Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
1.
J Inherit Metab Dis ; 45(2): 144-156, 2022 03.
Article in English | MEDLINE | ID: mdl-34595757

ABSTRACT

Inherited metabolic disorders (IMDs) are a heterogeneous group of rare disorders characterized by disruption of metabolic pathways. To date, data on incidence and prevalence of IMDs are limited. Taking advantage of a functioning network within the Austrian metabolic group, our registry research aimed to update the data of the "Registry for Inherited Metabolic Disorders" started between 1985 and 1995 with retrospectively retrieved data on patients with IMDs according to the Society for the Study of Inborn Errors of Metabolism International Classification of Diseases 11 (SSIEM ICD11) catalogue. Included in this retrospective register were 2631 patients with an IMD according to the SSIEM ICD11 Classification, who were treated in Austria. Thus, a prevalence of 1.8/10 000 for 2020 and a median minimal birth prevalence of 16.9/100 000 (range 0.7/100 000-113/100 000) were calculated for the period 1921 to February 2021. We detected a male predominance (m:f = 1.2:1) and a mean age of currently alive patients of 17.6 years (range 5.16 months-100 years). Most common diagnoses were phenylketonuria (17.7%), classical galactosaemia (6.6%), and biotinidase deficiency (4.2%). The most common diagnosis categories were disorders of amino acid and peptide metabolism (819/2631; 31.1%), disorders of energy metabolism (396/2631; 15.1%), and lysosomal disorders (395/2631; 15.0%). In addition to its epidemiological relevance, the "Registry for Inherited Metabolic Disorders" is an important tool for enhancing an exchange between care providers. Moreover, by pooling expertise it prospectively improves patient treatment, similar to pediatric oncology protocols. A substantial requirement for ful filling this goal is to regularly update the registry and provide nationwide coverage with inclusion of all medical specialties.


Subject(s)
Metabolic Diseases , Metabolism, Inborn Errors , Austria/epidemiology , Child , Female , Humans , Infant , Male , Metabolic Diseases/diagnosis , Metabolic Diseases/epidemiology , Metabolism, Inborn Errors/diagnosis , Metabolism, Inborn Errors/epidemiology , Prevalence , Registries , Retrospective Studies
2.
Diabetes Obes Metab ; 23(2): 589-598, 2021 02.
Article in English | MEDLINE | ID: mdl-33200501

ABSTRACT

AIM: To assess predictors of in-hospital mortality in people with prediabetes and diabetes hospitalized for COVID-19 infection and to develop a risk score for identifying those at the greatest risk of a fatal outcome. MATERIALS AND METHODS: A combined prospective and retrospective, multicentre, cohort study was conducted at 10 sites in Austria in 247 people with diabetes or newly diagnosed prediabetes who were hospitalized with COVID-19. The primary outcome was in-hospital mortality and the predictor variables upon admission included clinical data, co-morbidities of diabetes or laboratory data. Logistic regression analyses were performed to identify significant predictors and to develop a risk score for in-hospital mortality. RESULTS: The mean age of people hospitalized (n = 238) for COVID-19 was 71.1 ± 12.9 years, 63.6% were males, 75.6% had type 2 diabetes, 4.6% had type 1 diabetes and 19.8% had prediabetes. The mean duration of hospital stay was 18 ± 16 days, 23.9% required ventilation therapy and 24.4% died in the hospital. The mortality rate in people with diabetes was numerically higher (26.7%) compared with those with prediabetes (14.9%) but without statistical significance (P = .128). A score including age, arterial occlusive disease, C-reactive protein, estimated glomerular filtration rate and aspartate aminotransferase levels at admission predicted in-hospital mortality with a C-statistic of 0.889 (95% CI: 0.837-0.941) and calibration of 1.000 (P = .909). CONCLUSIONS: The in-hospital mortality for COVID-19 was high in people with diabetes but not significantly different to the risk in people with prediabetes. A risk score using five routinely available patient variables showed excellent predictive performance for assessing in-hospital mortality.


Subject(s)
COVID-19/mortality , Diabetes Mellitus, Type 2/mortality , Health Status Indicators , Patient Admission/statistics & numerical data , Prediabetic State/mortality , Aged , Austria , COVID-19/virology , Diabetes Mellitus, Type 2/virology , Female , Hospital Mortality , Hospitals , Humans , Length of Stay/statistics & numerical data , Male , Middle Aged , Prediabetic State/virology , Prospective Studies , Retrospective Studies , Risk Assessment , Risk Factors , SARS-CoV-2
3.
Liver Int ; 40(7): 1620-1633, 2020 07.
Article in English | MEDLINE | ID: mdl-32281248

ABSTRACT

BACKGROUND & AIMS: Osteopontin, a multifunctional protein and inflammatory cytokine, is overexpressed in adipose tissue and liver in obesity and contributes to the induction of adipose tissue inflammation and non-alcoholic fatty liver (NAFL). Studies performed in both mice and humans also point to a potential role for OPN in malignant transformation and tumour growth. To fully understand the role of OPN on the development of NAFL-derived hepatocellular carcinoma (HCC), we applied a non-alcoholic steatohepatitis (NASH)-HCC mouse model on osteopontin-deficient (Spp1-/- ) mice analysing time points of NASH, fibrosis and HCC compared to wild-type mice. METHODS: Two-day-old wild-type and Spp1-/- mice received a low-dose streptozotocin injection in order to induce diabetes, and were fed a high-fat diet starting from week 4. Different cohorts of mice of both genotypes were sacrificed at 8, 12 and 19 weeks of age to evaluate the NASH, fibrosis and HCC phenotypes respectively. RESULTS: Spp1-/- animals showed enhanced hepatic lipid accumulation and aggravated NASH, as also increased hepatocellular apoptosis and accelerated fibrosis. The worse steatotic and fibrotic phenotypes observed in Spp1-/- mice might be driven by enhanced hepatic fatty acid influx through CD36 overexpression and by a pathological accumulation of specific diacylglycerol species during NAFL. Lack of osteopontin lowered systemic inflammation, prevented HCC progression to less differentiated tumours and improved overall survival. CONCLUSIONS: Lack of osteopontin dissociates NASH-fibrosis severity from overall survival and HCC malignant transformation in NAFLD, and is therefore a putative therapeutic target only for advanced chronic liver disease.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Animals , Liver , Mice , Osteopontin/genetics
5.
Curr Atheroscler Rep ; 21(10): 37, 2019 07 27.
Article in English | MEDLINE | ID: mdl-31350625

ABSTRACT

PURPOSE OF REVIEW: A considerable body of data from genetic and epidemiological studies strongly support a causal relationship between high lipoprotein(a) [Lp(a)] levels, and the development of atherosclerosis and cardiovascular disease. This relationship is continuous, unrelated to Lp(a) threshold, and independent of low-density lipoprotein (LDL) and high-density lipoprotein (HDL) cholesterol levels. Unfortunately, the mechanism(s) through which Lp(a) promotes atherosclerosis are not clarified yet. Suggested hypotheses include: an increased Lp(a)-associated cholesterol entrapment in the arterial intima followed by inflammatory cell recruitment, abnormal upload of proinflammatory oxidized phospholipids, impaired fibrinolysis by inhibition of plasminogen activation, and enhanced coagulation, through inhibition of the tissue factor pathway inhibitor. This review is aimed at summarizing the available evidence on the topic. RECENT FINDINGS: There are two clinical forms, isolated hyperlipidemia(a) [HyperLp(a)] with acceptable LDL-C levels (< 70 mg/dL), and combined elevation of Lp(a) and LDL-C in plasma. To date, no drugs that selectively decrease Lp(a) are available. Some novel lipid-lowering drugs can lower Lp(a) levels, but to a limited extent, as their main effect is aimed at decreasing LDL-C levels. Significant Lp(a) lowering effects were obtained with nicotinic acid at high doses. However, adverse effects apart, nicotinic acid is no longer prescribed and available in Europe for clinical use, after European Agency of Medicines (EMA) ban. The only effective therapeutic option for now is Lipoprotein Apheresis (LA), albeit with some limitations. Lastly, it is to be acknowledged that the body of evidence confirming that reducing plasma isolated elevation of Lp(a) brings cardiovascular benefit is still insufficient. However, the growing bulk of clinical, genetic, mechanistic, and epidemiological available evidence strongly suggests that Lp(a) is likely to be the smoking gun.


Subject(s)
Atherosclerosis/blood , Lipoprotein(a)/blood , Adult , Antibodies, Monoclonal, Humanized/therapeutic use , Atherosclerosis/drug therapy , Blood Component Removal/methods , Cholesterol, LDL/blood , Humans , Hypolipidemic Agents/adverse effects , Hypolipidemic Agents/therapeutic use , Middle Aged , Niacin/adverse effects , Niacin/therapeutic use , Oligonucleotides, Antisense/therapeutic use , Risk Factors , Ubiquinone/analogs & derivatives , Ubiquinone/therapeutic use
6.
Hepatology ; 65(6): 1875-1890, 2017 06.
Article in English | MEDLINE | ID: mdl-28073161

ABSTRACT

The genetic polymorphism I148M of patatin-like phospholipase domain-containing 3 (PNPLA3) is robustly associated with hepatic steatosis and its progression to steatohepatitis, fibrosis, and cancer. Hepatic stellate cells (HSCs) are key players in the development of liver fibrosis, but the role of PNPLA3 and its variant I148M in this process is poorly understood. Here we analyzed the expression of PNPLA3 during human HSC activation and thereby explored how a PNPLA3 variant impacts hepatic fibrogenesis. We show that expression of PNPLA3 gene and protein increases during the early phases of activation and remains elevated in fully activated HSCs (P < 0.01). Knockdown of PNPLA3 significantly decreases the profibrogenic protein alpha-smooth muscle actin (P < 0.05). Primary human I148M HSCs displayed significantly higher expression and release of proinflammatory cytokines, such as chemokine (C-C motif) ligand 5 (P < 0.01) and granulocyte-macrophage colony-stimulating factor (P < 0.001), thus contributing to migration of immune cells (P < 0.05). Primary I148M HSCs showed reduced retinol (P < 0.001) but higher lipid droplet content (P < 0.001). In line with this, LX-2 cells stably overexpressing I148M showed augmented proliferation and migration, lower retinol, and abolished retinoid X receptor/retinoid A receptor transcriptional activities but more lipid droplets. Knockdown of I148M PNPLA3 (P < 0.001) also reduces chemokine (C-C motif) ligand 5 and collagen1α1 expression (P < 0.05). Notably, I148M cells display reduced peroxisome proliferator-activated receptor gamma transcriptional activity, and this effect was attributed to increased c-Jun N-terminal kinase, thereby inhibiting peroxisome proliferator-activated receptor gamma through serine 84 phosphorylation and promoting activator protein 1 transcription. Conversely, the c-Jun N-terminal kinase inhibitor SP600125 and the peroxisome proliferator-activated receptor gamma agonist rosiglitazone decreased activator protein 1 promoter activity. CONCLUSIONS: These data indicate that PNPLA3 is required for HSC activation and that its genetic variant I148M potentiates the profibrogenic features of HSCs, providing a molecular mechanism for the higher risk of progression and severity of liver diseases conferred to patients carrying the I148M variant. (Hepatology 2017;65:1875-1890).


Subject(s)
Cytokines/metabolism , Genetic Predisposition to Disease , Hepatic Stellate Cells/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Lipase/genetics , Blotting, Western , Cell Movement/genetics , Cell Proliferation , Cells, Cultured , Chromatography, Gas/methods , Flow Cytometry/methods , Gene Expression Regulation , Humans , Phenotype , Polymorphism, Genetic , Real-Time Polymerase Chain Reaction , Reference Values
7.
Horm Metab Res ; 50(6): 478-484, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29883974

ABSTRACT

Increased free fatty acids stimulate sympathetic nervous system activity, impair endothelium-dependent vasodilation, and increase regional blood flow. The aim of this study was to assess if fatty acids acutely elevated by infusion of intralipid/heparin affect cardiovascular reactivity employing two stressors eliciting either a cardiac (Stroop test) or vascular (Cold Face test) dominated pressor response. Two stress tasks were performed in 20 healthy subjects (10 women, 10 men) before and during a 180-min intralipid/heparin or saline infusion as placebo on alternate trial days in a randomized crossover study design. Blood pressure, heart rate, cardiac index, and total peripheral resistance index were measured. At baseline, the Stroop test did not affect hemodynamic parameters, and the Cold Face test had an impact on hemodynamic parameters except for heart rate. Plasma fatty acids concentrations increased to 810% (t=11.0, p<0.001) of baseline and C-peptide increased by 17% (t=4.66, p<0.001) during intralipid/heparin infusion. This was paralleled by increased cardiac index (F=9.98; p<0.005 vs. saline) and reduced total peripheral resistance index (F=4.46; p<0.05 vs saline). There was no effect of intralipid/heparin or saline infusion on Stroop test or Cold Face test reactivity of hemodynamic parameters. An acute increase in free fatty acids does not affect the magnitude or pattern of stress response in healthy volunteers, but primarily alter the underlying cardiovascular tone by decreasing total peripheral resistance index and increasing cardiac index to maintain a constant blood pressure.


Subject(s)
Cardiovascular Diseases/etiology , Fatty Acids, Nonesterified/administration & dosage , Stress, Psychological/complications , Adolescent , Adult , Cardiovascular Diseases/blood , Cross-Over Studies , Double-Blind Method , Female , Heart Rate , Humans , Male , Stress, Psychological/blood , Task Performance and Analysis , Young Adult
8.
Gynecol Endocrinol ; 34(9): 736-739, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29560763

ABSTRACT

McArdle disease or glycogen storage disease (GSD) type V is a rare autosomal recessive inherited disorder in skeletal muscle metabolism leading to exercise intolerance, muscle cramps and in some cases to rhabdomyolysis and acute renal failure due to elevated serum myoglobin levels. Albeit the uterine smooth muscle is not affected, pregnancy and delivery can be physically strenuous and may require specific anesthesiologic care. However, data on pregnancy progress and outcome and on special implications linked to anesthesia in women with McArdle's disease is scarce, thus posing a challenge to pre- and peripartal management. We report a case of a pregnant woman with Morbus McArdle who was monitored during her pregnancy and delivered a healthy male via cesarean section under spinal anesthesia. Pregnancy, delivery and recovery were uneventful. Our findings, combined with a literature review, lead to the conclusion that uncomplicated pregnancy and delivery can be expected.


Subject(s)
Glycogen Storage Disease Type V/therapy , Pregnancy Complications/therapy , Adult , Disease Management , Female , Humans , Pregnancy , Pregnancy Outcome
9.
Immunity ; 29(4): 565-77, 2008 Oct 17.
Article in English | MEDLINE | ID: mdl-18848473

ABSTRACT

The innate inflammatory immune response must be tightly controlled to avoid damage to the host. Here, we showed that the tuberous sclerosis complex-mammalian target of rapamycin (TSC-mTOR) pathway regulated inflammatory responses after bacterial stimulation in monocytes, macrophages, and primary dendritic cells. Inhibition of mTOR by rapamycin promoted production of proinflammatory cytokines via the transcription factor NF-kappaB but blocked the release of interleukin-10 via the transcription factor STAT3. Conversely, deletion of TSC2, the key negative regulator of mTOR, diminished NF-kappaB but enhanced STAT3 activity and reversed this proinflammatory cytokine shift. Rapamycin-hyperactivated monocytes displayed a strong T helper 1 (Th1) cell- and Th17 cell-polarizing potency. Inhibition of mTOR in vivo regulated the inflammatory response and protected genetically susceptible mice against lethal Listeria monocytogenes infection. These data identify the TSC2-mTOR pathway as a key regulator of innate immune homeostasis with broad clinical implications for infectious and autoimmune diseases, vaccination, cancer, and transplantation.


Subject(s)
Cytokines/immunology , Immunity, Innate , Listeriosis/immunology , Monocytes/immunology , Protein Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Cytokines/biosynthesis , Female , Humans , Inflammation/immunology , Inflammation/metabolism , Lipopolysaccharides/immunology , Listeria monocytogenes/immunology , Listeriosis/microbiology , Listeriosis/prevention & control , Mice , Mice, Inbred BALB C , Mice, Knockout , Monocytes/drug effects , Monocytes/metabolism , NF-kappa B/metabolism , Protein Kinases/immunology , STAT3 Transcription Factor/metabolism , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Th1 Cells/immunology , Th1 Cells/metabolism , Tuberous Sclerosis , Tuberous Sclerosis Complex 2 Protein
10.
J Gastroenterol Hepatol ; 32(1): 253-260, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27083512

ABSTRACT

BACKGROUND AND AIM: Obesity is a major risk factor for liver fibrosis and tightly associated with low levels of adiponectin. Adiponectin has antifibrogenic activity protecting from liver fibrosis, which is mainly driven by activated hepatic stellate cells (HSC). Aquaporins are transmembrane proteins that allow the movement of water and, in case of aquaglyceroporins (AQPs), of glycerol that is needed in quiescent HSC for lipogenesis. Expression of various AQPs in liver is altered by obesity; however, the mechanisms through which obesity influences HSCs activation and AQPs expression remain unclear. This study aimed to identify obesity-associated factors that are related to HSC AQPs expression activation and lipid storage. METHODS: Correlations between serum adipokine levels and hepatic AQPs gene expression were analyzed from a cohort of obese patients. AQP and fibrotic gene expression was determined in a HSC line (LX2) and in a hepatocyte cell line (HepG2) after stimulation with adiponectin using quantitative real-time polymerase chain reaction. RESULTS: We found that serum adiponectin significantly correlated with liver AQP3, AQP7, AQP9 gene expressions. In vitro, adiponectin induced upregulation of AQP3 gene and AQP3 protein expression in human HSCs, but not in hepatocytes, while AQP7, AQP9 remained undetectable. Accordingly, HSC stimulated with adiponectin increased glycerol uptake, lipogenic gene expression, and lipid storage while downregulating activation/fibrosis markers. CONCLUSIONS: These findings demonstrate that adiponectin is a potent inhibitor of HSC activation and induces AQPs expression. Thus, low serum levels of adiponectin could be a mechanism how obesity affects the functional state of HSC, thereby contributing to obesity-associated liver fibrosis.


Subject(s)
Adiponectin/physiology , Aquaglyceroporins/genetics , Aquaglyceroporins/metabolism , Gene Expression Regulation/genetics , Gene Expression/genetics , Hepatic Stellate Cells/metabolism , Adiponectin/deficiency , Adult , Body Mass Index , Female , Hep G2 Cells , Humans , Lipogenesis/genetics , Liver Cirrhosis/etiology , Liver Cirrhosis/prevention & control , Male , Obesity/genetics , Obesity/metabolism
11.
Eur J Clin Invest ; 46(11): 911-919, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27600500

ABSTRACT

BACKGROUND: Infiltration of white adipose tissue (WAT) by inflammatory cells in obesity is considered to be a key event in the development of insulin resistance. Recently, mast cells (MCs) have been identified as new players in the pathogenesis of obesity. We aimed to investigate the relationship between MCs and various inflammatory markers in serum and WAT and to determine the role of MCs in the aetiology of insulin resistance. MATERIALS AND METHODS: Gene expression was measured in WAT from 20 morbidly obese patients and 20 nonobese control subjects. Homoeostasis Model of Assessment-Insulin Resistance (HOMA-IR) was used to estimate insulin sensitivity. In addition, wild-type and mast cell-deficient mice were fed a high-fat or low-fat diet to study mast cell influence on inflammatory cell polarization in WAT and overall metabolic changes. RESULTS: WAT levels of MC-specific TPSb2 transcript were increased in obesity and significantly positively correlated with TNF, CCL2, CCL5 and CD68 gene expression levels in our study subjects after adjustment for sex, age and BMI. Accordingly, MC deficiency abrogated increase in expression of pro-inflammatory M1 macrophage marker genes in mouse WAT upon high-fat diet feeding. However, MCs accumulated in obese human WAT independent of insulin resistance and systemic changes in inflammatory mediators. CONCLUSIONS: Our results suggest that MCs contribute to the local pro-inflammatory state within WAT in obesity but do not play a primary role in causing insulin resistance.


Subject(s)
Insulin Resistance/physiology , Mast Cells/physiology , Obesity, Morbid/pathology , Adipose Tissue, White/pathology , Adult , Animals , Biomarkers/metabolism , Case-Control Studies , Cells, Cultured , Cytokines/metabolism , Female , Humans , Male , Mice, Inbred C57BL , Phenotype , Thinness/pathology , Tryptases/metabolism
12.
Eur J Nutr ; 55(6): 2011-9, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26266932

ABSTRACT

BACKGROUND: The inflammatory process associated with obesity mainly arises from white adipose tissue (WAT) alterations. In the last few years, nutritional-based strategies have been positioned as promising alternatives to pharmacological approaches against these pathologies. Our aim was to determine the potential of a rice bran enzymatic extract (RBEE)-supplemented diet in the prevention of metabolic, biochemical and functional adipose tissue and macrophage changes associated with a diet-induced obesity (DIO) in mice. METHODS: C57BL/6J mice were fed high-fat diet (HF), 1 and 5 % RBEE-supplemented high-fat diet (HF1 % and HF5 %, respectively) and standard diet as control. Serum cardiometabolic parameters, adipocytes size and mRNA expression of pro-inflammatory biomarkers and macrophage polarization-related genes from WAT and liver were evaluated. RESULTS: RBEE administration significantly decreased insulin resistance in obese mice. Serum triglycerides, total cholesterol, glucose, insulin, adiponectin and nitrites from treated mice were partially restored, mainly by 1 % RBEE-enriched diet. The incremented adipocytes size observed in HF group was reduced by RBEE treatment, being 1 % more effective than 5 % RBEE. Pro-inflammatory biomarkers in WAT such as IL-6 and IL-1ß were significantly decreased in RBEE-treated mice. Adiponectin, PPARγ, TNF-α, Emr1 or M1/M2 levels were significantly restored in WAT from HF1 % compared to HF mice. CONCLUSIONS: RBEE-supplemented diet attenuated insulin resistance, dyslipidemia and morphological and functional alterations of adipose tissue in DIO mice. These benefits were accompanied by a modulating effect in adipocytes secretion and some biomarkers associated with macrophage polarization. Therefore, RBEE may be considered an alternative nutritional complement over metabolic syndrome and its complications.


Subject(s)
Adipose Tissue, White/metabolism , Diet, High-Fat/adverse effects , Dietary Fiber/administration & dosage , Inflammation/diet therapy , Macrophages/metabolism , Oryza/chemistry , Adipocytes , Adiponectin/metabolism , Animals , Biomarkers/blood , Calcium-Binding Proteins , Cholesterol/blood , Dyslipidemias/diet therapy , Dyslipidemias/etiology , Inflammation/etiology , Insulin/blood , Insulin Resistance , Interleukin-1beta/blood , Interleukin-6/blood , Male , Mice , Mice, Inbred C57BL , Obesity/diet therapy , Obesity/etiology , PPAR gamma/metabolism , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled , Triglycerides/blood , Tumor Necrosis Factor-alpha/metabolism
13.
Am J Physiol Endocrinol Metab ; 308(8): E631-40, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25670825

ABSTRACT

Hypoglycemia, a major side effect of intensive glucose-lowering therapy, was recently linked to increased cardiovascular risk in patients with diabetes. Whether increased circulating free fatty acids (FFA) owing to catecholamine-induced lipolysis affect myocardial energy metabolism and thus link hypoglycemia to cardiac vulnerability is unclear. Therefore, this study investigated the impact of hypoglycemia counterregulation (± inhibition of lipolysis) on myocardial lipid content (MYCL) and left ventricular function in healthy subjects. Nine healthy men were studied in randomized order: 1) insulin/hypoglycemia test (IHT; ins+/aci-), 2) IHT during inhibition of adipose tissue lipolysis by acipimox (ins+/aci+), 3) normoglycemia with acipimox (ins-/aci+), and 4) normoglycemia with placebo (ins-/aci-). MYCL and cardiac function were assessed by employing magnetic resonance spectroscopy/imaging at baseline and at 2 and 6 h. In response to acute hypoglycemia, plasma FFA (P<0.0001) and ejection fraction (EF; from 63.2±5.5 to 69.6±6.3%, P=0.0001) increased significantly and were tightly correlated with each other (r=0.68, P=0.0002); this response was completely blunted by inhibition of adipose tissue lipolysis. In the presence of normoglycemia, inhibition of lipolysis was associated with a drop in EF (from 59.2±5.5 to 53.9±6.9%,P=0.005) and a significant decrease in plasma FFA, triglycerides, and MYCL (by 48.5%, P=0.0001). The present data indicate that an intact interorgan cross-talk between adipose tissue and the heart is a prerequisite for catecholamine-mediated myocardial contractility and preservation of myocardial lipid stores in response to acute hypoglycemia.


Subject(s)
Allostasis , Fatty Acids, Nonesterified/metabolism , Heart Ventricles/physiopathology , Hypoglycemia/physiopathology , Lipid Metabolism , Models, Biological , Ventricular Dysfunction, Left/etiology , Adenylyl Cyclase Inhibitors , Adenylyl Cyclases/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/enzymology , Adipose Tissue, White/metabolism , Adult , Allostasis/drug effects , Epinephrine/blood , Fatty Acids, Nonesterified/blood , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Humans , Hypoglycemia/blood , Hypoglycemia/chemically induced , Hypoglycemia/metabolism , Hypoglycemic Agents/adverse effects , Hypolipidemic Agents/pharmacology , Insulin/adverse effects , Lipid Metabolism/drug effects , Lipolysis/drug effects , Liver/drug effects , Liver/enzymology , Liver/metabolism , Male , Norepinephrine/blood , Stroke Volume/drug effects , Ventricular Dysfunction, Left/prevention & control , Young Adult
14.
Breast Cancer Res Treat ; 154(1): 63-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26482249

ABSTRACT

Breast and endometrial cancer are often estrogen dependent, and their incidence and mortality are increased by obesity in postmenopausal women. Osteopontin (OPN) is a cytokine strongly upregulated in adipose tissue (AT) in obesity. OPN function is potentiated by cleavage by matrix metalloproteinases (MMP). OPN and MMPs play a role in cancer development and are prognostic markers in breast cancer progression. While induction of the estrogen-synthesizing enzyme aromatase by TNFa and IL1 has been shown in preadipocytes, an impact of OPN on aromatase expression in AT has not been investigated yet. Gene expression was determined in AT samples of 21 morbidly obese and matched non-obese subjects. Primary human adipocytes were treated with full-length OPN or MMP-cleaved OPN (cOPN). Protein and mRNA expressions were analyzed from cell lysates, or cells were subsequently supplied with testosterone to determine estradiol production and for indirect co-culture with the estrogen-dependent MCF-7 cell line. Aromatase expression strongly correlated with gene expression of OPN and various MMPs in visceral and MMPs in subcutaneous AT, but not with TNFα expression in both tissues. In vitro, cOPN more effectively than full-length OPN upregulated aromatase mRNA in adipocytes and significantly increased aromatase protein level and estradiol production, leading to increased MCF-7 growth in indirect co-culture. OPN and MMPs are upregulated in AT in obesity, and MMP-cleaved OPN is particularly effective in inducing aromatase activity in human adipocytes. Thereby, obesity-induced OPN expression in AT may contribute to estradiol production and thus to the association of obesity with estrogen-dependent cancers.


Subject(s)
Adipocytes/metabolism , Aromatase/genetics , Estradiol/biosynthesis , Gene Expression Regulation , Osteopontin/metabolism , Adipocytes/drug effects , Adipogenesis/genetics , Adipose Tissue/metabolism , Aromatase/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Proliferation , Cells, Cultured , Female , Gene Expression , Gene Expression Regulation/drug effects , Humans , MCF-7 Cells , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Obesity/genetics , Obesity/metabolism , Osteopontin/genetics , Osteopontin/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism
15.
J Am Coll Nutr ; 34 Suppl 1: 39-41, 2015.
Article in English | MEDLINE | ID: mdl-26400433

ABSTRACT

Obesity is associated with chronic inflammation of the adipose tissue, which contributes to obesity-associated complications such as insulin resistance and type 2 diabetes. The increased inflammatory response seems to be directly related to modern nutrition, particularly aspects of fat quality and macronutrient composition. We have recently published an observational study investigating the practicability and effects of a combined dietary intervention with increased relative protein content and low-glycemic-index carbohydrates, supplemented with omega-3 polyunsaturated fatty acids (PUFAs), on metabolic control and inflammatory parameters in real-life situations in patients with type 2 diabetes. The primary efficacy parameter was the change in HbA1c, and secondary parameters included change in systemic inflammation (measured by ultrasensitive C-reactive protein), body weight, waist circumference, fat mass, and homeostasis model assessment-insulin resistance. Counseling a protein-enriched and low-glycemic-index diet supplemented with long-chain omega-3 PUFAs in a real-life clinical setting improved glycemic control, waist circumference, and silent inflammation in overweight or obese patients with type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/diet therapy , Dietary Supplements , Fatty Acids, Omega-3/administration & dosage , Inflammation/diet therapy , Overweight/diet therapy , Adiposity/drug effects , Body Weight/drug effects , C-Reactive Protein/drug effects , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/etiology , Dietary Carbohydrates/pharmacology , Dietary Proteins/pharmacology , Glycated Hemoglobin/analysis , Glycemic Index , Humans , Inflammation/blood , Inflammation/etiology , Insulin Resistance , Overweight/blood , Overweight/complications , Waist Circumference/drug effects
16.
Diabetologia ; 57(6): 1204-8, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24623100

ABSTRACT

AIMS/HYPOTHESIS: The newly identified liver- and fat-derived hormone, betatrophin, has recently been linked to insulin resistance and pancreatic beta cell growth in mice. These preclinical findings have suggested betatrophin as a potential candidate for novel glucose-lowering treatment concepts involving beta cell regeneration. However, the role of betatrophin in human insulin resistance and type 2 diabetes is currently unknown. Hence, the aim of this study was to investigate circulating betatrophin concentrations in two distinct cohorts with insulin resistance. METHODS: Betatrophin concentrations were analysed in (1) age- and sex-matched lean (n = 20) and morbidly obese individuals (n = 19), and (2) age-, sex- and BMI-matched non-diabetic (n = 19) and type 2 diabetic individuals (n = 18). RESULTS: Betatrophin concentrations did not differ between lean and morbidly obese or between non-diabetic and type 2 diabetic participants. No association was found with variables of beta cell function and glucose homeostasis. However, betatrophin did correlate significantly with plasma atherogenic lipids including total cholesterol, LDL-cholesterol and apolipoprotein B in morbidly obese and type 2 diabetic patients but not in controls. Insulin-resistant individuals with hypercholesterolaemia (≥5.2 mmol/l) had significantly higher betatrophin concentrations than those with normal cholesterol (<5.2 mmol/l). CONCLUSIONS/INTERPRETATION: Betatrophin is a recently identified hormone, the circulating concentrations of which are unaltered in human insulin resistance but correlate significantly with atherogenic lipid profiles in high-risk cohorts with morbid obesity or type 2 diabetes. Betatrophin could therefore be a novel pathomechanistic player in dysfunctional lipid metabolism associated with high cardiovascular risk.


Subject(s)
Diabetes Mellitus, Type 2/blood , Insulin/blood , Peptide Hormones/blood , Adult , Angiopoietin-Like Protein 8 , Angiopoietin-like Proteins , Blood Glucose/metabolism , Cholesterol, LDL/blood , Female , Humans , Insulin Resistance/physiology , Lipid Metabolism/physiology , Male , Middle Aged , Obesity, Morbid/blood
17.
Cardiovasc Diabetol ; 13: 23, 2014 Jan 17.
Article in English | MEDLINE | ID: mdl-24438079

ABSTRACT

BACKGROUND: Obesity and particularly the metabolic syndrome, which is often associated with obesity, combine a major risk for type 2 diabetes and cardiovascular disease. Emerging evidence indicate obesity-associated subclinical inflammation primarily originating from adipose tissue as a common cause for type 2 diabetes and cardiovascular disease. However, a suitable and well-characterized mouse model to simultaneously study obesity-associated metabolic disorders and atherosclerosis is not available yet. Here we established and characterized a murine model combining diet-induced obesity and associated adipose tissue inflammation and metabolic deteriorations as well as atherosclerosis, hence reflecting the human situation of cardio-metabolic disease. METHODS: We compared a common high-fat diet with 0.15% cholesterol (HFC), and a high-fat, high-sucrose diet with 0.15% cholesterol (HFSC) fed to LDL receptor-deficient (LDLR-/-) mice. Insulin resistance, glucose tolerance, atherosclerotic lesion formation, hepatic lipid accumulation, and inflammatory gene expression in adipose tissue and liver were assessed. RESULTS: After 12-16 weeks, LDLR-/- mice fed HFSC or HFC developed significant diet-induced obesity, adipose tissue inflammation, insulin resistance, and impaired glucose tolerance compared to lean controls. Notably, HFSC-fed mice developed significantly higher adipose tissue inflammation in parallel with significantly elevated atherosclerotic lesion area compared to those on HFC. Moreover, LDLR-/- mice on HFSC showed increased insulin resistance and impaired glucose tolerance relative to those on HFC. After prolonged feeding (20 weeks), however, no significant differences in inflammatory and metabolic parameters as well as atherosclerotic lesion formation were detectable any more between LDLR-/- mice fed HFSC or HFC. CONCLUSION: The use of high sucrose rather than more complex carbohydrates in high-fat diets significantly accelerates development of obesity-driven metabolic complications and atherosclerotic plaque formation parallel to obesity-induced adipose tissue inflammation in LDLR-/- mice. Hence LDLR-/- mice fed high-fat high-sucrose cholesterol-enriched diet appear to be a suitable and time-saving animal model for cardio-metabolic disease. Moreover our results support the suggested interrelation between adipose tissue inflammation and atherosclerotic plaque formation.


Subject(s)
Adipose Tissue/metabolism , Atherosclerosis/blood , Dietary Fats/adverse effects , Dietary Sucrose/adverse effects , Disease Models, Animal , Receptors, LDL/deficiency , Adipose Tissue/pathology , Animals , Atherosclerosis/etiology , Dietary Fats/administration & dosage , Dietary Sucrose/administration & dosage , Inflammation/blood , Inflammation/etiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Time Factors
18.
Gerontology ; 60(3): 189-96, 2014.
Article in English | MEDLINE | ID: mdl-24457898

ABSTRACT

Autoimmunity is a well-known pathogenic component in type 1 diabetes (T1DM). The assumption that the pathogenesis of type 2 diabetes (T2DM) also encompasses autoimmune aspects is recognized increasingly, based on the presence of circulating autoantibodies against ß cells, self-reactive T cells, but also on the glucose-lowering efficacy of some immunomodulatory therapies in T2DM. The identification of these autoantibodies in elderly patients with slowly progressive manifestation of diabetes led to the introduction of a distinct clinical entity termed latent autoimmune diabetes of the adult (LADA), which combines features of both T1DM and T2DM. The autoantibody cluster differs in patients with LADA from patients with T1DM, but their presence indicates steady progression towards ß-cell death and subsequent need for initiation of insulin treatment in a shorter period of time compared to autoantibody-negative T2DM patients. Autoimmune aspects in T2DM are not solely restricted to autoantibodies and thus LADA. They include the self-reactive T cells or defects in regulatory T cells (Tregs), which have been detected in autoantibody-negative T2DM patients as well. One contributor to the autoimmune activation in T2DM seems to be the chronic inflammatory state, characteristic of this disease. Upon inflammation-induced tissue destruction, cryptic 'self' antigens can trigger an autoimmune response, which in turn accelerates ß-cell death. Both innate and adaptive immune system components, specifically macrophages and self-reactive T cells, contribute to an increased secretion of inflammatory cytokines involved in inflammatory and autoimmune processes. However, the extent to which inflammation overlaps with autoimmunity is not known. Our review focuses on autoimmune involvement in T2DM, with an emphasis on LADA and the humoral immune response, on the involvement of chronic inflammation in autoimmunity, and specifically the role of B and T cells as links between inflammatory and autoimmune reactions. We will further stress the consequences of autoimmune activation for T2DM patients and present novel therapeutic approaches for T2DM management that rely on immune modulation.


Subject(s)
Autoimmunity , Diabetes Mellitus, Type 2/immunology , Adult , Aged , Animals , Autoimmune Diseases/etiology , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/therapy , Female , Glutamate Decarboxylase/immunology , Humans , Immunomodulation , Insulin-Secreting Cells/immunology , Lymphocytes/immunology , Male , Middle Aged , Models, Immunological
19.
Eur J Immunol ; 42(11): 2983-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22806614

ABSTRACT

Plasma lipoproteins such as LDL (low-density lipoprotein) are important therapeutic targets as they play a crucial role in macrophage biology and metabolic disorders. The impact of lipoprotein profiles on host defense pathways against Gram-positive bacteria is poorly understood. In this report, we discovered that human serum lipoproteins bind to lipoteichoic acid (LTA) from Staphylococcus aureus and thereby alter the immune response to these bacteria. Size-exclusion chromatography and solid-phase-binding analysis of serum revealed the direct interaction of LTA with apolipoproteins (Apo) B100, ApoA1, and ApoA2. Only ApoB100 and the corresponding LDL exerted biological effects as this binding significantly inhibited LTA-induced cytokine releases from human and murine immune cells. Serum from hypercholesterolemic mice or humans significantly diminished cytokine induction in response to S. aureus or its LTA. Sera taken from the patients with familial hypercholesterolemia before and after ApoB100-directed immuno-apheresis confirmed that ApoB100 inhibited LTA-induced inflammation in humans. In addition, mice in which LDL secretion was pharmacologically inhibited, displayed significantly increased serum cytokine levels upon infection with S. aureus in vivo. The present study identifies ApoB100 as an important suppressor of innate immune activation in response to S. aureus and its LTA.


Subject(s)
Apolipoprotein B-100/pharmacology , Lipopolysaccharides/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Teichoic Acids/immunology , Animals , Female , Humans , Hypercholesterolemia/immunology , Immunity, Innate/immunology , Leukocytes, Mononuclear/immunology , Lipopolysaccharides/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Mice, Knockout , Specific Pathogen-Free Organisms , Staphylococcal Infections/microbiology , Teichoic Acids/antagonists & inhibitors
20.
Wien Klin Wochenschr ; 135(Suppl 1): 161-163, 2023 Jan.
Article in German | MEDLINE | ID: mdl-37101038

ABSTRACT

Acute thrombotic complications as a key feature of accelerated atherothrombotic disease typically precipitate cardiovascular events and therefore strongly contribute to cardiovascular morbidity and mortality in patients with diabetes. Inhibition of platelet aggregation can reduce the risk for acute atherothrombosis. The present article represents the recommendations of the Austrian Diabetes Association for the use of antiplatelet drugs in patients with diabetes according to current scientific evidence.


Subject(s)
Diabetes Mellitus , Thrombosis , Humans , Platelet Aggregation , Platelet Aggregation Inhibitors/therapeutic use , Austria , Blood Platelets
SELECTION OF CITATIONS
SEARCH DETAIL