Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 835
Filtrar
1.
J Diabetes Res ; 2024: 8462987, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38712310

RESUMO

Background and Aims: This study is aimed at investigating the potential correlation of thyroid hormone sensitivity with visceral fat area (VFA), subcutaneous fat area (SFA), and body mass index (BMI) among euthyroid type 2 diabetes mellitus (T2DM) subjects. Methods: Thyroid hormone sensitivity indices were calculated by thyroid feedback quantile-based index (TFQI), TSH index (TSHI), thyrotropin thyroxine resistance index (TT4RI), and free thyroxine (fT4)/free triiodothyronine (fT3) ratio. These indices were then categorized into quartiles for analysis. The outcomes were the change rates in VFA, SFA, and BMI among the participants. Result: The present study included 921 patients, with a median follow-up of 2.2 years. In multivariate linear regression, when compared to the first quartile, SFA demonstrated a notable decline in the fourth quartile of TFQI, TSHI, and TT4RI (ß coefficient = -5.78, -7.83, and - 6.84 cm2 per year), while it significantly increased in the fourth quartile of fT4/fT3 ratio (ß coefficient = 6.13 cm2 per year). Similarly, in the fourth quartile of TFQI, TSHI, and TT4RI, VFA decreased significantly, evidenced by ß coefficients of -5.14, -4.80, and -4.08 cm2 per year. Yet, among the quartiles of the fT4/fT3 ratio, no discernible trend in VFA was observed. There was no significant association between indices of thyroid hormone sensitivity and change in BMI. Conclusion: Impaired central sensitivity to thyroid hormones was significantly associated with the reduction of VFA and SFA, while impaired peripheral sensitivity was associated with an increase of SFA in euthyroid individuals with T2DM.


Assuntos
Índice de Massa Corporal , Diabetes Mellitus Tipo 2 , Hormônios Tireóideos , Humanos , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/fisiopatologia , Pessoa de Meia-Idade , Masculino , Feminino , Estudos Retrospectivos , Hormônios Tireóideos/sangue , Idoso , Tiroxina/sangue , Gordura Intra-Abdominal/metabolismo , Tireotropina/sangue , Gordura Abdominal/metabolismo , Adulto , Tri-Iodotironina/sangue , Testes de Função Tireóidea
2.
Endocrinology ; 165(5)2024 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-38578949

RESUMO

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.


Assuntos
Gordura Abdominal , Metabolismo Energético , Proteína Adaptadora GRB7 , Insulina , Camundongos Knockout , Transdução de Sinais , Animais , Feminino , Masculino , Camundongos , Gordura Abdominal/metabolismo , Glicemia/metabolismo , Composição Corporal/genética , Dieta Hiperlipídica , Metabolismo Energético/genética , Proteína Adaptadora GRB7/genética , Proteína Adaptadora GRB7/metabolismo , Insulina/metabolismo , Resistência à Insulina/genética
3.
Environ Pollut ; 349: 123959, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38608855

RESUMO

The worldwide prevalence of obesity highlights the potential contribution of endocrine-disrupting chemicals (EDCs). However, common epidemiological measures such as body mass index and waist circumference may misrepresent the intricate obesity risks these chemicals pose across genders. This study delves deeper into abdominal fat by differentiating between subcutaneous and visceral regions by analyzing data from National Health and Nutrition Examination Surveys (NHANES). We particularly investigated the gender-specific associations between organophosphorus flame-retardant metabolites (mOPFRs), phthalates (mPAEs) and accumulated fat indexes from 2536 people. Aiding by Bayesian Kernel Machine Regression (BKMR), we found while co-exposure to mOPFRs and mPAEs was linked to general and abdominal obesity across the entire and gender-specific populations, a gender-specific fat distribution emerged. For women, urinary BDCPP and MBzP were linked to an increased subcutaneous fat index (SFI) [BDCPP OR: 1.12 (95% CI: 1.03-1.21), MBzP OR: 1.09 (95% CI: 1.01-1.18)], but not to visceral fat index (VFI). These metabolites had a combined linkage with SFI, with BDCPP (weighting 22.0%) and DPHP (weighting 31.0%) being the most influential in Quantile g-computation model (qgcomp) model. In men, BCEP exposure exclusively associated with the elevated VFI [OR: 1.14 (95% CI: 1.03-1.26)], a trend further highlighted in mixture models with BCEP as the predominant association. Intriguingly, only males displayed a marked correlation between these metabolites and insulin resistance in subpopulation. An attempted mediation analysis revealed that elevated C-reactive protein mediated 12.1% of the association between urinary BCEP and insulin resistance, suggesting a potential role of inflammation. In conclusion, the gender-specific fat distribution and insulin resistance that associated with mOPFRs represented the potential risk of these chemicals to man.


Assuntos
Exposição Ambiental , Resistência à Insulina , Ácidos Ftálicos , Humanos , Feminino , Masculino , Ácidos Ftálicos/metabolismo , Adulto , Pessoa de Meia-Idade , Exposição Ambiental/estatística & dados numéricos , Disruptores Endócrinos/metabolismo , Organofosfatos/metabolismo , Gordura Abdominal/metabolismo , Poluentes Ambientais/metabolismo , Ésteres/metabolismo , Retardadores de Chama/metabolismo , Adulto Jovem , Inquéritos Nutricionais , Fatores Sexuais
4.
Poult Sci ; 103(5): 103559, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38430780

RESUMO

Peroxisome proliferator-activated receptor gamma (PPARγ) is a master regulator of adipogenesis. Our previous study revealed that chicken PPARγ has 3 alternative promoters named as P1, P2, and P3, and the DNA methylation of promoter P3 was negatively associated with PPARγ mRNA expression in abdominal adipose tissue (AAT). However, the methylation status of promoters P1 and P2 is unclear. Here we assessed promoter P1 methylation status in AAT of Northeast Agricultural University broiler lines divergently selected for abdominal fat content (NEAUHLF). The results showed that promoter P1 methylation differed in AAT between the lean and fat lines of NEAUHLF at 7 wk of age (p < 0.05), and AAT expression of PPARγ transcript 1 (PPARγ1), which was derived from the promoter P1, was greatly higher in fat line than in lean line at 2 and 7 wk of age. The results of the correlation analysis showed that P1 methylation was positively correlated with PPARγ1 expression at 7 wk of age (Pearson's r = 0.356, p = 0.0242), suggesting P1 methylation promotes PPARγ1 expression. To explore the underlying molecular mechanism of P1 methylation on PPARγ1 expression, bioinformatics analysis, dual-luciferase reporter assay, pyrosequencing, and electrophoresis mobility shift assay (EMSA) were performed. The results showed that transcription factor NRF1 repressed the promoter activity of the unmethylated P1, but not the methylated P1. Of all the 4 CpGs (CpG48, CpG49, CpG50, and CpG51), which reside within or nearby the NRF1 binding sites of the P1, only CpG49 methylation in AAT was remarkably higher in the fat line than in lean line at 7 wk of age (3.18 to 0.57, p < 0.05), and CpG49 methylation was positively correlated with PPARγ1 expression (Pearson's r = 0.3716, p = 0.0432). Furthermore, EMSA showed that CpG49 methylation reduced the binding of NRF1 to the P1. Taken together, our findings illustrate that P1 methylation promotes PPARγ1 expression at least in part by preventing NRF1 from binding to the promoter P1.


Assuntos
Galinhas , Metilação de DNA , Fator 1 Nuclear Respiratório , PPAR gama , Regiões Promotoras Genéticas , Animais , PPAR gama/genética , PPAR gama/metabolismo , Galinhas/genética , Galinhas/metabolismo , Fator 1 Nuclear Respiratório/genética , Fator 1 Nuclear Respiratório/metabolismo , Proteínas Aviárias/genética , Proteínas Aviárias/metabolismo , Regulação da Expressão Gênica , Gordura Abdominal/metabolismo
5.
Int J Sport Nutr Exerc Metab ; 34(3): 145-153, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38330938

RESUMO

This study sought to investigate the effect of cold ambient temperature on subcutaneous abdominal adipose tissue (SCAAT) lipolysis and blood flow during steady-state endurance exercise in endurance-trained cyclists. Ten males (age: 23 ± 3 years; peak oxygen consumption: 60.60 ± 4.84 ml·kg-1·min-1; body fat: 18.4% ± 3.5%) participated in baseline lactate threshold (LT) and peak oxygen consumption testing, two familiarization trials, and two experimental trials. Experimental trials consisted of cycling in COLD (3 °C; 42% relative humidity) and neutral (NEU; 19 °C; 39% relative humidity) temperatures. Exercise consisted of 25 min cycling at 70% LT and 25 min at 90% LT. In situ SCAAT lipolysis and blood flow were measured via microdialysis. Heart rate, core temperature, carbohydrate and fat oxidation, blood glucose, and blood lactate were also measured. Heart rate, core temperature, oxygen consumption, and blood lactate increased with exercise but were not different between COLD and NEU. SCAAT blood flow did not change from rest to exercise or between COLD and NEU. Interstitial glycerol increased during exercise (p < .001) with no difference between COLD and NEU. Fat oxidation increased (p < .001) at the onset of exercise and remained elevated thereafter with no difference between COLD and NEU. Carbohydrate oxidation increased with increasing exercise intensity and was greater at 70% LT in COLD compared to NEU (p = .030). No differences were observed between conditions for any other variable. Cycling exercise increased SCAAT lipolysis but not blood flow. Ambient temperature did not alter SCAAT metabolism, SCAAT blood flow, or fat oxidation in well-trained cyclists, though cold exposure increased whole-body carbohydrate oxidation at lower exercise intensities.


Assuntos
Tecido Adiposo , Lipólise , Masculino , Humanos , Adulto Jovem , Adulto , Lipólise/fisiologia , Temperatura , Tecido Adiposo/metabolismo , Glicemia/metabolismo , Gordura Abdominal/metabolismo , Lactatos/metabolismo , Consumo de Oxigênio/fisiologia , Glicerol , Temperatura Baixa
6.
PLoS One ; 19(2): e0298662, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38394065

RESUMO

BACKGROUND: The aim was to evaluate the effect different types of abdominal fat have on NAFLD development and the effects of abdominal fat has on the association between Metabolic Syndrome (MetS) and NALFD. METHODS: Data was collected from the cross-sectional NHANES dataset (2017-2018 cycle). Using the controlled attenuation parameter (USG CAP, dB/m), which measures the level of steatosis, the cohort was stratified into two groups: NAFLD(+) (≥274 dB/m) and NAFLD(-). Using complex samples analyses, associations between liver steatosis or NAFLD and types of abdominal fat area [Total abdominal (TAFA), subcutaneous (SAT), and visceral (VAT)] were determined. Pearson's correlation coefficient (r) was calculated to evaluate the associations between adipose tissues and NAFLD. Logistic regression was used to determine the risk [odds ratio (OR) and 95% confidence interval (95%CI)]. Participants were also classified by MetS, using the Harmonizing Definition criteria. RESULTS: Using 1,980 participants (96,282,896 weighted), there was a significant (p<0.001) correlation between USG CAP and TAFA (r = 0.569), VAT (r = 0.645), and SAT (r = 0.479). Additionally, the risk of developing NAFLD was observed for total abdominal obesity (OR = 19.9, 95%CI: 5.1-77.8, p<0.001), visceral obesity (OR = 9.1, 95%CI: 6.2-13.5, p<0.001) and subcutaneous obesity (OR = 4.8, 95%CI: 3.2-6.9, p<0.001). Using 866 participants (44,399,696 weighted), for visceral obesity, participants with MetS and visceral obesity (OR = 18.1, 95%CI: 8.0-41.3, p<0.001) were shown to have a greater risk than participants with MetS only (OR = 6.3, 95%CI: 2.6-15.2, p<0.001). For subcutaneous obesity, again, participants with MetS and subcutaneous obesity (OR = 18.3, 95%CI: 8.0-41.9, p<0.001) were shown to have a greater risk than the MetS-only group (OR = 10.3, 95%CI: 4.8-22.4, p<0.001). CONCLUSION: TAFA, VAT, and SAT were positively associated with USG CAP values and increased the risk of developing NAFLD. Also, the type of abdominal fat depots did affect the association between MetS and NAFLD.


Assuntos
Síndrome Metabólica , Hepatopatia Gordurosa não Alcoólica , Humanos , Hepatopatia Gordurosa não Alcoólica/complicações , Hepatopatia Gordurosa não Alcoólica/epidemiologia , Hepatopatia Gordurosa não Alcoólica/metabolismo , Síndrome Metabólica/complicações , Síndrome Metabólica/epidemiologia , Síndrome Metabólica/metabolismo , Estudos Transversais , Obesidade Abdominal/complicações , Obesidade Abdominal/epidemiologia , Obesidade Abdominal/metabolismo , Inquéritos Nutricionais , Obesidade/complicações , Gordura Abdominal/metabolismo , Gordura Intra-Abdominal/metabolismo
7.
Cardiovasc Diabetol ; 22(1): 335, 2023 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-38066623

RESUMO

BACKGROUND: The assessment of obesity-related health risks has traditionally relied on the Body Mass Index and waist circumference, but their limitations have propelled the need for a more comprehensive approach. The differentiation between visceral (VIS) and subcutaneous (SC) fat provides a finer-grained understanding of these risks, yet practical assessment methods are lacking. We hypothesized that combining the SC-VIS fat ratio with non-invasive biomarkers could create a valuable tool for obesity-related risk assessment. METHODS AND RESULTS: A clinical study of 125 individuals with obesity revealed significant differences in abdominal fat distribution measured by CT-scan among genders and distinct models of obesity, including visceral, subcutaneous, and the SC/VIS ratio. Stratification based on these models highlighted various metabolic changes. The SC/VIS ratio emerged as an excellent metric to differentiate metabolic status. Gene expression analysis identified candidate biomarkers, with ISM1 showing promise. Subsequent validation demonstrated a correlation between ISM1 levels in SC and plasma, reinforcing its potential as a non-invasive biomarker for fat distribution. Serum adipokine levels also correlated with the SC/VIS ratio. The Receiver Operating Characteristic analysis revealed ISM1's efficacy in discriminating individuals with favorable metabolic profiles based on adipose tissue distribution. Correlation analysis also suggested that ISM1 was involved in glucose regulation pathways. CONCLUSION: The study's results support the hypothesis that the SC-VIS fat ratio and its derived non-invasive biomarkers can comprehensively assess obesity-related health risks. ISM1 could predict abdominal fat partitioning and be a potential biomarker for evaluating obesity-related health risks.


Assuntos
Adipocinas , Obesidade , Trombospondinas , Feminino , Humanos , Masculino , Gordura Abdominal/diagnóstico por imagem , Gordura Abdominal/metabolismo , Adipocinas/metabolismo , Tecido Adiposo/metabolismo , Biomarcadores/metabolismo , Índice de Massa Corporal , Gordura Intra-Abdominal/diagnóstico por imagem , Gordura Intra-Abdominal/metabolismo , Obesidade/metabolismo , Gordura Subcutânea/diagnóstico por imagem , Gordura Subcutânea/metabolismo , Trombospondinas/metabolismo
8.
PLoS One ; 18(12): e0295492, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38064530

RESUMO

BACKGROUND: Asian-Indians show thin fat phenotype, characterized by predominantly central deposition of excess fat. The roles of abdominal subcutaneous fat (SAT), intra-peritoneal adipose tissue, and fat depots surrounding the vital organs (IPAT-SV) and liver fat in insulin resistance (IR), type-2 diabetes (T2D) and metabolic syndrome (MetS) in this population are sparsely investigated. AIMS AND OBJECTIVES: Assessment of liver fat, SAT and IPAT-SV by MRI in subjects with T2D and MetS; and to investigate its correlation with IR, specifically according to different quartiles of HOMA-IR. METHODS: Eighty T2D and the equal number of age sex-matched normal glucose tolerant controls participated in this study. Abdominal SAT, IPAT-SV and liver fat were measured using MRI. IR was estimated by the Homeostatic Model Assessment for Insulin Resistance (HOMA-IR). RESULTS: T2D and MetS subjects have higher quantity liver fat and IPAT-SV fat than controls (P = 9 x 10-4 and 4 x 10-4 for T2D and 10-4 and 9 x 10-3 for MetS subjects respectively). MetS subjects also have higher SAT fat mass (P = 0.012), but not the BMI adjusted SAT fat mass (P = 0.48). Higher quartiles of HOMA-IR were associated with higher BMI, W:H ratio, waist circumference, and higher liver fat mass (ANOVA Test P = 0.020, 0.030, 2 x 10-6 and 3 x 10-3 respectively with F-values 3.35, 3.04, 8.82, 4.47 respectively). In T2D and MetS subjects, HOMA-IR showed a moderately strong correlation with liver fat (r = 0.467, P < 3 x 10-5 and r = 0.493, P < 10-7), but not with SAT fat and IPAT-SV. However, in MetS subjects IPAT-SV fat mass showed borderline correlation with IR (r = 0.241, P < 0.05), but not with the BMI adjusted IPAT-SV fat mass (r = 0.13, P = 0.26). In non-T2D and non-MetS subjects, no such correlation was seen. On analyzing the correlation between the three abdominal adipose compartment fat masses and IR according to its severity, the correlation with liver fat mass becomes stronger with increasing quartiles of HOMA-IR, and the strongest correlation is seen in the highest quartile (r = 0.59, P < 10-3). On the other hand, SAT fat mass tended to show an inverse relation with IR with borderline negative correlation in the highest quartile (r = -0.284, P < 0.05). IPAT-SV fat mass did not show any statistically significant correlation with HOMA-IR, but in the highest quartile it showed borderline, but statistically insignificant positive correlation (P = 0.07). CONCLUSION: In individuals suffering from T2D and MetS, IR shows a trend towards positive and borderline negative correlation with liver fat and SAT fat masses respectively. The positive trend with liver fat tends to become stronger with increasing quartile of IR. Therefore, these findings support the theory that possibly exhaustion of protective compartment's capacity to store excess fat results in its pathological deposition in liver as ectopic fat.


Assuntos
Diabetes Mellitus Tipo 2 , Resistência à Insulina , Síndrome Metabólica , Humanos , Diabetes Mellitus Tipo 2/metabolismo , Índice de Massa Corporal , Gordura Abdominal/diagnóstico por imagem , Gordura Abdominal/metabolismo
9.
Genes (Basel) ; 14(7)2023 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-37510361

RESUMO

Reducing abdominal fat (AF) accumulation and increasing the level of intramuscular fat (IMF) simultaneously is a major breeding goal in the poultry industry. To explore the different molecular mechanisms underlying AF and IMF, gene expression profiles in the breast muscle (BM) and AF from three chicken breeds were analyzed. A total of 4737 shared DEGs were identified between BM and AF, of which 2602 DEGs were upregulated and 2135 DEGs were downregulated in the BM groups compared with the AF groups. DEGs involved in glycerophospholipid metabolism and glycerolipid metabolism were potential regulators, resulting in the difference in lipid metabolite accumulation between IMF and AF. The PPAR signaling pathway was the most important pathway involved in tissue-specific lipid deposition. Correlation analysis showed that most representative DEGs enriched in the PPAR signaling pathway, such as FABP5, PPARG, ACOX1, and GK2, were negatively correlated with PUFA-enriched glycerophospholipid molecules. Most DEGs related to glycerophospholipid metabolism, such as GPD2, GPD1, PEMT, CRLS1, and GBGT1, were positively correlated with glycerophospholipid molecules, especially DHA- and arachidonic acid (ARA)-containing glycerophospholipid molecules. This study elucidated the molecular mechanism underlying tissue-specific lipid deposition and poultry meat quality.


Assuntos
Galinhas , Perfilação da Expressão Gênica , Animais , Perfilação da Expressão Gênica/métodos , Galinhas/genética , Galinhas/metabolismo , PPAR gama/genética , Gordura Abdominal/metabolismo , Lipídeos
10.
Front Endocrinol (Lausanne) ; 14: 1166961, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37361522

RESUMO

Background and purpose: Napping is a widespread practice worldwide and has in recent years been linked to increased abdominal adiposity. Lipase E or LIPE encodes the protein hormone-sensitive lipase (HSL), an enzyme that plays an important role in lipid mobilization and exhibits a circadian expression rhythm in human adipose tissue. We hypothesized that habitual napping may impact the circadian expression pattern of LIPE, which in turn may attenuate lipid mobilization and induce abdominal fat accumulation. Methods: Abdominal adipose tissue explants from participants with obesity (n = 17) were cultured for a 24-h duration and analyzed every 4 h. Habitual nappers (n = 8) were selected to match non-nappers (n = 9) in age, sex, BMI, adiposity, and metabolic syndrome traits. Circadian LIPE expression rhythmicity was analyzed using the cosinor method. Results: Adipose tissue explants exhibited robust circadian rhythms in LIPE expression in non-nappers. In contrast, nappers had a flattened rhythm. LIPE amplitude was decreased in nappers as compared with non-nappers (71% lower). The decrease in amplitude among nappers was related to the frequency of napping (times per week) where a lower rhythm amplitude was associated with a higher napping frequency (r = -0.80; P = 0.018). Confirmatory analyses in the activity of LIPE's protein (i.e., HSL) also showed a significant rhythm in non-nappers, whereas significance in the activity of HSL was lost among nappers. Conclusion: Our results suggest that nappers display dysregulated circadian LIPE expression as well as dysregulated circadian HSL activity, which may alter lipid mobilization and contribute to increased abdominal obesity in habitual nappers.


Assuntos
Tecido Adiposo , Lipase , Esterol Esterase , Humanos , Gordura Abdominal/metabolismo , Tecido Adiposo/metabolismo , Ritmo Circadiano , Obesidade/metabolismo , Esterol Esterase/metabolismo
11.
NPJ Biofilms Microbiomes ; 9(1): 28, 2023 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-37253749

RESUMO

Cecal microbiota plays an essential role in chicken health. However, its contribution to fat metabolism, particularly in abdominal fat deposition, which is a severe problem in the poultry industry, is still unclear. Here, chickens at 1, 4, and 12 months of age with significantly (p < 0.05) higher and lower abdominal fat deposition were selected to elucidate fat metabolism. A significantly (p < 0.05) higher mRNA expression of fat anabolism genes (ACSL1, FADS1, CYP2C45, ACC, and FAS), a significantly (p < 0.05) lower mRNA expression of fat catabolism genes (CPT-1 and PPARα) and fat transport gene APOAI in liver/abdominal fat of high abdominal fat deposition chickens indicated that an unbalanced fat metabolism leads to excessive abdominal fat deposition. Parabacteroides, Parasutterella, Oscillibacter, and Anaerofustis were found significantly (p < 0.05) higher in high abdominal fat deposition chickens, while Sphaerochaeta was higher in low abdominal fat deposition chickens. Further, Spearman correlation analysis indicated that the relative abundance of cecal Parabacteroides, Parasutterella, Oscillibacter, and Anaerofustis was positively correlated with abdominal fat deposition, yet cecal Sphaerochaeta was negatively correlated with fat deposition. Interestingly, transferring fecal microbiota from adult chickens with low abdominal fat deposition into one-day-old chicks significantly (p < 0.05) decreased Parabacteroides and fat anabolism genes, while markedly increased Sphaerochaeta (p < 0.05) and fat catabolism genes (p < 0.05). Our findings might help to assess the potential mechanism of cecal microbiota regulating fat deposition in chicken production.


Assuntos
Galinhas , Microbiota , Animais , Metabolismo dos Lipídeos , RNA Mensageiro/metabolismo , Gordura Abdominal/metabolismo
12.
PLoS One ; 18(4): e0285025, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37104286

RESUMO

In this study, we have compared frequencies, phenotype, function and metabolic requirements of B cells isolated from the breast and abdominal subcutaneous adipose tissue (AT) of women with obesity who underwent weight reduction surgeries. Results show that B cells from the abdominal AT are more inflammatory than those from the breast, characterized by higher frequencies of inflammatory B cell subsets and higher expression of RNA for inflammatory markers associated with senescence. Secretion of autoimmune antibodies is also higher in the abdominal AT as compared to the breast, and is associated with higher frequencies of autoimmune B cells with the membrane phenotype CD21lowCD95+ B cells expressing the transcription factor T-bet. Moreover, glucose uptake is higher in B cells from the abdominal AT as compared to the breast, thereby suggesting a better capacity to perform glycolysis, needed to support intrinsic B cell inflammation and autoimmune antibody secretion.


Assuntos
Adiposidade , Obesidade , Humanos , Feminino , Fenótipo , Gordura Subcutânea Abdominal , Gordura Abdominal/metabolismo , Autoanticorpos/metabolismo , Tecido Adiposo/metabolismo , Gordura Subcutânea/metabolismo
13.
Scand J Med Sci Sports ; 33(8): 1462-1472, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37081735

RESUMO

OBJECTIVES: Fat depots localization has a critical role in the metabolic health status of adults. Nevertheless, whether that is also the case in children remains under-studied. Therefore, the aims of this study were: (i) to examine the differences between metabolically healthy (MHO) and unhealthy (MUO) overweight/obesity phenotypes on specific abdominal fat depots, and (ii) to further explore whether cardiorespiratory fitness plays a major role in the differences between metabolic phenotypes among children with overweight/obesity. METHODS: A total of 114 children with overweight/obesity (10.6 ± 1.1 years, 62 girls) were included. Children were classified as MHO (n = 68) or MUO. visceral (VAT), abdominal subcutaneous (ASAT), intermuscular abdominal (IMAAT), psoas, hepatic, pancreatic, and lumbar bone marrow adipose tissues were measured by magnetic resonance imaging. Cardiorespiratory fitness was assessed using the 20 m shuttle run test. RESULTS: MHO children had lower VAT and ASAT contents and psoas fat fraction compared to MUO children (difference = 12.4%-25.8%, all p < 0.035). MUO-unfit had more VAT and ASAT content than those MUO-fit and MHO-fit (difference = 34.8%-45.3%, all p < 0.044). MUO-unfit shows also greater IMAAT fat fraction than those MUO-fit and MHO-fit peers (difference = 16.4%-13.9% respectively, all p ≤ 0.001). In addition, MHO-unfit presented higher IMAAT fat fraction than MHO-fit (difference = 13.4%, p < 0.001). MUO-unfit presented higher psoas fat fraction than MHO-fit (difference = 29.1%, p = 0.008). CONCLUSIONS: VAT together with ASAT and psoas fat fraction, were lower in MHO than in MUO children. Further, we also observed that being fit, regardless of metabolic phenotype, has a protective role over the specific abdominal fat depots among children with overweight/obesity.


Assuntos
Aptidão Cardiorrespiratória , Síndrome Metabólica , Humanos , Sobrepeso , Obesidade/metabolismo , Nível de Saúde , Gordura Abdominal/diagnóstico por imagem , Gordura Abdominal/metabolismo , Fenótipo , Síndrome Metabólica/metabolismo , Fatores de Risco , Índice de Massa Corporal
14.
Poult Sci ; 102(6): 102638, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37015160

RESUMO

This experiment was carried out to investigate the mechanism of action of mulberry leaf extract (MLE) in reducing abdominal fat accumulation in female broilers. A total of 192 one-day-old female Arbor Acres (AA) broilers were divided into 4 diet groups, with each group consisting of 8 replicates with 6 birds per replicate. The diets contained a basal diet and 3 test diets with supplementation of 400, 800, or 1,200 MLE mg/kg, respectively. The trial had 2 phases that lasted from 1 to 21 d and from 22 to 56 d, respectively. The growth performance, abdominal fat deposition, fatty acid composition, serum biochemistry and mRNA expression of genes related to fat metabolism in liver were determined. The results showed that, 1) dietary supplementation with MLE had no significant impact on broilers final body weight, average daily gain (ADG), or feed to gain ration (F/G) (P > 0.05), but linearly reduced abdominal fat accumulation in both experimental phases (P < 0.05); 2) the total contents of monounsaturated fatty acids (MUFA) and polyunsaturated fatty acids (PUFA), such as palmitoleic acid, oleic acid, and eicosadienoic acid, were increased quadratically as a result of dietary supplements of 400, 800, and 1,200 mg/kg MLE (P < 0.01), while the total contents of saturated fatty acids (SFA), such as teracosanoic acid were decreased (P < 0.01); 3) the addition of 800 or 1,200 MLE mg/kg to the diet linearly reduced total cholesterol (TC) in the serum and liver (P < 0.05). Adenosine-activated protein kinase (AMPK) mRNA expression in the liver was quadratically increased by the addition of 800 or 1,200 MLE mg/kg to the diet (P < 0.05), and the mRNA expression of sterol regulatory element binding protein-1c (SREBP-1c), acetyl-CoA carboxylase (ACC), and acetyl-CoA carboxylate), fatty acid synthase (FAS) were linearly decreased (P < 0.05). In conclusion, MLE can be employed as a viable fat loss feed supplement in fast-growing broiler diets since it reduces abdominal fat deposition in female AA broilers via the AMPK/SREBP-1c/ACC signaling pathway. MLE can also be utilized to modify the fatty acid profile in female broilers (AA) at varied inclusion levels.


Assuntos
Galinhas , Morus , Animais , Feminino , Galinhas/fisiologia , Acetil-CoA Carboxilase/genética , Acetil-CoA Carboxilase/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Adenosina Quinase/metabolismo , Ácidos Graxos/metabolismo , Fígado/metabolismo , Ácidos Graxos Insaturados/metabolismo , Transdução de Sinais , Gordura Abdominal/metabolismo , Extratos Vegetais/farmacologia , Extratos Vegetais/metabolismo , RNA Mensageiro/metabolismo
15.
Int J Mol Sci ; 24(2)2023 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-36675280

RESUMO

Prognostic factors for epithelial ovarian cancers (EOCs) are in particular clinical factors such as pathology staging at diagnosis (FIGO stages), genetic mutation, or histological phenotypes. In the present study, FIGO stage, tumor residue after surgery, and body mass index were clinical predictors of recurrence-free survival (RFS). Nonetheless, a number of studies support a lipid metabolism disorder in ovarian cancer patients. The objective of this pilot study was to explore whether fatty acid composition of adipose reflecting the qualitative dietary intake and fatty acids metabolism may be associated with RFS. Forty-six women with EOCs and six with borderline ovarian tumors between March 2017 and January 2020 were included in this prospective study at Tours university teaching hospital (central France). The patients involved in the present study are part of the METERMUS trial (clinicaltrials.gov NCT03027479). Adipose tissue specimens from four abdominal locations (superficial and deep subcutaneous, visceral (pericolic), and omental) were collected during surgery or exploratory laparoscopy. A fatty acid profile of adipose tissue triglycerides was established by gas chromatography. Fatty acids composition was compared among the four locations using nonparametric Friedman's ANOVA test for repeated measures. Median follow-up of EOC patients was 15 months and patients' RFS was analyzed using Kaplan−Meier survival curves and log-rank test by separating patients into two groups according to median fatty acid levels. The content of long-chain saturated fatty acids (SFAs) was increased and that of long-chain polyunsaturated fatty acids (PUFAs) decreased in deep versus superficial subcutaneous adipose tissue in EOC patients. Nevertheless, the content of total SFAs was ~28%, monounsaturated fatty acids (MUFAs) ~55%, PUFAs n-6 ~11.5%, and PUFAs n-3 about 1.3%, whatever the adipose tissue. When EOC patients were separated into two groups by median fatty acid content, total PUFAs (n-6+n-3) levels, whatever the adipose tissue, were positively and independently associated with RFS. RFS was about two times longer in EOC patients with high versus low total PUFA content (median survival: 12 vs. 27 months, p = 0.01 to <0.0001 according to the tissue). Content of total PUFAs (n-6+n-3) in abdominal adipose tissue (visceral and subcutaneous) are new prognostic factors in EOC.


Assuntos
Ácidos Graxos Insaturados , Neoplasias Ovarianas , Feminino , Humanos , Estudos Prospectivos , Projetos Piloto , Ácidos Graxos Insaturados/metabolismo , Ácidos Graxos/metabolismo , Tecido Adiposo/metabolismo , Gordura Abdominal/metabolismo , Neoplasias Ovarianas/metabolismo
16.
Clin Endocrinol (Oxf) ; 98(2): 229-237, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36267004

RESUMO

CONTEXT: 17ß-hydroxysteroid dehydrogenase type 2 (17ß-HSD2) may be involved in the local modulation of estradiol (E2) availability in adipose tissues. OBJECTIVE: To assess the conversion of E2 into estrone (E1) as well as the expression of 17ß-HSD2 and its localization in omental (OM) and subcutaneous (SC) adipose tissues obtained from women. METHODS: Rates of 14 C-E1 formation from 14 C-E2 were measured in OM and SC adipose tissue homogenates from 29 women. Specific 17ß-HSD2 inhibitor EM-919 was tested in OM and SC adipose tissue homogenates (n = 6). 17ß-HSD2 mRNA expression was measured in whole OM and SC adipose tissues (n = 14). Cellular localization of the enzyme was examined using immunohistochemistry. Anthropometric measurements were obtained and body composition as well as body fat distribution were measured. RESULTS: Significant 14 C-E1 formation from 14 C-E2 in OM and SC tissue homogenates was detected. The rate of 14 C-E1 formation was significantly higher in OM than SC adipose tissue (p < .0001). The conversion of 14 C-E2 to 14 C-E1 was significantly inhibited by EM-919 in OM (p < .05) and SC (p < .05) adipose tissues. Significantly higher expression of 17ß-HSD2 mRNA in OM versus SC fat was found (p = .03). 17ß-HSD2 was localized in the vasculature of OM and SC tissues. Significant negative associations were detected between OM 17ß-HSD2 activity and body mass index, WC, lean body mass as well as SC adipose tissue areas. CONCLUSION: 17ß-HSD2 converts E2 to E1 in OM and SC adipose tissues of women. The activity of this enzyme decreases with increasing adiposity.


Assuntos
17-Hidroxiesteroide Desidrogenases , Gordura Abdominal , Humanos , Feminino , 17-Hidroxiesteroide Desidrogenases/genética , Gordura Abdominal/metabolismo , Estradiol/metabolismo , RNA Mensageiro/genética
17.
Amyloid ; 30(1): 27-37, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35792725

RESUMO

BACKGROUND: Systemic AL amyloidosis arises from the misfolding of patient-specific immunoglobulin light chains (LCs). Potential drivers of LC amyloid formation are mutational changes and post-translational modifications (PTMs). However, little information is available on the exact primary structure of the AL proteins and their precursor LCs. OBJECTIVE: We analyse the exact primary structure of AL proteins extracted from 10 λ AL amyloidosis patients and their corresponding precursor LCs. MATERIALS AND METHODS: By cDNA sequencing of the precursor LC genes in combination with mass spectrometry of the AL proteins, the exact primary structure and PTMs were determined. This information was used to analyse their biochemical properties. RESULTS: All AL proteins comprise the VL and a small part of the CL with a common C-terminal truncation region. While all AL proteins retain the conserved native disulphide bond of the VL, we found no evidence for presence of other common PTMs. The analysis of the biochemical properties revealed that the isoelectric point of the VL is significantly increased due to introduced mutations. CONCLUSION: Our data imply that mutational changes influence the surface charge properties of the VL and that common proteolytic processes are involved in the generation of the cleavage sites of AL proteins.


Assuntos
Amiloidose , Amiloidose de Cadeia Leve de Imunoglobulina , Humanos , Amiloidose de Cadeia Leve de Imunoglobulina/genética , Amiloidose/genética , Amiloidose/metabolismo , Cadeias Leves de Imunoglobulina/metabolismo , Amiloide/genética , Amiloide/metabolismo , Espectrometria de Massas , Gordura Abdominal/metabolismo
18.
Arch Microbiol ; 204(11): 670, 2022 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-36241916

RESUMO

Bacteria capable of producing electricity in intestinal microbiota have been discovered. However, no studies have explored butyric acid which generated by electrogenic bacteria on the host organism have significant physiological impacts on certain organs. We found that the capacity for electrical current generation by the commensal gut Leuconostoc mesenteroides EH-1 (L. mesenteroides EH-1) during glucose fermentation. The electricity production was essential for the gut colonization of L. mesenteroides EH-1 since the inhibition of electricity production by cyclophilin A inhibitor (TMN355) significantly diminished the number of bacteria attached to the human gut epithelial cell surface. The adipocyte differentiation contributes to the increased 4-hydroxy-2-nonenal (4-HNE), considered as a biomarker of reactive oxygen species (ROS). The effect of intestinal electrogenic microbiota in the high-fat diet (HFD)-induced 4-HNE and abdominal fat accumulation in mice was investigated in this study. The oral administration of glucose with a butyric acid-producing L. mesenteroides EH-1 bacterium attenuated the expression of 4-HNE and abdominal fat. The level of 4-HNE and abdominal fat depot were markedly increased in mice administered with cyclophilin A inhibitor-pretreated bacteria or GLPG-0974, an antagonist of free fatty acid receptor 2 (Ffar2). Our studies suggest a novel means by which the probiotic bacteria can modulate fat mass deposition and oxidative stress via the cyclophilin A-mediated electron production and the butyric acid-activated Ffar2 pathway.


Assuntos
Leuconostoc mesenteroides , Gordura Abdominal/metabolismo , Animais , Bactérias/metabolismo , Ácido Butírico , Ciclofilina A/metabolismo , Dieta Hiperlipídica/efeitos adversos , Eletricidade , Ácidos Graxos não Esterificados/metabolismo , Fermentação , Glucose/metabolismo , Humanos , Leuconostoc mesenteroides/metabolismo , Camundongos , Espécies Reativas de Oxigênio/metabolismo
19.
Poult Sci ; 101(12): 102169, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36201879

RESUMO

Transforming growth factor beta receptor Ⅱ (TGFBR2), a core member of the transforming growth factor-ß (TGF-ß) signaling pathway. To date, chicken TGFBR2 (cTGFBR2) genomic structure has not been fully explored. Here, the complete sequences of cTGFBR2 transcript isoforms were determined by 5' and 3' rapid amplification of cDNA ends (5' & 3' RACE) and reverse transcription polymerase chain reaction (RT-PCR); the tissue expression profiling of cTGFBR2 transcript isoforms was performed using quantitative real-time polymerase chain reaction (qRT-PCR). The results showed that cTGFBR2 gene produced 3 transcript isoforms though alternative transcription initiation, splicing, and polyadenylation, which were designated as cTGFBR2-1, cTGFBR2-2, and cTGFBR2-3, respectively. These 3 cTGFBR2 transcript isoforms encoded 3 protein isoforms: cTGFBR2-1, cTGFBR2-2, and cTGFBR2-3. Duplication analysis revealed that, unlike other animal species, cTGFBR2 gene harbored a 5.5-kb intragenic tandem duplication. Tissue expression profiling in the 4-wk-old Arbor Acres (AA) broiler chickens showed that cTGFBR2-1 was ubiquitously expressed, with high expression in abdominal fat, subcutaneous fat, lung, gizzard, and muscle; cTGFBR2-2 was highly expressed in heart, kidney, gizzard, and muscle; cTGFBR2-3 was weakly expressed in all the tested chicken tissues. Tissue expression profiling in the 7-wk-old broiler chickens of the fat and lean lines of Northeast Agricultural University broiler lines divergently selected for abdominal fat content (NEAUHLF) showed that cTGFBR2-1 was significantly differentially expressed in all the tested tissues except heart, cTGFBR2-2 was significantly differentially expressed in all the tested tissues except subcutaneous fat and liver, and cTGFBR2-3 was significantly differentially expressed in all the tested tissues between the lean and fat lines. Intriguingly, in the fat line, the 3 cTGFBR2 transcript isoforms were expressed to varying degrees in all the 3 tested fat tissues, while in the lean line, only cTGFBR2-1 was expressed in all the 3 tested fat tissues. This is the first report of intragenic tandem duplication within TGFBR2 gene. Our findings pave the way for further studies on the functions and regulation of cTGFBR2 gene.


Assuntos
Gordura Abdominal , Galinhas , Animais , Receptor do Fator de Crescimento Transformador beta Tipo II/metabolismo , Gordura Abdominal/metabolismo , Isoformas de Proteínas/metabolismo , Genômica
20.
Poult Sci ; 101(12): 102216, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36279606

RESUMO

Glycoprotein nonmetastatic melanoma protein B (GPNMB) is a vital secreted factor that promotes the occurrence of obesity in mammals. However, the effects of GPNMB on abdominal fat deposition is still unknown in chickens. In this study, we looked into the genetic and expression association of GPNMB gene with abdominal fat traits in chicken, and found that a genetic variation rs31126482 in GPNMB promoter was significantly associated with abdominal fat weight (AFW, P < 0.05) and abdominal fat percentage (AFP, P < 0.01). Express profile analysis of the GPNMB indicated that the gene was mainly expressed in abdominal fat tissue, and its expression level was strongly positively correlated with AFW (R2 = 0.6356, P = 4.10E-05) and AFP (R2 = 0.6450, P = 2.90E-05). We then investigated biological function of GPNMB on adipogenesis in chicken, and found that GPNMB could inhibit abdominal preadipocyte proliferation, but promote abdominal preadipocyte differentiation and lipid deposition. Furthermore, we explored regulatory mechanism of GPNMB gene in chicken, and detected one nonclassical estrogen regulatory element (AP1) and one peroxisome proliferator-activated receptor α (PPARα) responsive element in the 2 kb promoter region of GPNMB gene, and demonstrated that estrogen could up-regulate GPNMB mRNA expression in adipose tissue and primary abdominal preadipocytes, while PPARα could down-regulate GPNMB expression in primary preadipocytes. Taken together, this study brings new insights into understanding the function and transcriptional control of GPNMB gene, and provides genetic markers for breeding selection to improve abdominal fat traits in chicken.


Assuntos
Galinhas , PPAR alfa , Animais , Galinhas/genética , Galinhas/metabolismo , PPAR alfa/genética , PPAR alfa/metabolismo , alfa-Fetoproteínas/genética , alfa-Fetoproteínas/metabolismo , Gordura Abdominal/metabolismo , Estrogênios/metabolismo , Variação Genética , Mamíferos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA