Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Mol Sci ; 25(5)2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38474262

ABSTRACT

Detrimental molecular processes in multiple sclerosis (MS) lead to the cellular accumulation of lipid peroxidation products and iron in the CNS, which represents the main driving force for ferroptosis. Ferroptosis is an iron-dependent form of regulated cell death, with proposed roles in neurodegeneration, oligodendrocyte loss and neuroinflammation in the pathogenesis of MS. Ferroptosis-related gene expression signature and molecular markers, which could reflect MS severity and progression, are currently understudied in humans. To tackle these challenges, we have applied a curated approach to create and experimentally analyze a comprehensive panel of ferroptosis-related genes covering a wide range of biological processes associated with ferroptosis. We performed the first ferroptosis-related targeted RNAseq on PBMCs from highly distinctive MS phenotype groups: mild relapsing-remitting (RR) (n = 24) and severe secondary progressive (SP) (n = 24), along with protein detection of GPX4 and products of lipid peroxidation (MDA and 4-HNE). Out of 138 genes, 26 were differentially expressed genes (DEGs), indicating changes in both pro- and anti-ferroptotic genes, representing a molecular signature associated with MS severity. The top three DEGs, as non-core ferroptosis genes, CDKN1A, MAP1B and EGLN2, were replicated by qPCR to validate findings in independent patient groups (16 RR and 16 SP MS). Co-expression and interactions of DEGs were presented as additional valuable assets for deeper understanding of molecular mechanisms and key targets related to MS severity. Our study integrates a wide genetic signature and biochemical markers related to ferroptosis in easily obtainable PBMCs of MS patients with clinical data and disease severity, thus providing novel molecular markers which can complement disease-related changes in the brain and undergo further research as potential therapeutic targets.


Subject(s)
Ferroptosis , Multiple Sclerosis , Humans , Transcriptome , Neoplasm Recurrence, Local , Patient Acuity , Iron , Hypoxia-Inducible Factor-Proline Dioxygenases
2.
Int J Mol Sci ; 25(12)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38928475

ABSTRACT

Increased fructose consumption and chronic stress, the major characteristics of modern lifestyle, impact human health; however, the consequences of their combination on the uterus remain understudied. In this study, we investigated contractile activity, morphology, and intracellular activity of antioxidant enzymes in uteri from virgin Wistar rats subjected to liquid fructose supplementation and/or unpredictable stress over 9 weeks. Contractile activity and uterine response to oxytocin or adrenaline were examined ex vivo using isolated bath chambers. Fructose supplementation, irrespective of stress, affected uterine morphology by increasing endometrium while decreasing myometrium volume density, attenuated uterine response to increasing doses of oxytocin, and increased glutathione peroxidase activity. Stress, irrespective of fructose, attenuated dose-dependent adrenaline-induced uterine relaxation. Stress, when applied solely, decreased mitochondrial superoxide dismutase activity. In the combined treatment, irregular estrous cycles and both reduced response to oxytocin and to adrenaline (as a consequence of fructose consumption and exposure to stress), along with fructose-related alteration of uterine morphology, were detected. In conclusion, fructose and stress affect uterine contractile activity, irrespective of each other, by inducing completely distinct responses in isolated uteri. In the combined treatment, the effects of both factors were evident, suggesting that the combination exerts more detrimental effects on the uterus than each factor individually.


Subject(s)
Fructose , Oxytocin , Rats, Wistar , Uterine Contraction , Uterus , Animals , Female , Fructose/adverse effects , Fructose/pharmacology , Rats , Uterine Contraction/drug effects , Oxytocin/pharmacology , Oxytocin/metabolism , Uterus/drug effects , Uterus/metabolism , Epinephrine/pharmacology , Stress, Physiological/drug effects , Stress, Psychological , Superoxide Dismutase/metabolism , Dietary Supplements , Myometrium/drug effects , Myometrium/metabolism , Antioxidants/pharmacology , Antioxidants/metabolism
3.
Int J Mol Sci ; 25(6)2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38542507

ABSTRACT

Prostate-specific membrane antigen (PSMA) and caveolin-1 are membrane proteins that are overexpressed in prostate cancer (PCa) and are involved in tumor growth and increase in aggressiveness. The aim of the present study is therefore to evaluate PSMA and caveolin-1 proteins from plasma exosomes as effective liquid biopsy biomarkers for PCa. This study included 39 patients with PCa and 33 with benign prostatic hyperplasia (BPH). The shape and size of the exosomes were confirmed by transmission electron microscopy (TEM) and scanning electron microscopy (SEM) analysis. Immunogold analysis showed that PSMA is localized to the membrane of exosomes isolated from the plasma of both groups of participants. The relative protein levels of PSMA and caveolin-1 in the plasma exosomes of PCa and BPH patients were determined by Western blot analysis. The relative level of the analyzed plasma exosomal proteins was compared between PCa and BPH patients and the relevance of the exosomal PSMA and caveoin-1 level to the clinicopathological parameters in PCa was investigated. The analysis performed showed an enrichment of exosomal PSMA in the plasma of PCa patients compared to the exosomes of men with BPH. The level of exosomal caveolin-1 in plasma was significantly higher in PCa patients with high PSA levels, clinical-stage T3 or T4 and in the group of PCa patients with aggressive PCa compared to favorable clinicopathological features or tumor aggressiveness. Plasma exosomes may serve as a suitable object for the identification of potential biomarkers for the early diagnosis and prognosis of PCa as well as carriers of therapeutic agents in precision medicine of PCa treatment.


