Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Telemed J E Health ; 2024 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-39049797

RESUMO

Introduction: The COVID-19 pandemic accelerated telemedicine adoption, impacting appointment no-show rates. This study examines neurology appointment preferences among individuals with previous no-shows. Methods: We analyzed transitions between in-person and telemedicine modalities at the Tulane Center for Clinical Neurosciences from August 2020 to February 2021 by race, sex, and insurance type. Logistic regression was used to assess which individual characteristics were associated with switching modalities. Results: A total of 118 patients were included. Transitions to telemedicine visits were significantly higher for female (odds ratio [OR] = 1.868, p = 0.051), Medicaid (OR = 0.433, p = 0.035), and Medicare (OR = 0.228, p = 0.001) beneficiaries compared with males and those with private coverage. Telemedicine to in-person transitions were significantly higher for Medicaid compared with private coverage (OR = 8.133, p = 0.018). Discussion: Females are more likely to switch to telemedicine following an in-person no-show, whereas Medicare beneficiaries are less likely. Medicaid beneficiaries are more likely to revert to in-person appointments. Telemedicine may enhance equitable neurological care, particularly because of its high utilization among females.

4.
Clin Epigenetics ; 15(1): 144, 2023 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-37679776

RESUMO

BACKGROUND: First-degree relatives of type 2 diabetics (FDR) exhibit a high risk of developing type 2 diabetes (T2D) and feature subcutaneous adipocyte hypertrophy, independent of obesity. In FDR, adipose cell abnormalities contribute to early insulin-resistance and are determined by adipocyte precursor cells (APCs) early senescence and impaired recruitment into the adipogenic pathway. Epigenetic mechanisms signal adipocyte differentiation, leading us to hypothesize that abnormal epigenetic modifications cause adipocyte dysfunction and enhance T2D risk. To test this hypothesis, we examined the genome-wide histone profile in APCs from the subcutaneous adipose tissue of healthy FDR. RESULTS: Sequencing-data analysis revealed 2644 regions differentially enriched in lysine 4 tri-methylated H3-histone (H3K4me3) in FDR compared to controls (CTRL) with significant enrichment in mitochondrial-related genes. These included TFAM, which regulates mitochondrial DNA (mtDNA) content and stability. In FDR APCs, a significant reduction in H3K4me3 abundance at the TFAM promoter was accompanied by a reduction in TFAM mRNA and protein levels. FDR APCs also exhibited reduced mtDNA content and mitochondrial-genome transcription. In parallel, FDR APCs exhibited impaired differentiation and TFAM induction during adipogenesis. In CTRL APCs, TFAM-siRNA reduced mtDNA content, mitochondrial transcription and adipocyte differentiation in parallel with upregulation of the CDKN1A and ZMAT3 senescence genes. Furthermore, TFAM-siRNA significantly expanded hydrogen peroxide (H2O2)-induced senescence, while H2O2 did not affect TFAM expression. CONCLUSIONS: Histone modifications regulate APCs ability to differentiate in mature cells, at least in part by modulating TFAM expression and affecting mitochondrial function. Reduced H3K4me3 enrichment at the TFAM promoter renders human APCs senescent and dysfunctional, increasing T2D risk.


Assuntos
Diabetes Mellitus Tipo 2 , Histonas , Humanos , Histonas/genética , Diabetes Mellitus Tipo 2/genética , Peróxido de Hidrogênio , Metilação de DNA , DNA Mitocondrial/genética , Proteínas de Ligação a DNA/genética , Fatores de Transcrição/genética , Proteínas Mitocondriais/genética
5.
Psychol Trauma ; 2023 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-37561435

RESUMO

OBJECTIVE: Currently, there is no single profile of mental health sequela in long-coronavirus disease (COVID) patients, impacting identification, treatment, and exacerbating stigma among this population. This article highlights the rationale for mental health professionals to consider a summary of mental health symptoms in long-COVID patients. METHOD: This article provides an overview of the existing literature regarding the health and mental health impact of long COVID on patients and proposes an approach to conceptualizing mental health symptoms in individuals living with long COVID. This article summarizes the health and mental health impacts of long COVID and underscores the limitations of the current approach to measuring and screening mental health symptoms in long-COVID patients. RESULTS: Long-COVID patients have reported new and worsening mental health symptoms; most frequently reported are depression, anxiety, posttraumatic stress disorder (PTSD), and insomnia. The article concludes by proposing the notion of long-COVID stress symptoms and calls for mental health researchers to identify the unique and complex mental health profiles emerging among this patient population. CONCLUSIONS: Though some long-COVID patients survived life-threatening illnesses and may, therefore, meet the formal criteria for PTSD, many will present with posttraumatic symptomology that mimics PTSD but may not arise from life-threatening medical trauma. A better understanding of the mental health burden of long-COVID stress symptoms is essential to providing efficient and effective mental health treatment, supporting physicians treating long-COVID patients, and enhancing access to and utilization of medical services. (PsycInfo Database Record (c) 2023 APA, all rights reserved).

6.
Cells ; 12(16)2023 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-37626900

RESUMO

The transcription factor HOXA5, from the HOX gene family, has long been studied due to its critical role in physiological activities in normal cells, such as organ development and body patterning, and pathological activities in cancer cells. Nonetheless, recent evidence supports the hypothesis of a role for HOXA5 in metabolic diseases, particularly in obesity and type 2 diabetes (T2D). In line with the current opinion that adipocyte and adipose tissue (AT) dysfunction belong to the group of primary defects in obesity, linking this condition to an increased risk of insulin resistance (IR) and T2D, the HOXA5 gene has been shown to regulate adipocyte function and AT remodeling both in humans and mice. Epigenetics adds complexity to HOXA5 gene regulation in metabolic diseases. Indeed, epigenetic mechanisms, specifically DNA methylation, influence the dynamic HOXA5 expression profile. In human AT, the DNA methylation profile at the HOXA5 gene is associated with hypertrophic obesity and an increased risk of developing T2D. Thus, an inappropriate HOXA5 gene expression may be a mechanism causing or maintaining an impaired AT function in obesity and potentially linking obesity to its associated disorders. In this review, we integrate the current evidence about the involvement of HOXA5 in regulating AT function, as well as its association with the pathogenesis of obesity and T2D. We also summarize the current knowledge on the role of DNA methylation in controlling HOXA5 expression. Moreover, considering the susceptibility of epigenetic changes to reversal through targeted interventions, we discuss the potential therapeutic value of targeting HOXA5 DNA methylation changes in the treatment of metabolic diseases.


Assuntos
Diabetes Mellitus Tipo 2 , Doenças Metabólicas , Humanos , Animais , Camundongos , Fatores de Transcrição/genética , Genes Homeobox , Diabetes Mellitus Tipo 2/genética , Tecido Adiposo , Doenças Metabólicas/genética , Obesidade/genética , Proteínas de Homeodomínio/genética
7.
Cells ; 12(13)2023 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-37443775

RESUMO

Adipose-derived stem cells (ADSCs) play a crucial role in angiogenesis and repair of damaged tissues. However, in pathological conditions including diabetes, ADSC function is compromised. This work aims at evaluating the effect of Methylglyoxal (MGO), a product of chronic hyperglycemia, on mouse ADSCs' (mADSCs) pro-angiogenic function and the molecular mediators involved. The mADSCs were isolated from C57bl6 mice. MGO-adducts and p-p38 MAPK protein levels were evaluated by Western Blot. Human retinal endothelial cell (hREC) migration was analyzed by transwell assays. Gene expression was measured by qRT-PCR, and SA-ßGal activity by cytofluorimetry. Soluble factor release was evaluated by multiplex assay. MGO treatment does not impair mADSC viability and induces MGO-adduct accumulation. hREC migration is reduced in response to both MGO-treated mADSCs and conditioned media from MGO-treated mADSCs, compared to untreated cells. This is associated with an increase of SA-ßGal activity, SASP factor release and p53 and p21 expression, together with a VEGF- and PDGF-reduced release from MGO-treated mADSCs and a reduced p38-MAPK activation in hRECs. The MGO-induced impairment of mADSC function is reverted by senolytics. In conclusion, MGO impairs mADSCs' pro-angiogenic function through the induction of a senescent phenotype, associated with the reduced secretion of growth factors crucial for hREC migration.


Assuntos
Diabetes Mellitus , Aldeído Pirúvico , Humanos , Camundongos , Animais , Aldeído Pirúvico/farmacologia , Aldeído Pirúvico/metabolismo , Óxido de Magnésio , Camundongos Endogâmicos C57BL , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Células-Tronco/metabolismo
9.
EMBO Rep ; 23(7): e52990, 2022 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-35620868

RESUMO

Tight control of glycemia is a major treatment goal for type 2 diabetes mellitus (T2DM). Clinical studies indicated that factors other than poor glycemic control may be important in fostering T2DM progression. Increased levels of methylglyoxal (MGO) associate with complications development, but its role in the early steps of T2DM pathogenesis has not been defined. Here, we show that MGO accumulation induces an age-dependent impairment of glucose tolerance and glucose-stimulated insulin secretion in mice knockdown for glyoxalase 1 (Glo1KD). This metabolic alteration associates with the presence of insular inflammatory infiltration (F4/80-positive staining), the islet expression of senescence markers, and higher levels of cytokines (MCP-1 and TNF-α), part of the senescence-activated secretory profile, in the pancreas from 10-month-old Glo1KD mice, compared with their WT littermates. In vitro exposure of INS832/13 ß-cells to MGO confirms its casual role on ß-cell dysfunction, which can be reverted by senolytic treatment. These data indicate that MGO is capable to induce early phenotypes typical of T2D progression, paving the way for novel prevention approaches to T2DM.


Assuntos
Diabetes Mellitus Tipo 2 , Intolerância à Glucose , Lactoilglutationa Liase/metabolismo , Animais , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Glucose/metabolismo , Intolerância à Glucose/genética , Lactoilglutationa Liase/genética , Óxido de Magnésio , Camundongos , Aldeído Pirúvico/metabolismo
11.
Aging Cell ; 21(3): e13557, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35146866

RESUMO

Senescence of adipose precursor cells (APC) impairs adipogenesis, contributes to the age-related subcutaneous adipose tissue (SAT) dysfunction, and increases risk of type 2 diabetes (T2D). First-degree relatives of T2D individuals (FDR) feature restricted adipogenesis, reflecting the detrimental effects of APC senescence earlier in life and rendering FDR more vulnerable to T2D. Epigenetics may contribute to these abnormalities but the underlying mechanisms remain unclear. In previous methylome comparison in APC from FDR and individuals with no diabetes familiarity (CTRL), ZMAT3 emerged as one of the top-ranked senescence-related genes featuring hypomethylation in FDR and associated with T2D risk. Here, we investigated whether and how DNA methylation changes at ZMAT3 promote early APC senescence. APC from FDR individuals revealed increases in multiple senescence markers compared to CTRL. Senescence in these cells was accompanied by ZMAT3 hypomethylation, which caused ZMAT3 upregulation. Demethylation at this gene in CTRL APC led to increased ZMAT3 expression and premature senescence, which were reverted by ZMAT3 siRNA. Furthermore, ZMAT3 overexpression in APC determined senescence and activation of the p53/p21 pathway, as observed in FDR APC. Adipogenesis was also inhibited in ZMAT3-overexpressing APC. In FDR APC, rescue of ZMAT3 methylation through senolytic exposure simultaneously downregulated ZMAT3 expression and improved adipogenesis. Interestingly, in human SAT, aging and T2D were associated with significantly increased expression of both ZMAT3 and the P53 senescence marker. Thus, DNA hypomethylation causes ZMAT3 upregulation in FDR APC accompanied by acquisition of the senescence phenotype and impaired adipogenesis, which may contribute to FDR predisposition for T2D.


Assuntos
Metilação de DNA , Diabetes Mellitus Tipo 2 , Adipócitos/metabolismo , Adipogenia/genética , Senescência Celular/genética , Metilação de DNA/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Proteína Supressora de Tumor p53/metabolismo
13.
Int J Mol Sci ; 22(21)2021 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-34769081

RESUMO

Diabetes is a severe threat to global health. Almost 500 million people live with diabetes worldwide. Most of them have type 2 diabetes (T2D). T2D patients are at risk of developing severe and life-threatening complications, leading to an increased need for medical care and reduced quality of life. Improved care for people with T2D is essential. Actions aiming at identifying undiagnosed diabetes and at preventing diabetes in those at high risk are needed as well. To this end, biomarker discovery and validation of risk assessment for T2D are critical. Alterations of DNA methylation have recently helped to better understand T2D pathophysiology by explaining differences among endophenotypes of diabetic patients in tissues. Recent evidence further suggests that variations of DNA methylation might contribute to the risk of T2D even more significantly than genetic variability and might represent a valuable tool to predict T2D risk. In this review, we focus on recent information on the contribution of DNA methylation to the risk and the pathogenesis of T2D. We discuss the limitations of these studies and provide evidence supporting the potential for clinical application of DNA methylation marks to predict the risk and progression of T2D.


Assuntos
Metilação de DNA , Diabetes Mellitus Tipo 2/genética , Animais , Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/patologia , Progressão da Doença , Epigênese Genética , Humanos , Medição de Risco
14.
FASEB J ; 35(4): e21357, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33710685

RESUMO

First-degree relatives (FDRs) of type 2 diabetics (T2D) feature dysfunction of subcutaneous adipose tissue (SAT) long before T2D onset. miRNAs have a role in adipocyte precursor cells (APC) differentiation and in adipocyte identity. Thus, impaired miRNA expression may contribute to SAT dysfunction in FDRs. In the present work, we have explored changes in miRNA expression associated with T2D family history which may affect gene expression in SAT APCs from FDRs. Small RNA-seq was performed in APCs from healthy FDRs and matched controls and omics data were validated by qPCR. Integrative analyses of APC miRNome and transcriptome from FDRs revealed down-regulated hsa-miR-23a-5p, -193a-5p and -193b-5p accompanied by up-regulated Insulin-like Growth Factor 2 (IGF2) gene which proved to be their direct target. The expression changes in these marks were associated with SAT adipocyte hypertrophy in FDRs. APCs from FDRs further demonstrated reduced capability to differentiate into adipocytes. Treatment with IGF2 protein decreased APC adipogenesis, while over-expression of hsa-miR-23a-5p, -193a-5p and -193b-5p enhanced adipogenesis by IGF2 targeting. Indeed, IGF2 increased the Wnt Family Member 10B gene expression in APCs. Down-regulation of the three miRNAs and IGF2 up-regulation was also observed in Peripheral Blood Leukocytes (PBLs) from FDRs. In conclusion, APCs from FDRs feature a specific miRNA/gene profile, which associates with SAT adipocyte hypertrophy and appears to contribute to impaired adipogenesis. PBL detection of this profile may help in identifying adipocyte hypertrophy in individuals at high risk of T2D.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Predisposição Genética para Doença , Fator de Crescimento Insulin-Like II/metabolismo , MicroRNAs/metabolismo , Adipogenia , Clonagem Molecular , Diabetes Mellitus Tipo 2/genética , Família , Regulação da Expressão Gênica , Humanos , Fator de Crescimento Insulin-Like II/genética , MicroRNAs/genética
15.
Cytokine Growth Factor Rev ; 58: 1-15, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33674185

RESUMO

SARS-CoV-2 is a novel coronavirus that severely affects the respiratory system, is the cause of the COVID-19 pandemic, and is projected to result in the deaths of 2 million people worldwide. Recent reports suggest that SARS-CoV-2 also affects the central nervous system along with other organs. COVID-19-associated complications are observed in older people with underlying neurological conditions like stroke, Alzheimer's disease, and Parkinson's disease. Hence, we discuss SARS-CoV-2 viral replication and its inflammation-mediated infection. This review also focuses on COVID-19 associated neurological complications in individuals with those complications as well as other groups of people. Finally, we also briefly discuss the current therapies available to treat patients, as well as ongoing available treatments and vaccines for effective cures with a special focus on the therapeutic potential of a small 5 amino acid peptide (PHSCN), ATN-161, that inhibits SARS-CoV-2 spike protein binding to both integrin α5ß1 and α5ß1/hACE2.


Assuntos
COVID-19/complicações , Doenças do Sistema Nervoso/virologia , Inflamação Neurogênica/virologia , SARS-CoV-2/patogenicidade , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , COVID-19/epidemiologia , Humanos , Doenças do Sistema Nervoso/epidemiologia , Inflamação Neurogênica/complicações , Neuroimunomodulação/fisiologia , Pandemias
16.
Nutrients ; 12(11)2020 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-33203037

RESUMO

Exposure to endocrine-disrupting chemicals such as Bisphenol-A (BPA) is associated with an increase in obesity prevalence. Diet is the primary cause of human exposure to this contaminant. BPA promotes obesity by inducing adipocyte dysfunction and altering adipogenesis. Contradictory evidence and unanswered questions are reported in the literature concerning the BPA effects on adipogenesis. To clarify this issue, we tested the effects of prolonged low-dose BPA exposure on different phases of adipogenesis in committed 3T3L1 and uncommitted NIH3T3 preadipocytes. Our findings show that BPA effects on the adipogenesis are mediated by epigenetic mechanisms by reducing peroxisome proliferator-activated receptor gamma (Pparγ) promoter methylation in preadipocytes. Nevertheless, in BPA-exposed 3T3L1, Pparγ expression only transiently increases as lipid accumulation at day 4 of differentiation, without altering the adipogenic potential of the precursor cells. In the absence of differentiation mix, BPA does not make the 3T3L1 an in vitro model of spontaneous adipogenesis and the effects on the Pparγ expression are still limited at day 4 of differentiation. Furthermore, BPA exposure does not commit the NIH3T3 to the adipocyte lineage, although Pparγ overexpression is more evident both in preadipocytes and during the adipocyte differentiation. Interestingly, termination of the BPA exposure restores the Pparγ promoter methylation and inflammatory profile of the 3T3L1 cells. This study shows that BPA induces epigenetic changes in a key adipogenic gene. These modifications are reversible and do not affect preadipocyte commitment and/or differentiation. We identify an alternative transcriptional mechanism by which BPA affects gene expression and demonstrate how the challenge of preventing exposure is fundamental for human health.


Assuntos
Adipócitos/efeitos dos fármacos , Tecido Adiposo/metabolismo , Compostos Benzidrílicos/farmacologia , Epigênese Genética/efeitos dos fármacos , PPAR gama/genética , PPAR gama/metabolismo , Fenóis/farmacologia , Regiões Promotoras Genéticas , Células 3T3-L1 , Adipogenia/efeitos dos fármacos , Animais , Diferenciação Celular , Disruptores Endócrinos , Poluentes Ambientais , Contaminação de Alimentos , Expressão Gênica , Inflamação , Resistência à Insulina , Camundongos , Células NIH 3T3 , Obesidade/metabolismo
17.
Epigenomics ; 12(10): 873-888, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32483983

RESUMO

Aim: First-degree relatives (FDR) of individuals with Type 2 diabetes (T2D) feature restricted adipogenesis, which render them more vulnerable to T2D. Epigenetics may contribute to these abnormalities. Methods: FDR pre-adipocyte Methylome and Transcriptome were investigated by MeDIP- and RNA-Seq, respectively. Results:Methylome analysis revealed 2841 differentially methylated regions (DMR) in FDR. Most DMR localized into gene-body and were hypomethylated. The strongest hypomethylation signal was identified in an intronic-DMR at the PTPRD gene. PTPRD hypomethylation in FDR was confirmed by bisulphite sequencing and was responsible for its upregulation. Interestingly, Ptprd-overexpression in 3T3-L1 pre-adipocytes inhibited adipogenesis. Notably, the validated PTPRD-associated DMR was significantly hypomethylated in peripheral blood leukocytes from the same FDR individuals. Finally, PTPRD methylation pattern was also replicated in obese individuals. Conclusion: Our findings indicated a previously unrecognized role of PTPRD in restraining adipogenesis. This abnormality may contribute to increase FDR proclivity toward T2D.


Assuntos
Adipogenia/genética , Metilação de DNA , Diabetes Mellitus Tipo 2/genética , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores/genética , Células 3T3-L1 , Adulto , Animais , Epigênese Genética , Feminino , Humanos , Masculino , Camundongos
18.
Front Oncol ; 10: 602153, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33634024

RESUMO

BACKGROUND: Thymic epithelial tumors (TETs) are rare thoracic malignancies, commonly divided into two different histopathological entities, thymoma (T) and thymic carcinoma (TC). To date, there are no specific biomarkers for monitoring the biological course of these rare tumors. We carried out a single center study aiming at the detection of circulating cell-free DNA (ccfDNA) and the correlation of its levels with metastatic dissemination and histological subtype in patients with TETs. METHODS: From July 2018 to January 2020, 5-ml blood samples from 26 patients with advanced TET (aTET) (11 patients with TC and 15 patients with T) and from six patients with completely resected TET (cr-TET), were prospectively obtained before the initiation of systemic therapy. Blood samples from 10 healthy donors were used as control. The QIAamp MinElute ccfDNA Kits was used for ccfDNA isolation from plasma; real-time PCR was used for cfDNA quantification. RESULTS: We found significantly higher ccfDNA amount in patients with T and TC compared to controls, with median ccfDNA level of 3.3 ng/µl, 11.4 ng/µl and 25.6 ng/µl, for healthy donors, T and TC patients, respectively (p<0.001). No significant difference was found between cr-TET and controls (p = 0.175). ccfDNA concentrations were higher in metastatic (M1a and M1b) compared to non-metastatic (M0) TETs (25.6 ng/µl versus 7.2 ng/µl; p= 0.037). No significant correlation was found either between ccfDNA and disease stage, according to both the Masaoka-Koga (p= 0.854) and the TNM 8th edition staging systems (p = 0.66), or between ccfDNA levels and overall tumor burden, estimated according RECIST 1.1 criteria (r = 0.07, p = 0.725). CONCLUSIONS: To the best of our knowledge, this is the first study that prospectively explores detection and quantification of ccfDNA in TETs. Higher baseline cfDNA levels have been observed in both advanced T and TC comparing to the control group.

19.
Clin Epigenetics ; 11(1): 181, 2019 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-31801613

RESUMO

BACKGROUND: Obesity is a major worldwide threat to human health. Increasing evidence indicates that epigenetic modifications have a major impact on the natural history of this disorder. Ankyrin Repeat Domain 26 (Ankrd26) is involved in the development of both obesity and diabetes in mice and is modulated by environmentally induced epigenetic modifications. This study aims at investigating whether impaired ANKRD26 gene expression and methylation occur in human obesity and whether they correlate to the phenotype of these subjects. RESULTS: We found that downregulation of ANKRD26 mRNA and hyper-methylation of a specific region of the ANKRD26 promoter, embedding the CpG dinucleotides - 689, - 659, and - 651 bp, occur in peripheral blood leukocytes from obese compared with the lean subjects. ANKRD26 gene expression correlates inversely to the percentage of DNA methylation at these 3 CpG sites. Luciferase assays reveal a cause-effect relationship between DNA methylation at the 3 CpG sites and ANKRD26 gene expression. Finally, both ANKRD26 mRNA levels and CpG methylation correlate to body mass index and to the pro-inflammatory status and the increased cardio-metabolic risk factors of these same subjects. CONCLUSION: Downregulation of the ANKRD26 gene and hyper-methylation at specific CpGs of its promoter are common abnormalities in obese patients. These changes correlate to the pro-inflammatory profile and the cardio-metabolic risk factors of the obese individuals, indicating that, in humans, they mark adverse health outcomes.


Assuntos
Metilação de DNA , Regulação para Baixo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Obesidade/sangue , Obesidade/genética , Adulto , Índice de Massa Corporal , Estudos de Casos e Controles , HDL-Colesterol/sangue , LDL-Colesterol/sangue , Ilhas de CpG , DNA/sangue , Epigênese Genética , Feminino , Humanos , Masculino , Obesidade/complicações , Regiões Promotoras Genéticas , Fatores de Risco , Triglicerídeos/sangue
20.
Int J Mol Sci ; 20(12)2019 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-31248068

RESUMO

A healthy diet improves life expectancy and helps to prevent common chronic diseases such as type 2 diabetes (T2D) and obesity. The mechanisms driving these effects are not fully understood, but are likely to involve epigenetics. Epigenetic mechanisms control gene expression, maintaining the DNA sequence, and therefore the full genomic information inherited from our parents, unchanged. An interesting feature of epigenetic changes lies in their dynamic nature and reversibility. Accordingly, they are susceptible to correction through targeted interventions. Here we will review the evidence supporting a role for nutritional factors in mediating metabolic disease risk through DNA methylation changes. Special emphasis will be placed on the potential of using DNA methylation traits as biomarkers to predict risk of obesity and T2D as well as on their response to dietary and pharmacological (epi-drug) interventions.


Assuntos
Metilação de DNA , Diabetes Mellitus Tipo 2/etiologia , Dieta , Suscetibilidade a Doenças , Obesidade/etiologia , Animais , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animais de Doenças , Epigênese Genética , Humanos , Camundongos , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Medição de Risco , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA