Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
J Reprod Immunol ; 163: 104234, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38479054

ABSTRACT

Combined oral contraceptives (COCs) are known to cause weight gain and alter metabolic and immunological pathways. However, modifications in arterial or venous thrombotic risk profiles of women of reproductive ages on COC remain unclear. The study aimed at assessing the impact of COC on immune activation in diet-induced obesity. We further established whether the dietary intervention of switching from a high-fat diet (HFD) to a low-fat diet (LFD) attenuates immunological responses. Twenty (n=20) five-week-old female Sprague Dawley rats were randomly divided into two diet groups of HFD (n=15) and LFD (n=5) and were monitored for eight weeks. After eight weeks, animals in the HFD group switched diets to LFD and were randomly assigned to receive high-dose COC (HCOC) or low-dose COC (LCOC) for six weeks. Animals on HFD significantly gained weight and had a higher lee index when compared to the LFD group (p < 0.05). Moreover, the triglyceride-glucose index, insulin, and other metabolic parameters also increased in the HFD group compared to the LFD group (p < 0.001). Consistently, the levels of interleukin (IL)-6 and tumor necrosis factor-alpha (TNF-α), were elevated in the HFD group when compared to the LFD group (p < 0.05). Upon switching from a high-fat to a low-fat diet, insulin levels persistently increased in animals receiving HCOC treatment compared to the LFD and HFD/LFD groups (p < 0.05). Thus, in a rat model of HFD-feeding, short-term HCOC treatment induces long-term metabolic dysregulation, which persists despite dietary intervention. However, further studies are recommended to confirm these findings.


Subject(s)
Diet, Fat-Restricted , Diet, High-Fat , Hyperinsulinism , Obesity , Rats, Sprague-Dawley , Animals , Female , Obesity/immunology , Rats , Diet, High-Fat/adverse effects , Hyperinsulinism/immunology , Hyperinsulinism/chemically induced , Humans , Insulin/blood , Insulin/metabolism , Contraceptives, Oral, Combined/administration & dosage , Contraceptives, Oral, Combined/adverse effects , Interleukin-6/metabolism , Interleukin-6/blood
2.
Diabetes ; 72(9): 1320-1329, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37358194

ABSTRACT

Congenital hyperinsulinism (HI) is a genetic disorder in which pancreatic ß-cell insulin secretion is excessive and results in hypoglycemia that, without treatment, can cause brain damage or death. Most patients with loss-of-function mutations in ABCC8 and KCNJ11, the genes encoding the ß-cell ATP-sensitive potassium channel (KATP), are unresponsive to diazoxide, the only U.S. Food and Drug Administration-approved medical therapy and require pancreatectomy. The glucagon-like peptide 1 receptor (GLP-1R) antagonist exendin-(9-39) is an effective therapeutic agent that inhibits insulin secretion in both HI and acquired hyperinsulinism. Previously, we identified a highly potent antagonist antibody, TB-001-003, which was derived from our synthetic antibody libraries that were designed to target G protein-coupled receptors. Here, we designed a combinatorial variant antibody library to optimize the activity of TB-001-003 against GLP-1R and performed phage display on cells overexpressing GLP-1R. One antagonist, TB-222-023, is more potent than exendin-(9-39), also known as avexitide. TB-222-023 effectively decreased insulin secretion in primary isolated pancreatic islets from a mouse model of hyperinsulinism, Sur1-/- mice, and in islets from an infant with HI, and increased plasma glucose levels and decreased the insulin to glucose ratio in Sur1-/- mice. These findings demonstrate that targeting GLP-1R with an antibody antagonist is an effective and innovative strategy for treatment of hyperinsulinism. ARTICLE HIGHLIGHTS: Patients with the most common and severe form of diazoxide-unresponsive congenital hyperinsulinism (HI) require a pancreatectomy. Other second-line therapies are limited in their use because of severe side effects and short half-lives. Therefore, there is a critical need for better therapies. Studies with the glucagon-like peptide 1 receptor (GLP-1R) antagonist, avexitide (exendin-(9-39)), have demonstrated that GLP-1R antagonism is effective at lowering insulin secretion and increasing plasma glucose levels. We have optimized a GLP-1R antagonist antibody with more potent blocking of GLP-1R than avexitide. This antibody therapy is a potential novel and effective treatment for HI.


Subject(s)
Congenital Hyperinsulinism , Glucagon-Like Peptide-1 Receptor , Hyperinsulinism , Animals , Mice , Antibodies/therapeutic use , Blood Glucose , Congenital Hyperinsulinism/drug therapy , Congenital Hyperinsulinism/genetics , Diazoxide/pharmacology , Glucagon-Like Peptide 1 , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Hyperinsulinism/immunology , Hyperinsulinism/therapy , Mutation , Sulfonylurea Receptors/genetics
3.
Neuropharmacology ; 205: 108920, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34902347

ABSTRACT

The 'apple-shaped' anatomical pattern that accompanies visceral adiposity increases risk for multiple chronic diseases, including conditions that impact the brain, such as diabetes and hypertension. However, distinguishing between the consequences of visceral obesity, as opposed to visceral adiposity-associated metabolic and cardiovascular pathologies, presents certain challenges. This review summarizes current literature on relationships between adipose tissue distribution and cognition in preclinical models and highlights unanswered questions surrounding the potential role of tissue- and cell type-specific insulin resistance in these effects. While gaps in knowledge persist related to insulin insensitivity and cognitive impairment in obesity, several recent studies suggest that cells of the neurovascular unit contribute to hippocampal synaptic dysfunction, and this review interprets those findings in the context of progressive metabolic dysfunction in the CNS. Signalling between cerebrovascular endothelial cells, astrocytes, microglia, and neurons has been linked with memory deficits in visceral obesity, and this article describes the cellular changes in each of these populations with respect to their role in amplification or diminution of peripheral signals. The picture emerging from these studies, while incomplete, implicates pro-inflammatory cytokines, insulin resistance, and hyperglycemia in various stages of obesity-induced hippocampal dysfunction. As in the parable of the five blind wanderers holding different parts of an elephant, considerable work remains in order to assemble a model for the underlying mechanisms linking visceral adiposity with age-related cognitive decline.


Subject(s)
Cognitive Dysfunction , Hippocampus , Hyperglycemia , Hyperinsulinism , Inflammation , Obesity, Abdominal , Animals , Cognitive Dysfunction/etiology , Cognitive Dysfunction/immunology , Cognitive Dysfunction/metabolism , Hippocampus/immunology , Hippocampus/metabolism , Hippocampus/physiopathology , Humans , Hyperglycemia/etiology , Hyperglycemia/immunology , Hyperglycemia/metabolism , Hyperinsulinism/etiology , Hyperinsulinism/immunology , Hyperinsulinism/metabolism , Inflammation/etiology , Inflammation/immunology , Inflammation/metabolism , Obesity, Abdominal/complications , Obesity, Abdominal/immunology , Obesity, Abdominal/metabolism
4.
Radiol Oncol ; 55(1): 7-17, 2021 Jan 12.
Article in English | MEDLINE | ID: mdl-33885236

ABSTRACT

BACKGROUND: Scientific evidence has shown that an increase in regular physical activity is associated with a decrease in the development of many types of cancer. Potential mechanisms that link physical activity to reduced cancer risk include a decrease in systemic inflammation, hyperinsulinemia, insulin-like growth factor (IGF-I), sex hormones, pro-inflammatory leptin and other obesity-related cytokines, and a significant increase in anti-inflammatory adiponectin levels. In addition, physical activity improves immune function and the composition and diversity of the gastrointestinal microbiota. Moderate physical activity is important for cancer protection, but the most significant changes in the inflammatory profile are conferred by physical activity performed at higher intensities. Thus, there is a need for further investigation into the type, intensity, and duration of physical activity for the prevention of some types of cancer and the development of effective recommendations. CONCLUSIONS: There is a strong evidence that physical activity of moderate to vigorous intensity protects against colon and breast cancer, and probably against cancer at all other sites.


Subject(s)
Exercise , Neoplasms/prevention & control , Biomarkers, Tumor/blood , Breast Neoplasms/immunology , Breast Neoplasms/prevention & control , Colonic Neoplasms/immunology , Colonic Neoplasms/prevention & control , Humans , Hyperinsulinism/immunology , Hyperinsulinism/prevention & control , Inflammation/immunology , Inflammation/prevention & control , Neoplasms/immunology , Risk
5.
Nutrients ; 12(12)2020 Dec 17.
Article in English | MEDLINE | ID: mdl-33348685

ABSTRACT

Consumption of red raspberries has been reported to exert acute beneficial effects on postprandial glycemia, insulinemia, triglyceridemia, and cytokine levels in metabolically disturbed subjects. In a two-arm parallel-group, randomized, controlled trial, 59 subjects with overweight or abdominal obesity and with slight hyperinsulinemia or hypertriglyceridemia were randomized to consume 280 g/day of frozen raspberries or to maintain their usual diet for 8 weeks. Primary analyses measured metabolic differences between the groups. Secondary analyses performed with omics tools in the intervention group assessed blood gene expression and plasma metabolomic changes following the raspberry supplementation. The intervention did not significantly affect plasma insulin, glucose, inflammatory marker concentrations, nor blood pressure. Following the supplementation, 43 genes were differentially expressed, and several functional pathways were enriched, a major portion of which were involved in the regulation of cytotoxicity, immune cell trafficking, protein signal transduction, and interleukin production. In addition, 10 serum metabolites were found significantly altered, among which ß-alanine, trimethylamine N-oxide, and bioactive lipids. Although the supplementation had no meaningful metabolic effects, these results highlight the impact of a diet rich in raspberry on the immune function and phospholipid metabolism, thus providing novel insights into potential immune-metabolic pathways influenced by regular raspberry consumption.


