Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Cell Mol Life Sci ; 80(4): 108, 2023 Mar 29.
Article in English | MEDLINE | ID: mdl-36988756

ABSTRACT

Episodes of chronic stress can result in psychic disorders like post-traumatic stress disorder, but also promote the development of metabolic syndrome and type 2 diabetes. We hypothesize that muscle, as main regulator of whole-body energy expenditure, is a central target of acute and adaptive molecular effects of stress in this context. Here, we investigate the immediate effect of a stress period on energy metabolism in Musculus gastrocnemius in our established C57BL/6 chronic variable stress (Cvs) mouse model. Cvs decreased lean body mass despite increased energy intake, reduced circadian energy expenditure (EE), and substrate utilization. Cvs altered the proteome of metabolic components but not of the oxidative phosphorylation system (OXPHOS), or other mitochondrial structural components. Functionally, Cvs impaired the electron transport chain (ETC) capacity of complex I and complex II, and reduces respiratory capacity of the ETC from complex I to ATP synthase. Complex I-OXPHOS correlated to diurnal EE and complex II-maximal uncoupled respiration correlated to diurnal and reduced nocturnal EE. Bioenergetics assessment revealed higher optimal thermodynamic efficiencies (ƞ-opt) of mitochondria via complex II after Cvs. Interestingly, transcriptome and methylome were unaffected by Cvs, thus excluding major contributions to supposed metabolic adaptation processes. In summary, the preclinical Cvs model shows that metabolic pressure by Cvs is initially compensated by adaptation of mitochondria function associated with high thermodynamic efficiency and decreased EE to manage the energy balance. This counter-regulation of mitochondrial complex II may be the driving force to longitudinal metabolic changes of muscle physiological adaptation as the basis of stress memory.


Subject(s)
Diabetes Mellitus, Type 2 , Mice , Animals , Diabetes Mellitus, Type 2/metabolism , Mice, Inbred C57BL , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Oxidative Phosphorylation , Energy Metabolism , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Mitochondria, Muscle/metabolism
2.
Nutr Metab Cardiovasc Dis ; 33(9): 1785-1796, 2023 09.
Article in English | MEDLINE | ID: mdl-37495452

ABSTRACT

BACKGROUND AND AIMS: Increased hepatocellular lipid content (HCL) is linked to insulin resistance, risk of type 2 diabetes and related complications. Conversely, a single-nucleotide polymorphism (TM6SF2EK; rs58542926) in the transmembrane 6 superfamily member 2-gene has been associated with nonalcoholic fatty liver disease (NAFLD), but lower cardiovascular risk. This case-control study tested the role of this polymorphism for tissue-specific insulin sensitivity during early course of diabetes. METHODS AND RESULTS: Males with recent-onset type 2 diabetes with (TM6SF2EK: n = 16) or without (TM6SF2EE: n = 16) the heterozygous TM6SF2-polymorphism of similar age and body mass index, underwent Botnia-clamps with [6,6-2H2]glucose to measure whole-body-, hepatic- and adipose tissue-insulin sensitivity. HCL was assessed with 1H-magnetic-resonance-spectroscopy. A subset of both groups (n = 24) was re-evaluated after 5 years. Despite doubled HCL, TM6SF2EK had similar hepatic- and adipose tissue-insulin sensitivity and 27% higher whole-body-insulin sensitivity than TM6SF2EE. After 5 years, whole-body-insulin sensitivity, HCL were similar between groups, while adipose tissue-insulin sensitivity decreased by 87% and 55% within both groups and circulating triacylglycerol increased in TM6SF2EE only. CONCLUSIONS: The TM6SF2-polymorphism rs58542926 dissociates HCL from insulin resistance in recent-onset type 2 diabetes, which is attenuated by disease duration. This suggests that diabetes-related metabolic alterations dominate over effects of the TM6SF2-polymorphism during early course of diabetes and NAFLD.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Humans , Male , Case-Control Studies , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/complications , Insulin Resistance/genetics , Liver/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/complications , Polymorphism, Single Nucleotide , Triglycerides/metabolism
3.
Int J Mol Sci ; 23(12)2022 Jun 20.
Article in English | MEDLINE | ID: mdl-35743314

ABSTRACT

Alterations in mitochondrial function are an important control variable in the progression of metabolic dysfunction-associated fatty liver disease (MAFLD), while also noted by increased de novo lipogenesis (DNL) and hepatic insulin resistance. We hypothesized that the organization and function of a mitochondrial electron transport chain (ETC) in this pathologic condition is a consequence of shifted substrate availability. We addressed this question using a transgenic mouse model with increased hepatic insulin resistance and DNL due to constitutively active human SREBP-1c. The abundance of ETC complex subunits and components of key metabolic pathways are regulated in the liver of these animals. Further omics approaches combined with functional assays in isolated liver mitochondria and primary hepatocytes revealed that the SREBP-1c-forced fatty liver induced a substrate limitation for oxidative phosphorylation, inducing enhanced complex II activity. The observed increased expression of mitochondrial genes may have indicated a counteraction. In conclusion, a shift of available substrates directed toward activated DNL results in increased electron flows, mainly through complex II, to compensate for the increased energy demand of the cell. The reorganization of key compounds in energy metabolism observed in the SREBP-1c animal model might explain the initial increase in mitochondrial function observed in the early stages of human MAFLD.