Subject(s)
Prostatic Hyperplasia , Prostatic Neoplasms , Male , Humans , Prostatic Hyperplasia/metabolism , Prostate/pathology , Caveolin 1/metabolism , Serbia , Biomarkers, Tumor/metabolism , Prostatic Neoplasms/metabolism , Prostate-Specific Antigen/metabolism
4.
Horm Behav ; 153: 105392, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37295324

ABSTRACT

Polycystic ovary syndrome (PCOS) is a complex disorder characterized by endocrine and metabolic abnormalities such as obesity and insulin resistance. PCOS is also associated with psychiatric disorders and cognitive impairment. The animal model of PCOS was induced by treating rats with 5α-dihydrotestosterone (5α-DHT) and additionally modified to induce adiposity by litter size reduction (LSR). Spatial learning and memory were assessed using the Barnes Maze test, and striatal markers of synaptic plasticity were analyzed. Striatal insulin signaling was estimated by the levels of insulin receptor substrate 1 (IRS1), its inhibitory phosphorylation at Ser307, and glycogen synthase kinase-3α/ß (GSK3α/ß) activity. Both LSR and DHT treatment significantly decreased striatal protein levels of IRS1, followed by increased GSK3α/ß activity in small litters. Results of the behavioral study showed that LSR had a negative effect on learning rate and memory retention, whereas DHT treatment did not induce impairment in memory formation. While protein levels of synaptophysin, GAP43, and postsynaptic density protein 95 (PSD-95) were not altered by the treatments, DHT treatment induced an increase in phosphorylation of PSD-95 at Ser295 in both normal and small litters. This study revealed that LSR and DHT treatment suppressed insulin signaling by downregulating IRS1 in the striatum. However, DHT treatment did not have an adverse effect on learning and memory, probably due to compensatory elevation in pPSD-95-Ser295, which had a positive effect on synaptic strength. This implies that hyperandrogenemia in this setting does not represent a threat to spatial learning and memory, opposite to the effect of overnutrition-related adiposity.


Subject(s)
Hyperandrogenism , Insulin Resistance , Polycystic Ovary Syndrome , Female , Humans , Rats , Animals , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/metabolism , Hyperandrogenism/complications , Hyperandrogenism/metabolism , Spatial Learning , Insulin Resistance/physiology , Insulin/metabolism , Dihydrotestosterone/pharmacology , Obesity/complications , Disease Models, Animal
5.
Int J Mol Sci ; 24(9)2023 May 08.
Article in English | MEDLINE | ID: mdl-37176160

ABSTRACT

Obesity is on the rise worldwide, and consequently, obesity-related non-communicable diseases are as well. Nutritional overload induces metabolic adaptations in an attempt to restore the disturbed balance, and the byproducts of the mechanisms at hand include an increased generation of reactive species. Obesity-related oxidative stress causes damage to vulnerable systems and ultimately contributes to neoplastic transformation. Dysfunctional obese adipose tissue releases cytokines and induces changes in the cell microenvironment, promoting cell survival and progression of the transformed cancer cells. Other than the increased risk of cancer development, obese cancer patients experience higher mortality rates and reduced therapy efficiency as well. The fact that obesity is considered the second leading preventable cause of cancer prioritizes the research on the mechanisms connecting obesity to cancerogenesis and finding the solutions to break the link. Oxidative stress is integral at different stages of cancer development and advancement in obese patients. Hypocaloric, balanced nutrition, and structured physical activity are some tools for relieving this burden. However, the sensitivity of simultaneously treating cancer and obesity poses a challenge. Further research on the obesity-cancer liaison would offer new perspectives on prevention programs and treatment development.


Subject(s)
Neoplasms , Obesity , Humans , Obesity/metabolism , Oxidative Stress , Adipose Tissue/metabolism , Neoplasms/etiology , Neoplasms/metabolism , Cytokines/metabolism , Tumor Microenvironment
6.
J Stroke Cerebrovasc Dis ; 31(1): 106212, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34814004

ABSTRACT

OBJECTIVES: Galectin-3 affects a variety of biological processes. It is encoded by LGALS-3, located in unique haplotype block in Caucasians. Most of the studies regarding the gal-3 role in atherosclerosis are focused exclusively on protein/mRNA levels. Genetic analyses of LGALS-3 are scarce. We sought to thoroughly examine the genetic background of gal-3 and to analyze tag variants that cover more than 80% variability of the LGALS-3 containing hap-block in association with carotid plaque presence (CPP). According to Tagger server, rs4040064 G/T, rs11628437 G/A and rs7159490 C/T cover 82% (r2 > 0.8) of the genetic variance of this hap-block. Our aims were to investigate possible association of rs4040064, rs11628437 and rs7159490 haplotypes with CPP in patients with advanced carotid atherosclerosis (CA) and to analyze their possible effect on LGALS-3 mRNA expression in carotid plaques. MATERIALS AND METHODS: Study group consisted of 468 patients and 296 controls. Rs4040064, rs11628437, rs7159490 and LGALS-3 mRNA expression were detected by TaqMan® technology. RESULTS: We have found that haplotype TAC was associated with the cerebrovascular insult (CVI) occurrence (OR = 1.68, 95% CI = 1.09-2.58, p = 0.02), compared to the referent haplotype. OR was adjusted for hypertension, age and BMI. TAC also showed higher, but not statistically significant, LGALS-3 expression in carotid plaques. CONCLUSIONS: Our results suggest that rs4040064, rs11628437 and rs7159490 bear no association with CPP, neither they affect LGALS-3 mRNA in carotid plaques. However, we showed a significant association of haplotype TAC with the CVI occurrence in CA patients from Serbia. Replication and validation of our results are required.


Subject(s)
Carotid Arteries/diagnostic imaging , Carotid Artery Diseases/genetics , Galectin 3/genetics , Plaque, Atherosclerotic/genetics , RNA, Messenger/metabolism , Adult , Aged , Aged, 80 and over , Blood Proteins , Carotid Artery Diseases/diagnostic imaging , Case-Control Studies , Female , Galectins , Genetic Predisposition to Disease , Haplotypes , Humans , Male , Middle Aged , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Serbia
7.
Int J Mol Sci ; 23(16)2022 Aug 11.
Article in English | MEDLINE | ID: mdl-36012206

ABSTRACT

Polycystic ovary syndrome (PCOS) is a well-known reproductive syndrome usually associated with obesity, insulin resistance, and hyperinsulinemia. Although the first signs of PCOS begin early in adolescence, it is underexplored whether peripubertal obesity predisposes women to PCOS metabolic disturbances. To highlight that, we examined the impact of postnatal overfeeding-induced obesity, achieved by litter size reduction during the suckling period, on metabolic disturbances associated with visceral and subcutaneous adipose tissue (VAT and SAT) function in the 5α-dihydrotestosterone (5α-DHT)-induced animal model of PCOS. We analyzed markers of insulin signaling, lipid metabolism, and energy sensing in the VAT and SAT. Our results showed that postnatally overfed DHT-treated Wistar rats had increased VAT mass with hypertrophic adipocytes, together with hyperinsulinemia and increased HOMA index. In the VAT of these animals, insulin signaling remained unchanged while lipogenic markers decreased, which was accompanied by increased AMPK activation. In the SAT of the same animals, markers of lipogenesis and lipolysis increased, while the activity of AMPK decreased. Taken together, obtained results showed that postnatal overfeeding predisposes development of PCOS systemic insulin resistance, most likely as a result of worsened metabolic function of SAT, while VAT preserved its tissue insulin sensitivity through increased activity of AMPK.


Subject(s)
Insulin Resistance , Polycystic Ovary Syndrome , AMP-Activated Protein Kinases/metabolism , Adipose Tissue/metabolism , Animals , Female , Humans , Insulin/metabolism , Insulin Resistance/physiology , Obesity/metabolism , Polycystic Ovary Syndrome/metabolism , Rats , Rats, Wistar , Subcutaneous Fat/metabolism
8.
Int J Mol Sci ; 23(16)2022 Aug 12.
Article in English | MEDLINE | ID: mdl-36012287

ABSTRACT

The effects of a fructose-rich diet and chronic stress on copper metabolism in the kidneys are still understudied. We investigated whether fructose and/or chronic unpredictable stress modulate copper metabolism in a way that affects redox homeostasis, thus contributing to progression of metabolic disturbances in the kidney. We determined protein level of copper transporters, chaperones, and cuproenzymes including cytochrome c oxidase, as well as antioxidant enzymes function in the kidneys of male Wistar rats subjected to 20% liquid fructose supplementation and/or chronic stress. Liquid fructose supplementation increased level of copper chaperone of superoxide dismutase and decreased metallothionein level, while rendering the level of copper importer and copper chaperones involved in copper delivery to mitochondria and trans Golgi network unaffected. Stress had no effect on renal copper metabolism. The activity and expression of renal antioxidant enzymes remained unaltered in all experimental groups. In conclusion, fructose, independently of stress, decreased renal copper level, and modulated renal copper metabolism as to preserve vital cellular function including mitochondrial energy production and antioxidative defense, at the expense of intracellular copper storage.


Subject(s)
Antioxidants , Fructose , Animals , Antioxidants/pharmacology , Copper/pharmacology , Fructose/metabolism , Male , Oxidative Stress , Rats , Rats, Wistar
9.
J Nutr ; 151(12): 3661-3670, 2021 12 03.
Article in English | MEDLINE | ID: mdl-34510217

ABSTRACT

BACKGROUND: Both fructose consumption and chronic stress contribute to the development of metabolic disorders. The consequences of such combination are not fully understood. OBJECTIVE: We investigated whether fructose supplementation and chronic stress synergistically disturb hepatic lipid and glucose metabolism. The role of energy sensing, redox, and inflammatory status during development of metabolic disturbances was investigated. METHODS: Female Wistar rats, aged 2.5 mo, were divided into 4 experimental groups: control (C) fed a standard diet (commercial food and drinking water); fructose (F) fed the same food and 10% fructose solution; stress (S) fed the standard diet and subjected to chronic unpredictable stress and, stress + fructose (SF) combining conditions F and S as above. Stress included daily stressors: cold water forced swimming, physical restraint, cold room, wet bedding, rocking, switching, or tilting cages. After 9 wk, hepatic enzymes and transcription factors involved in gluconeogenesis, lipogenesis, fatty acid oxidation, antioxidative defence, energy sensing, and cytokines were assessed by qPCR, Western blotting, and spectrophotometry and analyzed by 2-factor ANOVA. RESULTS: Fructose increased AMP-activated protein kinase (AMPK) phosphorylation (40%; P < 0.05) and the ratio of inhibitory phosphorylation to total acetyl-CoA carboxylase (46%; P < 0.01), and decreased sterol regulatory element binding protein 1c nuclear translocation by 30% (P < 0.05) in F and SF compared with C rats. Increased phosPck (phoenolpyruvate carboxykinase) (85%) and G6pase(glucose-6-phosphatase) (55%) was observed in S rats (P < 0.05). A 40% decrease in Apob (apolipoprotein B-100) and an increase in hepatic lipids (P < 0.05), together with a double increase in TNF-α (P < 0.001), were observed in S rats, but without liver histopathological changes. These stress effects on lipid accumulation and TNF-α were abolished in SF rats (P < 0.05). CONCLUSIONS: Fructose does not enhance stress effects on hepatic lipid and glucose metabolism but attenuates its effects on hepatic lipid accumulation and inflammation, suggesting that, in female rats, AMPK activation prevails over stress-induced effects.


Subject(s)
Fructose , Liver , Animals , Diet , Female , Fructose/adverse effects , Fructose/metabolism , Lipogenesis , Liver/metabolism , Rats , Rats, Wistar
10.
Int J Mol Sci ; 22(13)2021 Jul 04.
Article in English | MEDLINE | ID: mdl-34281257

ABSTRACT

The modern lifestyle brings both excessive fructose consumption and daily exposure to stress which could lead to metabolic disturbances and type 2 diabetes. Muscles are important points of glucose and lipid metabolism, with a crucial role in the maintenance of systemic energy homeostasis. We investigated whether 9-week fructose-enriched diet, with and without exposure to 4-week unpredictable stress, disturbs insulin signaling in the skeletal muscle of male rats and evaluated potential contributory roles of muscle lipid metabolism, glucocorticoid signaling and inflammation. The combination of fructose-enriched diet and stress increased peroxisome proliferator-activated receptors-α and -δ and stimulated lipid uptake, lipolysis and ß-oxidation in the muscle of fructose-fed stressed rats. Combination of treatment also decreased systemic insulin sensitivity judged by lower R-QUICKI, and lowered muscle protein content and stimulatory phosphorylations of insulin receptor supstrate-1 and Akt, as well as the level of 11ß-hydroxysteroid dehydrogenase type 1 and glucocorticoid receptor. At the same time, increased levels of protein tyrosine phosphatase-1B, nuclear factor-κB, tumor necrosis factor-α, were observed in the muscle of fructose-fed stressed rats. Based on these results, we propose that decreased glucocorticoid signaling in the skeletal muscle can make a setting for lipid-induced inflammation and the development of insulin resistance in fructose-fed stressed rats.


Subject(s)
Fructose/administration & dosage , Glucocorticoids/metabolism , Inflammation/metabolism , Lipid Metabolism , Muscle, Skeletal/metabolism , Stress, Physiological/physiology , Animals , Fructose/adverse effects , Humans , Inflammation/etiology , Insulin Resistance/physiology , Male , Models, Biological , Rats , Rats, Wistar , Receptors, Glucocorticoid/metabolism , Signal Transduction
11.
Int J Food Sci Nutr ; 71(7): 815-825, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32070154

ABSTRACT

Overconsumption of fructose-enriched beverages and everyday stress are involved in the pathogenesis of metabolic disorders through modulation of hepatic glucose metabolism. The aim of the study was to investigate whether interaction of high-fructose diet and chronic stress alter insulin and glucocorticoid signalling thus affecting hepatic glucose homeostasis. High-fructose diet led to hyperinsulinemia, increased glucose transporter 2 level, elevated protein kinase B (Akt) phosphorylation, increased glucokinase mRNA and phospho-to-total glycogen synthase kinase 3 ratio and decreased expression of gluconeogenic genes. Fructose diet also led to stimulated glucocorticoid prereceptor metabolism, but downstream signalling remained unchanged due to increased glucocorticoid clearance. Stress did not affect hepatic insulin and glucocorticoid signalling nor glucose metabolism, while the interaction of the factors was observed only for glucokinase expression. The results suggest that, under conditions of fructose-induced hyperinsulinemia, suppression of gluconeogenesis and glycogen synthase activation contribute to the maintenance of glucose homeostasis. The increased glucocorticoid inactivation may represent an adaptive mechanism to prevent hyperglycaemia.


Subject(s)
Dietary Sugars/administration & dosage , Fructose/administration & dosage , Glucose/metabolism , Homeostasis/drug effects , Liver/drug effects , Animals , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Glucocorticoids/metabolism , Insulin/genetics , Insulin/metabolism , Liver/metabolism , Male , Rats , Rats, Wistar , Signal Transduction/drug effects , Stress, Physiological
12.
Eur J Nutr ; 58(5): 1829-1845, 2019 Aug.
Article in English | MEDLINE | ID: mdl-29845385

ABSTRACT

PURPOSE: High-fructose consumption and chronic stress are both associated with metabolic inflammation and insulin resistance. Recently, disturbed activity of energy sensor AMP-activated protein kinase (AMPK) was recognized as mediator between nutrient-induced stress and inflammation. Thus, we analyzed the effects of high-fructose diet, alone or in combination with chronic stress, on glucose homeostasis, inflammation and expression of energy sensing proteins in the rat liver. METHODS: In male Wistar rats exposed to 9-week 20% fructose diet and/or 4-week chronic unpredictable stress we measured plasma and hepatic corticosterone level, indicators of glucose homeostasis and lipid metabolism, hepatic inflammation (pro- and anti-inflammatory cytokine levels, Toll-like receptor 4, NLRP3, activation of NFκB, JNK and ERK pathways) and levels of energy-sensing proteins AMPK, SIRT1 and peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α). RESULTS: High-fructose diet led to glucose intolerance, activation of NFκB and JNK pathways and increased intrahepatic IL-1ß, TNFα and inhibitory phosphorylation of insulin receptor substrate 1 on Ser307. It also decreased phospho-AMPK/AMPK ratio and increased SIRT1 expression. Stress alone increased plasma and hepatic corticosterone but did not influence glucose tolerance, nor hepatic inflammatory or energy-sensing proteins. After the combined treatment, hepatic corticosterone was increased, glucose tolerance remained preserved, while hepatic inflammation was partially prevented despite decreased AMPK activity. CONCLUSION: High-fructose diet resulted in glucose intolerance, hepatic inflammation, decreased AMPK activity and reduced insulin sensitivity. Chronic stress alone did not exert such effects, but when applied together with high-fructose diet it could partially prevent fructose-induced inflammation, presumably due to increased hepatic glucocorticoids.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Diet/methods , Fructose/administration & dosage , Inflammation/physiopathology , Stress, Physiological/physiology , Animals , Chronic Disease , Disease Models, Animal , Liver , Male , Rats , Rats, Wistar
13.
Mol Biol Rep ; 45(6): 2227-2236, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30229476

ABSTRACT

Post-infarct left ventricular remodeling (LVR) process increases the risk of heart failure (HF). Circulating galectin-3 has been associated with fibrosis, inflammation and cardiac dysfunction during the remodeling process after myocardial infarction (MI). The aims of this prospective case study were to investigate the association of potentially functional variants in the vicinity of LGALS-3 locus, rs2274273 and rs17128183 with maladaptive LVR and whether these variants could affect LGALS-3 mRNA expression in peripheral blood mononuclear cells of patients 6 months after the first MI. This study encompassed 167 patients with acute MI that were followed up for 6 months. Evidence of LVR was obtained by repeated 2D Doppler echocardiography. Rs2274273, rs17128183 and LGALS-3 mRNA expression were detected by TaqMan® technology. Rs2274273 and rs17128183 rare allele bearing genotypes, according to the dominant model (CT+TT vs. CC and AG+GG vs. AA, respectively), were significantly and independently associated with maladaptive LVR (adjusted OR = 3.02, P = 0.016; adjusted OR = 3.14, P = 0.019, respectively) and higher LGALS-3 mRNA expression (fold induction 1.203, P = 0.03 and 1.214, P = 0.03, respectively). Our exploratory results suggest that rs2274273 and rs17128183 variants affect LGALS-3 mRNA and bear the risk for maladaptive LVR post-MI remodeling. Further replication and validation in a larger group of patients is inevitable.


Subject(s)
Galectin 3/genetics , Ventricular Function/genetics , Ventricular Remodeling/genetics , Adult , Aged , Blood Proteins , Female , Galectin 3/physiology , Galectins , Heart Failure/genetics , Heart Ventricles , Humans , Male , Middle Aged , Myocardial Infarction/genetics , Prospective Studies , RNA, Messenger , Serbia , Transcriptome
14.
Horm Behav ; 96: 95-103, 2017 11.
Article in English | MEDLINE | ID: mdl-28919555

ABSTRACT

Macrophage migration inhibitory factor (MIF) is a multifunctional cytokine well known for its role in inflammation enhancement. However, a growing body of evidence is emerging on its role in energy metabolism in insulin sensitive tissues such as hippocampus, a brain region implicated in cognition, learning and memory. We hypothesized that genetic deletion of MIF may result in the specific behavioral changes, which may be linked tо impairments in brain or systemic insulin sensitivity by possible changes of the hippocampal synaptic plasticity. To assess memory, exploratory behavior and anxiety, three behavioral tests were applied on Mif gene-deficient (MIF-/-) and "wild type" C57BL/6J mice (WT). The parameters of systemic and hippocampal insulin sensitivity were also determined. The impact of MIF deficiency on hippocampal plasticity was evaluated by analyzing the level of synaptosomal polysialylated-neural cell adhesion molecule (PSA-NCAM) plasticity marker and mRNA levels of different neurotrophic factors. The results showed that MIF-/- mice exhibit emphasized anxiety-like behaviors, as well as impaired recognition memory, which may be hippocampus-dependent. This behavioral phenotype was associated with impaired systemic insulin sensitivity and attenuated hippocampal insulin sensitivity, characterized by increased inhibitory Ser307 phosphorylation of insulin receptor substrate 1 (IRS1). Finally, MIF-/- mice displayed a decreased hippocampal PSA-NCAM level and unchanged Bdnf, NT-3, NT-4 and Igf-1 mRNA levels. The results suggest that the lack of MIF leads to disturbances of systemic and hippocampal insulin sensitivity, which are possibly responsible for memory deficits and anxiety, most likely through decreased PSA-NCAM-mediated neuroplasticity rather than through neurotrophic factors.


Subject(s)
Behavior, Animal/physiology , Hippocampus/metabolism , Insulin Resistance/genetics , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Neural Cell Adhesion Molecule L1/metabolism , Sialic Acids/metabolism , Animals , Anxiety/genetics , Anxiety/metabolism , Exploratory Behavior/physiology , Male , Maze Learning/physiology , Memory Disorders/genetics , Memory Disorders/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity/genetics
15.
J Natl Compr Canc Netw ; 15(2): 205-212, 2017 02.
Article in English | MEDLINE | ID: mdl-28188190

ABSTRACT

Background: Rapid advancements in the field of metastatic breast cancer (mBC) add to the complexity of managing patients with this disease. An educational needs assessment of multidisciplinary mBC clinicians was executed to identify practice performance gaps and recommend educational strategies aimed at closing these gaps. Methods: To ensure a collection of reliable data for assessment, a systematic process was used to design, develop, and validate the tools that were used. This grounded theory approach included assessment and confirmation by clinical experts and validation testing within the target audiences. A mixed-methods approach was used to identify practice performance gaps in care, using both qualitative in-depth interviews and quantitative surveying. The quantitative survey assessment consisted of 2 main sections: the Clinician Change Readiness Inventory tool and a Clinical Knowledge and Practice Assessment. Results: The study included 42 clinicians in the interview phase and 186 clinicians in the survey phase from 36 different states. Five key practice performance gaps were identified: (1) selecting optimal treatment, (2) personalizing therapy, (3) monitoring mBC, (4) engaging in effective communication, and (5) balancing patient access and time. Most of the gaps overlap and are related to the integral role communication plays in management decision-making in mBC. Conclusions: Awareness of the key practice performance gaps is critical to inform improvements in quality care.


Subject(s)
Breast Neoplasms/therapy , Clinical Competence , Educational Measurement/methods , Interdisciplinary Communication , Medical Oncology/education , Breast Neoplasms/pathology , Cohort Studies , Communication , Education, Medical, Continuing/methods , Female , Grounded Theory , Health Personnel/education , Humans , Surveys and Questionnaires , United States
16.
J Clin Lab Anal ; 30(6): 1150-1157, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27207566

ABSTRACT

BACKGROUND: Previous research has shown that there is an association between galectin-3 (gal-3) protein and cardiovascular pathology. The aim of this study was to investigate the effects of rs2274273 and rs17128183 on genetic susceptibility to advanced carotid atherosclerosis (CA) and its complications. The rs2274273 has been singled out as the lead SNP of the haplotype block containing LGALS-3 (gal-3 gene) associated with gal-3 circulating levels, while rs17128183 constitutes a potentially functional SNP of the same hap-block. We further sought to determine whether these genetic variants have an impact on the expression of LGALS-3 mRNA in human carotid atherosclerotic plaque tissue. METHODS: The study encompassed 300 control subjects and 485 patients with advanced CA who had undergone carotid endarterectomy. Rs2274273, rs17128183, and LGALS-3 relative mRNA expression was detected by means of real-time PCR (TaqMan® technology). RESULTS: There were no statistically significant associations of the investigated genetic variants with susceptibility to advanced CA, nor did we find any associations in terms of ultrasonographically defined plaque phenotypes. The relative expression of LGALS-3 mRNA proved to be significantly higher in carriers of the rare alleles (P = 0.039) for both genetic variants. CONCLUSION: Our exploratory results suggest that while rs2274273 and rs17128183 bear no association with the risk of advanced CA or CA-related complications, these genetic variants are likely to affect LGALS-3 expression levels. In order to reach a definitive conclusion on the role played by rs2274273 and rs17128183 in advanced CA, our results should be further validated.


Subject(s)
Carotid Artery Diseases/genetics , Galectin 3/genetics , Gene Expression/genetics , Genetic Predisposition to Disease/genetics , Polymorphism, Single Nucleotide/genetics , RNA, Messenger/metabolism , Adult , Aged , Blood Proteins , Carotid Arteries/diagnostic imaging , Carotid Artery Diseases/pathology , DNA Mutational Analysis , Female , Galectins , Genetic Association Studies , Genotype , Humans , Male , Middle Aged , Statistics, Nonparametric , Ultrasonography
17.
Nutr Neurosci ; 18(2): 66-75, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24257416

ABSTRACT

OBJECTIVES: High fructose diet has been shown to have damaging effects on the hippocampus, a brain region critical for learning and memory. Fructose-induced hippocampal dysfunction may arise from insulin resistance and inflammation, and from concomitant changes in plasticity-related presynaptic proteins. We hypothesized that long-term access to fructose (10% and 60% solutions over a period of 9 weeks) affects insulin sensitivity, hippocampal inflammation, and synaptic plasticity in male Wistar rats. METHODS: We used the area under curve (AUC) glucose value and inhibitory Ser³°7 phosphorylation of hippocampal insulin receptor substrate 1 (IRS-1) as hallmarks of insulin resistance. To examine inflammatory state, we analysed protein levels and intracellular redistribution of glucocorticoid receptor and nuclear factor-κB (NFκB), as well as mRNA levels of tumour necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-1ß (IL-1ß). Polysialylated neural cell adhesion molecule (PSA-NCAM) protein was used as a synaptic plasticity marker. RESULTS: The results indicate different impacts of diverse fructose-enriched diets on insulin sensitivity and hippocampal inflammation and plasticity. Long-term ingestion of 10% fructose solution led to increase in AUC glucose value, as well as to elevation in hippocampal IRS-1 Ser³°7 phosphorylation and increase in IL-6 mRNA. In rats consuming 60% fructose, the level of PSA-NCAM was reduced, in parallel with augmented glucocorticoid signalization. DISCUSSION: The results showed that long-term consumption of 10% fructose solution induces hippocampal insulin resistance and inflammation, with no concomitant plasticity changes. Interestingly, rats fed with higher concentrations of fructose displayed impaired plastic response of the hippocampus, coinciding with augmented glucocorticoid signalling, which may provide a basis for cognitive deficits associated with metabolic syndrome.


Subject(s)
Dietary Carbohydrates/adverse effects , Fructose/adverse effects , Hippocampus/metabolism , Insulin Resistance , Neural Cell Adhesion Molecule L1/metabolism , Neuronal Plasticity , Neurons/metabolism , Sialic Acids/metabolism , Animals , Biomarkers/metabolism , Cytokines/genetics , Cytokines/metabolism , Dietary Carbohydrates/administration & dosage , Fructose/administration & dosage , Hippocampus/immunology , Insulin Receptor Substrate Proteins/metabolism , Male , NF-kappa B/metabolism , Nerve Tissue Proteins/metabolism , Neurons/immunology , Phosphorylation , Protein Processing, Post-Translational , Protein Transport , Random Allocation , Rats, Wistar , Receptors, Glucocorticoid/metabolism
18.
Eur J Nutr ; 53(6): 1393-402, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24389792

ABSTRACT

PURPOSE: High fructose consumption provokes metabolic perturbations that result in chronic low-grade inflammation and insulin resistance. Glucocorticoids, potent anti-inflammatory hormones, have important role in pathogenesis of diet-induced metabolic disturbances. The aim of this study was to examine the link between glucocorticoid metabolism and inflammation in the liver of fructose-fed rats. METHODS: Fructose-fed male Wistar rats consumed 60% fructose solution for 9 weeks. Glucocorticoid prereceptor metabolism and signaling were analyzed by measuring the level of 11ß-hydroxysteroid dehydrogenase type 1 (11ßHSD1) and hexose-6-phosphate dehydrogenase expression, as well as via determination of intracellular corticosterone concentration, glucocorticoid receptor subcellular distribution and expression of its target gene, phosphoenolpyruvate carboxykinase. Nuclear factor kappa B (NFκB), tumor necrosis factor alpha (TNFα) and the level of inhibitory phosphorylation of insulin receptor substrate-1 (IRS-1) on Ser(307) were analyzed as markers of hepatic inflammation. The protein and/or mRNA levels of all examined molecules were assessed by Western blot and/or qPCR. RESULTS: Fructose-rich diet led to an enhancement of 11ßHSD1 protein level in the liver, without affecting intracellular level of corticosterone and downstream glucocorticoid signaling. On the other hand, proinflammatory state was achieved through NFκB activation and increased TNFα expression, while elevated level of inhibitory phosphorylation of IRS-1 was observed as an early hallmark of insulin resistance. CONCLUSION: High-fructose diet does not influence hepatic glucocorticoid signaling downstream of the receptor, permitting development of NFκB-driven inflammation. The alteration in 11ßHSD1 expression is most likely the consequence of enhanced inflammation, finally leading to disruption of insulin signaling in the rat liver.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Fructose/administration & dosage , Liver/physiopathology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , Adipose Tissue/drug effects , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Carbohydrate Dehydrogenases/genetics , Carbohydrate Dehydrogenases/metabolism , Energy Intake , Fatty Acids, Nonesterified/blood , Fructose/adverse effects , Glucocorticoids/metabolism , Inflammation/etiology , Inflammation/pathology , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance , Liver/drug effects , Liver/enzymology , Male , NF-kappa B/metabolism , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Glucocorticoid/metabolism , Triglycerides/blood , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
19.
Biofactors ; 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38318672

ABSTRACT

Obesity is a pressing problem worldwide for which standard therapeutic strategies have limited effectiveness. The use of natural products seems to be a promising approach to alleviate obesity and its associated complications. The tepals of Crocus sativus (Cr) plant, usually wasted in saffron production, are an unexplored source of bioactive compounds. Our aim was to elucidate the mechanisms of Cr tepals extract in obesity by investigating its effects on adipocyte differentiation, visceral adipose tissue (VAT) and subcutaneous adipose tissue (SAT) hypertrophy, and lipid metabolism in an animal model of diet-induced obesity. To this end, mouse 3T3-F442A preadipocytes were treated with Cr tepals extract and the expression of adipocyte differentiation genes was determined. Caloric intake, body mass, triglycerides, systemic insulin sensitivity, histology, insulin signaling, and lipid metabolism in VAT and SAT were analyzed in mice fed a 60% fat diet for 14 weeks and treated orally with Cr tepals extract during the last 5 weeks of the diet. We demonstrated for the first time that Cr tepals extract inhibits adipocyte differentiation in vitro. The animal model confirmed that oral treatment with Cr tepals extract results in weight loss, improved systemic insulin sensitivity, lower triglycerides, and improved lipid peroxidation. The suppressive effect of Cr tepals extract on adipocyte hypertrophy and inflammation was observed only in SAT, which, together with preserved SAT insulin signaling, most likely contributed to improved systemic insulin sensitivity. Our results suggest the functionality of SAT as a possible target for the treatment of obesity and its complications.

20.
Br J Nutr ; 110(3): 456-65, 2013 Aug 28.
Article in English | MEDLINE | ID: mdl-23286672

ABSTRACT

High fructose consumption is commonly associated with insulin resistance, disturbed glucose homeostasis and low-grade inflammation. Increased glucocorticoid production within adipose tissue has been implicated in the pathogenesis of fructose-induced metabolic syndrome. Immunosuppressive actions of glucocorticoids can be counter-regulated by macrophage migration inhibitory factor (MIF), which is recognised as a key molecule in metabolic inflammation. In the present study, we hypothesised that coordinated action of glucocorticoids and MIF can mediate the effects of a high-fructose diet on adipose tissue and liver inflammation. We examined the effects of long-term consumption of a 10% fructose solution on corticosterone (CORT) and MIF levels in rat blood plasma, liver and adipose tissue, as well as MIF and TNF-a mRNA expression and NF-kB activation in the same tissues. The high-fructose diet led to an increase in both CORT and MIF in the adipose tissue, and a highly significant positive correlation between their levels was observed. The attenuated NF-kB activation and unaltered TNF-a mRNA expression noticed in the adipose tissue could be interpreted as an outcome of the opposing actions of CORT and MIF. In contrast to adipose tissue, inflammation in the liver was characterised by NF-kB activation, an increased TNF-a mRNA level and unchanged levels of MIF protein, MIF mRNA and CORT. Overall, these findings suggest that a high-fructose diet differently affects the levels of glucocorticoids and MIF in the adipose tissue and liver, implicating that fructose over-consumption has tissue-specific effects on regulation of metabolic inflammation.


Subject(s)
Adipose Tissue/drug effects , Corticosterone/metabolism , Diet/adverse effects , Fructose/adverse effects , Inflammation/etiology , Liver/drug effects , Macrophage Migration-Inhibitory Factors/metabolism , Adipose Tissue/metabolism , Animals , Dietary Sucrose/adverse effects , Inflammation/metabolism , Leukocyte L1 Antigen Complex/genetics , Leukocyte L1 Antigen Complex/metabolism , Liver/metabolism , Macrophages/metabolism , Male , Metabolic Syndrome/etiology , Metabolic Syndrome/metabolism , NF-kappa B/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL