Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Eur J Pharmacol ; 970: 176476, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38493915

ABSTRACT

BACKGROUND: Nowadays type 2 diabetes mellitus (T2DM) leads to population mortality growth. Today glucagon-like peptide type 1 receptor agonists (GLP-1 RA) are one of the most promising glucose-lowered drugs with anorexigenic and cardioprotective effects. The present study aims to determine the effects of GLP-1 RA semaglutide 6-month therapy on T2DM patient metabolic parameters and adipose progenitor cell health. METHODS: T2DM patients (N = 8) underwent clinical characterization and subcutaneous fat biopsy at start point and after semaglutide 6-month therapy. Adipose-derived stem cells (ADSC) were isolated by enzymatic method. Cell proliferation analysis was performed by MTT and immunocytochemistry. White and beige adipogenesis was analyzed by BODIPY493/503 staining and confocal microscopy. Adipocyte's metabolic properties were estimated by 3H- and 14C-based metabolic assays. Thermogenesis analysis was performed by ERthermAC staining and confocal microscopy. Protein markers were assessed by Western blotting. RESULTS: Semaglutide 6-month therapy demonstrated significant anorexigenic and glucose-lowering effects. However, insulin sensitivity (HOMA-IR and M-index) was unchanged after therapy. Semaglutide 6-month therapy increased ADSC proliferation and white and beige adipogenesis. Moreover, lipid droplets fragmentation was observed in beige adipocytes. Both white and beige adipocytes after semaglutide therapy demonstrated 2-3 fold growth of glucose uptake without changes in insulin sensitivity. Newly formed white adipocytes demonstrated glucose utilization for active ATP synthesis, whereas beige adipocytes for canonical thermogenesis. CONCLUSIONS: Our study has revealed that semaglutide 6-month therapy has not only systemic anorexigenic effects, but can markedly improve adipose tissue health. We have demonstrated critical restoration of ADSC renewal functions, which potentially can be involved in semaglutide based weight loss.


Subject(s)
Diabetes Mellitus, Type 2 , Glucagon-Like Peptides , Insulin Resistance , Humans , Adipose Tissue, White/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Adipose Tissue, Brown/metabolism , Insulin Resistance/physiology , Obesity/metabolism , Adipocytes, White/metabolism , Glucose/metabolism , Glucagon-Like Peptide 1/metabolism
2.
Cells ; 11(23)2022 Nov 29.
Article in English | MEDLINE | ID: mdl-36497083

ABSTRACT

BACKGROUND: Combined non-viral gene therapy (GT) of ischemia and cardiovascular disease is a promising tool for potential clinical translation. In previous studies our group has developed combined gene therapy by vascular endothelial growth factor 165 (VEGF165) + hepatocyte growth factor (HGF). Our recent works have demonstrated that a bicistronic pDNA that carries both human HGF and VEGF165 coding sequences has a potential for clinical application in peripheral artery disease (PAD). The present study aimed to test HGF/VEGF combined plasmid efficacy in ischemic skeletal muscle comorbid with predominant complications of PAD-impaired glucose tolerance and type 2 diabetes mellitus (T2DM). METHODS: Male C57BL mice were housed on low-fat (LFD) or high-fat diet (HFD) for 10 weeks and metabolic parameters including FBG level, ITT, and GTT were evaluated. Hindlimb ischemia induction and plasmid administration were performed at 10 weeks with 3 weeks for post-surgical follow-up. Limb blood flow was assessed by laser Doppler scanning at 7, 14, and 21 days after ischemia induction. The necrotic area of m.tibialis anterior, macrophage infiltration, angio- and neuritogenesis were evaluated in tissue sections. The mitochondrial status of skeletal muscle (total mitochondria content, ETC proteins content) was assessed by Western blotting of muscle lysates. RESULTS: At 10 weeks, the HFD group demonstrated impaired glucose tolerance in comparison with the LFD group. HGF/VEGF plasmid injection aggravated glucose intolerance in HFD conditions. Blood flow recovery was not changed by HGF/VEGF plasmid injection either in LFD or HFD conditions. GT in LFD, but not in HFD conditions, enlarged the necrotic area and CD68+ cells infiltration. However, HGF/VEGF plasmid enhanced neuritogenesis and enlarged NF200+ area on muscle sections. In HFD conditions, HGF/VEGF plasmid injection significantly increased mitochondria content and ETC proteins content. CONCLUSIONS: The current study demonstrated a significant role of dietary conditions in pre-clinical testing of non-viral GT drugs. HGF/VEGF combined plasmid demonstrated a novel aspect of potential participation in ischemic skeletal muscle regeneration, through regulation of innervation and bioenergetics of muscle. The obtained results made HGF/VEGF combined plasmid a very promising tool for PAD therapy in impaired glucose tolerance conditions.


Subject(s)
Diabetes Mellitus, Type 2 , Glucose Intolerance , Mice , Male , Humans , Animals , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Glucose Intolerance/complications , Glucose Intolerance/genetics , Glucose Intolerance/therapy , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/therapy , Mice, Inbred C57BL , Ischemia/metabolism , Genetic Therapy/methods , Muscle, Skeletal/metabolism
3.
Ter Arkh ; 94(10): 1131-1135, 2022 Nov 22.
Article in Russian | MEDLINE | ID: mdl-36468985

ABSTRACT

Obesity and type 2 diabetes mellitus (DM 2) are two interrelated metabolic diseases widespread throughout the developed world. However, up to 30% of individuals with a long history of obesity do not have a carbohydrate metabolism disorder. This article presents the results of a multi-year study of adipose tissue biology in obese individuals with DM 2 compared with individuals with the same history of obesity without DM 2. Comparative analysis of hormonal, cellular, and genetic factors in two groups of patients showed that DM 2 occurs in individuals with abnormal proliferation and adipogenic differentiation of mesenchymal stem cells (MSCs) of adipose tissue. It leads to adipocyte hypertrophy and inflammatory infiltration of adipose tissue macrophages, resulting in increased insulin resistance and diabetogenic effects. These disorders are due to abnormal expression of genes responsible for the proliferation and adipogenic differentiation of MSCs. The study of the possible reversibility of abnormal changes in adipose tissue MSCs in obese patients after significant weight loss and DM 2 remission appears to be a promising research direction. The ability to control adipose tissue progenitor cells may represent a new target for treating and preventing metabolic disorders in obesity.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Humans , Adipocytes/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/metabolism , Obesity/complications , Obesity/epidemiology , Obesity/metabolism , Adipose Tissue/metabolism
4.
Bull Exp Biol Med ; 171(4): 532-540, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34546443

ABSTRACT

Murine peritoneal macrophages isolated from the lavage fluid after administration of thioglycolate and concanavalin A are presented by two populations of cells of different diameters. Polarization of macrophages into a proinflammatory (M1) phenotype is accompanied by an increase in number of small cells. Macrophages obtained after administration of thioglycolate demonstrate higher tendency to anti-inflammatory (M2) phenotype, while macrophages isolated after administration of concanavalin A are committed in the proinflammatory direction. Lactate level is increased in M1 macrophages in comparison with M2 cells, which indicates predominance of glycolytic metabolism. Macrophages obtained after administration of concanavalin A have reduced mitochondrial potential, which reflects a tendency to apoptosis. Autophagy activation and inhibition neutralize the differences in pro- and anti-inflammatory properties of polarized macrophages obtained after thioglycolate administration, but have less pronounced effect on macrophages obtained after administration concanavalin A. Autophagy inhibitor increases mitochondrial potential in non-polarized macrophages obtained after administration of concanavalin A. These results demonstrate divergent properties of macrophages obtained after administration of glycolate and concanavalin A due to the difference in the mechanisms of experimental peritonitis.


Subject(s)
Concanavalin A/pharmacology , Macrophage Activation/drug effects , Macrophages, Peritoneal/drug effects , Thioglycolates/pharmacology , Animals , Cell Polarity/drug effects , Disease Models, Animal , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/physiology , Male , Mice , Mice, Inbred C57BL , Peritonitis/immunology , Peritonitis/pathology
5.
Anal Biochem ; 628: 114268, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34090857

ABSTRACT

In the modern world obesity and insulin resistance contribute to a high impact on the structure of mortality. Basic research and pharmacological screenings for the search of new targets and insulin sensitizers require relevant cell models of adipocytes. Today the 3T3-L1 preadipocytes cell line is a widely used mouse-based model for investigation of adipocyte biology. Nonetheless, animal studies cannot be transferred directly in human research and nowadays the search for relevant and renewable cell models of human adipocyte is of undeniable importance. In the present study, we have compared pooled culture of human adipose-derived stem cells (ADSC) with immortalized ADSC cell line ASC52Telo. Both cell types had mesenchymal stem cell phenotype verified by flow cytometry. However, the efficacy of adipogenic differentiation, stimulation of FABP4 and PPARg protein expressions, and glucose uptake stimulation by insulin were reduced for ASC52Telo-derived adipocytes in comparison with ADSC-derived adipocytes. In addition, the analysis of insulin signaling has shown impaired phosphorylation of IRS1 and AS160 in ASC52Telo-derived cells. In summary, we have shown that immortalized cell line of human ADSC ASC52Telo have mesenchymal stem cell phenotype. Nevertheless, ASC52Telo-derived adipocytes demonstrate impaired adipogenesis and insulin sensitivity that are the main properties of healthy adipocytes.


Subject(s)
Adipose Tissue, White/metabolism , Stem Cells/metabolism , Telomerase/metabolism , Adolescent , Adult , Cell Line , Healthy Volunteers , Humans , Male , Young Adult
6.
Mitochondrion ; 59: 157-168, 2021 07.
Article in English | MEDLINE | ID: mdl-34010673

ABSTRACT

Non-shivering thermogenesis takes place in brown and beige adipocytes and facilitates cold tolerance and acclimation. However, thermogenesis in adipose tissue also was found to be activated in metabolic overload states for fast utilization of nutrients excess. This observation spurred research interest in mechanisms of thermogenesis regulation for metabolic overload and obesity prevention. One of proposed regulators of thermogenic efficiency in adipocytes is the dynamics of mitochondria, where thermogenesis takes place. Indeed, brown and beige adipocytes exhibit fragmented round-shaped mitochondria, while white adipocytes have elongated organelles with high ATP synthesis. Mitochondrial morphology can determine uncoupling protein 1 (UCP1) content, efficiency of catabolic pathways and electron transport chain, supplying thermogenesis. This review will highlight the co-regulation of mitochondrial dynamics and thermogenesis and formulate hypothetical ways for excessive nutrients burning in response to mitochondrial morphology manipulation.


Subject(s)
Mitochondria/metabolism , Thermogenesis , Uncoupling Protein 1/metabolism , Adipose Tissue/metabolism , Animals , Energy Metabolism , Humans , Mitochondrial Dynamics
7.
Biochemistry (Mosc) ; 84(11): 1329-1345, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31760921

ABSTRACT

Obesity is a major risk factor for type 2 diabetes and metabolic syndrome and an essential medical and social problem. In the first part of the review, we briefly highlight the biochemical basis of metabolic disbalance in obesity and evolution of our views on the mechanisms of insulin resistance development in insulin-sensitive tissues. Because obesity relates to the disturbance in the normal physiology of fat tissue, the second part of the review focuses on latent inflammation that develops in obesity and is supported by immune cells. Finally, the problem of adipocyte hypertrophy, reduced regenerative potential of fat progenitor cells, and impaired renewal of fat depots is discussed in the context of type 2 diabetes pathogenesis.


Subject(s)
Inflammation/pathology , Insulin Resistance , Obesity/pathology , Adipogenesis , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Humans , Inflammation/metabolism , Lymphocytes/cytology , Lymphocytes/immunology , Lymphocytes/metabolism , Metabolic Syndrome/etiology , Metabolic Syndrome/metabolism , Metabolic Syndrome/pathology , Obesity/complications , Obesity/metabolism
8.
Bull Exp Biol Med ; 167(4): 492-495, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31515638

ABSTRACT

TNFα mediates the expression of MMP-9 in THP-1 monocytes induced by urokinase (uPA). Upregulation of MMP-9 caused by uPA and TNFα is suppressed by etanercept, a TNFα inhibitor. In addition, uPA stimulates TNFα mRNA expression. Both uPA and TNFα induce ROS generation in monocytes, while MMP-9 secretion induced by uPA and TNFα is inhibited by antioxidants. Inhibitors of NFκB, ligands of PPARα and PPARγ receptors, and SIRT1 activators negatively affect MMP-9 secretion induced by uPA. MMP-9 secretion during monocyte differentiation into macrophages is downregulated by etanercept and antioxidants. These factors as well as MMP inhibitor GM6001 reduce the number of macrophages attached to substrate during cell differentiation, indicating the role of urokinase, TNFα, and ROS in MMP expression in monocytes and MMP involvement in macrophage maturation.


Subject(s)
Matrix Metalloproteinase 9/metabolism , Monocytes/metabolism , Tumor Necrosis Factor-alpha/metabolism , Urokinase-Type Plasminogen Activator/pharmacology , Cell Line , Humans , Monocytes/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Superoxides/metabolism , THP-1 Cells
9.
Bull Exp Biol Med ; 167(3): 413-417, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31350657

ABSTRACT

A stimulating effect of a combination of hepatocyte growth factor (HGF) and glial neurotrophic factor (GDNF) on the growth of neurites in the spinal ganglion model was demonstrated. The mechanism of neurite growth in the spinal ganglion model is associated with transactivation of HGF c-met receptor in the presence of both HGF and GDNF. The combination of HGF and GDNF significantly activated mitogenic signaling cascade mediated by protein kinases ERK1/2, which can be a mechanism for increasing the number of neurites. Our findings can be used for developing effective methods for restoring impaired peripheral nerve function after traumatic and ischemic injury using a combination of GDNF and HGF.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/metabolism , Hepatocyte Growth Factor/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurites/metabolism , Animals , Cell Line, Tumor , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred C57BL , Nerve Regeneration/physiology , Phosphorylation , Proto-Oncogene Proteins c-met/metabolism
10.
Biochemistry (Mosc) ; 84(5): 553-561, 2019 May.
Article in English | MEDLINE | ID: mdl-31234769

ABSTRACT

Obesity is accompanied by dyslipidemia, hypoxia, endoplasmic reticulum (ER) stress, and inflammation, representing the major risk factor for the development of insulin resistance (IR) and type 2 diabetes. We modeled these conditions in cultured 3T3-L1 adipocytes and studied their effect on insulin signaling, glucose uptake, and inflammatory response via activation of stress-dependent JNK1/2 kinases. Decreased insulin-induced phosphorylation of the insulin cascade components IRS, Akt, and AS160 was observed under all tested conditions (lipid overloading of cells by palmitate, acute inflammation induced by bacterial lipopolysaccharide, hypoxia induced by Co2+, and ER stress induced by brefeldin A). In all the cases, except the acute inflammation, glucose uptake by adipocytes was reduced, and the kinetics of JNK1/2 activation was bi-phasic exhibiting sustained activation for 24 h. By contrast, in acute inflammation, JNK1/2 phosphorylation increased transiently and returned to the basal level within 2-3 h of stimulation. These results suggest a critical role of sustained (latent) vs. transient (acute) inflammation in the induction of IR and impairment of glucose utilization by adipose tissue. The components of the inflammatory signaling can be promising targets in the development of new therapeutic approaches for preventing IR and type 2 diabetes.


Subject(s)
Inflammation , Insulin Resistance , Obesity/pathology , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Endoplasmic Reticulum Stress/drug effects , Fatty Acids, Nonesterified/pharmacology , Inflammation/etiology , Insulin/pharmacology , Lipopolysaccharides/pharmacology , Mice , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Obesity/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
11.
J Diabetes Complications ; 33(2): 148-159, 2019 02.
Article in English | MEDLINE | ID: mdl-30482492

ABSTRACT

BACKGROUND: Obesity and type 2 diabetes mellitus (T2DM) are among the most important morbidity factors. In this study we tested the hypothesis that low proliferative potential of adipose derived stromal cells (ADSC) associates with reduced formation of new fat depots, excess accumulation of fat in the functional adipocytes and their hypertrophy, resulting in fat inflammation and insulin resistance. METHODS: We screened two groups of obese patients with or without T2DM, matched for BMI, age, and duration of obesity to test the hypothesis that hypertrophy and decreased renewal of adipocytes may underlie transition from obesity to T2DM. All patients were matched for carbohydrate metabolism (fasting blood glucose level, glycated hemoglobin, HOMA-IR index and M-index). The subcutaneous and omental fat tissue biopsies were obtained during bariatric surgery from obese individuals with or without T2DM. The morphology and immunophenotype of subcutaneous and omental fat was assessed in frozen tissue sections. ADSC were isolated from both types of fat tissue biopsies and screened for morphology, proliferative potential and inflammatory status. RESULTS: The non-diabetic patients had normal carbohydrate metabolism and moderate insulin resistance measured by HOMA-IR and hyperinsulinemic clamp (M-index), while T2DM patients were extremely insulin resistant by both indexes. The average size of diabetic adipocytes was higher than that of non-diabetic in both subcutaneous and omental fat tissues, indicating adipocyte hypertrophy in T2DM. Both these tissues contained higher level of macrophage infiltration and increased M1-like to M2-like ratio of macrophage subpopulations, suggesting increased fat inflammation in T2DM. This was confirmed by increased activatory phosphorylation of stress-induced JNK1/2 in diabetic ADSC. CONCLUSION: These results suggest that blunted proliferation and increased hypertrophy of diabetic ADSC may lead to reduced insulin sensitivity via increased inflammation mediated by M1 macrophages and JNK1/2 pathway.


Subject(s)
Abdominal Fat/pathology , Cell Proliferation/physiology , Diabetes Mellitus, Type 2/pathology , Inflammation/etiology , Mesenchymal Stem Cells/physiology , Omentum/pathology , Subcutaneous Fat/pathology , Adipose Tissue/cytology , Adult , Case-Control Studies , Cells, Cultured , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Female , Humans , Hypertrophy/etiology , Hypertrophy/pathology , Inflammation/pathology , Insulin Resistance/physiology , Male , Middle Aged , Obesity/complications , Obesity/metabolism , Obesity/pathology , Obesity/physiopathology
12.
Biochemistry (Mosc) ; 83(5): 498-506, 2018 May.
Article in English | MEDLINE | ID: mdl-29738684

ABSTRACT

Obesity and latent inflammation in adipose tissue significantly contribute to the development of insulin resistance (IR) and type 2 diabetes. Here we studied whether the antiinflammatory interleukin-4 (IL-4) can restore insulin sensitivity in cultured 3T3-L1 adipocytes. The activity of key components of the insulin signaling cascade was assessed by immunoblotting using phospho-specific antibodies to insulin receptor substrate IRS1 (Tyr612), Akt (Thr308 and Ser473), and AS160 (Ser318) protein that regulates translocation of the GLUT4 glucose transporter to the plasma membrane. IR was induced in mature adipocytes with albumin-conjugated palmitate. IR significantly reduced phosphorylation levels of all the above-mentioned proteins. Addition of IL-4 to the culturing medium during IR induction led to a dose-dependent stimulation of the insulin-promoted phosphorylation of IRS1, Akt, and AS160. At the optimal concentration of 50 ng/ml, IL-4 fully restored activation of the insulin cascade in IR cells, but it did not affect insulin signaling activation in the control cells. IL-4 neither upregulated expression of key adipogenesis markers GLUT4 and PPARγ nor caused lipid accumulation in the adipocytes. These results demonstrate that IL-4 can restore insulin sensitivity in adipocytes via mechanisms not associated with induced adipogenesis or de novo formation of lipid depots.


Subject(s)
Adipocytes/metabolism , Insulin Resistance , Interleukin-4/metabolism , Lipids , 3T3-L1 Cells , Animals , Cells, Cultured , Mice
13.
Biomed Pharmacother ; 101: 682-690, 2018 May.
Article in English | MEDLINE | ID: mdl-29518615

ABSTRACT

Peripheral nerve injury remains a common clinical problem with no satisfactory treatment options. Numerous studies have shown that hepatocyte growth factor (HGF) exerts neurotrophic effect in motor, sensory, and parasympathetic neurons in addition to mitogenic, morphogenic, angiogenic, antiapoptotic, antifibrotic, and anti-inflammatory effect on various tissues and cells. In our study we examined efficacy of gene therapy with HGF-bearing plasmid (pC4W-hHGF) to improve consequences of traumatic nerve injury in mice. Treatment by pC4W-hHGF led to restoration of nerve structure and functional recovery compared to similar parameters in control animals. Compound action potentials (CAP) in experimental groups treated with 100 or 200 µg of pC4W-hHGF demonstrated increased amplitude and latency decrease compared to spontaneous recovery control group. In HGF-treated mice histological analysis showed a three-fold increase in axon number in nerve portion located distal to the lesion site compared to control. Moreover, significant functional recovery of n. peroneus communis triggered by pC4W-hHGF gene therapy was observed using the footprints analysis. Obtained results provide evidence for plasmid-based HGF gene therapy as a potential treatment for traumatic injury of peripheral nerve.


Subject(s)
Genetic Therapy/methods , Hepatocyte Growth Factor/administration & dosage , Nerve Regeneration/drug effects , Peripheral Nerve Injuries/drug therapy , Plasmids/administration & dosage , Sciatic Nerve/drug effects , Animals , Hepatocyte Growth Factor/genetics , Humans , Injections, Intramuscular , Male , Mice , Mice, Inbred C57BL , Nerve Regeneration/genetics , Peripheral Nerve Injuries/genetics , Plasmids/genetics , Sciatic Nerve/injuries , Sciatic Nerve/physiology
14.
Int J Endocrinol ; 2017: 5076732, 2017.
Article in English | MEDLINE | ID: mdl-28912810

ABSTRACT

Obesity is a growing problem in modern society and medicine. It closely associates with metabolic disorders such as type 2 diabetes mellitus (T2DM) and hepatic and cardiovascular diseases such as nonalcoholic fatty liver disease, atherosclerosis, myocarditis, and hypertension. Obesity is often associated with latent inflammation; however, the link between inflammation, obesity, T2DM, and cardiovascular diseases is still poorly understood. Insulin resistance is the earliest feature of metabolic disorders. It mostly develops as a result of dysregulated insulin signaling in insulin-sensitive cells, as compared to inactivating mutations in insulin receptor or signaling proteins that occur relatively rare. Here, we argue that inflammatory signaling provides a link between latent inflammation, obesity, insulin resistance, and metabolic disorders. We further hypothesize that insulin-activated PI3-kinase pathway and inflammatory signaling mediated by several IκB kinases may constitute negative feedback leading to insulin resistance at least in the fat tissue. Finally, we discuss perspectives for anti-inflammatory therapies in treating the metabolic diseases.

15.
Kardiologiia ; 57(12): 53-59, 2017 Dec.
Article in Russian | MEDLINE | ID: mdl-29466211

ABSTRACT

A new trend in modern experimental cardiology is the development of approaches to correction of reparation after myocardial infarction (MI) with the use of specific effects on immune cells. One of the main targets for such interventions is the process of macrophage's polarization in the infarction zone. Proinflammatory M1­macrophages contribute to hampered myocardial repair, in contrast to M2­macrophages that promote regeneration. Currently, there are two main ways of targeted delivery of agents necessary for macrophage reprogramming - inlipoid and inglycan-encapsulated particles. As modulating agents, small interfering RNA and other genetic constructions are usually used. Both these approaches are currently awaiting their translation into cardiology. The most physiological approach to reprogramming of immune cells may consist in attempts to switch the metabolism of the immune cell from glycolytic to oxidative, which allows macrophages to switch from M1 to M2 phenotype. Among possible targets for macrophage reprogramming, it is worthwhile to isolate the protein complex mTORC1, the blocking of which promotes oxidative metabolism, and the transcription factor HIF-1α, the blocking of which also facilitates the switching of the metabolism from glycolytic to oxidative one.


Subject(s)
Infarction , Myocardial Infarction , Humans , Macrophages , Myocardium , Phenotype
16.
Biochemistry (Mosc) ; 80(10): 1217-26, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26567565

ABSTRACT

The problem of metabolic syndrome is one of the most important in medicine today. The main hazard of metabolic syndrome is development of latent inflammation in adipose tissue, which promotes atherosclerosis, non-alcoholic fatty liver disease, myocarditis, and a number of other illnesses. Therefore, understanding of molecular mechanisms of latent inflammation in adipose tissue is very important for treatment of metabolic syndrome. Three main components that arise during hypertrophy and hyperplasia of adipocytes underlie such inflammation: endoplasmic reticulum stress, oxidative stress, and hypoxia. Each of these components mediates activation in different ways of the key factor of inflammation - NF-κB. For metabolic syndrome therapy, it is suggested to influence a number of inflammatory signaling components by activating other cell factors to suppress development of inflammation. Such potential factors are peroxisome proliferator-activated receptors type γ that suppress transcription factor NF-κB through direct contact or via kinase of a NF-κB inhibitor (IKK), and also the antiinflammatory transcription factor AP-1. Other possible targets are type 3 NAD+-dependent histone deacetylases (sirtuins). There are mutually antagonistic relationships between NF-κB and sirtuin type 1 that prevent development of inflammation in metabolic syndrome. Moreover, sirtuin type 1 inhibits the antiinflammatory transcription factor AP-1. Study of the influence of these factors on the relationship between macrophages and adipocytes, macrophages, and adipose tissue-derived stromal cells can help to understand mechanisms of signaling and development of latent inflammation in metabolic syndrome.


Subject(s)
Metabolic Syndrome/metabolism , PPAR gamma/metabolism , Sirtuins/metabolism , Animals , Humans , Inflammation/enzymology , Inflammation/metabolism , Metabolic Syndrome/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...