Subject(s)
Fatty Liver , Insulin Resistance , Animals , Fatty Liver/metabolism , Lipogenesis/genetics , Liver/metabolism , Mice , Oxidative Phosphorylation , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism
4.
Diabetologia ; 64(2): 458-468, 2021 02.
Article in English | MEDLINE | ID: mdl-33084971

ABSTRACT

AIMS/HYPOTHESIS: Emerging evidence suggests that in addition to hyperglycaemia, dyslipidaemia could represent a contributing pathogenetic factor to diabetic neuropathy, while obesity and insulin resistance play a role in the development of diabetic cardiac autonomic neuropathy (CAN) characterised by reduced heart rate variability (HRV), particularly in type 2 diabetes. We hypothesised that distinct lipid metabolites are associated with diminished HRV in recent-onset type 2 diabetes rather than type 1 diabetes. METHODS: We analysed 127 plasma lipid metabolites (11 acylcarnitines, 39 NEFA, 12 sphingomyelins (SMs), 56 phosphatidylcholines and nine lysophosphatidylcholines) using MS in participants from the German Diabetes Study baseline cohort recently diagnosed with type 1 (n = 100) and type 2 diabetes (n = 206). Four time-domain HRV indices (number of normal-to-normal (NN) intervals >50 ms divided by the number of all NN intervals [pNN50]; root mean square of successive differences [RMSSD]; SD of NN intervals [SDNN]; and SD of differences between adjacent NN intervals) and three frequency-domain HRV indices (very-low-frequency [VLF], low-frequency [LF] and high-frequency [HF] power spectrum) were computed from NN intervals recorded during a 3 h hyperinsulinaemic-euglycaemic clamp at baseline and in subsets of participants with type 1 (n = 60) and type 2 diabetes (n = 95) after 5 years. RESULTS: In participants with type 2 diabetes, after Bonferroni correction and rigorous adjustment, SDNN was inversely associated with higher levels of diacyl-phosphatidylcholine (PCaa) C32:0, PCaa C34:1, acyl-alkyl-phosphatidylcholine (PCae) C36:0, SM C16:0 and SM C16:1. SD of differences between NN intervals was inversely associated with PCaa C32:0, PCaa C34:1, PCaa C34:2, PCae C36:0 and SM C16:1, and RMSSD with PCae C36:0. For VLF power, inverse associations were found with PCaa C30:0, PCaa C32:0, PCaa C32:1, PCaa C34:2 and SM C16:1, and for LF power inverse associations were found with PCaa C32:0 and SM C16:1 (r = -0.242 to r = -0.349; p ≤ 0.0005 for all correlations). In contrast, no associations of lipid metabolites with measures of cardiac autonomic function were noted in participants recently diagnosed with type 1 diabetes. After 5 years, HRV declined due to ageing rather than diabetes, whereby prediction analyses for lipid metabolites were hampered. CONCLUSIONS/INTERPRETATION: Higher plasma levels of specific lipid metabolites are closely linked to cardiac autonomic dysfunction in recent-onset type 2 diabetes but not type 1 diabetes, suggesting a role for perturbed lipid metabolism in the early development of CAN in type 2 diabetes. Graphical abstract.


Subject(s)
Autonomic Nervous System Diseases/blood , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 2/blood , Diabetic Neuropathies/blood , Lipidomics , Adult , Autonomic Nervous System Diseases/etiology , Autonomic Nervous System Diseases/physiopathology , Carnitine/analogs & derivatives , Carnitine/blood , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 2/complications , Diabetic Neuropathies/etiology , Diabetic Neuropathies/physiopathology , Dyslipidemias/blood , Fatty Acids, Nonesterified/blood , Female , Glucose Clamp Technique , Heart Rate , Humans , Insulin Resistance , Lipid Metabolism , Lysophosphatidylcholines/blood , Male , Middle Aged , Obesity/blood , Phosphatidylcholines/blood , Sphingomyelins/blood , Young Adult
5.
Int J Mol Sci ; 21(22)2020 Nov 20.
Article in English | MEDLINE | ID: mdl-33233602

ABSTRACT

Lipodystrophies are a heterogeneous group of physiological changes characterized by a selective loss of fatty tissue. Here, no fat cells are present, either through lack of differentiation, loss of function or premature apoptosis. As a consequence, lipids can only be stored ectopically in non-adipocytes with the major health consequences as fatty liver and insulin resistance. This is a crucial difference to being slim where the fat cells are present and store lipids if needed. A simple clinical classification of lipodystrophies is based on congenital vs. acquired and generalized vs. partial disturbance of fat distribution. Complications in patients with lipodystrophy depend on the clinical manifestations. For example, in diabetes mellitus microangiopathic complications such as nephropathy, retinopathy and neuropathy may develop. In addition, due to ectopic lipid accumulation in the liver, fatty liver hepatitis may also develop, possibly with cirrhosis. The consequences of extreme hypertriglyceridemia are typically acute pancreatitis or eruptive xanthomas. The combination of severe hyperglycemia with dyslipidemia and signs of insulin resistance can lead to premature atherosclerosis with its associated complications of coronary heart disease, peripheral vascular disease and cerebrovascular changes. Overall, lipodystrophy is rare with an estimated incidence for congenital (<1/1.000.000) and acquired (1-9/100.000) forms. Due to the rarity of the syndrome and the phenotypic range of metabolic complications, only studies with limited patient numbers can be considered. Experimental animal models are therefore useful to understand the molecular mechanisms in lipodystrophy and to identify possible therapeutic approaches.


Subject(s)
Atherosclerosis/genetics , Coronary Disease/genetics , Diabetes Mellitus/genetics , Fatty Liver/genetics , Hypertriglyceridemia/genetics , Lipodystrophy/genetics , Acyltransferases/deficiency , Acyltransferases/genetics , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Atherosclerosis/etiology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Body Fat Distribution , Coronary Disease/etiology , Coronary Disease/metabolism , Coronary Disease/pathology , Diabetes Mellitus/etiology , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Disease Models, Animal , Fatty Liver/complications , Fatty Liver/metabolism , Fatty Liver/pathology , Humans , Hypertriglyceridemia/complications , Hypertriglyceridemia/metabolism , Hypertriglyceridemia/pathology , Insulin Resistance , Lamin Type A/deficiency , Lamin Type A/genetics , Lipid Metabolism/genetics , Lipodystrophy/complications , Lipodystrophy/metabolism , Lipodystrophy/pathology , Pancreatitis/etiology , Pancreatitis/genetics , Pancreatitis/metabolism , Pancreatitis/pathology , Xanthomatosis/etiology , Xanthomatosis/genetics , Xanthomatosis/metabolism , Xanthomatosis/pathology
6.
Int J Mol Sci ; 21(11)2020 Jun 10.
Article in English | MEDLINE | ID: mdl-32532003

ABSTRACT

Fatty liver occurs from simple steatosis with accumulated hepatic lipids and hepatic insulin resistance to severe steatohepatitis, with aggravated lipid accumulation and systemic insulin resistance, but this progression is still poorly understood. Analyses of hepatic gene expression patterns from alb-SREBP-1c mice with moderate, or aP2-SREBP-1c mice with aggravated, hepatic lipid accumulation revealed IGFBP2 as key nodal molecule differing between moderate and aggravated fatty liver. Reduced IGFBP2 expression in aggravated fatty liver was paralleled with promoter hypermethylation, reduced hepatic IGFBP2 secretion and IGFBP2 circulating in plasma. Physiologically, the decrease of IGFBP2 was accompanied with reduced fatty acid oxidation and increased de novo lipogenesis potentially mediated by IGF1 in primary hepatocytes. Furthermore, methyltransferase and sirtuin activities were enhanced. In humans, IGFBP2 serum concentration was lower in obese men with non-alcoholic fatty liver disease (NAFLD) and steatohepatitis (NASH) compared to non-obese controls, and liver fat reduction by weight-loss intervention correlated with an increase of IGFBP2 serum levels. In conclusion, hepatic IGFBP2 abundance correlates to its circulating level and is related to hepatic energy metabolism and de novo lipogenesis. This designates IGFBP2 as non-invasive biomarker for fatty liver disease progression and might further provide an additional variable for risk prediction for pathogenesis of fatty liver in diabetes subtype clusters.


Subject(s)
Energy Metabolism/physiology , Insulin-Like Growth Factor Binding Protein 2/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Adult , Animals , Body Weight , Case-Control Studies , Energy Metabolism/genetics , Hepatocytes/metabolism , Humans , Insulin Resistance , Insulin-Like Growth Factor Binding Protein 2/blood , Insulin-Like Growth Factor Binding Protein 2/metabolism , Insulin-Like Growth Factor Binding Protein 3/blood , Insulin-Like Growth Factor I/analysis , Male , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Non-alcoholic Fatty Liver Disease/genetics , Obesity/complications , Obesity/metabolism , Obesity/surgery , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism
7.
Diabetologia ; 62(2): 286-291, 2019 02.
Article in English | MEDLINE | ID: mdl-30413829

ABSTRACT

AIMS/HYPOTHESIS: The aim of this study was to investigate the modifying effect of the glucose transporter (GLUT2) gene SLC2A2 (rs8192675) variant on the glycaemic response to metformin in individuals recently diagnosed with type 2 diabetes. METHODS: Individuals with type 2 diabetes (n = 508) from the prospective German Diabetes Study (age [mean ± SD] 53 ± 10 years; 65% male; BMI 32 ± 6 kg/m2, metformin use 57%) underwent detailed metabolic characterisation (hyperinsulinaemic-euglycaemic clamp, IVGTT) during the first year after diagnosis. Participants provided self-reported data from the time of diagnosis. The change in fasting glucose was assessed in relation to SLC2A2 genotype and glucose-lowering treatment using two-way ANCOVA with gene×treatment interactions adjusted for age, sex, BMI and diabetes duration. RESULTS: The C variant allele of rs8192675 was associated with a higher prevalence of diabetes symptoms at diabetes diagnosis. In the metformin monotherapy group only, patients with a C allele showed a larger adjusted blood glucose reduction during the first year after diabetes diagnosis than patients with the TT genotype (6.3 mmol/l vs 3.9 mmol/l; genotype difference 2.4 mmol/l, p = 0.02; p value for genotype interaction [metformin monotherapy vs non-pharmacological therapy] <0.01). The greater decline in fasting glucose (CC/CT vs TT) in metformin monotherapy persisted after further adjusting for glucose values at diagnosis (genotype difference 1.0 mmol/l, p = 0.01; genotype×treatment interaction p = 0.06). CONCLUSIONS/INTERPRETATION: The variant rs8192675 in the SLC2A2 gene (C allele) is associated with an improved glucose response to metformin monotherapy during the first year after diagnosis in type 2 diabetes. TRIAL REGISTRATION: ClinicalTrials.gov NCT01055093.


Subject(s)
Blood Glucose/genetics , Diabetes Mellitus, Type 2/genetics , Glucose Transporter Type 2/genetics , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Adult , Alleles , Diabetes Mellitus, Type 2/drug therapy , Female , Genotype , Glucose Clamp Technique , Humans , Male , Middle Aged , Pharmacogenetics , Polymorphism, Single Nucleotide
8.
Int J Mol Sci ; 20(10)2019 May 24.
Article in English | MEDLINE | ID: mdl-31137678

ABSTRACT

Adipocyte and hepatic lipid metabolism govern whole-body metabolic homeostasis, whereas a disbalance of de novo lipogenesis (DNL) in fat and liver might lead to obesity, with severe co-morbidities. Nevertheless, some obese people are metabolically healthy, but the "protective" mechanisms are not yet known in detail. Especially, the adipocyte-derived molecular mediators that indicate adipose functionality are poorly understood. We studied transgenic mice (alb-SREBP-1c) with a "healthy" obese phenotype, and obob mice with hyperphagia-induced "sick" obesity to analyze the impact of the tissue-specific DNL on the secreted proteins, i.e., the adipokinome, of the primary adipose cells by label-free proteomics. Compared to the control mice, adipose DNL is reduced in both obese mouse models. In contrast, the hepatic DNL is reduced in obob but elevated in alb-SREBP-1c mice. To investigate the relationship between lipid metabolism and adipokinomes, we formulated the "liver-to-adipose-tissue DNL" ratio. Knowledge-based analyses of these results revealed adipocyte functionality with proteins, which was involved in tissue remodeling or metabolism in the alb-SREBP-1c mice and in the control mice, but mainly in fibrosis in the obob mice. The adipokinome in "healthy" obesity is similar to that in a normal condition, but it differs from that in "sick" obesity, whereas the serum lipid patterns reflect the "liver-to-adipose-tissue DNL" ratio and are associated with the adipokinome signature.


Subject(s)
Adipokines/metabolism , Adipose Tissue/metabolism , Fatty Acids, Nonesterified/blood , Obesity/metabolism , Adipokines/genetics , Animals , Lipogenesis , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/blood , Obesity/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sterol Regulatory Element Binding Protein 1/genetics
9.
Int J Obes (Lond) ; 42(5): 1039-1050, 2018 06.
Article in English | MEDLINE | ID: mdl-29467502

ABSTRACT

BACKGROUND: CDH13, an atypical member of the cadherin superfamily, has been identified in adipocyte secretomes of lean mouse models. CDH13 abundance differs in mouse models according to their susceptibility to develop metabolic disorders, but the role of CDH13 in adipose tissue is unknown. METHODS: Secreted CDH13 protein levels and mRNA levels in visceral adipose tissue were determined in lean and obese mouse models. In vitro studies were performed in 3T3-L1 adipocytes to determine the role of CDH13 in adipocyte differentiation. The pathophysiological impact of visceral adipose tissue CDH13 mRNA and circulating CDH13 levels were determined in humans (normal-weight men n = 37, obese men n = 109 including n = 51 type 2 diabetes patients) and in obese patients (n = 14) pre- and post-metabolic surgery. RESULTS: This study shows that in visceral adipose tissue CDH13 protein secretion and mRNA levels were decreased in obese mouse models. Mechanistically, CDH13 affects lipid metabolism during adipogenesis but not in mature adipocytes. CDH13 knockdown during adipogenesis reduced fatty acid uptake and lipid content in developing adipocytes. Furthermore, CDH13 depletion during adipogenesis lowered the induction of PPARγ and C/EBPα expression. These observations are of pathophysiological impact since visceral adipose tissue CDH13 mRNA and circulating CDH13 levels were decreased in obese men compared to normal-weight controls. Weight loss induced by bariatric surgery restored circulating CDH13 to levels found in normal-weight controls. CONCLUSIONS: CDH13 levels in adipose tissue and the circulation are affected by obesity in mouse models and humans and are restored by weight loss in humans. CDH13 interferes with the differentiation potential of adipocytes and therefore is a marker for plasticity of fat tissue that might reflect the health status of adipose tissue.


Subject(s)
Adipocytes/cytology , Adipose Tissue/chemistry , Cadherins/metabolism , Cell Differentiation/physiology , Obesity/metabolism , Adipocytes/metabolism , Adipogenesis/drug effects , Adipogenesis/physiology , Adipose Tissue/metabolism , Adult , Animals , Biomarkers/analysis , Biomarkers/metabolism , Cadherins/analysis , Cadherins/genetics , Cadherins/pharmacology , Cell Differentiation/drug effects , Female , Humans , Male , Mice , Mice, Obese , Middle Aged , Obesity/blood , Obesity/physiopathology , RNA, Messenger/analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism
10.
Int J Mol Sci ; 19(4)2018 Mar 25.
Article in English | MEDLINE | ID: mdl-29587401

ABSTRACT

The key lipid metabolism transcription factor sterol regulatory element-binding protein (SREBP)-1a integrates gene regulatory effects of hormones, cytokines, nutrition and metabolites as lipids, glucose, or cholesterol via phosphorylation by different mitogen activated protein kinase (MAPK) cascades. We have previously reported the impact of SREBP-1a phosphorylation on the phenotype in transgenic mouse models with liver-specific overexpression of the N-terminal transcriptional active domain of SREBP-1a (alb-SREBP-1a) or a MAPK phosphorylation site-deficient variant (alb-SREBP-1a∆P; (S63A, S117A, T426V)), respectively. In this report, we investigated the molecular basis of the systemic observations by holistic analyses of gene expression in liver and of proteome patterns in lipid-degrading organelles involved in the pathogenesis of metabolic syndrome, i.e., peroxisomes, using 2D-DIGE and mass spectrometry. The differences in hepatic gene expression and peroxisomal protein patterns were surprisingly small between the control and alb-SREBP-1a mice, although the latter develop a severe phenotype with visceral obesity and fatty liver. In contrast, phosphorylation site-deficient alb-SREBP-1a∆P mice, which are protected from fatty liver disease, showed marked differences in hepatic gene expression and peroxisomal proteome patterns. Further knowledge-based analyses revealed that disruption of SREBP-1a phosphorylation resulted in massive alteration of cellular processes, including signs for loss of targeting lipid pathways.


Subject(s)
Disease Models, Animal , Fatty Liver/metabolism , Mice , Proteome/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Animals , Fatty Liver/genetics , Gene Deletion , Humans , Male , Metabolic Syndrome/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Obesity, Abdominal/metabolism , Peroxisomes/pathology , Phosphorylation , Sterol Regulatory Element Binding Protein 1/genetics , Transcriptome
11.
Mol Biol Rep ; 44(1): 51-61, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27878529

ABSTRACT

Polygenic diseases with a broad phenotypic spectrum, such as polycystic ovary syndrome (PCOS), present a particular challenge in terms of identifying the underlying genetic mechanisms, nevertheless genetic variants have impact on the individual phenotype. We aimed to determine if next to genetic variations like SNPs further mechanisms might play a role in the pathogenesis of PCOS. We examined the effect of copy-number variations (CNVs) on metabolic phenotypes in PCOS. The intragenic rs1244979, rs2815752 in NEGR1 gene, and rs780094 in GCKR gene were genotyped and CNVs were determined by droplet digital polymerase chain reaction (ddPCR) in PCOS patients (n = 153) and controls without metabolic syndrome (n = 142). The study indicated that SNPs are not associated with the pathogenesis of PCOS but affect metabolic phenotypes. The CNVs investigated show a lower variability in PCOS than in CON. Furthermore, we provided direct evidence that the copy number, but not the genotype of the CNV in the genomic regions of rs780094(GCKR) is associated with low level of high-density lipoprotein cholesterol in PCOS. This study supports the hypothesis that not only genetic variants, but also CNVs in metabolically relevant genes, have an effect on metabolic phenotypes in our group of PCOS patients.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cell Adhesion Molecules, Neuronal/genetics , Cholesterol, HDL/metabolism , DNA Copy Number Variations , Metabolic Syndrome/genetics , Polycystic Ovary Syndrome/genetics , Adult , Bone Density , Case-Control Studies , Female , GPI-Linked Proteins/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Middle Aged , Polycystic Ovary Syndrome/metabolism , Polymorphism, Single Nucleotide , Prospective Studies , Young Adult
12.
Int J Mol Sci ; 18(9)2017 Sep 08.
Article in English | MEDLINE | ID: mdl-28885548

ABSTRACT

The group of adipokines comprises hundreds of biological active proteins and peptides released from adipose tissue. Alterations of those complex protein signatures are suggested to play a crucial role in the pathophysiology of multifactorial, metabolic diseases. We hypothesized that also the pathophysiology of type-2-diabetes is linked to the dysregulation of the adipocyte secretome. To test this, we investigated mouse models with monogenic defects in leptin signaling which are susceptible to adipositas (C57BL/6 Cg-Lepob (obob)) or adipositas with diabetes (C57BL/KS Cg-Leprdb (dbdb)) according to their genetic background. At the age of 17 weeks, visceral fat was obtained and primary murine adipocytes were isolated to harvest secretomes. Quantitative proteome analyses (LC-ESI-MS/MS) identified more than 800 potential secreted proteins. The secretome patterns revealed significant differences connected to the pathophysiology of obese mice. Pathway analyses indicated that these differences focus on exosome modelling, but failed to provide more precise specifications. To investigate the relationship of secretome data to insulin sensitivity, we examined the content of diabetogenic lipids, i.e., diacylglycerols (DAGs), identified as key players in lipid-induced insulin resistance. In contrast to obob mice, fat tissue of dbdb mice showed elevated DAG content, especially of DAG species with saturated fatty acid C16:0 and C18:0, while unsaturated fatty acid C16:1 were only changed in obob. Furthermore, DAG signatures of the models specifically correlate to secreted regulated adipokines indicating specific pathways. In conclusion, our data further support the concept that the fat tissue is an endocrine organ that releases bioactive factors corresponding to adipose tissue health status.


Subject(s)
Adipokines/metabolism , Diabetes Mellitus, Type 2/metabolism , Obesity/metabolism , Proteomics , Adipocytes/metabolism , Adipose Tissue/metabolism , Animals , Biomarkers , Cluster Analysis , Disease Models, Animal , Lipid Metabolism , Male , Mass Spectrometry , Mice , Mice, Obese , Proteome , Proteomics/methods
13.
Diabetologia ; 59(10): 2203-7, 2016 10.
Article in English | MEDLINE | ID: mdl-27376543

ABSTRACT

AIMS/HYPOTHESIS: Both inherited and acquired insulin resistance have been associated with abnormal muscle mitochondrial function. At whole-body level, maximal oxygen uptake ([Formula: see text]) and/or metabolic flexibility (as given by ΔRQ) reflect certain features of mitochondrial function. This study tests the hypotheses (1) that [Formula: see text] and ΔRQ correlate tightly with each other and with insulin sensitivity and (2) that glycaemia, lipidaemia or subclinical inflammation would explain such relationships. METHODS: Near-normoglycaemic individuals with type 2 diabetes mellitus (n = 136) with a short known disease duration (<12 months) underwent cycling spiroergometry, indirect calorimetry and hyperinsulinaemic-euglycaemic clamp tests. RESULTS: Both [Formula: see text] (r = 0.39, p < 0.0001) and ΔRQ (r = 0.32, p < 0.0001) correlated positively with whole-body insulin sensitivity, even after adjusting for anthropometric variables, glycaemia and glucose-lowering medication, but not after adjusting for NEFA. [Formula: see text] further correlated negatively with circulating high-sensitivity C-reactive protein concentration. However, [Formula: see text] did not relate to ΔRQ, even after adjusting for whole-body insulin sensitivity. CONCLUSIONS/INTERPRETATION: Oxidative capacity and metabolic flexibility are independent determinants of insulin sensitivity but are influenced by circulating NEFA in recent-onset type 2 diabetes. ClinicalTrial.gov registration no: NCT01055093.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Adolescent , Adult , Aged , C-Reactive Protein/metabolism , Calorimetry, Indirect , Diabetes Mellitus, Type 2/metabolism , Female , Glucose Clamp Technique , Humans , Insulin/metabolism , Insulin Resistance/physiology , Male , Middle Aged , Muscle, Skeletal/metabolism , Obesity/blood , Obesity/metabolism , Overweight/blood , Overweight/metabolism , Oxidation-Reduction , Oxygen Consumption/physiology , Young Adult
14.
Biochim Biophys Acta ; 1851(7): 965-76, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25790917

ABSTRACT

UNLABELLED: Major causes of lipid accumulation in liver are increased import or synthesis or decreased catabolism of fatty acids. The latter is caused by dysfunction of cellular organelles controlling energy homeostasis, i.e., mitochondria. Peroxisomes also appear to be an important organelle in lipid metabolism of hepatocytes, but little is known about their role in the development of non-alcoholic fatty liver disease (NAFLD). To investigate the role of peroxisomes alongside mitochondria in excessive hepatic lipid accumulation, we used leptin-resistant db/db mice on C57BLKS background, a mouse model that develops hyperphagia-induced diabetes with obesity and NAFLD. Proteome and gene expression analyses along with lipid analyses in the liver revealed differential expression of genes related to lipid metabolism and ß-oxidation, whereas genes for peroxisomal proteins were predominantly regulated. CONCLUSION: Our investigations show that in fatty liver disease in combination with obesity and diabetes, the hepatocyte-protecting organelle peroxisome is altered. Hence, peroxisomes might indicate a stage of pre-NAFLD, play a role in the early development of NAFLD and appear to be a potential target for treatment and prevention of NAFLD.


Subject(s)
Lipid Metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Peroxisomes/physiology , Animals , Gene Expression , Lipid Metabolism/genetics , Liver/pathology , Male , Metabolic Networks and Pathways/genetics , Mice , Mice, Inbred C57BL , Mice, Obese , Mice, Transgenic , Mitochondrial Proteins/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Peroxisomes/metabolism
15.
BMC Med Genet ; 17(1): 36, 2016 May 04.
Article in English | MEDLINE | ID: mdl-27142837

ABSTRACT

BACKGROUND: Maturity onset diabetes of the young (MODY) is an autosomal dominant form of non-insulin-dependent diabetes mellitus caused by mutations in at least 13 different genes. The hepatocyte nuclear factor (HNF)-1α gene is affected in the most common form (HNF1A-MODY [MODY3]). CASE PRESENTATION: We describe the co-inheritance of a novel heterozygous missense mutation c.1761C > G (p.Pro588Ala) with a novel complex deletion insertion mutation (c.1765_1766delinsGCCCGfs86*) in the HNF-1α gene among affected members of one family. Both mutations were present in the affected patients and neither was present in unaffected family members. The family had not only inheritance of MODY but also increased susceptibility to type 2 diabetes. Therefore one family member had classical type 2 diabetes including metabolic syndrome aggravated by a genetic predisposition in the form of HNF1A-MODY. CONCLUSION: The presence of common type 2 diabetes features should not detract from the possibility of MODY in patients with a striking autosomal-dominant family history.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Hepatocyte Nuclear Factor 1-alpha/genetics , INDEL Mutation , Mutation, Missense , Adult , Female , Genetic Predisposition to Disease , Heterozygote , Humans , Male , Pedigree , Phenotype , Sequence Analysis, DNA/methods
16.
Cardiovasc Diabetol ; 15(1): 138, 2016 Sep 27.
Article in English | MEDLINE | ID: mdl-27677442

ABSTRACT

AIMS: In patients with type 2 diabetes (T2D), responsiveness of serum lipid concentrations to dietary patterns may vary by genotype. The aims of the present study were to identify explorative dietary patterns and to examine their independent associations with serum lipid levels and interactions with apolipoprotein (Apo)A5 and ApoE variants among patients recently diagnosed with T2D. METHODS: Within a cross-sectional analysis, participants of the German Diabetes Study (n = 348) with mean T2D duration of 6 months were investigated for fasting serum lipid levels, ApoA5 and ApoE genotypes; food consumption frequencies were assessed by a food propensity questionnaire. Dietary patterns were derived using principal component analysis (PCA) and reduced rank regression (RRR), which extracts patterns explaining variation in serum lipid concentrations. RESULTS: PCA yielded interpretable dietary patterns which were, however, not related to serum lipid levels. Relevance of the RRR patterns varied by genotype: a preferred consumption of fruit gum, fruit juice, and potato dumpling, whilst avoiding fruits and vegetables independently associated with higher triglyceride levels among ApoA5*2. Patients in the highest compared to the lowest tertile of pattern adherence had 99 % higher triglycerides. Lower consumption frequencies of butter, cream cake, French fries, or high-percentage alcoholic beverages were independently related to lower LDL-cholesterol among ApoE2 carriers, with those in the highest compared to the lowest tertile of pattern adherence having 40 % lower LDL-cholesterol (both Pinteraction < 0.05). CONCLUSIONS: Our explorative data analyses suggest that associations of dietary patterns with triglycerides and LDL-cholesterol differ by ApoA5 and ApoE haplotype in recently diagnosed T2D. Trial registration Clinicaltrials.gov: NCT01055093. Date of registration: January 22, 2010 (retrospectively registered). Date of enrolment of first participant to the trial: September 2005.

17.
Biochim Biophys Acta ; 1844(5): 1011-7, 2014 May.
Article in English | MEDLINE | ID: mdl-23994228

ABSTRACT

The skeletal muscle is a metabolically active tissue that secretes various proteins. These so-called myokines have been proposed to affect muscle physiology and to exert systemic effects on other tissues and organs. Yet, changes in the secretory profile may participate in the pathophysiology of metabolic diseases. The present study aimed at characterizing the secretome of differentiated primary human skeletal muscle cells (hSkMC) derived from healthy, adult donors combining three different mass spectrometry based non-targeted approaches as well as one antibody based method. This led to the identification of 548 non-redundant proteins in conditioned media from hSkmc. For 501 proteins, significant mRNA expression could be demonstrated. Applying stringent consecutive filtering using SignalP, SecretomeP and ER_retention signal databases, 305 proteins were assigned as potential myokines of which 12 proteins containing a secretory signal peptide were not previously described. This comprehensive profiling study of the human skeletal muscle secretome expands our knowledge of the composition of the human myokinome and may contribute to our understanding of the role of myokines in multiple biological processes. This article is part of a Special Issue entitled: Biomarkers: A Proteomic Challenge.


Subject(s)
Biomarkers/analysis , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Myoblasts/metabolism , Proteome/analysis , Proteomics/methods , Adult , Cells, Cultured , Chromatography, Liquid , Computational Biology , Culture Media, Conditioned/pharmacology , Electrophoresis, Gel, Two-Dimensional , Female , Humans , Male , Mass Spectrometry , Muscle Proteins/genetics , Muscle, Skeletal/cytology , Myoblasts/cytology , Transcriptome
18.
iScience ; 27(3): 109276, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38450153

ABSTRACT

Chronic stress episodes increase metabolic disease risk even after recovery. We propose that persistent stress detrimentally impacts hepatic metabolic reprogramming, particularly mitochondrial function. In male C57BL/6 mice chronic variable stress (Cvs) reduced energy expenditure (EE) and body mass despite increased energy intake versus controls. This coincided with decreased glucose metabolism and increased lipid ß-oxidation, correlating with EE. After Cvs, mitochondrial function revealed increased thermodynamic efficiency (ƞ-opt) of complex CI, positively correlating with blood glucose and NEFA and inversely with EE. After Cvs recovery, the metabolic flexibility of hepatocytes was lost. Reduced CI-driving NAD+/NADH ratio, and diminished methylation-related one-carbon cycle components hinted at epigenetic regulation. Although initial DNA methylation differences were minimal after Cvs, they diverged during the recovery phase. Here, the altered enrichment of mitochondrial DNA methylation and linked transcriptional networks were observed. In conclusion, Cvs rapidly initiates the reprogramming of hepatic energy metabolism, supported by lasting epigenetic modifications.

19.
Diabetes ; 73(7): 1058-1071, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38608276

ABSTRACT

The Rab-GTPase-activating protein (RabGAP) TBC1D4 (AS160) represents a key component in the regulation of glucose transport into skeletal muscle and white adipose tissue (WAT) and is therefore crucial during the development of insulin resistance and type 2 diabetes. Increased daily activity has been shown to be associated with improved postprandial hyperglycemia in allele carriers of a loss-of-function variant in the human TBC1D4 gene. Using conventional Tbc1d4-deficient mice (D4KO) fed a high-fat diet, we show that moderate endurance exercise training leads to substantially improved glucose and insulin tolerance and enhanced expression levels of markers for mitochondrial activity and browning in WAT from D4KO animals. Importantly, in vivo and ex vivo analyses of glucose uptake revealed increased glucose clearance in interscapular brown adipose tissue and WAT from trained D4KO mice. Thus, chronic exercise is able to overcome the genetically induced insulin resistance caused by Tbc1d4 depletion. Gene variants in TBC1D4 may be relevant in future precision medicine as determinants of exercise response.


Subject(s)
Adipose Tissue, White , GTPase-Activating Proteins , Insulin Resistance , Mice, Knockout , Physical Conditioning, Animal , Insulin Resistance/genetics , Insulin Resistance/physiology , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Animals , Mice , Physical Conditioning, Animal/physiology , Adipose Tissue, White/metabolism , Diet, High-Fat , Male , Adipose Tissue, Brown/metabolism , Muscle, Skeletal/metabolism , Glucose/metabolism , Mice, Inbred C57BL
20.
Mol Biol Rep ; 39(6): 6835-42, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22311022

ABSTRACT

The polycystic ovary syndrome (PCOS) is a complex endocrine-metabolic disorder consisting of reproductive disturbances associated with all aspects of the metabolic syndrome and genetic components in the pathology of this complex disease is very likely. Accordingly, variations in single genes might affect specific features of PCOS and thereby help to define different subgroups. SREBP-1 or LXRα have been shown to be genetically linked to lipid metabolism or insulin sensitivity. As these are two major aspects of the PCOS phenotype, we evaluated both genes in a cohort of 153 PCOS patients. Analyses of both genes revealed in SREBF-1, i.e. SREBP-1a and SREBP-1c, not any variation and in the LXRα gene no novel sequence variations. Common variants of LXRα (rs2279238:G; all:0.8658; PCOS:0.8627; controls: 0.8686 or A: all:0.13412; PCOS:0.1373; controls:0.1314; (OR (95% CI) 0.9508 (0.4226-2.1385); rs11039155: G: all:0.8767; PCOS:0.8663; controls:0.8857 and A all:0.1233; PCOS:0.1337; controls:0.1143; (OR (95% CI) 0.8383 (0.3618-1.9371)) were also not directly associated to PCOS. Combined analyses of both polymorphism revealed that there was no difference of distribution between the groups. In contrast, analyses of the impact of these polymorphisms on metabolic parameters of the syndrome indicated significant differences related to genotypes. The data indicated that rs11039155 increases metabolic risk, whereas rs2279238 has a protective effect on the overall metabolic risk. The investigation of the PCOS group presented indicates that the combined analyses of variations in putative candidate genes allowed a genotype-phenotype correlation for metabolic features.


Subject(s)
Orphan Nuclear Receptors/genetics , Polycystic Ovary Syndrome/genetics , Sterol Regulatory Element Binding Protein 1/genetics , Adult , Blood Glucose , Body Mass Index , Case-Control Studies , Female , Gene Frequency , Genetic Association Studies , Genetic Variation , Humans , Insulin/blood , Linkage Disequilibrium , Liver X Receptors , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/physiopathology , Polymorphism, Single Nucleotide , Sequence Analysis, DNA , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL