Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Hormones (Athens) ; 2024 May 31.
Article in English | MEDLINE | ID: mdl-38819743

ABSTRACT

INTRODUCTION: Somatic mutations in ubiquitin-specific protease-8 (USP8), encoding a deubiquinating protein, are found in approximately 30% of corticotroph-derived pituitary adenomas (CPAs). Stratifin, a protein encoded by SFN, inhibits USP8 catalytic activity. USP8 has immunomodulating properties that have been demonstrated in non-tumoral diseases. METHODS: We assessed the influence of USP8 on the immune landscape of CPA and validated this effect and its dependency on stratifin in large cohorts of non-pituitary tumors. We analyzed data of CPA samples (n = 20) and additional non-pituitary tumors from the TCGA database, using transcriptome signature-recognition algorithms. Immune tumor microenvironment (iTME) was compared both by USP8 and SFN expression levels (n = 843) and by USP8 mutation status and SFN expression (n = 12,389). RESULTS: CPA with activating USP8 mutations was associated with "cold" iTME compared with wild-type USP8 CPA, as reflected by lower fractions of immune cells, including B cells, CD4, regulatory and gamma/delta T cells, natural killer cells, M0 and M1 macrophages, dendritic cells, and eosinophils (p < 0.05 for all comparisons). Pathways altered by the presence of USP8 mutation, based on the most differentially expressed genes (3061 genes), included microglia pathogen phagocytosis and multiple toll-like receptor signaling pathways (p < 0.0001). In a validation analysis based on large cohorts of non-pituitary tumors, high expression of USP8 was associated with a suppressed iTME effect that was augmented by a low SFN expression. CONCLUSIONS: Our data demonstrate for the first time, to our knowledge, a distinct immune landscape of tumors based on USP8 status and expression and the dependency of this immunological effect on SFN expression.

2.
Article in English | MEDLINE | ID: mdl-38317059

ABSTRACT

BACKGROUND: Familial Mediterranean Fever (FMF) is a genetic disorder characterized by recurrent episodes of fever and inflammation in various organs, including the joints. Traditionally, the arthritis of FMF has been considered relatively harmless. However, anecdotal evidence has suggested that it may contribute to long-term joint damage, which may necessitate surgical joint replacement. This study aimed to investigate the rates of arthroplasty among FMF patients and compare it to the general population. METHODS: The study used the electronic database of the largest healthcare organization in Israel to identify 9,769 FMF patients diagnosed between 2000 and 2016. A similar number of age-, gender-, and residency-matched controls were also identified. The rates of arthroplasty were compared between the two groups. A logistic regression model predicting the need for arthroplasty within the FMF group was formed to identify potential risk factors. RESULTS: Of the 9,769 FMF patients, 114 (1.2%) underwent arthroplasty, compared with 64 (0.7%) of the control group [unadjusted odds ratio (OR)=1.79, 95% confidence interval (CI) 1.32-2.43; partially adjusted OR = 1.97, 95% CI 1.40-2.77; fully adjusted OR = 1.92, 95% CI 1.35-2.72]. Within the FMF cohort, those of North African origin had a significantly higher risk of arthroplasty (OR = 6.89, 95% CI 5.09-9.33; p< 0.001). CONCLUSION: FMF patients can experience long-term joint damage that may require arthroplasty. Although this complication is relatively uncommon in FMF patients, it occurs almost twice as frequently as compared with the general population. FMF patients of North African origin are at an even higher risk.

4.
Cells ; 11(19)2022 09 29.
Article in English | MEDLINE | ID: mdl-36231008

ABSTRACT

Objective: Up-regulated expression of transcription-factor E2F1 in human visceral adipose tissue (VAT) characterizes a dysmetabolic obesity sub-phenotype. An E2F1-miRNA network has been described in multiple cancers. Here we investigated whether elevated VAT-E2F1 in obesity is associated with VAT-miRNA alterations similar to, or distinct from, those described in cancer. Furthermore, we assessed if E2F1-associated miRNA changes may contribute to the link between high- VAT-E2F1 and a dysmetabolic obesity phenotype. Methods: We assembled a cohort of patients with obesity and high-VAT-E2F1, matched by age, sex, ±BMI to patients with low-VAT-E2F1, with and without obesity (8 patients/groupX3 groups). We performed Nanostring©-based miRNA profiling of VAT samples from all 24 patients. Candidate E2F1-related miRNAs were validated by qPCR in an independent cohort of patients with extreme obesity, with or without type-2-diabetes (T2DM) (n = 20). Bioinformatic tools and manipulation of E2F1 expression in cells were used to establish the plausibility of the functional VAT-E2F1-miRNA network in obesity. Results: Among n = 798 identified miRNAs, 17 were differentially expressed in relation to E2F1 and not to obesity itself. No evidence for the cancer-related E2F1-miRNA network was identified in human VAT in obesity. In HEK293-cells, overexpression/downregulation of E2F1 correspondingly altered the expression of miRNA-206 and miRNA-210-5p, two miRNAs with reported metabolic functions consistent with those of E2F1. In VAT from both cohorts, the expression of both miRNA-206 and 210-5p intercorrelated, and correlated with the expression of E2F1. In cohort 1 we did not detect significant associations with biochemical parameters. In cohort 2 of patients with extreme obesity, all those with high VAT-E2F1 showed a diabetes-complicated obesity phenotype and higher expression of miRNA-206 and miRNA-210-5p, which also correlated with fasting glucose levels (both miRNAs) and fasting insulin (miRNA-210-5p). Conclusions: Whilst the previously described cancer-related E2F1-miRNA network does not appear to operate in VAT in obesity, miRNAs-206 and 210-5p may link high-E2F1 expression in VAT with diabetes-complicated extreme obesity phenotype.


Subject(s)
Diabetes Mellitus, Type 2 , MicroRNAs , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Glucose/metabolism , HEK293 Cells , Humans , Insulin/metabolism , Intra-Abdominal Fat/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Obesity/genetics , Obesity/metabolism
5.
Lung Cancer ; 172: 136-141, 2022 10.
Article in English | MEDLINE | ID: mdl-36087486

ABSTRACT

OBJECTIVE: Despite major progress over the past decade in the field of lung cancer care, only mild advances have occurred in Small Cell Lung Cancer (SCLC), with prognosis remaining poor. Based on two randomized clinical trials (RCTs), two checkpoint-inhibitors have recently been approved in extensive-SCLC with moderate improvements in median overall survival (OS). However, only limited data exist regarding the impact of immunotherapy in real-world SCLC patients. This study reports the efficacy of immunotherapy in the first-line treatment of extensive-stage SCLC patients in a real-world setting. METHODS: a retrospective cohort study of all patients treated for extensive-SCLC with chemotherapy with or without immunotherapy, at a single center in Israel between October-2017 and July-2021. Patient characteristics, adverse-events and survival analyses were conducted. RESULTS: Of 102 patients identified, 54 patients (53%) received immunotherapy in addition to chemotherapy. 34.7% of patients had a performance status (PS) of 2-4. Patients that received only chemotherapy were older, had more liver metastasis and a poorer PS. In the whole cohort, patients receiving immunotherapy had a significantly longer median OS (353 days vs 194 days, HR = 0.40p < 0.0001). After stratification by PS groups, survival analysis remained significantly longer in the PS 0-1 group (HR 0.43, p = 0.0036), with a trend for better survival in the PS 2-3 group. Multivariate analysis validated an OS advantage with immunotherapy (HR = 0.46, p = 0.004). CONCLUSION: We present evidence for the efficacy of immunotherapy in SCLC in a real-world setting. Although treatment groups differ in their baseline characteristics, it appears that even some patients not included in RCTs, such as poor PS, may benefit from the addition of immunotherapy to their treatment protocol.


Subject(s)
Lung Neoplasms , Small Cell Lung Carcinoma , Humans , Immunotherapy/methods , Lung Neoplasms/drug therapy , Prognosis , Small Cell Lung Carcinoma/drug therapy , Treatment Outcome
6.
Front Endocrinol (Lausanne) ; 13: 860799, 2022.
Article in English | MEDLINE | ID: mdl-35574032

ABSTRACT

Obesity is a heterogenous condition that affects the life and health of patients to different degrees and in different ways. Yet, most approaches to treat obesity are not currently prescribed, at least in a systematic manner, based on individual obesity sub-phenotypes or specifically-predicted health risks. Adipose tissue is one of the most evidently affected tissues in obesity. The degree of adipose tissue changes - "adiposopathy", or as we propose to relate to herein as Obesity-related Adipose tissue Disease (OrAD), correspond, at least cross-sectionally, to the extent of obesity-related complications inflicted on an individual patient. This potentially provides an opportunity to better personalize anti-obesity management by utilizing the information that can be retrieved by assessing OrAD. This review article will summarize current knowledge on histopathological OrAD features which, beyond cross-sectional analyses, had been shown to predict future obesity-related endpoints and/or the response to specific anti-obesity interventions. In particular, the review explores adipocyte cell size, adipose tissue inflammation, and fibrosis. Rather than highly-specialized methods, we emphasize standard pathology laboratory approaches to assess OrAD, which are readily-available in most clinical settings. We then discuss how OrAD assessment can be streamlined in the obesity/weight-management clinic. We propose that current studies provide sufficient evidence to inspire concerted efforts to better explore the possibility of predicting obesity related clinical endpoints and response to interventions by histological OrAD assessment, in the quest to improve precision medicine in obesity.


Subject(s)
Avitaminosis , Precision Medicine , Adipocytes/pathology , Adipose Tissue/pathology , Cross-Sectional Studies , Humans , Obesity/complications , Obesity/pathology , Obesity/therapy
7.
Am J Physiol Endocrinol Metab ; 321(5): E702-E713, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34632797

ABSTRACT

In chronic obesity, activated adipose tissue proinflammatory cascades are tightly linked to metabolic dysfunction. Yet, close temporal analyses of the responses to obesogenic environment such as high-fat feeding (HFF) in susceptible mouse strains question the causal relationship between inflammation and metabolic dysfunction, and/or raises the possibility that certain inflammatory cascades play adaptive/homeostatic, rather than pathogenic roles. Here, we hypothesized that CTRP6, a C1QTNF family member, may constitute an early responder to acute nutritional changes in adipose tissue, with potential physiological roles. Both 3-days high-fat feeding (3dHFF) and acute obesity reversal [2-wk switch to low-fat diet after 8-wk HFF (8wHFF)] already induced marked changes in whole body fuel utilization. Although adipose tissue expression of classical proinflammatory cytokines (Tnf-α, Ccl2, and Il1b) exhibited no, or only minor, change, C1qtnf6 uniquely increased, and decreased, in response to 3dHFF and acute obesity reversal, respectively. CTRP6 knockout (KO) mouse embryonic fibroblasts (MEFs) exhibited increased adipogenic gene expression (Pparg, Fabp4, and Adipoq) and markedly reduced inflammatory genes (Tnf-α, Ccl2, and Il6) compared with wild-type MEFs, and recombinant CTRP6 induced the opposite gene expression signature, as assessed by RNA sequencing. Consistently, 3dHFF of CTRP6-KO mice induced a greater whole body and adipose tissue weight gain compared with wild-type littermates. Collectively, we propose CTRP6 as a gene that rapidly responds to acute changes in caloric intake, acting in acute overnutrition to induce a "physiological inflammatory response" that limits adipose tissue expansion.NEW & NOTEWORTHY CTRP6 (C1qTNF6), a member of adiponectin gene family, regulates inflammation and metabolism in established obesity. Here, short-term high-fat feeding in mice is shown to increase adipose tissue expression of CTRP6 before changes in the expression of classical inflammatory genes occur. Conversely, CTRP6 expression in adipose tissue decreases early in the course of obesity reversal. Gain- and loss-of-function models suggest CTRP6 as a positive regulator of inflammatory cascades, and a negative regulator of adipogenesis and adipose tissue expansion.


Subject(s)
Adipokines/physiology , Adipose Tissue/pathology , Inflammation/genetics , Nutritional Physiological Phenomena/genetics , Adipogenesis/genetics , Adipokines/genetics , Adipose Tissue/metabolism , Animals , Cells, Cultured , Diet, High-Fat , Embryo, Mammalian , Female , HEK293 Cells , Humans , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Size/genetics , Overnutrition/genetics , Overnutrition/metabolism , Overnutrition/pathology , Pregnancy
8.
Physiol Rep ; 9(18): e15044, 2021 09.
Article in English | MEDLINE | ID: mdl-34553504

ABSTRACT

In humans, exercise-induced thermogenesis is a markedly variable component of total energy expenditure, which had been acutely affected worldwide by COVID-19 pandemic-related lockdowns. We hypothesized that dietary macronutrient composition may affect metabolic adaptation/fuel selection in response to an acute decrease in voluntary activity. Using mice fed short-term high-fat diet (HFD) compared to low-fat diet (LFD)-fed mice, we evaluated whole-body fuel utilization by metabolic cages before and 3 days after omitting a voluntary running wheel in the cage. Short-term (24-48 h) HFD was sufficient to increase energy intake, fat oxidation, and decrease carbohydrate oxidation. Running wheel omission did not change energy intake, but resulted in a significant 50% decrease in total activity and a ~20% in energy expenditure in the active phase (night-time), compared to the period with wheel, irrespective of the dietary composition, resulting in significant weight gain. Yet, while in LFD wheel omission significantly decreased active phase fat oxidation, thereby trending to increase respiratory exchange ratio (RER), in HFD it diminished active phase carbohydrate oxidation. In conclusion, acute decrease in voluntary activity resulted in positive energy balance in mice on both diets, and decreased oxidation of the minor energy (macronutrient) fuel source, demonstrating that dietary macronutrient composition determines fuel utilization choices under conditions of acute changes in energetic demand.


Subject(s)
Diet, Fat-Restricted , Diet, High-Fat , Dietary Fats/administration & dosage , Energy Metabolism , Adaptation, Physiological , Animal Feed , Animal Nutritional Physiological Phenomena , Animals , Dietary Fats/metabolism , Energy Intake , Male , Mice, Inbred C57BL , Nutritional Status , Nutritive Value , Running , Time Factors
9.
Obesity (Silver Spring) ; 29(11): 1857-1867, 2021 11.
Article in English | MEDLINE | ID: mdl-34472713

ABSTRACT

OBJECTIVE: Orexin/hypocretin (Ox) and its receptors (OxR), a neuroendocrine system centrally regulating sleep/wakefulness, were implicated in the regulation of peripheral metabolism. It was hypothesized that human adipose tissue constitutes a direct target of the OxA/OxR system that associates with distinct metabolic profile(s). METHODS: Serum Ox levels and abdominal subcutaneous and visceral adipose tissue expression of Ox/HCRT, OxR1/HCRTR1, and OxR2/HCRTR2 were measured in n = 81 patients. RESULTS: Higher morning circulating Ox levels were associated with improved lipid profile and insulin sensitivity, independently of BMI (ß = -0.363, p = 0.018 for BMI-adjusted homeostatic model of insulin resistance). Adipose HCRT mRNA was detectable in <20% of patients. Visceral HCRT expressers were mostly (80%) males and, compared with nonexpressers, had lower total and LDL cholesterol. HCRTR1 was readily detectable, and HCRTR2 was undetectable. HCRTR1 mRNA and OxR1 protein expression were higher in subcutaneous than visceral adipose tissue, and among nonobese patients, patients with obesity, and patients with obesity and T2DM were 3.4 (1.0), 0.7 (0.1), 0.6 (0.1) (AU) (p < 0.001) and 1.0 (0.2), 0.5 (0.1), 0.4 (0.1) (AU) (p = NS), respectively. Higher visceral HCRTR1 expression was associated with lower fasting insulin and homeostatic model of insulin resistance, also after adjusting for BMI. In human adipocytes, HCRTR1 expression did not exhibit significant oscillation. CONCLUSIONS: Human adipose tissue is a putative direct target of the OxA-OxR1 system, with higher morning input being associated with improved metabolic profile.


Subject(s)
Adipose Tissue , Insulin Resistance , Orexin Receptors , Orexins/genetics , Cross-Sectional Studies , Female , Humans , Intra-Abdominal Fat , Male , Orexin Receptors/genetics
10.
Int J Gynaecol Obstet ; 154(2): 297-303, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33872397

ABSTRACT

OBJECTIVE: To evaluate the effect of the coronavirus disease 2019 (COVID-19) mask-wearing on hematological laboratory components and obstetrical outcomes among women delivering during the COVID-19 pandemic. METHODS: Laboratory results and obstetrical outcomes of women with singleton gestations, admitted for delivery during the COVID-19 mask-wearing period (April-June 2020) were compared with those of women delivering during the parallel period in 2019 and with a larger cohort derived from nine pre-pandemic years (March 2011-April 2020). RESULTS: Overall, 1838 women delivered during the COVID-19 pandemic. Compared with the pre-pandemic period, mean hemoglobin and fibrinogen levels were significantly higher during the mask-wearing period (12.15 ± 1.1 vs 11.96 ± 1.2, P < 0.001 and 472 ± 103.6 vs 448 ± 85.1 mg/dl, P < 0.001, respectively). Platelet levels were lower (200 ± 56.0 vs 206 ± 57.5 K/µl, P < 0.001). The rate of delivery at <34 weeks of gestation was lower during the mask-wearing period (1.1% vs 2%, odds ratio [OR] 0.57, 95% confidence intervals [CI] 0.37-0.88, P = 0.01), whereas cesarean delivery and postpartum hemorrhage rates were higher (26.7% vs 24.4%, OR 1.13, 95% CI 1.02-1.25, P = 0.022 and 4.1% vs 2.8%, OR 1.5, 95% CI 1.2-1.8, P = 0.001, respectively). CONCLUSION: A hard-to-ventilate space created by wearing a mask during the COVID-19 era may be the underlying cause of the observed higher hemoglobin level among pregnant women, possibly affecting obstetrical outcomes.


Subject(s)
COVID-19/prevention & control , Cesarean Section/statistics & numerical data , Masks , Pregnant Women/psychology , Term Birth , Adult , Blood Platelets , COVID-19/epidemiology , Cohort Studies , Female , Fibrinogen/analysis , Hemoglobins/analysis , Hospitalization , Humans , Pandemics , Pregnancy , SARS-CoV-2
12.
Cell Rep ; 33(3): 108295, 2020 10 20.
Article in English | MEDLINE | ID: mdl-33086065

ABSTRACT

TMEM18 is the strongest candidate for childhood obesity identified from GWASs, yet as for most GWAS-derived obesity-susceptibility genes, the functional mechanism remains elusive. We here investigate the relevance of TMEM18 for adipose tissue development and obesity. We demonstrate that adipocyte TMEM18 expression is downregulated in children with obesity. Functionally, downregulation of TMEM18 impairs adipocyte formation in zebrafish and in human preadipocytes, indicating that TMEM18 is important for adipocyte differentiation in vivo and in vitro. On the molecular level, TMEM18 activates PPARG, particularly upregulating PPARG1 promoter activity, and this activation is repressed by inflammatory stimuli. The relationship between TMEM18 and PPARG1 is also evident in adipocytes of children and is clinically associated with obesity and adipocyte hypertrophy, inflammation, and insulin resistance. Our findings indicate a role of TMEM18 as an upstream regulator of PPARG signaling driving healthy adipogenesis, which is dysregulated with adipose tissue dysfunction and obesity.


Subject(s)
Membrane Proteins/genetics , Obesity/genetics , 3T3-L1 Cells , Adipocytes/metabolism , Adipogenesis/genetics , Adipose Tissue/metabolism , Animals , Cell Differentiation/genetics , Cell Line , Female , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Humans , Insulin Resistance/genetics , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Obesity/metabolism , PPAR gamma/metabolism , Signal Transduction , Zebrafish
13.
Diabetes ; 69(11): 2310-2323, 2020 11.
Article in English | MEDLINE | ID: mdl-32732304

ABSTRACT

Elevated expression of E2F1 in adipocyte fraction of human visceral adipose tissue (hVAT) associates with a poor cardiometabolic profile. We hypothesized that beyond directly activating autophagy and MAP3K5 (ASK)-MAP kinase signaling, E2F1 governs a distinct transcriptome that contributes to adipose tissue and metabolic dysfunction in obesity. We performed RNA sequencing of hVAT samples from age-, sex-, and BMI-matched patients, all obese, whose visceral E2F1 protein expression was either high (E2F1high) or low (E2F1low). Tumor necrosis factor superfamily (TNFSF) members, including TRAIL (TNFSF10), TL1A (TNFSF15), and their receptors, were enriched in E2F1high While TRAIL was equally expressed in adipocytes and stromal vascular fraction (SVF), TL1A was mainly expressed in SVF, and TRAIL-induced TL1A was attributed to CD4+ and CD8+ subclasses of hVAT T cells. In human adipocytes, TL1A enhanced basal and impaired insulin-inhibitable lipolysis and altered adipokine secretion, and in human macrophages it induced foam cell biogenesis and M1 polarization. Two independent human cohorts confirmed associations between TL1A and TRAIL expression in hVAT and higher leptin and IL6 serum concentrations, diabetes status, and hVAT-macrophage lipid content. Jointly, we propose an intra-adipose tissue E2F1-associated TNFSF paracrine loop engaging lymphocytes, macrophages, and adipocytes, ultimately contributing to adipose tissue dysfunction in obesity.


Subject(s)
Adipocytes/physiology , E2F1 Transcription Factor/metabolism , Lymphocytes/physiology , Macrophages/physiology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism , Adipose Tissue/physiology , Adolescent , Adult , Aged , Aged, 80 and over , Cells, Cultured , Coculture Techniques , E2F1 Transcription Factor/genetics , Female , Gene Expression Regulation , Humans , Male , Middle Aged , Paracrine Communication , TNF-Related Apoptosis-Inducing Ligand/genetics , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics , Young Adult
14.
Adipocyte ; 8(1): 51-60, 2019 12.
Article in English | MEDLINE | ID: mdl-30676227

ABSTRACT

Obesity, a condition most commonly associated with hyper-leptinemia, is also characterized by increased expression of autophagy genes and likely autophagic activity in human adipose tissue (AT). Indeed, circulating leptin levels were previously shown to positively associate with the expression levels of autophagy genes such as Autophagy related gene-5 (ATG5). Here we hypothesized that leptin acts in an autocrine-paracrine manner to increase autophagy in two major AT cell populations, adipocytes and macrophages. We followed the dynamics of autophagosomes following acute leptin administration with or without a leptin receptor antagonist (SMLA) using high-throughput live-cell imaging in murine epididymal adipocyte and macrophage (RAW264.7) cell-lines. In macrophages leptin exerted only a mild effect on autophagy dynamics, tending to attenuate autophagosomes growth rate. In contrast, leptin-treated adipocytes exhibited a moderate, ~20% increase in the rate of autophagosome growth, an effect that was blocked by SMLA. This finding corresponded to mild increases in mRNA and protein expression of key autophagy genes. Interestingly, a long-lived proteins degradation assay uncovered a robust, >2-fold leptin-mediated stimulation of the autophagy/lysosome-related (bafilomycin-inhibited) activity, which was entirely blocked by SMLA. Collectively, leptin regulates autophagy in a cell-type specific manner. In adipocytes, autophagosome dynamics is moderately enhanced, but even more pronounced stimulation is seen in autophagy-related long-lived protein degradation. These findings suggest a causal link between obesity-associated hyperleptinemia and elevated adipocyte and AT autophagy-related processes.


Subject(s)
Adipocytes/metabolism , Leptin/metabolism , Proteolysis/drug effects , Adipose Tissue/metabolism , Animals , Autophagosomes/physiology , Autophagy/physiology , Gene Expression/genetics , Leptin/physiology , Lysosomes/metabolism , Macrophages , Mice , Obesity/metabolism , RAW 264.7 Cells , RNA, Messenger/genetics , Receptors, Leptin/metabolism
15.
Physiology (Bethesda) ; 31(4): 270-82, 2016 07.
Article in English | MEDLINE | ID: mdl-27252162

ABSTRACT

There is growing interest in understanding how dysregulated autophagy may contribute to pathogenesis of disease. Most frequently, disease states are associated with diminished autophagy, mostly attributed to genetic variation in autophagy genes and/or to dysfunctional posttranscriptional mechanisms. In human adipose tissue (AT), in obesity, expression of autophagy genes is upregulated and autophagy is likely activated, associating with adipose dysfunction. This review explores the emerging role of transcriptional mechanisms regulating AT autophagy in obesity.


Subject(s)
Adipose Tissue/metabolism , Autophagy , Obesity/genetics , Obesity/metabolism , Transcription, Genetic , Animals , Cell Cycle , Humans , Mice , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...