Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 5.857
Filtrer
1.
Sci Rep ; 14(1): 15717, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38977823

RÉSUMÉ

Obesity is a global health concern and independent risk factor for cancers including hepatocellular carcinoma (HCC). However, evidence on the causal links between obesity and HCC is limited and inconclusive. This study aimed to investigate the causal relationship between obesity-related traits and HCC risk and explore underlying mechanisms using bioinformatics approaches. Two-sample Mendelian randomization analysis was conducted leveraging publicly available genome-wide association study summary data on obesity traits (body mass index, body fat percentage, waist circumference, waist-to-hip ratio, visceral adipose tissue volume) and HCC. Associations of obesity with primary mechanisms (insulin resistance, adipokines, inflammation) and their effects on HCC were examined. Differentially expressed genes in obesity and HCC were identified and functional enrichment analyses were performed. Correlations with tumor microenvironment (TME) and immunotherapy markers were analyzed. Genetically predicted higher body mass index and body fat percentage showed significant causal relationships with increased HCC risk. Overall obesity also demonstrated causal links with insulin resistance, circulating leptin levels, C-reactive protein levels and risk of severe insulin resistant type 2 diabetes. Four differentially expressed genes (ESR1, GCDH, FAHD2A, DCXR) were common in obesity and HCC. Enrichment analyses indicated their roles in processes like RNA capping, viral transcription, IL-17 signaling and endocrine resistance. They exhibited negative correlations with immune cell infiltration and immunotherapy markers in HCC. Overall obesity likely has a causal effect on HCC risk in Europeans, possibly via influencing primary mechanisms. The identified differentially expressed genes may be implicated in obesity-induced hepatocarcinogenesis through regulating cell cycle, inflammation and immune evasion. Further research on precise mechanisms is warranted.


Sujet(s)
Carcinome hépatocellulaire , Étude d'association pangénomique , Tumeurs du foie , Obésité , Humains , Carcinome hépatocellulaire/génétique , Tumeurs du foie/génétique , Obésité/complications , Obésité/génétique , Indice de masse corporelle , Facteurs de risque , Insulinorésistance/génétique , Microenvironnement tumoral/génétique , Analyse de randomisation mendélienne
2.
Zhonghua Liu Xing Bing Xue Za Zhi ; 45(7): 932-940, 2024 Jul 10.
Article de Chinois | MEDLINE | ID: mdl-39004964

RÉSUMÉ

Objective: To explore the association between insulin resistance (IR) and genome-wide DNA methylation based on Shanghai twin study. Methods: Monozygotic twins (MZ) from Shanghai were recruited during 2012-2013, 2017-2018, and 2022-2023. Data were collected by questionnaire survey, physical examination and laboratory tests. Genome-wide DNA methylation was quantified. Generalized linear mixed effect model was applied to analyze the association between methylation level at each site and homeostatic model assessment 2-insulin resistance (HOMA2-IR). Non-paired and paired designs were used to assess the association between DNA methylation and phenotype of IR. Cluster analysis was conducted to identify the clusters of top significant sites. Generalized linear regression was performed to examine the differential methylation patterns from clusters. Results: A total of 100 MZ pairs were included in this study. Hypermethylated cg10535199-2q23.1 (ß=0.74%, P=1.51×10-7, OR=1.06, 95%CI: 1.03-1.09) and ch.17.49619327-SPOP (ß=0.23%, P=7.54×10-7, OR=1.17, 95%CI: 1.08-1.28) were identified with suggestive significance. After correcting for multiple testing, no sites reached genome-wide significance. There was no statistical significance in the paired analysis. Two clusters with hypomethylated (ß=-0.39%, P<0.001) and hypermethylated (ß=0.47%, P<0.001) patterns were observed for HOMA2-IR. Conclusions: IR was significantly associated with DNA methylation, and genetic factors might contribute to the association.


Sujet(s)
Méthylation de l'ADN , Insulinorésistance , Femelle , Humains , Mâle , Chine/épidémiologie , Étude d'association pangénomique , Insulinorésistance/génétique , Jumeaux monozygotes
3.
Nutr Diabetes ; 14(1): 48, 2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-38951151

RÉSUMÉ

BACKGROUND: This study aimed to assess whether the Haptoglobin (Hp) genotype influences the relationship between hemoglobin (Hb) levels and the development of gestational diabetes mellitus (GDM). Additionally, it sought to evaluate the interaction and joint association of Hb levels and Hp genotype with GDM risk. METHODS: This retrospective study involved 358 women with GDM and 1324 women with normal glucose tolerance (NGT). Peripheral blood leukocytes were collected from 360 individuals at 14-16 weeks' gestation for Hp genotyping. GDM was diagnosed between 24-28 weeks' gestation. Interactive moderating effect, joint analysis, and mediation analysis were performed to evaluate the crosslink of Hb levels and Hp genotype with GDM risk. RESULTS: Women who developed GDM had significantly higher Hb levels throughout pregnancy compared to those with NGT. Increase first-trimester Hb concentration was associated with a progressive rise in GDM incidence, glucose levels, glycosylated hemoglobin levels, Homeostasis Model Assessment for Insulin Resistance (HOMA-IR) values, cesarean delivery rates, and composite neonatal outcomes. Spline regression showed a significant linear association of GDM incidence with continuous first-trimester Hb level when the latter exceeded 122 g/L. Increased first-trimester Hb concentration was an independent risk factor for GDM development after adjusting for potential confounding factors in both the overall population and a matched case-control group. The Hp2-2 genotype was more prevalent among pregnant women with GDM when first-trimester Hb exceeded 122 g/L. Significant multiplicative and additive interactions were identified between Hb levels and Hp genotype for GDM risk, adjusted for age and pre-pregnancy BMI. The odds ratio (OR) for GDM development increased incrementally when stratified by Hb levels and Hp genotype. Moreover, first-trimester Hb level partially mediated the association between Hp genotype and GDM risk. CONCLUSION: Increased first-trimester Hb levels were closely associated with the development of GDM and adverse pregnancy outcomes, with this association moderated by the Hp2-2 genotype.


Sujet(s)
Diabète gestationnel , Génotype , Haptoglobines , Hémoglobines , Premier trimestre de grossesse , Humains , Femelle , Grossesse , Diabète gestationnel/génétique , Diabète gestationnel/sang , Diabète gestationnel/épidémiologie , Haptoglobines/génétique , Études rétrospectives , Adulte , Hémoglobines/analyse , Chine/épidémiologie , Facteurs de risque , Asiatiques/génétique , Hémoglobine glyquée/analyse , Glycémie/analyse , Glycémie/métabolisme , Insulinorésistance/génétique , Peuples d'Asie de l'Est
4.
Skin Res Technol ; 30(7): e13811, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38923681

RÉSUMÉ

BACKGROUND: Acne vulgaris (AV) is a chronic inflammatory skin condition affecting the pilosebaceous unit, commonly presenting as comedones, papules, pustules, or nodules on the face, upper limbs, torso, and back, with comedones formation being the primary pathology leading to disfiguring inflammation, hyperpigmentation, scarring, and psychological impact. AIM: The purpose of this study was to investigate the significance of two genetic variants in the promoter region of the tumor necrosis factor-alpha (TNF-α) gene and their association with insulin resistance (IR) in acne patients. To understand how these variants contribute to AV and its associated IR. SUBJECTS AND METHODS: An analytical cross-sectional study with a case-control design and research evaluation was carried out on 87 AV patients and 73 healthy volunteers. The medical histories of both groups were obtained, as well as the severity and duration of inflammation among acne sufferers, as well as demographic data. Biochemical analysis was performed on both sets of participants, including fasting blood glucose levels, insulin levels while fasting, IR, and serum TNF-α. PCR-RFLP analysis identified -863 G > A (rs1800630) and -308 G > A (rs1800629) variations, and real-time PCR analysis evaluated TNF-α gene expression in both patients and healthy people. RESULTS: Acne patients exhibited significantly higher levels of IR, fasting glucose, fasting insulin, serum TNF-α, and TNF-α folding change, when compared to healthy controls. The co-dominant model for -863 G > A and -308 G > A variants exhibited significant variations between the two groups. Severe acne patients who had the A/A genotype for -308 variants exhibited higher levels of IR, serum TNF-α, and TNF-α folding change. Highly significant positive linear correlation between IR, serum TNF-α, and TNF-α folding change in severe AV. CONCLUSION: There is a correlation between AV, especially severe acne, and the -863 G > A and -308 G > A polymorphism, which influences TNF-α gene expression and serum TNF-α levels.


Sujet(s)
Acné juvénile , Insulinorésistance , Facteur de nécrose tumorale alpha , Humains , Acné juvénile/génétique , Acné juvénile/sang , Insulinorésistance/génétique , Facteur de nécrose tumorale alpha/génétique , Facteur de nécrose tumorale alpha/sang , Mâle , Femelle , Études cas-témoins , Études transversales , Adulte , Jeune adulte , Adolescent , Indice de gravité de la maladie , Polymorphisme de nucléotide simple , Prédisposition génétique à une maladie/génétique
5.
Front Endocrinol (Lausanne) ; 15: 1384103, 2024.
Article de Anglais | MEDLINE | ID: mdl-38938516

RÉSUMÉ

Insulin resistance (IR) and beta cell dysfunction are the major drivers of type 2 diabetes (T2D). Genome-Wide Association Studies (GWAS) on IR have been predominantly conducted in European populations, while Middle Eastern populations remain largely underrepresented. We conducted a GWAS on the indices of IR (HOMA2-IR) and beta cell function (HOMA2-%B) in 6,217 non-diabetic individuals from the Qatar Biobank (QBB; Discovery cohort; n = 2170, Replication cohort; n = 4047) with and without body mass index (BMI) adjustment. We also developed polygenic scores (PGS) for HOMA2-IR and compared their performance with a previously derived PGS for HOMA-IR (PGS003470). We replicated 11 loci that have been previously associated with HOMA-IR and 24 loci that have been associated with HOMA-%B, at nominal statistical significance. We also identified a novel locus associated with beta cell function near VEGFC gene, tagged by rs61552983 (P = 4.38 × 10-8). Moreover, our best performing PGS (Q-PGS4; Adj R2 = 0.233 ± 0.014; P = 1.55 x 10-3) performed better than PGS003470 (Adj R2 = 0.194 ± 0.014; P = 5.45 x 10-2) in predicting HOMA2-IR in our dataset. This is the first GWAS on HOMA2 and the first GWAS conducted in the Middle East focusing on IR and beta cell function. Herein, we report a novel locus in VEGFC that is implicated in beta cell dysfunction. Inclusion of under-represented populations in GWAS has potentials to provide important insights into the genetic architecture of IR and beta cell function.


Sujet(s)
Diabète de type 2 , Étude d'association pangénomique , Insulinorésistance , Hérédité multifactorielle , Humains , Insulinorésistance/génétique , Femelle , Mâle , Adulte d'âge moyen , Diabète de type 2/génétique , Adulte , Qatar/épidémiologie , Polymorphisme de nucléotide simple , Cellules à insuline/métabolisme , Sujet âgé , Indice de masse corporelle , Études de cohortes , Prédisposition génétique à une maladie
6.
Int J Mol Sci ; 25(12)2024 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-38928056

RÉSUMÉ

We aimed to provide an in-depth analysis with respect to three turning points in pancreas involvement in primary hyperparathyroidism (PHP): hypercalcemia-induced pancreatitis (HCa-P), MEN1 (multiple endocrine neoplasia)-related neuroendocrine tumors (NETs), and insulin resistance (IR). This was a comprehensive review conducted via a PubMed search between January 2020 and January 2024. HCa-P (n = 9 studies, N = 1375) involved as a starting point parathyroid NETs (n = 7) or pancreatitis (n = 2, N = 167). Case report-focused analysis (N = 27) showed five cases of pregnancy PHP-HCa-P and three reports of parathyroid carcinoma (female/male ratio of 2/1, ages of 34 in women, men of 56). MEN1-NET studies (n = 7) included MEN1-related insulinomas (n = 2) or MEN1-associated PHP (n = 2) or analyses of genetic profile (n = 3), for a total of 877 MEN1 subjects. In MEN1 insulinomas (N = 77), the rate of associated PHP was 78%. Recurrence after parathyroidectomy (N = 585 with PHP) was higher after less-than-subtotal versus subtotal parathyroidectomy (68% versus 45%, p < 0.001); re-do surgery was 26% depending on surgery for pancreatic NETs (found in 82% of PHP patients). MEN1 pathogenic variants in exon 10 represented an independent risk factor for PHP recurrence. A single pediatric study in MEN1 (N = 80) revealed the following: a PHP rate of 80% and pancreatic NET rate of 35% and 35 underlying germline MEN1 pathogenic variants (and 3/35 of them were newly detected). The co-occurrence of genetic anomalies included the following: CDC73 gene variant, glucokinase regulatory protein gene pathogenic variant (c.151C>T, p.Arg51*), and CAH-X syndrome. IR/metabolic feature-focused analysis identified (n = 10, N = 1010) a heterogeneous spectrum: approximately one-third of adults might have had prediabetes, almost half displayed some level of IR as reflected by HOMA-IR > 2.6, and serum calcium was positively correlated with HOMA-IR. Vitamin D deficiency was associated with a higher rate of metabolic syndrome (n = 1). Normocalcemic and mildly symptomatic hyperparathyroidism (n = 6, N = 193) was associated with a higher fasting glucose and some improvement after parathyroidectomy. This multilayer pancreas/parathyroid analysis highlighted a complex panel of connections from pathogenic factors, including biochemical, molecular, genetic, and metabolic factors, to a clinical multidisciplinary panel.


Sujet(s)
Hypercalcémie , Hyperparathyroïdie primitive , Insulinorésistance , Pancréatite , Humains , Hyperparathyroïdie primitive/génétique , Hyperparathyroïdie primitive/chirurgie , Hyperparathyroïdie primitive/complications , Insulinorésistance/génétique , Hypercalcémie/génétique , Hypercalcémie/étiologie , Pancréatite/génétique , Pancréatite/étiologie , Femelle , Mâle , Protéines proto-oncogènes/génétique , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/complications , Néoplasie endocrinienne multiple de type 1/génétique , Néoplasie endocrinienne multiple de type 1/complications , Tumeurs de la parathyroïde/génétique , Tumeurs de la parathyroïde/complications , Tumeurs de la parathyroïde/chirurgie , Adulte , Parathyroïdectomie , Tumeurs neuroendocrines/génétique , Tumeurs neuroendocrines/complications , Tumeurs neuroendocrines/anatomopathologie , Pancréas/anatomopathologie , Pancréas/chirurgie , Pancréas/métabolisme
7.
Commun Biol ; 7(1): 692, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38862620

RÉSUMÉ

Organismal adaptations to spaceflight have been characterized at the molecular level in model organisms, including Drosophila and C. elegans. Here, we extend molecular work to energy metabolism and sex hormone signaling in mice and humans. We found spaceflight induced changes in insulin and estrogen signaling in rodents and humans. Murine changes were most prominent in the liver, where we observed inhibition of insulin and estrogen receptor signaling with concomitant hepatic insulin resistance and steatosis. Based on the metabolic demand, metabolic pathways mediated by insulin and estrogen vary among muscles, specifically between the soleus and extensor digitorum longus. In humans, spaceflight induced changes in insulin and estrogen related genes and pathways. Pathway analysis demonstrated spaceflight induced changes in insulin resistance, estrogen signaling, stress response, and viral infection. These data strongly suggest the need for further research on the metabolic and reproductive endocrinologic effects of space travel, if we are to become a successful interplanetary species.


Sujet(s)
Oestrogènes , Insuline , Vol spatial , Animaux , Insuline/métabolisme , Oestrogènes/métabolisme , Humains , Souris , Mâle , Femelle , Transcriptome , Transduction du signal , Souris de lignée C57BL , Métabolisme énergétique/génétique , Insulinorésistance/génétique , Foie/métabolisme , Adulte , Régulation de l'expression des gènes
8.
Nat Commun ; 15(1): 5394, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38918428

RÉSUMÉ

Adipose tissue macrophages (ATMs) influence obesity-associated metabolic dysfunction, but the mechanisms by which they do so are not well understood. We show that miR-6236 is a bona fide miRNA that is secreted by ATMs during obesity. Global or myeloid cell-specific deletion of miR-6236 aggravates obesity-associated adipose tissue insulin resistance, hyperglycemia, hyperinsulinemia, and hyperlipidemia. miR-6236 augments adipocyte insulin sensitivity by inhibiting translation of negative regulators of insulin signaling, including PTEN. The human genome harbors a miR-6236 homolog that is highly expressed in the serum and adipose tissue of obese people. hsa-MIR-6236 expression negatively correlates with hyperglycemia and glucose intolerance, and positively correlates with insulin sensitivity. Together, our findings establish miR-6236 as an ATM-secreted miRNA that potentiates adipocyte insulin signaling and protects against metabolic dysfunction during obesity.


Sujet(s)
Adipocytes , Hyperglycémie , Insulinorésistance , Insuline , microARN , Obésité , Phosphohydrolase PTEN , Transduction du signal , microARN/métabolisme , microARN/génétique , Obésité/métabolisme , Obésité/génétique , Animaux , Adipocytes/métabolisme , Hyperglycémie/métabolisme , Hyperglycémie/génétique , Humains , Insuline/métabolisme , Insulinorésistance/génétique , Souris , Mâle , Phosphohydrolase PTEN/métabolisme , Phosphohydrolase PTEN/génétique , Souris de lignée C57BL , Macrophages/métabolisme , Tissu adipeux/métabolisme , Cellules myéloïdes/métabolisme , Souris knockout , Hyperinsulinisme/métabolisme , Hyperinsulinisme/génétique
9.
Cells ; 13(11)2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38891115

RÉSUMÉ

The 9p21.3 genomic locus is a hot spot for disease-associated single-nucleotide polymorphisms (SNPs), and its strongest associations are with coronary artery disease (CAD). The disease-associated SNPs are located within the sequence of a long noncoding RNA ANRIL, which potentially contributes to atherogenesis by regulating vascular cell stress and proliferation, but also affects pancreatic ß-cell proliferation. Altered expression of a neighboring gene, CDKN2B, has been also recognized to correlate with obesity and hepatic steatosis in people carrying the risk SNPs. In the present study, we investigated the impact of 9p21.3 on obesity accompanied by hyperlipidemia in mice carrying a deletion of the murine ortholog for the 9p21.3 (Chr4Δ70/Δ70) risk locus in hyperlipidemic Ldlr-/-ApoB100/100 background. The Chr4Δ70/Δ70 mice showed decreased mRNA expression of insulin receptors in white adipose tissue already at a young age, which developed into insulin resistance and obesity by aging. In addition, the Sirt1-Ppargc1a-Ucp2 pathway was downregulated together with the expression of Cdkn2b, specifically in the white adipose tissue in Chr4Δ70/Δ70 mice. These results suggest that the 9p21.3 locus, ANRIL lncRNA, and their murine orthologues may regulate the key energy metabolism pathways in a white adipose tissue-specific manner in the presence of hypercholesterolemia, thus contributing to the pathogenesis of metabolic syndrome.


Sujet(s)
Hypercholestérolémie , Insulinorésistance , Obésité , Animaux , Obésité/génétique , Obésité/métabolisme , Insulinorésistance/génétique , Hypercholestérolémie/génétique , Hypercholestérolémie/métabolisme , Hypercholestérolémie/complications , Souris , Humains , Chromosomes humains de la paire 9/génétique , Mâle , Délétion de gène , Locus génétiques , Souris de lignée C57BL , Tissu adipeux blanc/métabolisme , ARN long non codant/génétique , ARN long non codant/métabolisme
10.
J Mol Endocrinol ; 73(2)2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38832641

RÉSUMÉ

The objective was to assess the potential differential effects of human versus mouse growth hormone in vivo, given that human unlike mouse growth hormone can bind prolactin as well as the growth hormone receptor. To this end, a transgenic CD-1 mouse expressing human but not mouse growth hormone was generated, and the phenotypes of male mice fed with a regular chow or high-fat diet were assessed. Pancreas and epididymal white adipose tissue gene expression and/or related function were targeted as the pancreas responds to both prolactin and growth hormone receptor signaling, and catabolic effects like lipolytic activity are more directly attributable to growth hormone and growth hormone receptor signaling. The resulting human growth hormone-expressing mice are smaller than wild-type CD-1 mice, despite higher body fat and larger adipocytes, but both mouse types grow at the same rate with similar bone densities. Unlike wild-type mice, there was no significant delay in glucose clearance in human growth hormone-expressing mice when assessed at 8 versus 24 weeks on a high-fat diet. However, both mouse types showed signs of hepatic steatosis that correlated with elevated prolactin but not growth hormone RNA levels. The larger adipocytes in human growth hormone-expressing mice were associated with modified leptin (higher) and adiponectin (lower) RNA levels. Thus, while limited to observations in the male, the human growth hormone-expressing mice exhibit signs of growth hormone insufficiency and adipocyte dysfunction as well as an initial resistance to the negative effects of high-fat diet on glucose clearance.


Sujet(s)
Tissu adipeux , Alimentation riche en graisse , Stéatose hépatique , Glucose , Homéostasie , Insulinorésistance , Souris transgéniques , Animaux , Humains , Alimentation riche en graisse/effets indésirables , Insulinorésistance/génétique , Stéatose hépatique/métabolisme , Stéatose hépatique/étiologie , Stéatose hépatique/génétique , Souris , Mâle , Glucose/métabolisme , Tissu adipeux/métabolisme , Hormone de croissance humaine/métabolisme , Hormone de croissance humaine/génétique , Hormone de croissance/métabolisme , Hormone de croissance/génétique , Prolactine/métabolisme , Leptine/métabolisme , Adipocytes/métabolisme , Tissu adipeux blanc/métabolisme
11.
Endocrinology ; 165(7)2024 May 27.
Article de Anglais | MEDLINE | ID: mdl-38836615

RÉSUMÉ

About half of the world population carries at least one allele of the Ala92-DIO2, which slows down the activity of the type 2 deiodinase (D2), the enzyme that activates T4 to T3. Carrying the Ala92-DIO2 allele has been associated with increased body mass index and insulin resistance, but this has not been reproduced in all populations. To test if the genetic background affects the impact of this polymorphism, here we studied the genetically distant C57Bl/6J (B6) and FVB/N (FVB) mice carrying the Ala92-Dio2 allele as compared to control mice carrying the Thr92-Dio2 allele. Whereas B6-Ala92-Dio2 and B6-Thr92-Dio2 mice-fed chow or high-fat diet-behaved metabolically similar in studies using indirect calorimetry, glucose- and insulin tolerance tests, and measuring white adipose tissue (WAT) weight and liver steatosis, major differences were observed between FVB-Ala92-Dio2 and FVB-Thr92-Dio2 mice: carrying the Ala92-Dio2 allele (on a chow diet) resulted in hypercholesterolemia, smaller WAT pads, hepatomegaly, steatosis, and transcriptome changes in the interscapular brown adipose tissue (iBAT) typical of ER stress and apoptosis. Acclimatization at thermoneutrality (30 °C) eliminated most of the metabolic phenotype, indicating that impaired adaptive (BAT) thermogenesis can be involved. In conclusion, the metabolic impact of carrying the Ala92-Dio2 allele depends greatly on the genetic background of the mouse, varying from no phenotype in B6 mice to a major phenotype in FVB mice. These results will help the planning of future clinical trials studying the Thr92Ala-DIO2 polymorphism and may explain why some clinical studies performed in different populations across the globe have obtained inconsistent results.


Sujet(s)
Iodide peroxidase , , Souris de lignée C57BL , Animaux , Mâle , Iodide peroxidase/génétique , Souris , Alimentation riche en graisse , Contexte génétique , Tissu adipeux blanc/métabolisme , Tissu adipeux brun/métabolisme , Polymorphisme génétique , Insulinorésistance/génétique , Stéatose hépatique/génétique
12.
Nutrients ; 16(9)2024 Apr 25.
Article de Anglais | MEDLINE | ID: mdl-38732523

RÉSUMÉ

The presence of vitamin D3 deficiency associated with the presence of metabolic syndrome (MS) has important public health effects. This study aims to investigate the relationship between vitamin D3 deficiency, MS and vitamin D3 receptor (VDR), GC Vitamin D binding protein (GC), and cytochrome P450 family 2 subfamily R member 1 (CYP2R1) gene polymorphisms, and genes whose encoded proteins are responsible for vitamin D3 metabolism and transport. A total of 58 participants were included in this study (age 39 ± 12 years) and were selected over a 12-month period. They were divided into four groups, depending on the presence of polymorphisms in VDR, GC, and CYP2R1 genes and their weight status. At baseline, in months 3, 6, and 12, biochemical parameters including 25(OH)D3, total cholesterol, LDL cholesterol, HDL cholesterol, triglycerides, and homeostatic model assessment (HOMA index), the insulin resistance indicator were measured. Our results show that all subjects in the polymorphism group supplemented with vitamin D3 reached an optimal level of vitamin D3 associated with high concentrations of 25(OH)D3. Weight loss was most significant in patients in the POW group (overweight patients).


Sujet(s)
Cholécalciférol , Cholestanetriol 26-monooxygenase , Syndrome métabolique X , Récepteur calcitriol , Carence en vitamine D , Protéine de liaison à la vitamine D , Adulte , Femelle , Humains , Mâle , Adulte d'âge moyen , Cholécalciférol/sang , Cholestanetriol 26-monooxygenase/génétique , Famille-2 de cytochromes P450/génétique , Insulinorésistance/génétique , Syndrome métabolique X/génétique , Polymorphisme génétique , Récepteur calcitriol/génétique , Carence en vitamine D/génétique , Carence en vitamine D/sang , Protéine de liaison à la vitamine D/génétique
13.
Am J Physiol Endocrinol Metab ; 327(1): E69-E80, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38717361

RÉSUMÉ

Acylglycerophosphate acyltransferases (AGPATs) catalyze the de novo formation of phosphatidic acid to synthesize glycerophospholipids and triglycerides. AGPATs demonstrate unique physiological roles despite a similar biochemical function. AGPAT3 is highly expressed in the testis, kidney, and liver, with intermediate expression in adipose tissue. Loss of AGPAT3 is associated with reproductive abnormalities and visual dysfunction. However, the role of AGPAT3 in adipose tissue and whole body metabolism has not been investigated. We found that male Agpat3 knockout (KO) mice exhibited reduced body weights with decreased white and brown adipose tissue mass. Such changes were less pronounced in the female Agpat3-KO mice. Agpat3-KO mice have reduced plasma insulin growth factor 1 (IGF1) and insulin levels and diminished circulating lipid metabolites. They manifested intact glucose homeostasis and insulin sensitivity despite a lean phenotype. Agpat3-KO mice maintained an energy balance with normal food intake, energy expenditure, and physical activity, except for increased water intake. Their adaptive thermogenesis was also normal despite reduced brown adipose mass and triglyceride content. Mechanistically, Agpat3 was elevated during mouse and human adipogenesis and enriched in adipocytes. Agpat3-knockdown 3T3-L1 cells and Agpat3-deficient mouse embryonic fibroblasts (MEFs) have impaired adipogenesis in vitro. Interestingly, pioglitazone treatment rescued the adipogenic deficiency in Agpat3-deficient cells. We conclude that AGPAT3 regulates adipogenesis and adipose development. It is possible that adipogenic impairment in Agpat3-deficient cells potentially leads to reduced adipose mass. Findings from this work support the unique role of AGPAT3 in adipose tissue.NEW & NOTEWORTHY AGPAT3 deficiency results in male-specific growth retardation. It reduces adipose tissue mass but does not significantly impact glucose homeostasis or energy balance, except for influencing water intake in mice. Like AGPAT2, AGPAT3 is upregulated during adipogenesis, potentially by peroxisome proliferator-activated receptor gamma (PPARγ). Loss of AGPAT3 impairs adipocyte differentiation, which could be rescued by pioglitazone. Overall, AGPAT3 plays a significant role in regulating adipose tissue mass, partially involving its influence on adipocyte differentiation.


Sujet(s)
1-Acylglycerol-3-phosphate O-acyltransferase , Adipocytes , Souris knockout , Animaux , Femelle , Mâle , Souris , 1-Acylglycerol-3-phosphate O-acyltransferase/génétique , 1-Acylglycerol-3-phosphate O-acyltransferase/métabolisme , Adipocytes/métabolisme , Adipogenèse/génétique , Adipogenèse/physiologie , Tissu adipeux brun/métabolisme , Différenciation cellulaire , Métabolisme énergétique/génétique , Insulinorésistance/génétique , Souris de lignée C57BL , Phénotype , Thermogenèse/génétique , Maigreur/métabolisme , Maigreur/génétique
14.
Am J Physiol Endocrinol Metab ; 327(1): E81-E88, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38809511

RÉSUMÉ

Nicotinamide adenine dinucleotide (NAD+) is a universal coenzyme regulating cellular energy metabolism in many cell types. Recent studies have demonstrated the close relationships between defective NAD+ metabolism and aging and age-associated metabolic diseases. The major purpose of the present study was to test the hypothesis that NAD+ biosynthesis, mediated by a rate-limiting NAD+ biosynthetic enzyme, nicotinamide phosphoribosyltransferase (NAMPT), is essential for maintaining normal adipose tissue function and whole body metabolic health during the aging process. To this end, we provided in-depth and comprehensive metabolic assessments for female adipocyte-specific Nampt knockout (ANKO) mice during aging. We first evaluated body fat mass in young (≤4-mo-old), middle aged (10-14-mo-old), and old (≥18-mo-old) mice. Intriguingly, adipocyte-specific Nampt deletion protected against age-induced obesity without changing energy balance. However, data obtained from the hyperinsulinemic-euglycemic clamp procedure (HECP) demonstrated that, despite the lean phenotype, old ANKO mice had severe insulin resistance in skeletal muscle, heart, and white adipose tissue (WAT). Old ANKO mice also exhibited hyperinsulinemia and hypoadiponectinemia. Mechanistically, loss of Nampt caused marked decreases in WAT gene expression of lipogenic targets of peroxisome proliferator-activated receptor gamma (PPAR-γ) in an age-dependent manner. In addition, administration of a PPAR-γ agonist rosiglitazone restored fat mass and improved metabolic abnormalities in old ANKO mice. In conclusion, these findings highlight the importance of the NAMPT-NAD+-PPAR-γ axis in maintaining functional integrity and quantity of adipose tissue, and whole body metabolic function in female mice during aging.NEW & NOTEWORTHY Defective NAD+ metabolism is associated with aging and age-associated metabolic diseases. In the present study, we provided in-depth metabolic assessments in female mice with adipocyte-specific inactivation of a key NAD+ biosynthetic enzyme NAMPT and revealed an unexpected role of adipose tissue NAMPT-NAD+-PPAR-γ axis in maintaining functional integrity and quantity of adipose tissue and whole body metabolic health during the aging process.


Sujet(s)
Adipocytes , Vieillissement , Cytokines , Souris knockout , NAD , Nicotinamide phosphoribosyltransferase , Animaux , Nicotinamide phosphoribosyltransferase/métabolisme , Nicotinamide phosphoribosyltransferase/génétique , Femelle , Vieillissement/métabolisme , Souris , Adipocytes/métabolisme , NAD/métabolisme , Cytokines/métabolisme , Phénotype , Insulinorésistance/génétique , Métabolisme énergétique/génétique , Obésité/métabolisme , Obésité/génétique , Récepteur PPAR gamma/métabolisme , Récepteur PPAR gamma/génétique , Souris de lignée C57BL
15.
Lipids Health Dis ; 23(1): 115, 2024 Apr 20.
Article de Anglais | MEDLINE | ID: mdl-38643148

RÉSUMÉ

BACKGROUND: The triglyceride to high-density lipoprotein cholesterol (TG/HDL-C) ratio and triglyceride-glucose (TyG) index are novel indexes for insulin resistance (IR). We aimed to evaluate associations of TG/HDL-C and TyG with arterial stiffness risk. METHODS: We enrolled 1979 participants from the Rural Chinese Cohort Study, examining arterial stiffness by brachial-ankle pulse wave velocity (baPWV). Logistic and linear regression models were employed to calculate effect estimates. For meta-analysis, we searched relevant articles from PubMed, Embase and Web of Science up to August 26, 2023. The fixed-effects or random-effects models were used to calculate the pooled estimates. We evaluated dose-response associations using restricted cubic splines. RESULTS: For cross-sectional studies, the adjusted ORs (95%CIs) for arterial stiffness were 1.12 (1.01-1.23) and 1.78 (1.38-2.30) for per 1 unit increment in TG/HDL-C and TyG. In the meta-analysis, the pooled ORs (95% CIs) were 1.26 (1.14-1.39) and 1.57 (1.36-1.82) for per 1 unit increment of TG/HDL-C and TyG. Additionally, both TG/HDL-C and TyG were positively related to PWV, with ß of 0.09 (95% CI 0.04-0.14) and 0.57 (95% CI 0.35-0.78) m/s. We also found linear associations of TG/HDL-C and TyG with arterial stiffness risk. CONCLUSIONS: High TG/HDL-C and TyG were related to increased arterial stiffness risk, indicating TG/HDL-C and TyG may be convincing predictors of arterial stiffness.


Sujet(s)
Insulinorésistance , Rigidité vasculaire , Humains , Glucose , Triglycéride , Études de cohortes , Index de pression systolique cheville-bras , Rigidité vasculaire/physiologie , Cholestérol HDL , Études transversales , Analyse de l'onde de pouls , Insulinorésistance/génétique , Glycémie , Marqueurs biologiques
16.
Int J Mol Sci ; 25(8)2024 Apr 09.
Article de Anglais | MEDLINE | ID: mdl-38673735

RÉSUMÉ

Experimental animal models of diabetes can be useful for identifying novel targets related to disease, for understanding its physiopathology, and for evaluating emerging antidiabetic treatments. This study aimed to characterize two rat diabetes models: HFD + STZ, a high-fat diet (60% fat) combined with streptozotocin administration (STZ, 35 mg/kg BW), and a model with a single STZ dose (65 mg/kg BW) in comparison with healthy rats. HFD + STZ- induced animals demonstrated a stable hyperglycemia range (350-450 mg/dL), whereas in the STZ-induced rats, we found glucose concentration values with a greater dispersion, ranging from 270 to 510 mg/dL. Moreover, in the HFD + STZ group, the AUC value of the insulin tolerance test (ITT) was found to be remarkably augmented by 6.2-fold higher than in healthy animals (33,687.0 ± 1705.7 mg/dL/min vs. 5469.0 ± 267.6, respectively), indicating insulin resistance (IR). In contrast, a more moderate AUC value was observed in the STZ group (19,059.0 ± 3037.4 mg/dL/min) resulting in a value 2.5-fold higher than the average exhibited by the control group. After microarray experiments on liver tissue from all animals, we analyzed genes exhibiting a fold change value in gene expression <-2 or >2 (p-value <0.05). We found 27,686 differentially expressed genes (DEG), identified the top 10 DEGs and detected 849 coding genes that exhibited opposite expression patterns between both diabetes models (491 upregulated genes in the STZ model and 358 upregulated genes in HFD + STZ animals). Finally, we performed an enrichment analysis of the 849 selected genes. Whereas in the STZ model we found cellular pathways related to lipid biosynthesis and metabolism, in the HFD + STZ model we identified pathways related to immunometabolism. Some phenotypic differences observed in the models could be explained by transcriptomic results; however, further studies are needed to corroborate these findings. Our data confirm that the STZ and the HFD + STZ models are reliable experimental models for human T1D and T2D, respectively. These results also provide insight into alterations in the expression of specific liver genes and could be utilized in future studies focusing on diabetes complications associated with impaired liver function.


Sujet(s)
Diabète expérimental , Diabète de type 2 , Foie , Animaux , Foie/métabolisme , Rats , Diabète expérimental/génétique , Diabète expérimental/métabolisme , Diabète de type 2/génétique , Diabète de type 2/métabolisme , Mâle , Diabète de type 1/génétique , Diabète de type 1/métabolisme , Alimentation riche en graisse/effets indésirables , Transcriptome , Insulinorésistance/génétique , Analyse de profil d'expression de gènes , Streptozocine , Modèles animaux de maladie humaine , Glycémie/métabolisme
17.
J Ovarian Res ; 17(1): 79, 2024 Apr 12.
Article de Anglais | MEDLINE | ID: mdl-38610028

RÉSUMÉ

OBJECTIVE: IR emerges as a feature in the pathophysiology of PCOS, precipitating ovulatory anomalies and endometrial dysfunctions that contribute to the infertility challenges characteristic of this condition. Despite its clinical significance, a consensus on the precise mechanisms by which IR exacerbates PCOS is still lacking. This study aims to harness bioinformatics tools to unearth key IR-associated genes in PCOS patients, providing a platform for future therapeutic research and potential intervention strategies. METHODS: We retrieved 4 datasets detailing PCOS from the GEO, and sourced IRGs from the MSigDB. We applied WGCNA to identify gene modules linked to insulin resistance, utilizing IR scores as a phenotypic marker. Gene refinement was executed through the LASSO, SVM, and Boruta feature selection algorithms. qPCR was carried out on selected samples to confirm findings. We predicted both miRNA and lncRNA targets using the ENCORI database, which facilitated the construction of a ceRNA network. Lastly, a drug-target network was derived from the CTD. RESULTS: Thirteen genes related to insulin resistance in PCOS were identified via WGCNA analysis. LASSO, SVM, and Boruta algorithms further isolated CAPN2 as a notably upregulated gene, corroborated by biological verification. The ceRNA network involving lncRNA XIST and hsa-miR-433-3p indicated a possible regulatory link with CAPN2, supported by ENCORI database. Drug prediction analysis uncovered seven pharmacological agents, most being significant regulators of the endocrine system, as potential candidates for addressing insulin resistance in PCOS. CONCLUSIONS: This study highlights the pivotal role of CAPN2 in insulin resistance within the context of PCOS, emphasizing its importance as both a critical biomarker and a potential therapeutic target. By identifying CAPN2, our research contributes to the expanding evidence surrounding the CAPN family, particularly CAPN10, in insulin resistance studies beyond PCOS. This work enriches our understanding of the mechanisms underlying insulin resistance, offering insights that bridge gaps in the current scientific landscape.


Sujet(s)
Insulinorésistance , microARN , Syndrome des ovaires polykystiques , ARN long non codant , Humains , Femelle , Insulinorésistance/génétique , Syndrome des ovaires polykystiques/génétique , ARN long non codant/génétique , Algorithmes , Biologie informatique , Calpain/génétique
18.
Lipids Health Dis ; 23(1): 103, 2024 Apr 13.
Article de Anglais | MEDLINE | ID: mdl-38615017

RÉSUMÉ

BACKGROUND: Previous studies have demonstrated the relationship between adipocyte factors, insulin resistance, and other indicators with telomere length. However, these studies did not consider the influence of changes in different indicators on telomere length over time. Therefore, the aim of this study is to elucidate the impact of changes in adipocyte factors, HOMA-IR, and other indicators on the dynamic variation of telomere length. METHODS: The data were from a cohort study conducted in Ningxia, China. A total of 1624 subjects were analyzed. Adipokines and relative leukocyte telomere length (RLTL) were measured, and changes in Homeostatic Model Assessment for Insulin Resistance (HOMA-IR), Homeostatic Model Assessment for ß-Cell Function (HOMA-ß), and Quantitative Insulin Sensitivity Check Index (QUICKI) were calculated. Generalized linear models evaluated associations between changes in adipokines and RLTL changes. Furthermore, univariate analyses examined the effects of changes in adipokines and insulin resistance indicators on ΔRLTL. RESULTS: The research findings indicate that females generally have shorter telomeres compared to males. In comparison to the low-level group of Δleptin (LEP), the high-level group of ΔLEP shows a negative correlation with ΔRLTL (B=-1.32, 95% CI (-2.38, -0.27)). Even after multivariable adjustments, this relationship persists (B=-1.31, 95% CI (-2.24, -0.23)). Further analysis reveals that after adjusting for ΔHOMA-IR, ΔHOMA-ß, and ΔQUICKI, the high-level group of ΔLEP still exhibits a significant negative correlation with ΔRLTL (B=-1.37, 95% CI (-2.43, -0.31)). However, the interaction effects between ΔHOMA-IR, ΔHOMA-ß, ΔQUICKI, and ΔLEP do not affect ΔRLTL. CONCLUSIONS: Elevated levels of leptin were significantly correlated with shortened telomere length. This suggests that increased leptin levels may impact overall individual health by affecting telomere length, underscoring the importance of measures to reduce leptin levels to mitigate the onset and progression of related diseases.


Sujet(s)
Insulinorésistance , Leptine , Femelle , Mâle , Humains , Leptine/génétique , Études de cohortes , Insulinorésistance/génétique , Population rurale , Raccourcissement des télomères , Télomère/génétique , Adipokines , Chine , Leucocytes
19.
Gene ; 919: 148504, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38670392

RÉSUMÉ

OBJECTIVES: To evaluate the correlation between serum vitamin D, homocysteine and the methylene tetrahydrofolate reductase (MTHFR) C677T polymorphism in women with polycystic ovary syndrome (PCOS). Study design We retrospectively compared the serum homocysteine and vitamin D levels and the MTHFR C677T polymorphism in 104 PCOS patients and 104 controls. Parameters related to PCOS were statistically analysed. RESULTS: Comparative analysis revealed that women with PCOS had significantly greater serum homocysteine levels (P = 0.002) and lower vitamin D concentrations (P = 0.040) than controls. The distribution frequency of the MTHFR C677T genotype did not significantly differ between the PCOS group and the control group. (P > 0.05). In the PCOS group, the serum level of homocysteine in the TT group was significantly greater than that in the CT (P = 0.003) and CC (P = 0.002) groups and the level of vitamin D in the TT group was significantly less than that in the CC (P < 0.001) and CT (P = 0.172) groups. The results were similar when the PCOS and control groups were divided according to whether they had insulin resistance. Vitamin D levels were significantly negatively correlated with homocysteine levels in all PCOS patients (r = -0.281, P = 0.004), similarly, vitamin D levels were negatively correlated with homocysteine levels in the CC, CT and TT of PCOS patients. According to multivariate analysis, vitamin D concentration was an independent risk factor for hyperhomocysteinaemia (adjusted OR 1.372, 95 % CI: 1.100-1.712). CONCLUSIONS: No significant differences were found in the distributions of MTHFR C677T genotypes between the PCOS and control groups but these genotypes affected the patients' serum homocysteine and vitamin D concentrations. Women with the TT genotype have significantly lower vitamin D levels and higher homocysteine levels than women with the CC and CT genotypes. However, because of the limitations of this investigation, large-sample, high-quality prospective studies are needed to further verify these results in the future.


Sujet(s)
Homocystéine , Methylenetetrahydrofolate reductase (NADPH2) , Syndrome des ovaires polykystiques , Vitamine D , Humains , Syndrome des ovaires polykystiques/génétique , Syndrome des ovaires polykystiques/sang , Femelle , Methylenetetrahydrofolate reductase (NADPH2)/génétique , Homocystéine/sang , Vitamine D/sang , Adulte , Études rétrospectives , Études cas-témoins , Polymorphisme de nucléotide simple , Jeune adulte , Génotype , Prédisposition génétique à une maladie , Insulinorésistance/génétique
20.
Endocrinology ; 165(5)2024 Mar 29.
Article de Anglais | MEDLINE | ID: mdl-38578949

RÉSUMÉ

OBJECTIVES: Growth factor receptor bound protein 7 (GRB7) is a multidomain signaling adaptor. Members of the Grb7/10/14 family, specifically Gbrb10/14, have important roles in metabolism. We ablated the Grb7 gene in mice to examine its metabolic function. METHODS: Global ablation of Grb7 in FVB/NJ mice was generated. Growth, organ weight, food intake, and glucose homeostasis were measured. Insulin signaling was examined by Western blotting. Fat and lean body mass was measured by nuclear magnetic resonance, and body composition after fasting or high-fat diet was assessed. Energy expenditure was measured by indirect calorimetry. Expression of adiposity and lipid metabolism genes was measured by quantitative PCR. RESULTS: Grb7-null mice were viable, fertile, and without obvious phenotype. Grb7 ablation improved glycemic control and displayed sensitization to insulin signaling in the liver. Grb7-null females but not males had increased gonadal white adipose tissue mass. Following a 12-week high-fat diet, Grb7-null female mice gained fat body mass and developed relative insulin resistance. With fasting, there was less decrease in fat body mass in Grb7-null female mice. Female mice with Grb7 ablation had increased baseline food intake, less energy expenditure, and displayed a decrease in the expression of lipolysis and adipose browning genes in gonadal white adipose tissue by transcript and protein analysis. CONCLUSION: Our study suggests that Grb7 is a negative regulator of glycemic control. Our results reveal a role for Grb7 in female mice in the regulation of the visceral adipose tissue mass, a powerful predictor of metabolic dysfunction in obesity.


Sujet(s)
Graisse abdominale , Métabolisme énergétique , Protéine adaptatrice GRB7 , Insuline , Souris knockout , Transduction du signal , Animaux , Femelle , Mâle , Souris , Graisse abdominale/métabolisme , Glycémie/métabolisme , Composition corporelle/génétique , Alimentation riche en graisse , Métabolisme énergétique/génétique , Protéine adaptatrice GRB7/génétique , Protéine adaptatrice GRB7/métabolisme , Insuline/métabolisme , Insulinorésistance/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...