Subject(s)
Diet/methods , Hyperinsulinism/complications , Hypertriglyceridemia/complications , Metabolic Syndrome/prevention & control , Overweight/complications , Rubus/immunology , Rubus/metabolism , Adult , Biomarkers/blood , Blood Glucose/drug effects , Blood Glucose/immunology , Cytokines/blood , Cytokines/drug effects , Cytokines/immunology , Female , Fruit/immunology , Fruit/metabolism , Humans , Hyperinsulinism/blood , Hyperinsulinism/immunology , Hypertriglyceridemia/blood , Hypertriglyceridemia/immunology , Insulin/blood , Insulin/immunology , Lipids/blood , Lipids/immunology , Male , Metabolic Syndrome/blood , Metabolic Syndrome/immunology , Middle Aged , Overweight/blood , Overweight/immunology , Young Adult
6.
PLoS One ; 15(9): e0239261, 2020.
Article in English | MEDLINE | ID: mdl-32991593

ABSTRACT

Currently, there are no registered veterinary drugs for the treatment of endocrinopathic equine laminitis, and although this form of the disease is known to be caused by prolonged hyperinsulinaemia, the mechanism of insulin toxicity is unclear. One possibility is that high concentrations of insulin activate IGF-1 receptors (IGF-1R) in lamellar tissue, leading to uncontrolled cell proliferation and epidermal lamellar dysregulation. An equinized version of a human anti-IGF-1R therapeutic monoclonal antibody (mAb11) was generated to test this theory, using a modification of the prolonged euglycaemic-hyperinsulinaemic clamp technique. Healthy Standardbred horses were infused for 48 h with 0.9% saline (negative-control, n = 6), a combination of insulin (4.5 mIU/kgBW/min) and a variable infusion of 50% glucose to maintain euglycaemia (positive-control, n = 6), or insulin and glucose, preceded by a low dose of mAb11 (20 mg), designed to treat one foot only and delivered by retrograde infusion into one forelimb (mAb-treated, n = 7). Maximum insulin concentrations were 502 ± 54.4 and 435 ± 30.4 µIU/mL in the positive-control and mAb11-treated groups, respectively (P = 0.33). While the control group remained healthy, all the insulin-treated horses developed laminitis within 30 h, as judged by clinical examination, foot radiographs and histological analysis. Some effects of insulin were not attenuated by the antibody, however, relative to the positive-control group, horses treated with mAb11 showed less sinking of the distal phalanx (P < 0.05) and milder histological changes, with markedly less elongation at the tips of the secondary epidermal lamellae (P < 0.05). These differences were apparent in both front feet and were statistically significant when the values for both feet were combined. The results confirm that IGF-1R may have a role in insulin-induced laminitis and suggest that mAb11 warrants further research as a potential agent to prevent or treat the disease.


Subject(s)
Horse Diseases/drug therapy , Hyperinsulinism/drug therapy , Insulin/metabolism , Receptor, IGF Type 1/immunology , Animals , Antibodies, Anti-Idiotypic/immunology , Antibodies, Anti-Idiotypic/pharmacology , Antibodies, Monoclonal/pharmacology , Cell Proliferation/drug effects , Epidermis/drug effects , Glucose/metabolism , Horse Diseases/immunology , Horses , Hyperinsulinism/immunology , Hyperinsulinism/pathology , Hyperinsulinism/veterinary , Insulin/immunology , Receptor, IGF Type 1/antagonists & inhibitors
7.
Cancer Epidemiol Biomarkers Prev ; 29(5): 974-981, 2020 05.
Article in English | MEDLINE | ID: mdl-32094199

ABSTRACT

BACKGROUND: Energy balance-related factors, such as body mass index (BMI), diet, and physical activity, may influence colorectal cancer etiology through interconnected metabolic pathways, but their combined influence is less clear. METHODS: We used reduced rank regression to derive three energy balance scores that associate lifestyle factors with combinations of prediagnostic, circulating levels of high-sensitivity C-reactive protein (hsCRP), C-peptide, and hemoglobin A1c (HbA1c) among 2,498 participants in the Cancer Prevention Study-II Nutrition Cohort. Among 114,989 participants, we verified 2,228 colorectal cancer cases. We assessed associations of each score with colorectal cancer incidence and by tumor molecular phenotypes using Cox proportional hazards regression. RESULTS: The derived scores comprised BMI, physical activity, screen time, and 14 food groups, and explained 5.1% to 10.5% of the variation in biomarkers. The HR and 95% confidence interval (CI) for quartile 4 versus 1 of the HbA1c+C peptide-based score and colorectal cancer was 1.30 (1.15-1.47), the hsCRP-based score was 1.35 (1.19-1.53), and the hsCRP, C-peptide, and HbA1c-based score was 1.35 (1.19-1.52). The latter score was associated with non-CIMP tumors (HRQ4vsQ1: 1.59; 95% CI: 1.17-2.16), but not CIMP-positive tumors (P heterogeneity = 0.04). CONCLUSIONS: These results further support hypotheses that systemic biomarkers of metabolic health-inflammation and abnormal glucose homeostasis-mediate part of the relationship between several energy balance-related modifiable factors and colorectal cancer risk. IMPACT: Results support cancer prevention guidelines for maintaining a healthful body weight, consuming a healthful diet, and being physically active. More research is needed on these clusters of exposures with molecular phenotypes of tumors.


Subject(s)
Colorectal Neoplasms/epidemiology , Energy Metabolism/physiology , Hyperglycemia/diagnosis , Hyperinsulinism/diagnosis , Aged , Biomarkers/blood , Body Mass Index , C-Peptide/blood , Colorectal Neoplasms/blood , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Exercise/physiology , Feeding Behavior/physiology , Female , Follow-Up Studies , Glycated Hemoglobin/analysis , Humans , Hyperglycemia/blood , Hyperglycemia/immunology , Hyperglycemia/metabolism , Hyperinsulinism/immunology , Hyperinsulinism/metabolism , Incidence , Inflammation/blood , Inflammation/diagnosis , Inflammation/immunology , Inflammation/metabolism , Male , Middle Aged , Prospective Studies , Receptors, Immunologic/blood , Risk Assessment/methods , Risk Assessment/statistics & numerical data
8.
Mol Med Rep ; 20(4): 3951-3958, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31485663

ABSTRACT

Polygonatum sibiricum polysaccharide (PSP) has been shown to alleviate hyperglycemia and reduce oxidative stress to delay the progression of diabetic retinopathy and cataracts. However, its role and underlying mechanisms in regulating type 2 diabetes mellitus (T2DM) remain unclear. Nuclear factor erythroid 2­related factor 2 (Nrf2) activation plays a protective role in T2DM. The present study focused on the effect of PSP on inflammatory cytokine secretion and Nrf2 expression in the adipocytes of T2DM patients. In this study, high­glucose­ and high­insulin­induced 3T3­L1 adipocytes were used to mimic insulin­resistant (IR)­3T3­L1 adipocytes. Furthermore, the effect and underlying mechanisms of PSP on inflammation and glucose uptake in IR­3T3­L1 adipocytes were investigated. The present study found that proliferation after 50, 100 and 250 µg/ml PSP treatment had no significant change in normal 3T3­L1 adipocytes. A total of 50, 100 and 250 µg/ml of PSP also alleviated IL­1ß, IL­6, and TNF­α levels and promoted proliferation, glucose uptake, and glucose transporter 4 expression in IR­3T3­L1 adipocytes. Furthermore, 50, 100 and 250 µg/ml PSP promoted Nrf2 and HO­1 expression. However, silencing Nrf2 expression reversed the effect of 100 µg/ml PSP in IR­3T3­L1 adipocytes. In conclusion, these results suggest that PSP alleviates inflammatory cytokines and promotes glucose uptake in IR­3T3­L1 adipocytes by promoting Nrf2 expression. PSP may be a potential therapeutic agent for T2DM treatment by promoting Nrf2 expression.


Subject(s)
Adipocytes/drug effects , Anti-Inflammatory Agents/pharmacology , Cytokines/immunology , NF-E2-Related Factor 2/genetics , Polysaccharides/pharmacology , 3T3-L1 Cells , Adipocytes/immunology , Animals , Anti-Inflammatory Agents/chemistry , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/immunology , Humans , Hyperglycemia/drug therapy , Hyperglycemia/immunology , Hyperinsulinism/drug therapy , Hyperinsulinism/immunology , Mice , Polygonatum/chemistry , Polysaccharides/chemistry , Up-Regulation/drug effects
9.
Arch Physiol Biochem ; 124(4): 306-312, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29113509

ABSTRACT

CONTEXT: We have previously shown that an antidepressant-like effect of probiotics in rats was associated with a higher plasma level of the microbial tryptophan metabolite indole-3-propionic acid (IPA). OBJECTIVE: We therefore wanted to study the isolated effect of IPA on behaviour and glucose metabolism in rats. METHODS: Male Sprague-Dawley rats were fed control or IPA-enriched diet for six weeks (n = 12 per group) and assessed in the elevated plus maze, open field and forced swim test. Blood glucose, metabolic hormones and the white blood cell (WBC) composition were analysed. RESULTS: IPA (mean intake 27.3 mg/kg/day) significantly lowered fasting blood glucose level by 0.42 mM (95% CI 0.11-0.73). Similarly, fasting plasma insulin levels and the homeostatic model assessment (HOMA) index of insulin resistance were reduced, whereas plasma metabolic hormones, behaviour and WBC composition remained unaffected by IPA. CONCLUSIONS: Our findings highlight IPA as a promising candidate for treatment of metabolic disorders associated with insulin resistance.


Subject(s)
Dietary Supplements , Hyperinsulinism/prevention & control , Hypoglycemic Agents/therapeutic use , Indoles/therapeutic use , Insulin Resistance , Prediabetic State/prevention & control , Propionates/therapeutic use , Animals , Anxiety/blood , Anxiety/immunology , Anxiety/metabolism , Anxiety/prevention & control , Behavior, Animal , Blood Glucose/analysis , Depression/blood , Depression/immunology , Depression/metabolism , Depression/prevention & control , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/prevention & control , Dietary Supplements/adverse effects , Hyperinsulinism/blood , Hyperinsulinism/immunology , Hyperinsulinism/metabolism , Hypoglycemic Agents/adverse effects , Indoles/adverse effects , Insulin/blood , Leukocyte Count , Male , Prediabetic State/blood , Prediabetic State/immunology , Prediabetic State/metabolism , Propionates/adverse effects , Random Allocation , Rats, Sprague-Dawley
10.
BMC Syst Biol ; 11(1): 64, 2017 Jun 26.
Article in English | MEDLINE | ID: mdl-28651594

ABSTRACT

BACKGROUND: Obesity is frequently linked to insulin resistance, high insulin levels, chronic inflammation, and alterations in the behaviour of CD4+ T cells. Despite the biomedical importance of this condition, the system-level mechanisms that alter CD4+ T cell differentiation and plasticity are not well understood. RESULTS: We model how hyperinsulinemia alters the dynamics of the CD4+ T regulatory network, and this, in turn, modulates cell differentiation and plasticity. Different polarizing microenvironments are simulated under basal and high levels of insulin to assess impacts on cell-fate attainment and robustness in response to transient perturbations. In the presence of high levels of insulin Th1 and Th17 become more stable to transient perturbations, and their basin sizes are augmented, Tr1 cells become less stable or disappear, while TGFß producing cells remain unaltered. Hence, the model provides a dynamic system-level framework and explanation to further understand the documented and apparently paradoxical role of TGFß in both inflammation and regulation of immune responses, as well as the emergence of the adipose Treg phenotype. Furthermore, our simulations provide new predictions on the impact of the microenvironment in the coexistence of the different cell types, suggesting that in pro-Th1, pro-Th2 and pro-Th17 environments effector and regulatory cells can coexist, but that high levels of insulin severely diminish regulatory cells, especially in a pro-Th17 environment. CONCLUSIONS: This work provides a first step towards a system-level formal and dynamic framework to integrate further experimental data in the study of complex inflammatory diseases.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , Hyperinsulinism/immunology , Models, Biological , Acute Disease , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Inflammation/immunology , Insulin/metabolism , Transforming Growth Factor beta/metabolism
11.
Sci Rep ; 7(1): 491, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28352127

ABSTRACT

Hyperinsulinaemia, obesity and dyslipidaemia are independent and collective risk factors for many cancers. Here, the long-term effects of a 23% Western high-fat diet (HFD) in two immunodeficient mouse strains (NOD/SCID and Rag1 -/-) suitable for engraftment with human-derived tissue xenografts, and the effect of diet-induced hyperinsulinaemia on human prostate cancer cell line xenograft growth, were investigated. Rag1 -/-and NOD/SCID HFD-fed mice demonstrated diet-induced impairments in glucose tolerance at 16 and 23 weeks post weaning. Rag1 -/- mice developed significantly higher fasting insulin levels (2.16 ± 1.01 ng/ml, P = 0.01) and increased insulin resistance (6.70 ± 1.68 HOMA-IR, P = 0.01) compared to low-fat chow-fed mice (0.71 ± 0.12 ng/ml and 2.91 ± 0.42 HOMA-IR). This was not observed in the NOD/SCID strain. Hepatic steatosis was more extensive in Rag1 -/- HFD-fed mice compared to NOD/SCID mice. Intramyocellular lipid storage was increased in Rag1 -/- HFD-fed mice, but not in NOD/SCID mice. In Rag1 -/- HFD-fed mice, LNCaP xenograft tumours grew more rapidly compared to low-fat chow-fed mice. This is the first characterisation of the metabolic effects of long-term Western HFD in two mouse strains suitable for xenograft studies. We conclude that Rag1 -/- mice are an appropriate and novel xenograft model for studying the relationship between cancer and hyperinsulinaemia.


Subject(s)
Disease Models, Animal , Disease Susceptibility , Hyperinsulinism/etiology , Hyperinsulinism/metabolism , Adipose Tissue/metabolism , Animals , Blood Glucose , Body Weight , Diet, High-Fat , Female , Heterografts , Homeodomain Proteins/genetics , Humans , Hyperinsulinism/immunology , Insulin/blood , Insulin/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Muscle, Skeletal/metabolism , Organ Specificity , Pancreas/metabolism
12.
Eur J Nutr ; 56(2): 705-713, 2017 Mar.
Article in English | MEDLINE | ID: mdl-26621632

ABSTRACT

PURPOSE: Obesity is usually associated with low-grade inflammation, which impairs insulin action. The amino acid, taurine (TAU), regulates glucose homeostasis and lipid metabolism and presents anti-inflammatory actions. Here, we evaluated whether inflammatory markers are altered in the serum and retroperitoneal adipose tissue of monosodium glutamate (MSG) obese rats, supplemented or not with TAU. METHODS: Male Wistar rats received subcutaneous injections of MSG (4 mg/kg body weight/day, MSG group) or hypertonic saline (CTL) during the first 5 days of life. From 21 to 120 days of age, half of each of the MSG and CTL groups received 2.5 % TAU in their drinking water (CTAU and MTAU). RESULTS: At 120 days of age, MSG rats were obese and hyperinsulinemic. TAU supplementation reduced fat deposition without affecting insulinemia in MTAU rats. MSG rats presented increased pIκ-Bα/Iκ-Bα protein expression in the retroperitoneal adipose tissue. TAU supplementation decreased the ratio of pIκ-Bα/Iκ-Bα protein, possibly contributing to the increased Iκ-Bα content in MTAU adipose tissue. Furthermore, MSG obesity or supplementation did not alter TNF-α, IL-1ß or IL-6 content in adipose tissue. In contrast, MSG rats presented lower serum TNF-α, IL-4 and IL-10 concentrations, and these alterations were prevented by TAU treatment. CONCLUSION: MSG obesity in rats was not associated with alterations in pro-inflammatory markers in retroperitoneal fat stores; however, reductions in the serum concentrations of anti-inflammatory cytokines and of TNF-α were observed. TAU treatment decreased adiposity, and this effect was associated with the normalization of circulating TNF-α and IL-4 concentrations in MTAU rats.


Subject(s)
Anti-Obesity Agents/therapeutic use , Dietary Supplements , Gene Expression Regulation , Intra-Abdominal Fat/metabolism , NF-KappaB Inhibitor alpha/metabolism , Obesity/diet therapy , Taurine/therapeutic use , Adiposity , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Biomarkers/blood , Biomarkers/metabolism , Hyperinsulinism/diet therapy , Hyperinsulinism/etiology , Hyperinsulinism/immunology , Hyperinsulinism/metabolism , I-kappa B Proteins/agonists , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Injections, Subcutaneous , Interleukin-4/antagonists & inhibitors , Interleukin-4/blood , Interleukin-4/metabolism , Intra-Abdominal Fat/immunology , Male , NF-KappaB Inhibitor alpha/agonists , NF-KappaB Inhibitor alpha/genetics , Obesity/etiology , Obesity/immunology , Obesity/metabolism , Phosphorylation , Protein Processing, Post-Translational , Rats, Wistar , Sodium Glutamate/administration & dosage , Sodium Glutamate/adverse effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/metabolism
13.
Toxicol Lett ; 258: 46-54, 2016 Sep 06.
Article in English | MEDLINE | ID: mdl-27220267

ABSTRACT

This study tested whether the maternal transport of dexamethasone (DEXA) may affect the development of the neuroendocrine system. DEXA (0.2mg/kg b.w., subcutaneous injection) was administered to pregnant rats from gestation day (GD) 1-20. In the DEXA-treated group, a decrease in maternal serum thyroxine (T4), triiodothyronine (T3), and increase in thyrotropin (TSH) levels (hypothyroid status) were observed at GDs 15 & 20 with respect to control group. The reverse pattern (hyperthyroid status) was observed in their fetuses at embryonic days (EDs) 15 & 20. Although the maternal body weight was diminished, the weight of the thyroid gland was increased at studied GDs as compared to the control group. The fetal growth retardation, hyperleptinemia, hyperinsulinism, and cytokines distortions (transforming growth factor-beta; TGF-ß, tumor necrosis factor-alpha; TNF-α, and interferon-γ; IFN-γ) were noticed at examined EDs if compared to the control group. Alternatively, the maternofetal thyroid dysfunctions due to the maternal DEXA administration attenuated the levels of fetal cerebral norepinephrine (NE) and epinephrine (E), and elevated the levels of dopamine (DA) and 5-hydroxytryptamine (5-HT) at considered days. These alterations were age-dependent and might damage the nerve transmission. Finally, maternal DEXA might act as neuroendocrine disruptor causing dyshormonogenesis and fetal cerebral dysfunction.


Subject(s)
Anti-Inflammatory Agents/adverse effects , Dexamethasone/adverse effects , Endocrine Disruptors/adverse effects , Fetal Development/drug effects , Hypothyroidism/chemically induced , Maternal-Fetal Exchange , Neurosecretory Systems/drug effects , Animals , Anti-Inflammatory Agents/administration & dosage , Cytokines/agonists , Cytokines/blood , Dexamethasone/administration & dosage , Endocrine Disruptors/administration & dosage , Female , Fetal Growth Retardation/blood , Fetal Growth Retardation/chemically induced , Fetal Growth Retardation/immunology , Fetal Growth Retardation/pathology , Hyperinsulinism/chemically induced , Hyperinsulinism/embryology , Hyperinsulinism/immunology , Hyperinsulinism/pathology , Hypothyroidism/embryology , Hypothyroidism/immunology , Hypothyroidism/pathology , Injections, Subcutaneous , Leptin/agonists , Leptin/blood , Neurosecretory Systems/embryology , Organ Size/drug effects , Pregnancy , Rats, Wistar , Thyroid Gland/drug effects , Thyroid Gland/embryology , Thyroid Gland/immunology , Thyroid Gland/pathology , Thyrotropin/blood , Thyroxine/blood , Triiodothyronine/blood , Weight Gain/drug effects
14.
Intern Med ; 55(9): 1125-9, 2016.
Article in English | MEDLINE | ID: mdl-27150866

ABSTRACT

The older of a pair of sisters experienced hypoglycemia after the start of thiamazole (MMI) treatment. Based on a high insulin antibody level, she was diagnosed with insulin autoimmune syndrome (IAS). HLA-DNA typing identified DRB1*04:06. Although a 75-g oral glucose tolerance test (OGTT) showed biphasic insulin secretion, the secretion pattern became monophasic after discontinuation of the MMI. The younger sister was diagnosed with IAS after the start of MMI treatment. HLA-DNA typing identified DRB1*04:06. The 75-g OGTT showed biphasic insulin secretion, but it became monophasic after discontinuation of the MMI. According to the similar insulin secretion kinetics in the two sisters with IAS, we suspect that a genetic predisposition may be associated with the features of anti-insulin antibodies.


Subject(s)
Antithyroid Agents/adverse effects , Autoimmune Diseases/chemically induced , Graves Disease/drug therapy , Insulin Antibodies/blood , Methimazole/adverse effects , Adult , Antithyroid Agents/therapeutic use , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Female , Genetic Predisposition to Disease , Glucose Tolerance Test , Graves Disease/complications , Graves Disease/genetics , HLA-DRB1 Chains/genetics , Humans , Hyperinsulinism/chemically induced , Hyperinsulinism/genetics , Hyperinsulinism/immunology , Hypoglycemia/chemically induced , Insulin/metabolism , Insulin Secretion , Methimazole/therapeutic use , Siblings
15.
Thromb Haemost ; 113(4): 750-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25653143

ABSTRACT

Diabetes mellitus (DM) patients have an increased incidence of cardiovascular events. Blood tissue factor-procoagulant activity (TF-PCA), the initiating mechanism for blood coagulation, is elevated in DM. We have shown that hyperglycaemia (HG), hyperinsulinaemia (HI) and combined HG+HI (induced using 24-hour infusion clamps) increases TF-PCA in healthy and type 2 DM (T2DM) subjects, but not in type 1 DM (T1DM) subjects. The mechanisms for this are unknown. DM patients have elevated plasma lipopolysaccharide (LPS), a toll-like receptor (TLR) 4 ligand. We postulated that TLR4 plays a role in modulating TF levels. We studied the effect of HG+HI on TLR4 and TF-PCA in vivo during 24-hour HG+HI infusion clamps in healthy subjects, and T1DM and T2DM subjects, and in vitro in blood. In vivo, in healthy subjects, 24-hour HG + HI infusion increased TLR4 six-fold, which correlated with TF-PCA (r= 0.91, p<0.0001). T2DM patients showed smaller increases in both. In T1DM subjects, TLR4 declined (50%, p<0.05) and correlated with TF-PCA (r=0.55; p<0.05). In vitro, HG (200 mg/dl added glucose) and HI (1-100 nM added insulin) increased TF-PCA in healthy subjects (~2-fold, 2-4 hours). Insulin inhibited by ~30% LPS-induced increase in TF-PCA and high glucose reversed it. TLR4 levels paralleled TF-PCA (r=0.71, p<0.0001); HG and HI increased TLR4 and insulin inhibited LPS-induced TLR4 increase. This is first evidence that even in healthy subjects, HG of short duration increases TLR4 and TF-PCA, key players in inflammation and thrombosis. TLR4-TF interplay is strikingly different in non-diabetic, T1DM and T2DM subjects.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 2/blood , Hyperglycemia/blood , Hyperinsulinism/blood , Insulin/blood , Thromboplastin/metabolism , Toll-Like Receptor 4/blood , Adult , Biomarkers/blood , Blood Coagulation , Blood Coagulation Tests , Case-Control Studies , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/immunology , Female , Glucose Clamp Technique , Humans , Hyperglycemia/diagnosis , Hyperglycemia/immunology , Hyperinsulinism/diagnosis , Hyperinsulinism/immunology , Inflammation/blood , Inflammation/immunology , Lipopolysaccharides/pharmacology , Male , Middle Aged , Thrombosis/blood , Thrombosis/immunology , Time Factors , Toll-Like Receptor 4/agonists
17.
Int J Obes (Lond) ; 39(2): 270-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24854430

ABSTRACT

BACKGROUND: Obesity is a severe health problem worldwide, which leads to multiple comorbidities including type 2 diabetes mellitus and cardiovascular diseases. Inflammation has been found to be an important characteristic of adipose tissue in obese subjects. However, obesity is also associated with compromised immune responses to infections and the impact of obesity on immune function has not been fully understood. SUBJECTS/METHODS: To clarify the role of obesity in the immune responses, we investigated the Toll-like receptor (TLR)-induced cytokine secretion by leukocytes from obese and lean subjects. We also investigated the relationship between insulin-induced intracellular signaling and cytokine production using peripheral blood mononuclear cells (PBMCs) and a monocytic cell line THP-1. RESULTS: We found decreased TLR-induced interferon-γ, interleukin-6 (IL-6) and tumor necrosis factor-α secretions and elevated IL-10 secretion by leukocytes from obese subjects when compared with lean controls. PBMCs from obese subjects showed enhanced basal Akt and glycogen synthase kinase-3ß (GSK-3ß) phosphorylation, which did not further increase with insulin and lipopolysaccharide (LPS) stimulation. We also found that LPS-induced IκBα degradation was inhibited in PBMCs from obese subjects. By using THP-1 cells with GSK-3ß knockdown or cells treated with hyperinsulinemic and high-fatty acid conditions, we found that LPS-induced nuclear factor κB (NF-κB) activation was inhibited and cyclic adenosine monophosphate response element-binding protein (CREB) activation was enhanced. CONCLUSIONS: These findings indicate that GSK-3ß is important in the regulation of NF-κB and CREB activation in leukocytes under the metabolic condition of obesity. Our study revealed a key mechanism through which metabolic abnormalities compromise leukocyte functions in people with obesity.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Hyperinsulinism/metabolism , Hyperlipidemias/metabolism , Interleukin-10/metabolism , NF-kappa B/antagonists & inhibitors , Obesity/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Hyperinsulinism/immunology , Hyperlipidemias/immunology , I-kappa B Proteins/metabolism , Inflammation/immunology , Leukocytes, Mononuclear/metabolism , NF-KappaB Inhibitor alpha , Obesity/immunology , Phosphorylation , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
18.
J Immunol ; 192(2): 623-9, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24323581

ABSTRACT

Chronic inflammation is known to promote metabolic dysregulation in obesity and type 2 diabetes. Although the precise origin of the unchecked inflammatory response in obesity is unclear, it is known that overproduction of proinflammatory cytokines by innate immune cells affects metabolism. For example, TNF-α contributes to the inability of cells to respond to insulin and to the increase in levels of insulin. Whether this hyperinsulinemia itself is part of a feedback loop that affects the progression of chronic adipose inflammation is unknown. In this article, we show that regulatory T cells (Tregs) express the insulin receptor, and that high levels of insulin impair the ability of Tregs to suppress inflammatory responses via effects on the AKT/mTOR signaling pathway. Insulin activated AKT signaling in Tregs, leading to inhibition of both IL-10 production and the ability of Tregs to suppress the production of TNF-α by macrophages in a contact-independent manner. The effect of insulin on Treg suppression was limited to IL-10 production and it did not alter the expression of other proteins associated with Treg function, including CTLA-4, CD39, and TGF-ß. In a model of diet-induced obesity, Tregs from the visceral adipose tissue of hyperinsulinemic, obese mice showed a similar specific decrease in IL-10 production, as well as a parallel increase in production of IFN-γ. These data suggest that hyperinsulinemia may contribute to the development of obesity-associated inflammation via a previously unknown effect of insulin on the IL-10-mediated function of Tregs.


Subject(s)
Insulin/immunology , Insulin/metabolism , Interleukin-10/immunology , Interleukin-10/metabolism , Obesity/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Apyrase/immunology , Apyrase/metabolism , CTLA-4 Antigen/immunology , CTLA-4 Antigen/metabolism , Cells, Cultured , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelium/immunology , Epithelium/metabolism , Hyperinsulinism/immunology , Hyperinsulinism/metabolism , Inflammation/immunology , Inflammation/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-10/antagonists & inhibitors , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Obesity/metabolism , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/immunology , Receptor, Insulin/metabolism , Signal Transduction/immunology , TOR Serine-Threonine Kinases/immunology , TOR Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
19.
Vet Immunol Immunopathol ; 157(1-2): 78-86, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24246153

ABSTRACT

Equine laminitis, a disease of the lamellar structure of the horse's hoof, can be incited by numerous factors that include inflammatory and metabolic aetiologies. However, the role of inflammation in hyperinsulinaemic laminitis has not been adequately defined. Toll-like receptor (TLR) activation results in up-regulation of inflammatory pathways and the release of pro-inflammatory cytokines, including interleukin-6 (IL-6) and tumour necrosis factor-alpha (TNF-α), and may be a pathogenic factor in laminitis. The aim of this study was to determine whether TLR4 expression and subsequent pro-inflammatory cytokine production is increased in lamellae and skeletal muscle during equine hyperinsulinaemia. Standardbred horses were treated with either a prolonged, euglycaemic hyperinsulinaemic clamp (p-EHC) or a prolonged, glucose infusion (p-GI), which induced marked and moderate hyperinsulinaemia, respectively. Age-matched control horses were treated simultaneously with a balanced electrolyte solution. Treated horses developed clinical (p-EHC) or subclinical (p-GI) laminitis, whereas controls did not. Skeletal muscle and lamellar protein extracts were analysed by Western blotting for TLR4, IL-6, TNF-α and suppressor of cytokine signalling 3 (SOCS3) expression. Lamellar protein expression of TLR4 and TNF-α, but not IL-6, was increased by the p-EHC, compared to control horses. A significant positive correlation was found between lamellar TLR4 and SOCS3. Skeletal muscle protein expression of TLR4 signalling parameters did not differ between control and p-EHC-treated horses. Similarly, the p-GI did not result in up-regulation of lamellar protein expression of any parameter. The results suggest that insulin-sensitive tissues may not accurately reflect lamellar pathology during hyperinsulinaemia. While TLR4 is present in the lamellae, its activation appears unlikely to contribute significantly to the developmental pathogenesis of hyperinsulinaemic laminitis. However, inflammation may have a role to play in the later stages (e.g., repair or remodelling) of the disease.


Subject(s)
Foot Diseases/veterinary , Gene Expression Regulation/immunology , Hoof and Claw/immunology , Horse Diseases/immunology , Hyperinsulinism/veterinary , Toll-Like Receptor 4/immunology , Animals , Blotting, Western/veterinary , Cytokines/analysis , Cytokines/immunology , Foot Diseases/etiology , Foot Diseases/genetics , Foot Diseases/immunology , Horse Diseases/etiology , Horse Diseases/genetics , Horses , Hyperinsulinism/immunology , Muscle, Skeletal/immunology , Random Allocation , Statistics, Nonparametric , Toll-Like Receptor 4/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...