivadas , Femenino , Humanos , Insulina/metabolismo , Insulina/farmacología , Resistencia a la Insulina/fisiología , Masculino , Fosforilación , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
5.
J Invest Dermatol ; 135(12): 3144-3152, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26302069

RESUMEN

Notch is a family of transmembrane receptors that participate in the regulation of cell differentiation, proliferation, and stemness. Notch pathway activation has also been found associated with different human cancers including primary cutaneous T-cell lymphomas (CTCL). The elucidation of the mechanisms driving Notch activation in these particular diseases has remained elusive. Here we studied the possibility that DNA methylation at Notch pathway gene promoters and/or deregulation of Notch-associated microRNAs contribute to activate Notch in mycosis fungoides (MF). By genome-wide DNA methylation analysis, we failed to detect any consistent methylation at the Notch1, the Notch-ligand Jagged1, or the Notch-target Hes1 gene promoters, but found a significant methylation of the Notch-related microRNAs, in particular miR-200c and miR-124. Downregulation of miR-200c is associated with overexpression of Jagged1, concomitant to Notch1 activation. CTCL cell lines were infected with lentiviral vector encoding for miR-200c and ectopic expression of miR-200c in CTCL lines resulted in Jagged1 protein downregulation associated with a reduction in the levels of active Notch1. Our study deciphers an epigenetic mechanism regulating the Notch pathway in (MF) that might contribute to the future design of more specific therapeutic strategies.


Asunto(s)
Epigénesis Genética , Linfoma Cutáneo de Células T/genética , MicroARNs/fisiología , Micosis Fungoide/genética , Receptor Notch1/fisiología , Transducción de Señal/fisiología , Neoplasias Cutáneas/genética , Proteínas de Unión al Calcio/fisiología , Línea Celular Tumoral , Metilación de ADN , Humanos , Péptidos y Proteínas de Señalización Intercelular/fisiología , Proteína Jagged-1 , Proteínas de la Membrana/fisiología , Estudios Retrospectivos , Proteínas Serrate-Jagged
6.
J Endocrinol ; 210(2): R1-7, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21690170

RESUMEN

This study aimed to investigate the role of preproghrelin-derived peptides in adipogenesis. Immunocytochemical analysis of 3T3-L1 adipocyte cells showed stronger preproghrelin expression compared with that observed in 3T3-L1 preadipocyte cells. Insulin promoted this expression throughout adipogenesis identifying mTORC1 as a critical downstream substrate for this profile. The role of preproghrelin-derived peptides on the differentiation process was supported by preproghrelin knockdown experiments, which revealed its contribution to adipogenesis. Neutralization of endogenous O-acyl ghrelin (acylated ghrelin), unacylated ghrelin, and obestatin by specific antibodies supported their adipogenic potential. Furthermore, a parallel increase in the expression of ghrelin-associated enzymatic machinery, prohormone convertase 1/3 (PC1/3) and membrane-bound O-acyltransferase 4 (MBOAT4), was dependent on the expression of preproghrelin in the course of insulin-induced adipogenesis. The coexpression of preproghrelin system and their receptors, GHSR1a and GPR39, during adipogenesis supports an autocrine/paracrine role for these peptides. Preproghrelin, PC1/3, and MBOAT4 exhibited dissimilar expression depending on the white fat depot, revealing their regulation in a positive energy balance situation in mice. The results underscore a key role for preproghrelin-derived peptides on adipogenesis through an autocrine/paracrine mechanism.


Asunto(s)
Adipocitos/metabolismo , Adipogénesis/fisiología , Insulina/metabolismo , Hormonas Peptídicas/metabolismo , Precursores de Proteínas/metabolismo , Células 3T3-L1 , Adipocitos/citología , Animales , Masculino , Ratones , ARN Interferente Pequeño
7.
Arch Med Res ; 40(4): 306-10, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19608021

RESUMEN

BACKGROUND AND AIMS: Carrying variants on the leptin receptor gene (LEPR) may have an impact on inflammatory and stress markers. Thus, the aim of the study was to analyze the role of the Lys109Arg LEPR gene polymorphism on inflammatory (leptin and IL-6) and stress (cortisol) markers in obese subjects who followed a hypocaloric diet designed to lose weight. METHODS: One hundred and seventy (80 females/90 males) Caucasian subjects (body mass index: 30.8 +/- 2.4 kg/m(2)), were genotyped for the Lys109Arg polymorphism by a PCR/RFLP procedure. Anthropometric measurements were assessed and blood samples were drawn in all the volunteers before and after an 8-week energy-restricted diet (-30% E). Plasma levels of leptin as well as interleukin-6 (IL-6) as proinflammatory markers and circulating cortisol concentrations as a stress hormone were measured. RESULTS: Weight loss (-6.1 +/- 2.7%; p <0.001) induced significant changes in anthropometric and biochemical determinations. The AA genotype group showed a higher fat mass loss as well as greater total cholesterol decrease compared with the minor allele carriers. Moreover, the G allele carriers were associated with a higher basal risk of inflammation (OR = 2.5; p = 0.042) and stress (OR = 3.3; p = 0.011), which were reduced after weight lowering (p >0.05). CONCLUSIONS: The Arg allele carriers of the Lys109Arg LEPR gene polymorphism were associated with an increased proinflammatory state and stress condition at baseline. These obesity-related markers were importantly decreased after following a hypocaloric diet.


Asunto(s)
Inflamación/genética , Obesidad/genética , Receptores de Leptina/genética , Estrés Fisiológico/genética , Pérdida de Peso/genética , Adulto , Alelos , Índice de Masa Corporal , Restricción Calórica , Femenino , Genotipo , Humanos , Hidrocortisona/sangre , Interleucina-6/sangre , Leptina/sangre , Masculino , Persona de Mediana Edad , Polimorfismo Genético
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA