36 results on '"Karalis KP"'
Search Results
2. cGMP-dependent protein kinase contributes to hydrogen sulfide-stimulated vasorelaxation
- Author
-
Valentina Vellecco, Altaany Zaid, Robert Feil, Rui Wang, Katia Karalis, Mariarosaria Bucci, Anna Cantalupo, Andreas Papapetropoulos, Giuseppe Cirino, Panagiotis Giannogonas, Zongmin Zhou, Sandeep Dhayade, Bucci, Mariarosaria, Papapetropoulos, A, Vellecco, Valentina, Zhou, Z, Zaid, A, Giannogonas, P, Cantalupo, A, Dhayade, S, Karalis, Kp, Wang, R, Feil, R, and Cirino, Giuseppe
- Subjects
Male ,Anatomy and Physiology ,Mouse ,lcsh:Medicine ,Vasodilation ,030204 cardiovascular system & hematology ,Signal transduction ,Cardiovascular ,Cardiovascular System ,Biochemistry ,Mice ,0302 clinical medicine ,Molecular cell biology ,Hydrogen Sulfide ,lcsh:Science ,Mesenteric arteries ,Aorta ,Cells, Cultured ,Mice, Knockout ,Peripheral Vascular Diseases ,0303 health sciences ,Multidisciplinary ,Cystathionine gamma-lyase ,Neurochemistry ,Animal Models ,Potassium channel ,medicine.anatomical_structure ,Hypertension ,cardiovascular system ,Medicine ,Female ,Neurochemicals ,Research Article ,medicine.medical_specialty ,Cell Physiology ,Endothelium ,Signaling in cellular processes ,Biology ,Nitric Oxide ,Cardiovascular Pharmacology ,03 medical and health sciences ,Model Organisms ,Vascular Biology ,Internal medicine ,medicine ,Cyclic GMP-Dependent Protein Kinases ,Animals ,Phosphodiesterase inhibitor ,Rats, Wistar ,Protein kinase A ,030304 developmental biology ,Cyclic Nucleotide Phosphodiesterases, Type 5 ,lcsh:R ,Phosphodiesterase 5 Inhibitors ,equipment and supplies ,Rats ,Endocrinology ,cGMP signaling ,lcsh:Q ,Endothelium, Vascular ,cGMP-dependent protein kinase - Abstract
A growing body of evidence suggests that hydrogen sulfide (H₂S) is a signaling molecule in mammalian cells. In the cardiovascular system, H₂S enhances vasodilation and angiogenesis. H₂S-induced vasodilation is hypothesized to occur through ATP-sensitive potassium channels (K(ATP)); however, we recently demonstrated that it also increases cGMP levels in tissues. Herein, we studied the involvement of cGMP-dependent protein kinase-I in H₂S-induced vasorelaxation. The effect of H₂S on vessel tone was studied in phenylephrine-contracted aortic rings with or without endothelium. cGMP levels were determined in cultured cells or isolated vessel by enzyme immunoassay. Pretreatment of aortic rings with sildenafil attenuated NaHS-induced relaxation, confirming previous findings that H₂S is a phosphodiesterase inhibitor. In addition, vascular tissue levels of cGMP in cystathionine gamma lyase knockouts were lower than those in wild-type control mice. Treatment of aortic rings with NaHS, a fast releasing H₂S donor, enhanced phosphorylation of vasodilator-stimulated phosphoprotein in a time-dependent manner, suggesting that cGMP-dependent protein kinase (PKG) is activated after exposure to H₂S. Incubation of aortic rings with a PKG-I inhibitor (DT-2) attenuated NaHS-stimulated relaxation. Interestingly, vasodilatory responses to a slowly releasing H₂S donor (GYY 4137) were unaffected by DT-2, suggesting that this donor dilates mouse aorta through PKG-independent pathways. Dilatory responses to NaHS and L-cysteine (a substrate for H₂S production) were reduced in vessels of PKG-I knockout mice (PKG-I⁻/⁻). Moreover, glibenclamide inhibited NaHS-induced vasorelaxation in vessels from wild-type animals, but not PKG-I⁻/⁻, suggesting that there is a cross-talk between K(ATP) and PKG. Our results confirm the role of cGMP in the vascular responses to NaHS and demonstrate that genetic deletion of PKG-I attenuates NaHS and L-cysteine-stimulated vasodilation.
- Published
- 2012
3. Correction: Hippocampal neural stem cells and microglia response to experimental inflammatory bowel disease (IBD).
- Author
-
Gampierakis IA, Koutmani Y, Semitekolou M, Morianos I, Polissidis A, Katsouda A, Charalampopoulos I, Xanthou G, Gravanis A, and Karalis KP
- Published
- 2021
- Full Text
- View/download PDF
4. Hippocampal neural stem cells and microglia response to experimental inflammatory bowel disease (IBD).
- Author
-
Gampierakis IA, Koutmani Y, Semitekolou M, Morianos I, Polissidis A, Katsouda A, Charalampopoulos I, Xanthou G, Gravanis A, and Karalis KP
- Subjects
- Animals, Humans, Mice, Cytokines metabolism, Dextran Sulfate toxicity, Disease Models, Animal, Hippocampus metabolism, Mice, Inbred C57BL, Microglia metabolism, Colitis chemically induced, Inflammatory Bowel Diseases, Neural Stem Cells metabolism
- Abstract
Inflammatory bowel disease (IBD), including Crohn's disease (CD) and ulcerative colitis (UC), is a disease associated with dysbiosis, resulting in compromised intestinal epithelial barrier and chronic mucosal inflammation. Patients with IBD present with increased incidence of psychiatric disorders and cognitive impairment. Hippocampus is a brain region where adult neurogenesis occurs with functional implications in mood control and cognition. Using a well-established model of experimental colitis based on the administration of dextran sodium sulfate (DSS) in the drinking water, we sought to characterize the short and long-term effects of colitis on neurogenesis and glia responses in the hippocampus. We show that acute DSS colitis enhanced neurogenesis but with deficits in cell cycle kinetics of proliferating progenitors in the hippocampus. Chronic DSS colitis was characterized by normal levels of neurogenesis but with deficits in the migration and integration of newborn neurons in the functional circuitry of the DG. Notably, we found that acute DSS colitis-induced enhanced infiltration of the hippocampus with macrophages and inflammatory myeloid cells from the periphery, along with elevated frequencies of inflammatory M1-like microglia and increased release of pro-inflammatory cytokines. In contrast, increased percentages of tissue-repairing M2-like microglia, along with elevated levels of the anti-inflammatory cytokine, IL-10 were observed in the hippocampus during chronic DSS colitis. These findings uncover key effects of acute and chronic experimental colitis on adult hippocampal neurogenesis and innate immune cell responses, highlighting the potential mechanisms underlying cognitive and mood dysfunction in patients with IBD.
- Published
- 2021
- Full Text
- View/download PDF
5. CRH Promotes the Neurogenic Activity of Neural Stem Cells in the Adult Hippocampus.
- Author
-
Koutmani Y, Gampierakis IA, Polissidis A, Ximerakis M, Koutsoudaki PN, Polyzos A, Agrogiannis G, Karaliota S, Thomaidou D, Rubin LL, Politis PK, and Karalis KP
- Subjects
- Animals, Cell Line, Cells, Cultured, Corticotropin-Releasing Hormone genetics, Hippocampus cytology, Hippocampus physiology, Humans, Male, Mice, Mice, Inbred C57BL, Neural Stem Cells cytology, Receptors, Corticotropin-Releasing Hormone genetics, Receptors, Corticotropin-Releasing Hormone metabolism, Spatial Memory, Corticotropin-Releasing Hormone metabolism, Hippocampus metabolism, Neural Stem Cells metabolism, Neurogenesis
- Abstract
Local cues in the adult neurogenic niches dynamically regulate homeostasis in neural stem cells, whereas their identity and associated molecular mechanisms remain poorly understood. Here, we show that corticotropin-releasing hormone (CRH), the major mediator of mammalian stress response and a key neuromodulator in the adult brain, is necessary for hippocampal neural stem cell (hiNSC) activity under physiological conditions. In particular, we demonstrate functionality of the CRH/CRH receptor (CRHR) system in mouse hiNSCs and conserved expression in humans. Most important, we show that genetic deficiency of CRH impairs hippocampal neurogenesis, affects spatial memory, and compromises hiNSCs' responsiveness to environmental stimuli. These deficits have been partially restored by virus-mediated CRH expression. Additionally, we provide evidence that local disruption of the CRH/CRHR system reduces neurogenesis, while exposure of adult hiNSCs to CRH promotes neurogenic activity via BMP4 suppression. Our findings suggest a critical role of CRH in adult neurogenesis, independently of its stress-related systemic function., (Copyright © 2019 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
6. CD8+ T cells in beige adipogenesis and energy homeostasis.
- Author
-
Moysidou M, Karaliota S, Kodela E, Salagianni M, Koutmani Y, Katsouda A, Kodella K, Tsakanikas P, Ourailidou S, Andreakos E, Kostomitsopoulos N, Skokos D, Chatzigeorgiou A, Chung KJ, Bornstein S, Sleeman MW, Chavakis T, and Karalis KP
- Subjects
- Adipose Tissue, Beige cytology, Adipose Tissue, Beige immunology, Adipose Tissue, White cytology, Adipose Tissue, White immunology, Adipose Tissue, White metabolism, Adoptive Transfer, Animals, CD8 Antigens genetics, CD8 Antigens metabolism, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes transplantation, Cell Differentiation physiology, Disease Models, Animal, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, Humans, Interferon-gamma genetics, Interferon-gamma metabolism, Lipid Metabolism physiology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Models, Animal, Obesity genetics, Obesity immunology, Adipogenesis physiology, Adipose Tissue, Beige metabolism, CD8-Positive T-Lymphocytes metabolism, Energy Metabolism immunology, Obesity metabolism
- Abstract
Although accumulation of lymphocytes in the white adipose tissue (WAT) in obesity is linked to insulin resistance, it remains unclear whether lymphocytes also participate in the regulation of energy homeostasis in the WAT. Here, we demonstrate enhanced energy dissipation in Rag1-/- mice, increased catecholaminergic input to subcutaneous WAT, and significant beige adipogenesis. Adoptive transfer experiments demonstrated that CD8+ T cell deficiency accounts for the enhanced beige adipogenesis in Rag1-/- mice. Consistently, we identified that CD8-/- mice also presented with enhanced beige adipogenesis. The inhibitory effect of CD8+ T cells on beige adipogenesis was reversed by blockade of IFN-γ. All together, our findings identify an effect of CD8+ T cells in regulating energy dissipation in lean WAT, mediated by IFN-γ modulation of the abundance of resident immune cells and of local catecholaminergic activity. Our results provide a plausible explanation for the clinical signs of metabolic dysfunction in diseases characterized by altered CD8+ T cell abundance and suggest targeting of CD8+ T cells as a promising therapeutic approach for obesity and other diseases with altered energy homeostasis.
- Published
- 2018
- Full Text
- View/download PDF
7. Mapping metabolic changes by noninvasive, multiparametric, high-resolution imaging using endogenous contrast.
- Author
-
Liu Z, Pouli D, Alonzo CA, Varone A, Karaliota S, Quinn KP, Münger K, Karalis KP, and Georgakoudi I
- Subjects
- Adipose Tissue, Brown drug effects, Adipose Tissue, Brown metabolism, Animals, Carbonyl Cyanide m-Chlorophenyl Hydrazone pharmacology, Cell Line, Fatty Acids biosynthesis, Flavin-Adenine Dinucleotide metabolism, Fluorescence, Glutamine metabolism, Glycolysis, Humans, Mice, Mitochondria drug effects, Mitochondria metabolism, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, NAD metabolism, Oxidation-Reduction drug effects, Imaging, Three-Dimensional methods, Metabolism drug effects
- Abstract
Monitoring subcellular functional and structural changes associated with metabolism is essential for understanding healthy tissue development and the progression of numerous diseases, including cancer, diabetes, and cardiovascular and neurodegenerative disorders. Unfortunately, established methods for this purpose either are destructive or require the use of exogenous agents. Recent work has highlighted the potential of endogenous two-photon excited fluorescence (TPEF) as a method to monitor subtle metabolic changes; however, mechanistic understanding of the connections between the detected optical signal and the underlying metabolic pathways has been lacking. We present a quantitative approach to detecting both functional and structural metabolic biomarkers noninvasively, relying on endogenous TPEF from two coenzymes, NADH (reduced form of nicotinamide adenine dinucleotide) and FAD (flavin adenine dinucleotide). We perform multiparametric analysis of three optical biomarkers within intact, living cells and three-dimensional tissues: cellular redox state, NADH fluorescence lifetime, and mitochondrial clustering. We monitor the biomarkers in cells and tissues subjected to metabolic perturbations that trigger changes in distinct metabolic processes, including glycolysis and glutaminolysis, extrinsic and intrinsic mitochondrial uncoupling, and fatty acid oxidation and synthesis. We demonstrate that these optical biomarkers provide complementary insights into the underlying biological mechanisms. Thus, when used in combination, these biomarkers can serve as a valuable tool for sensitive, label-free identification of changes in specific metabolic pathways and characterization of the heterogeneity of the elicited responses with single-cell resolution.
- Published
- 2018
- Full Text
- View/download PDF
8. Strain-specific Differences in the Effects of Lymphocytes on the Development of Insulin Resistance and Obesity in Mice.
- Author
-
Kodela E, Moysidou M, Karaliota S, Koutmani Y, Tsakanikas P, Kodella K, Karavia EA, Kypreos KE, Kostomitsopoulos N, and Karalis KP
- Subjects
- Adipose Tissue metabolism, Animals, Diet, High-Fat, Disease Susceptibility, Mice, Inbred BALB C, Mice, Inbred C57BL, Uncoupling Protein 1 metabolism, Insulin Resistance immunology, Lymphocytes physiology, Obesity immunology
- Abstract
Obesity is characterized as a chronic, low-grade inflammatory disease owing to the infiltration of the adipose tissue by macrophages. Although the role of macrophages in this process is well established, the role of lymphocytes in the development of obesity and metabolism remains less well defined. In the current study, we fed WT and Rag1-/- male mice, of C57BL/6J and BALB/c backgrounds, high-fat diet (HFD) or normal diet for 15 wk. Compared with WT mice, Rag1-/- mice of either of the examined strains were found less prone to insulin resistance after HFD, had higher metabolic rates, and used lipids more efficiently, as shown by the increased expression of genes related to fatty acid oxidation in epidydimal white adipose tissue. Furthermore, Rag1-/- mice had increased Ucp1 protein expression and associated phenotypic characteristics indicative of beige adipose tissue in subcutaneous white adipose tissue and increased Ucp1 expression in brown adipose tissue. As with inflammatory and other physiologic responses previously reported, the responses of mice to HFD show strain-specific differences, with increased susceptibility of C57BL/6J as compared with BALB/c strain. Our findings unmask a crucial role for lymphocytes in the development of obesity and insulin resistance, in that lymphocytes inhibit efficient dissipation of energy by adipose tissue. These strain-associated differences highlight important metabolic factors that should be accommodated in disease modeling and drug testing.
- Published
- 2018
9. A self-sustained loop of inflammation-driven inhibition of beige adipogenesis in obesity.
- Author
-
Chung KJ, Chatzigeorgiou A, Economopoulou M, Garcia-Martin R, Alexaki VI, Mitroulis I, Nati M, Gebler J, Ziemssen T, Goelz SE, Phieler J, Lim JH, Karalis KP, Papayannopoulou T, Blüher M, Hajishengallis G, and Chavakis T
- Subjects
- 3T3-L1 Cells, Adipocytes immunology, Adipocytes metabolism, Adult, Aged, Aged, 80 and over, Animals, Cell Adhesion immunology, Diet, High-Fat, Down-Regulation, Extracellular Signal-Regulated MAP Kinases metabolism, Feedback, Female, Gene Knockdown Techniques, Humans, Immunoblotting, Integrin alpha4 genetics, Macrophages metabolism, Male, Mice, Middle Aged, Monocytes immunology, Obesity metabolism, RNA, Messenger metabolism, Real-Time Polymerase Chain Reaction, Subcutaneous Fat, T-Lymphocytes immunology, Uncoupling Protein 1 genetics, Uncoupling Protein 1 metabolism, Vascular Cell Adhesion Molecule-1 genetics, Vascular Cell Adhesion Molecule-1 metabolism, Young Adult, Adipocytes, Beige, Adipogenesis immunology, Adipose Tissue, White immunology, Cell Differentiation immunology, Inflammation immunology, Macrophages immunology, Obesity immunology
- Abstract
In obesity, inflammation of white adipose tissue (AT) is associated with diminished generation of beige adipocytes ('beige adipogenesis'), a thermogenic and energy-dissipating function mediated by beige adipocytes that express the uncoupling protein UCP1. Here we delineated an inflammation-driven inhibitory mechanism of beige adipogenesis in obesity that required direct adhesive interactions between macrophages and adipocytes mediated by the integrin α
4 and its counter-receptor VCAM-1, respectively; expression of the latter was upregulated in obesity. This adhesive interaction reciprocally and concomitantly modulated inflammatory activation of macrophages and downregulation of UCP1 expression dependent on the kinase Erk in adipocytes. Genetic or pharmacological inactivation of the integrin α4 in mice resulted in elevated expression of UCP1 and beige adipogenesis of subcutaneous AT in obesity. Our findings, established in both mouse systems and human systems, reveal a self-sustained cycle of inflammation-driven impairment of beige adipogenesis in obesity.- Published
- 2017
- Full Text
- View/download PDF
10. Metformin protects against infection-induced myocardial dysfunction.
- Author
-
Tzanavari T, Varela A, Theocharis S, Ninou E, Kapelouzou A, Cokkinos DV, Kontaridis MI, and Karalis KP
- Subjects
- Adenosine Triphosphate metabolism, Animals, Autophagy drug effects, Cardiomyopathies physiopathology, Echocardiography, Fatty Acids metabolism, Glucose metabolism, Heart Diseases chemically induced, Heart Diseases diagnostic imaging, Heart Diseases prevention & control, Inflammation Mediators metabolism, Lipopolysaccharides pharmacology, Male, Mice, Mice, Inbred C57BL, Myocardium metabolism, Cardiomyopathies etiology, Cardiomyopathies prevention & control, Gram-Negative Bacterial Infections complications, Hypoglycemic Agents therapeutic use, Metformin therapeutic use
- Abstract
Background and Purpose: Metformin administration is associated with myocardial protection during ischemia and/or reperfusion, possibly via inhibition of inflammatory responses in the heart. Exposure to pathogens, in addition to the activation of the immune system and the associated metabolic dysfunction, often results in compromised myocardial function. We examined whether metformin administration could maintain the normal myocardial function in experimental moderate Gram negative infection, induced by lipopolysaccharide (LPS) administration., Experimental Approach: 129xC57BL/6 mice were divided into control groups that received either vehicle or a single intraperitoneal (i.p.) injection of low dose LPS (5mg/kg body wt), and metformin treated groups that received either daily metformin (4mg/kg/animal) i.p. injections for five days prior to LPS administration [Experiment 1], or a single metformin injection following same dose of LPS [Experiment 2]., Key Results: LPS alone caused cardiac dysfunction, as confirmed by echocardiography, whereas metformin administration, either before or after LPS, rescued myocardial function. LPS caused marked reduction of the cardiac metabolism-related genes tested, including Prkaa2, Cpt1b, Ppargc1a and Ppargc1b; reduction of fatty acid oxidation, as reflected by the regulation of Ppara, Acaca and Acacb; increased glucose transport, as shown by Slc2a4 levels; reduction of ATP synthesis; significant increase of inflammatory markers, in particular IL6; and reduction of autophagy. Pretreatment with metformin normalized the levels of all these factors., Conclusions and Implications: We show for the first time that metformin protects the myocardium from LPS-associated myocardial dysfunction mainly by supporting its metabolic activity and allowing efficient energy utilization. Metformin can be a potential cardioprotective agent in individuals susceptible to exposure to pathogens., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
11. Two-photon excited fluorescence of intrinsic fluorophores enables label-free assessment of adipose tissue function.
- Author
-
Alonzo CA, Karaliota S, Pouli D, Liu Z, Karalis KP, and Georgakoudi I
- Subjects
- Humans, Adipocytes physiology, Adipose Tissue physiology, Optical Imaging methods
- Abstract
Current methods for evaluating adipose tissue function are destructive or have low spatial resolution. These limit our ability to assess dynamic changes and heterogeneous responses that occur in healthy or diseased subjects, or during treatment. Here, we demonstrate that intrinsic two-photon excited fluorescence enables functional imaging of adipocyte metabolism with subcellular resolution. Steady-state and time-resolved fluorescence from intracellular metabolic co-factors and lipid droplets can distinguish the functional states of excised white, brown, and cold-induced beige fat. Similar optical changes are identified when white and brown fat are assessed in vivo. Therefore, these studies establish the potential of non-invasive, high resolution, endogenous contrast, two-photon imaging to identify distinct adipose tissue types, monitor their functional state, and characterize heterogeneity of induced responses.
- Published
- 2016
- Full Text
- View/download PDF
12. Identification of a novel interaction between corticotropin releasing hormone (Crh) and macroautophagy.
- Author
-
Giannogonas P, Apostolou A, Manousopoulou A, Theocharis S, Macari SA, Psarras S, Garbis SD, Pothoulakis C, and Karalis KP
- Subjects
- Adenine analogs & derivatives, Adenine pharmacology, Animals, Autophagy drug effects, Cells, Cultured, Colitis chemically induced, Colitis genetics, Disease Models, Animal, Fibroblasts cytology, Fibroblasts drug effects, Fibroblasts metabolism, Gastrointestinal Tract metabolism, Gene Knockout Techniques, Male, Mice, Proteomics methods, RAW 264.7 Cells, Colitis metabolism, Corticotropin-Releasing Hormone genetics, Corticotropin-Releasing Hormone metabolism, Dextran Sulfate toxicity
- Abstract
In inflammatory bowel disease (IBD), compromised restitution of the epithelial barrier contributes to disease severity. Owing to the complexity in the pathogenesis of IBD, a variety of factors have been implicated in its progress. In this study, we report a functional interaction between macroautophagy and Corticotropin Releasing Hormone (Crh) in the gut. For this purpose we used DSS colitis model on Crh -/- or wild-type (wt) with pharmacological inhibition of autophagy. We uncovered sustained basal autophagy in the gut of Crh -/- mice, which persisted over the course of DSS administration. Autophagy inhibition resulted in partial rescue of Crh -/- mice, while it increased the expression of Crh in the wt gut. Similarly, Crh deficiency was associated with sustained activation of base line autophagy. In vitro models of amino acid deprivation- and LPS-induced autophagy confirmed the in vivo findings. Our results indicate a novel role for Crh in the intestinal epithelium that involves regulation of autophagy, while suggesting the complementary action of the two pathways. These data suggest the intriguing possibility that targeting Crh stimulation in the intestine may provide a novel therapeutic approach to support the integrity of the epithelial barrier and to protect from chronic colitis.
- Published
- 2016
- Full Text
- View/download PDF
13. Neural stem cells respond to stress hormones: distinguishing beneficial from detrimental stress.
- Author
-
Koutmani Y and Karalis KP
- Abstract
Neural stem cells (NSCs), the progenitors of the nervous system, control distinct, position-specific functions and are critically involved in the maintenance of homeostasis in the brain. The responses of these cells to various stressful stimuli are shaped by genetic, epigenetic, and environmental factors via mechanisms that are age and developmental stage-dependent and still remain, to a great extent, elusive. Increasing evidence advocates for the beneficial impact of the stress response in various settings, complementing the extensive number of studies on the detrimental effects of stress, particularly in the developing brain. In this review, we discuss suggested mechanisms mediating both the beneficial and detrimental effects of stressors on NSC activity across the lifespan. We focus on the specific effects of secreted factors and we propose NSCs as a "sensor," capable of distinguishing among the different stressors and adapting its functions accordingly. All the above suggest the intriguing hypothesis that NSCs are an important part of the adaptive response to stressors via direct and indirect, specific mechanisms.
- Published
- 2015
- Full Text
- View/download PDF
14. Dual role of B7 costimulation in obesity-related nonalcoholic steatohepatitis and metabolic dysregulation.
- Author
-
Chatzigeorgiou A, Chung KJ, Garcia-Martin R, Alexaki VI, Klotzsche-von Ameln A, Phieler J, Sprott D, Kanczkowski W, Tzanavari T, Bdeir M, Bergmann S, Cartellieri M, Bachmann M, Nikolakopoulou P, Androutsellis-Theotokis A, Siegert G, Bornstein SR, Muders MH, Boon L, Karalis KP, Lutgens E, and Chavakis T
- Subjects
- Animals, B7 Antigens deficiency, B7 Antigens genetics, Cell Communication physiology, Disease Models, Animal, Liver pathology, Male, Mice, Mice, Knockout, Phenotype, T-Lymphocytes, Regulatory pathology, B7 Antigens physiology, Metabolic Syndrome physiopathology, Non-alcoholic Fatty Liver Disease physiopathology, Obesity physiopathology
- Abstract
Unlabelled: The low-grade inflammatory state present in obesity contributes to obesity-related metabolic dysregulation, including nonalcoholic steatohepatitis (NASH) and insulin resistance. Intercellular interactions between immune cells or between immune cells and hepatic parenchymal cells contribute to the exacerbation of liver inflammation and steatosis in obesity. The costimulatory molecules, B7.1 and B7.2, are important regulators of cell-cell interactions in several immune processes; however, the role of B7 costimulation in obesity-related liver inflammation is unknown. Here, diet-induced obesity (DIO) studies in mice with genetic inactivation of both B7.1 and B7.2 (double knockout; DKO) revealed aggravated obesity-related metabolic dysregulation, reduced insulin signalling in the liver and adipose tissue (AT), glucose intolerance, and enhanced progression to steatohepatitis resulting from B7.1/B7.2 double deficiency. The metabolic phenotype of B7.1/B7.2 double deficiency upon DIO was accompanied by increased hepatic and AT inflammation, associated with largely reduced numbers of regulatory T cells (Tregs) in these organs. In order to assess the role of B7 costimulation in DIO in a non-Treg-lacking environment, we performed antibody (Ab)-mediated inhibition of B7 molecules in wild-type mice in DIO. Antibody-blockade of both B7.1 and B7.2 improved the metabolic phenotype of DIO mice, which was linked to amelioration of hepatic steatosis and reduced inflammation in liver and AT., Conclusion: Our study demonstrates a dual role of B7 costimulation in the course of obesity-related sequelae, particularly NASH. The genetic inactivation of B7.1/B7.2 deteriorates obesity-related liver steatosis and metabolic dysregulation, likely a result of the intrinsic absence of Tregs in these mice, rendering DKO mice a novel murine model of NASH. In contrast, inhibition of B7 costimulation under conditions where Tregs are present may provide a novel therapeutic approach for obesity-related metabolic dysregulation and, especially, NASH., (© 2014 by the American Association for the Study of Liver Diseases.)
- Published
- 2014
- Full Text
- View/download PDF
15. The complement anaphylatoxin C5a receptor contributes to obese adipose tissue inflammation and insulin resistance.
- Author
-
Phieler J, Chung KJ, Chatzigeorgiou A, Klotzsche-von Ameln A, Garcia-Martin R, Sprott D, Moisidou M, Tzanavari T, Ludwig B, Baraban E, Ehrhart-Bornstein M, Bornstein SR, Mziaut H, Solimena M, Karalis KP, Economopoulou M, Lambris JD, and Chavakis T
- Subjects
- Adipocytes immunology, Adipocytes metabolism, Animals, Complement C5a metabolism, Dietary Fats immunology, Dietary Fats metabolism, Female, Fibrosis immunology, Inflammation immunology, Insulin-Secreting Cells metabolism, Interleukin-10 biosynthesis, Macrophage Activation immunology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Obesity immunology, Obesity metabolism, Receptor, Anaphylatoxin C5a biosynthesis, Receptor, Anaphylatoxin C5a immunology, Up-Regulation, Adipose Tissue immunology, Adipose Tissue metabolism, Insulin Resistance immunology, Macrophages immunology, Receptor, Anaphylatoxin C5a metabolism
- Abstract
Obese adipose tissue (AT) inflammation contributes critically to development of insulin resistance. The complement anaphylatoxin C5a receptor (C5aR) has been implicated in inflammatory processes and as regulator of macrophage activation and polarization. However, the role of C5aR in obesity and AT inflammation has not been addressed. We engaged the model of diet-induced obesity and found that expression of C5aR was significantly upregulated in the obese AT, compared with lean AT. In addition, C5a was present in obese AT in the proximity of macrophage-rich crownlike structures. C5aR-sufficient and -deficient mice were fed a high-fat diet (HFD) or a normal diet (ND). C5aR deficiency was associated with increased AT weight upon ND feeding in males, but not in females, and with increased adipocyte size upon ND and HFD conditions in males. However, obese C5aR(-/-) mice displayed improved systemic and AT insulin sensitivity. Improved AT insulin sensitivity in C5aR(-/-) mice was associated with reduced accumulation of total and proinflammatory M1 macrophages in the obese AT, increased expression of IL-10, and decreased AT fibrosis. In contrast, no difference in β cell mass was observed owing to C5aR deficiency under an HFD. These results suggest that C5aR contributes to macrophage accumulation and M1 polarization in the obese AT and thereby to AT dysfunction and development of AT insulin resistance.
- Published
- 2013
- Full Text
- View/download PDF
16. Corticotropin-releasing hormone exerts direct effects on neuronal progenitor cells: implications for neuroprotection.
- Author
-
Koutmani Y, Politis PK, Elkouris M, Agrogiannis G, Kemerli M, Patsouris E, Remboutsika E, and Karalis KP
- Subjects
- Animals, Apoptosis physiology, Brain metabolism, Cell Proliferation drug effects, Corticotropin-Releasing Hormone antagonists & inhibitors, Corticotropin-Releasing Hormone genetics, Corticotropin-Releasing Hormone metabolism, Dexamethasone antagonists & inhibitors, Dexamethasone toxicity, Humans, Mice, Mice, Knockout, Neurogenesis drug effects, Pyrimidines pharmacology, Pyrroles pharmacology, Receptors, Corticotropin-Releasing Hormone agonists, Receptors, Corticotropin-Releasing Hormone metabolism, Receptors, Corticotropin-Releasing Hormone physiology, Signal Transduction drug effects, Signal Transduction physiology, Stem Cells drug effects, Brain drug effects, Corticotropin-Releasing Hormone pharmacology, Neurogenesis physiology, Neuroprotective Agents pharmacology, Stem Cells physiology
- Abstract
Neurogenesis during embryonic and adult life is tightly regulated by a network of transcriptional, growth and hormonal factors. Emerging evidence indicates that activation of the stress response, via the associated glucocorticoid increase, reduces neurogenesis and contributes to the development of adult diseases.As corticotrophin-releasing hormone (CRH) or factor is the major mediator of adaptive response to stressors, we sought to investigate its involvement in this process. Accordingly, we found that CRH could reverse the damaging effects of glucocorticoid on neural stem/progenitor cells (NS/PCs), while its genetic deficiency results in compromised proliferation and enhanced apoptosis during neurogenesis. Analyses in fetal and adult mouse brain revealed significant expression of CRH receptors in proliferating neuronal progenitors. Furthermore, by using primary cultures of NS/PCs, we characterized the molecular mechanisms and identified CRH receptor-1 as the receptor mediating the neuroprotective effects of CRH. Finally, we demonstrate the expression of CRH receptors in human fetal brain from early gestational age, in areas of active neuronal proliferation. These observations raise the intriguing possibility for CRH-mediated pharmacological applications in diseases characterized by altered neuronal homeostasis, including depression, dementia, neurodegenerative diseases, brain traumas and obesity.
- Published
- 2013
- Full Text
- View/download PDF
17. Lymphocytes in obesity-related adipose tissue inflammation.
- Author
-
Chatzigeorgiou A, Karalis KP, Bornstein SR, and Chavakis T
- Subjects
- Adipose Tissue, White pathology, Animals, B-Lymphocyte Subsets physiology, Disease Models, Animal, Humans, Inflammation pathology, Leptin physiology, Mice, Obesity pathology, T-Lymphocyte Subsets physiology, Adipose Tissue, White physiopathology, Inflammation physiopathology, Lymphocytes physiology, Obesity physiopathology
- Abstract
Inflammation in the white adipose tissue (WAT) is considered a major player in the development of insulin resistance. The role of macrophages accumulating in the WAT during obesity, promoting WAT inflammation and insulin resistance is well established. In contrast, less is known about the role of lymphocytes. Recent studies have implicated different lymphocyte subsets in WAT inflammation. For instance, cytotoxic CD8(+) T cells infiltrating the WAT may contribute to the recruitment, differentiation and activation of macrophages. On the other hand, a differential role for CD4(+) Th1 and CD4(+) Th2 cells has been suggested. Levels of WAT regulatory T cells decrease during the course of obesity and may represent a crucial factor for the maintenance of insulin sensitivity. Moreover, activation of natural killer T cells, an innate-like T cell population, which recognises lipid antigens, promotes insulin resistance and WAT inflammation. Finally, B cells may infiltrate WAT very early in response to high-fat feeding and worsen glucose metabolism through modulation of T cells and the production of pathogenic antibodies. These interesting new findings however bear controversies and introduce novel, yet unanswered, questions. Here, we review and discuss the impact of the different lymphocyte subsets in obesity-related WAT inflammation and attempt to identify the open questions to be answered by future studies.
- Published
- 2012
- Full Text
- View/download PDF
18. cGMP-dependent protein kinase contributes to hydrogen sulfide-stimulated vasorelaxation.
- Author
-
Bucci M, Papapetropoulos A, Vellecco V, Zhou Z, Zaid A, Giannogonas P, Cantalupo A, Dhayade S, Karalis KP, Wang R, Feil R, and Cirino G
- Subjects
- Animals, Aorta drug effects, Aorta physiology, Cells, Cultured, Cyclic GMP-Dependent Protein Kinases antagonists & inhibitors, Cyclic GMP-Dependent Protein Kinases genetics, Cyclic GMP-Dependent Protein Kinases metabolism, Cyclic Nucleotide Phosphodiesterases, Type 5 metabolism, Cyclic Nucleotide Phosphodiesterases, Type 5 physiology, Endothelium, Vascular drug effects, Endothelium, Vascular physiology, Female, Male, Mice, Mice, Knockout, Phosphodiesterase 5 Inhibitors pharmacology, Rats, Rats, Wistar, Vasodilation genetics, Cyclic GMP-Dependent Protein Kinases physiology, Hydrogen Sulfide pharmacology, Vasodilation drug effects
- Abstract
A growing body of evidence suggests that hydrogen sulfide (H₂S) is a signaling molecule in mammalian cells. In the cardiovascular system, H₂S enhances vasodilation and angiogenesis. H₂S-induced vasodilation is hypothesized to occur through ATP-sensitive potassium channels (K(ATP)); however, we recently demonstrated that it also increases cGMP levels in tissues. Herein, we studied the involvement of cGMP-dependent protein kinase-I in H₂S-induced vasorelaxation. The effect of H₂S on vessel tone was studied in phenylephrine-contracted aortic rings with or without endothelium. cGMP levels were determined in cultured cells or isolated vessel by enzyme immunoassay. Pretreatment of aortic rings with sildenafil attenuated NaHS-induced relaxation, confirming previous findings that H₂S is a phosphodiesterase inhibitor. In addition, vascular tissue levels of cGMP in cystathionine gamma lyase knockouts were lower than those in wild-type control mice. Treatment of aortic rings with NaHS, a fast releasing H₂S donor, enhanced phosphorylation of vasodilator-stimulated phosphoprotein in a time-dependent manner, suggesting that cGMP-dependent protein kinase (PKG) is activated after exposure to H₂S. Incubation of aortic rings with a PKG-I inhibitor (DT-2) attenuated NaHS-stimulated relaxation. Interestingly, vasodilatory responses to a slowly releasing H₂S donor (GYY 4137) were unaffected by DT-2, suggesting that this donor dilates mouse aorta through PKG-independent pathways. Dilatory responses to NaHS and L-cysteine (a substrate for H₂S production) were reduced in vessels of PKG-I knockout mice (PKG-I⁻/⁻). Moreover, glibenclamide inhibited NaHS-induced vasorelaxation in vessels from wild-type animals, but not PKG-I⁻/⁻, suggesting that there is a cross-talk between K(ATP) and PKG. Our results confirm the role of cGMP in the vascular responses to NaHS and demonstrate that genetic deletion of PKG-I attenuates NaHS and L-cysteine-stimulated vasodilation.
- Published
- 2012
- Full Text
- View/download PDF
19. Modulation of pancreatic islets-stress axis by hypothalamic releasing hormones and 11beta-hydroxysteroid dehydrogenase.
- Author
-
Schmid J, Ludwig B, Schally AV, Steffen A, Ziegler CG, Block NL, Koutmani Y, Brendel MD, Karalis KP, Simeonovic CJ, Licinio J, Ehrhart-Bornstein M, and Bornstein SR
- Subjects
- Animals, Corticotropin-Releasing Hormone, Humans, Insulin biosynthesis, Insulinoma pathology, RNA, Messenger, Rats, Receptors, Corticotropin-Releasing Hormone metabolism, Receptors, Neuropeptide metabolism, Receptors, Pituitary Hormone-Regulating Hormone metabolism, 11-beta-Hydroxysteroid Dehydrogenases physiology, Hypothalamo-Hypophyseal System metabolism, Islets of Langerhans metabolism, Pituitary Hormone-Releasing Hormones physiology, Pituitary-Adrenal System metabolism
- Abstract
Corticotropin-releasing hormone (CRH) and growth hormone-releasing hormone (GHRH), primarily characterized as neuroregulators of the hypothalamic-pituitary-adrenal axis, directly influence tissue-specific receptor-systems for CRH and GHRH in the endocrine pancreas. Here, we demonstrate the expression of mRNA for CRH and CRH-receptor type 1 (CRHR1) and of protein for CRHR1 in rat and human pancreatic islets and rat insulinoma cells. Activation of CRHR1 and GHRH-receptor significantly increased cell proliferation and reduced cell apoptosis. CRH stimulated both cellular content and release of insulin in rat islet and insulinoma cells. At the ultrastructural level, CRHR1 stimulation revealed a more active metabolic state with enlarged mitochondria. Moreover, glucocorticoids that promote glucose production are balanced by both 11b-hydroxysteroid dehydrogenase (11β-HSD) isoforms; 11β-HSD-type-1 and 11β-HSD-type-2. We demonstrated expression of mRNA for 11β-HSD-1 and 11β-HSD-2 and protein for 11β-HSD-1 in rat and human pancreatic islets and insulinoma cells. Quantitative real-time PCR revealed that stimulation of CRHR1 and GHRH-receptor affects the metabolism of insulinoma cells by down-regulating 11β-HSD-1 and up-regulating 11β-HSD-2. The 11β-HSD enzyme activity was analyzed by measuring the production of cortisol from cortisone. Similarly, activation of CRHR1 resulted in reduced cortisol levels, indicating either decreased 11β-HSD-1 enzyme activity or increased 11β-HSD-2 enzyme activity; thus, activation of CRHR1 alters the glucocorticoid balance toward the inactive form. These data indicate that functional receptor systems for hypothalamic-releasing hormone agonists exist within the endocrine pancreas and influence synthesis of insulin and the pancreatic glucocorticoid shuttle. Agonists of CRHR1 and GHRH-receptor, therefore, may play an important role as novel therapeutic tools in the treatment of diabetes mellitus.
- Published
- 2011
- Full Text
- View/download PDF
20. A novel role of peripheral corticotropin-releasing hormone (CRH) on dermal fibroblasts.
- Author
-
Rassouli O, Liapakis G, Lazaridis I, Sakellaris G, Gkountelias K, Gravanis A, Margioris AN, Karalis KP, and Venihaki M
- Subjects
- Animals, Antibodies, Neutralizing immunology, Apoptosis, Cell Movement, Cell Proliferation, Corticotropin-Releasing Hormone deficiency, Fibroblasts cytology, Humans, Interleukin-6 biosynthesis, Interleukin-6 immunology, Mice, Receptors, Corticotropin-Releasing Hormone metabolism, Transforming Growth Factor beta1 biosynthesis, Transforming Growth Factor beta1 immunology, Corticotropin-Releasing Hormone metabolism, Dermis cytology, Fibroblasts metabolism
- Abstract
Corticotropin-releasing hormone, or factor, (CRH or CRF) exerts important biological effects in multiple peripheral tissues via paracrine/autocrine actions. The aim of our study was to assess the effects of endogenous CRH in the biology of mouse and human skin fibroblasts, the primary cell type involved in wound healing. We show expression of CRH and its receptors in primary fibroblasts, and we demonstrate the functionality of fibroblast CRH receptors by induction of cAMP. Fibroblasts genetically deficient in Crh (Crh-/-) had higher proliferation and migration rates and compromised production of IL-6 and TGF-β1 compared to the wildtype (Crh+/+) cells. Human primary cultures of foreskin fibroblasts exposed to the CRF(1) antagonist antalarmin recapitulated the findings in the Crh-/- cells, exhibiting altered proliferative and migratory behavior and suppressed production of IL-6. In conclusion, our findings show an important role of fibroblast-expressed CRH in the proliferation, migration, and cytokine production of these cells, processes associated with the skin response to injury. Our data suggest that the immunomodulatory effects of CRH may include an important, albeit not explored yet, role in epidermal tissue remodeling and regeneration and maintenance of tissue homeostasis.
- Published
- 2011
- Full Text
- View/download PDF
21. Corticotropin-releasing factor regulates TLR4 expression in the colon and protects mice from colitis.
- Author
-
Chaniotou Z, Giannogonas P, Theoharis S, Teli T, Gay J, Savidge T, Koutmani Y, Brugni J, Kokkotou E, Pothoulakis C, and Karalis KP
- Subjects
- Acute Disease, Animals, Colitis chemically induced, Colitis mortality, Dextran Sulfate toxicity, Disease Models, Animal, Kaplan-Meier Estimate, Male, Mice, Mice, Inbred C57BL, Mice, Mutant Strains, Morbidity, Stress, Physiological immunology, Colitis immunology, Colon immunology, Corticotropin-Releasing Hormone genetics, Corticotropin-Releasing Hormone immunology, Corticotropin-Releasing Hormone metabolism, Toll-Like Receptor 4 genetics, Toll-Like Receptor 4 immunology, Toll-Like Receptor 4 metabolism
- Abstract
Background & Aims: Defects in the colonic innate immune response have been associated with inflammatory bowel disease (IBD). Corticotropin-releasing hormone (CRH, or corticotropin-releasing factor [CRF]) is a neuropeptide that mediates the stress response in humans, is an immunomodulatory factor with proinflammatory effects, and regulates transcription of Toll-like receptors (TLR)-2 and TLR4. We investigated the role of CRF in an innate immunity-dependent mouse model of IBD., Methods: Crh(-/-) and wild-type (Crh(+/+)) mice, which are glucocorticoid insufficient, were given dextran sodium sulfate in their drinking water to induce colitis; in some experiments, mice were also given glucocorticoids. Phenotypes of mice were compared; tissues were analyzed by histology and for expression of immune mediators., Results: Crh(-/-) mice had more colonic inflammation than Crh(+/+) mice, characterized by reduced numbers of crypts and severe epithelial damage and ulcerations. Colonic tissue levels of the proinflammatory factors interleukin-12 and prostaglandin E(2) were increased in the Crh(-/-) mice. Colons of Crh(-/-) mice expressed lower levels of Tlr4 than wild-type mice before, but not after, colitis was induced. Administration of glucocorticoid at low levels did not prevent Crh(-/-) mice from developing severe colitis. Crh(-/-) mice were unable to recover from acute colitis, as indicated by their increased death rate., Conclusions: Mice deficient in CRF down-regulate TLR4 and are more susceptible to dextran sodium sulfate-induced colitis. CRF has anti-inflammatory effects in innate immunity-dependent colitis and its recovery phase; these are independent of glucocorticoid administration. CRF might therefore be developed as a therapeutic target for patients with IBD., (Copyright © 2010 AGA Institute. Published by Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
22. TNF-alpha and obesity.
- Author
-
Tzanavari T, Giannogonas P, and Karalis KP
- Subjects
- Adipose Tissue immunology, Adipose Tissue physiopathology, Animals, Gene Expression, Humans, Insulin Resistance immunology, Insulin Resistance physiology, Mice, Models, Genetic, Obesity drug therapy, Obesity physiopathology, Tumor Necrosis Factor-alpha antagonists & inhibitors, Tumor Necrosis Factor-alpha genetics, Tumor Necrosis Factor-alpha physiology, Obesity immunology, Tumor Necrosis Factor-alpha immunology
- Abstract
Obesity, an epidemic of our times with rates rising to alarming levels, is associated with comorbidities including cardiovascular diseases, arthritis, certain cancers, and degenerative diseases of the brain and other organs. Importantly, obesity is a leading cause of insulin resistance and type 2 diabetes. As emerging evidence has shown over the last decade, inflammation is one of the critical processes associated with the development of insulin resistance, diabetes and related diseases, and obesity is now considered as a state of chronic low-grade inflammation. Adipose tissue, apart from its classical role as an energy storage depot, is also a major endocrine organ secreting many factors, whose local and circulating levels are affected by the degree of adiposity. Obesity leads to infiltration of the expanded adipose tissue by macrophages and increased levels in proinflammatory cytokines. The first indication for increased cytokine release in obesity was provided by the identification of increased expression of TNF-alpha, a proinflammatory cytokine, in the adipose tissue of obese mice in the early 1990s. TNF-alpha is expressed in and secreted by adipose tissue, its levels correlating with the degree of adiposity and the associated insulin resistance. Targeting TNF-alpha and/or its receptors has been suggested as a promising treatment for insulin resistance and type 2 diabetes. This review will summarize the available knowledge on the role of TNF-alpha in obesity and related processes and the potential implications of the above in the development of new therapeutic approaches for obesity and insulin resistance. Recent data from clinical studies will also be described together with late findings on the pathogenesis of obesity and insulin resistance., (Copyright (c) 2010 S. Karger AG, Basel.)
- Published
- 2010
- Full Text
- View/download PDF
23. Activation of phosphatidylinositol 3-kinase/protein kinase B by corticotropin-releasing factor in human monocytes.
- Author
-
Chandras C, Koutmani Y, Kokkotou E, Pothoulakis C, and Karalis KP
- Subjects
- Cell Line, Enzyme Activation, Extracellular Signal-Regulated MAP Kinases metabolism, Humans, MAP Kinase Signaling System, NF-kappa B metabolism, Corticotropin-Releasing Hormone metabolism, Monocytes enzymology, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism, Receptors, Corticotropin-Releasing Hormone metabolism
- Abstract
Corticotropin-releasing factor (CRF) exerts proinflammatory effects in peripheral tissues, whereas the intracellular pathways mediating these effects have not been completely characterized yet. We have previously shown that CRF induces nuclear factor-kappaB DNA-binding activity in mouse and human leukocytes. Here we demonstrate that in the human monocytic THP-1 cells, CRF activates the phosphatidylinositol 3-kinase (PI3K)/Akt and ERK1/2 pathways. These effects of CRF are mediated by corticotropin-releasing factor receptor 2 (CRF2), as suggested by their abolishment after treatment with the specific CRF2 antagonist, astressin 2B. The CRF-mediated PI3K/Akt activation induces cell survival as suggested by the stimulation of the antiapoptotic factor Bcl-2. ERK1/2 activation results in up-regulation of IL-8 expression, an effect inhibited by the CRF-induced activation of PI3K/Akt. These studies demonstrate novel effects of CRF in human monocytes mediated by the activation of PI3K/Akt. Moreover, they reveal pathway-specific effects of the CRF/CRF2 system in chemokine activation and cell survival that may be of importance for the development of novel therapeutics for inflammatory diseases.
- Published
- 2009
- Full Text
- View/download PDF
24. Mechanisms of obesity and related pathology: linking immune responses to metabolic stress.
- Author
-
Karalis KP, Giannogonas P, Kodela E, Koutmani Y, Zoumakis M, and Teli T
- Subjects
- Animals, Endoplasmic Reticulum metabolism, Humans, I-kappa B Kinase metabolism, JNK Mitogen-Activated Protein Kinases metabolism, Obesity pathology, Oxidative Stress immunology, Oxidative Stress radiation effects, Signal Transduction, Obesity immunology, Obesity metabolism
- Abstract
There is a tightly regulated interaction, which is well-conserved in evolution, between the metabolic and immune systems that is deranged in states of over- or under-nutrition. Obesity, an energy-rich condition, is characterized by the activation of an inflammatory process in metabolically active sites such as adipose tissue, liver and immune cells. The consequence of this response is a sharp increase in circulating levels of proinflammatory cytokines, adipokines and other inflammatory markers. Activation of the immune response in obesity is mediated by specific signaling pathways, with Jun N-terminal kinase and IkappaB kinase beta/nuclear factor kappa-light-chain-enhancer of activated B cells being the most well studied. It is known that the above events modify insulin signaling and result in the development of insulin resistance. The nutrient overload characterizing obesity is a metabolic stressor associated with intracellular organelle (e.g. the endoplasmic reticulum) stress. The exact characterization of the series of events and the mechanisms that integrate the inflammatory response with metabolic homeostasis at the cellular and systemic level is a very active research field. In this minireview, we discuss the signaling pathways and molecules associated with the development of obesity-induced inflammation, as well as the evidence that supports a critical role for the stress response in this process.
- Published
- 2009
- Full Text
- View/download PDF
25. Corticotropin-releasing hormone deficiency is associated with reduced local inflammation in a mouse model of experimental colitis.
- Author
-
Gay J, Kokkotou E, O'Brien M, Pothoulakis C, and Karalis KP
- Subjects
- Animals, Anorexia blood, Anorexia genetics, Anorexia pathology, Colitis, Ulcerative chemically induced, Colitis, Ulcerative genetics, Corticotropin-Releasing Hormone genetics, Disease Models, Animal, Female, Gene Expression, Inflammation genetics, Inflammation metabolism, Interleukin-6 blood, Leptin blood, Male, Mice, Mice, Inbred C57BL, Mice, Inbred Strains, Mice, Knockout, Reverse Transcriptase Polymerase Chain Reaction, Trinitrobenzenesulfonic Acid toxicity, Weight Loss, Colitis, Ulcerative pathology, Corticotropin-Releasing Hormone deficiency, Inflammation pathology
- Abstract
CRH, the hypothalamic component of the hypothalamic-pituitary adrenal axis, attenuates inflammation through stimulation of glucocorticoid release, whereas peripherally expressed CRH acts as a proinflammatory mediator. CRH is expressed in the intestine and up-regulated in patients with ulcerative colitis. However, its pathophysiological significance in intestinal inflammatory diseases has just started to emerge. In a mouse model of acute, trinitrobenzene sulfonic acid-induced experimental colitis, we demonstrate that, despite low glucocorticoid levels, CRH-deficient mice develop substantially reduced local inflammatory responses. These effects were shown by histological scoring of tissue damage and neutrophil infiltration. At the same time, CRH deficiency was found to be associated with higher serum leptin and IL-6 levels along with sustained anorexia and weight loss, although central CRH has been reported to be a strong appetite suppressor. Taken together, our results support an important proinflammatory role for CRH during mouse experimental colitis and possibly in inflammatory bowel disease in humans. Moreover, the results suggest that CRH is involved in homeostatic pathways that link inflammation and metabolism.
- Published
- 2008
- Full Text
- View/download PDF
26. Antiproliferative effect of adiponectin on MCF7 breast cancer cells: a potential hormonal link between obesity and cancer.
- Author
-
Arditi JD, Venihaki M, Karalis KP, and Chrousos GP
- Subjects
- Adenocarcinoma pathology, Apoptosis drug effects, Breast Neoplasms pathology, Cell Line, Tumor, Female, Humans, Receptors, Adiponectin, Receptors, Cell Surface metabolism, Adenocarcinoma etiology, Adiponectin pharmacology, Adiponectin physiology, Breast Neoplasms etiology, Cell Proliferation drug effects, Obesity etiology
- Abstract
Adiponectin, a hormone secreted by adipose tissue, circulates at high concentrations in human plasma. Paradoxically, plasma levels of adiponectin are approximately 50% lower in obese than in lean subjects. An association between low plasma levels of adiponectin and higher risk of developing breast and other cancers was recently reported. Obesity and overweight have also been associated with increased mortality from cancer. To test the hypothesis that adiponectin exerts direct antiproliferative and/or pro-apoptotic effects on cancer cells, we used the MCF7 human breast adenocarcinoma cell line. The proliferation rate of the MCF7 cells was measured using the MTT method, while apoptosis was examined by quantifying the DNA fragmentation using an ELISA assay. In addition, adiponectin receptor 1 (AdipoR1) and AdipoR2 mRNA expression was detected using RT-PCR. Adiponectin diminished the proliferation rate of MCF7 cells; this effect was significant after 48-96 hours of treatment. The presence of receptor expression suggested that the effect of adiponectin on cell proliferation was most likely specific and adiponectin receptor-mediated. Adiponectin induced no apoptosis of MCF7 cells over 48 hours. We conclude that adiponectin inhibits proliferation but causes no apoptosis of MCF7 breast cancer cells. These data suggest that adiponectin may represent a direct hormonal link between obesity and cancer.
- Published
- 2007
- Full Text
- View/download PDF
27. Regulation of appetite and insulin signaling in inflammatory states.
- Author
-
Teli T, Xanthaki D, and Karalis KP
- Subjects
- Animals, Cytokines metabolism, Humans, Hyperglycemia metabolism, Inflammation blood, Inflammation immunology, Mice, Obesity metabolism, Appetite physiology, Inflammation metabolism, Insulin metabolism, Insulin Resistance, Signal Transduction
- Abstract
Inflammatory states are characterized by decreased food intake, hyperglycemia, and insulin resistance. The contribution of cytokines in this phenotype is important and is exerted through activation of SOCS proteins and inhibition of insulin signaling, as well as through direct stimulation of the ob gene. Obesity, a condition that has reached epidemic rates, is characterized by hyperglycemia, hyperlipidemia, insulin resistance and increased food intake, and body weight. In the following article we summarize the current views of the mechanisms underlying insulin resistance in obesity and the other inflammatory states. We also discuss the regulation of appetite in inflammatory states, and we provide evidence on the cytokine-independent induction of anorexia following immune activation in mice. Understanding of the exact mechanisms regulating these processes may provide important insights for the control of this group of diseases that compromise to a great extent the quality of life and are associated with high mortality.
- Published
- 2006
- Full Text
- View/download PDF
28. Interleukin-6 genetic ablation protects from trinitrobenzene sulfonic acid-induced colitis in mice. Putative effect of antiinflammatory cytokines.
- Author
-
Gay J, Kokkotou E, O'Brien M, Pothoulakis C, and Karalis KP
- Subjects
- Animals, Body Weight drug effects, Body Weight immunology, Chemotaxis, Leukocyte drug effects, Chemotaxis, Leukocyte immunology, Colitis chemically induced, Colon pathology, Colon physiopathology, Disease Models, Animal, Eating drug effects, Eating immunology, Female, Interleukin-10 immunology, Mice, Mice, Inbred C57BL, Mice, Knockout, Transforming Growth Factor beta immunology, Trinitrobenzenesulfonic Acid adverse effects, Up-Regulation drug effects, Up-Regulation immunology, Colitis genetics, Colitis immunology, Colon immunology, Interleukin-10 metabolism, Interleukin-6 genetics, Transforming Growth Factor beta metabolism
- Abstract
Background: Interleukin (IL)-6 is a proinflammatory cytokine implicated in the pathogenesis of inflammatory bowel disease. IL-6 is locally upregulated in inflammatory bowel disease patients and in murine models of experimental colitis. Treatment with anti-IL-6 receptor antibody ameliorates intestinal inflammation., Objective: It was the aim of this study to investigate the role of genetic IL-6 deficiency in trinitrobenzene sulfonic acid (TNBS)-mediated colitis, an experimental model inflammation that shares several features with Crohn's disease in humans., Methods: Acute colitis was induced in wild-type and IL-6-deficient (Il-6(-/-)) mice by intracolonic administration of TNBS. Forty-eight hours after treatment, the local and systemic features of inflammation, i.e. food intake, weight loss, histological markers of colitis, cytokine expression by real-time PCR, food intake and body weight changes, were assessed., Results: In wild-type mice, TNBS administration increased both IL-6 serum levels and local expression of IL-6 by 36 and 9 fold, respectively, compared with control, vehicle-injected mice. Compared with the wild-type mice, the Il-6(-/-) mice had significantly reduced intestinal inflammation as evidenced by epithelial damage, neutrophil infiltration, colon thickness and proinflammatory cytokine expression, following treatment with TNBS. Moreover, baseline levels of the antiinflammatory cytokines IL-10 and tumor growth factor-beta were significantly elevated in Il-6(-/-)compared with the wild-type mice., Conclusions: Our results demonstrate that Il-6(-/-)are partially protected from the development of TNBS-induced acute experimental colitis, most likely via their significantly elevated baseline levels of antiinflammatory cytokines.
- Published
- 2006
- Full Text
- View/download PDF
29. Corticotropin-releasing hormone contributes to the peripheral inflammatory response in experimental autoimmune encephalomyelitis.
- Author
-
Benou C, Wang Y, Imitola J, VanVlerken L, Chandras C, Karalis KP, and Khoury SJ
- Subjects
- Animals, Antigen Presentation genetics, Antigen Presentation immunology, Antigen-Presenting Cells immunology, Antigen-Presenting Cells pathology, Cell Differentiation genetics, Cell Differentiation immunology, Cells, Cultured, Corticotropin-Releasing Hormone antagonists & inhibitors, Corticotropin-Releasing Hormone deficiency, Corticotropin-Releasing Hormone genetics, Cytokines biosynthesis, Encephalomyelitis, Autoimmune, Experimental genetics, Encephalomyelitis, Autoimmune, Experimental immunology, Encephalomyelitis, Autoimmune, Experimental prevention & control, Glucocorticoids biosynthesis, I-kappa B Proteins metabolism, Immunity, Innate genetics, Inflammation Mediators antagonists & inhibitors, Lymphocyte Count, Mice, Mice, Inbred C57BL, Mice, Knockout, NF-KappaB Inhibitor alpha, Phosphorylation, Receptors, Antigen, T-Cell physiology, Severity of Illness Index, Spleen immunology, Spleen metabolism, Spleen pathology, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism, T-Lymphocyte Subsets pathology, Th1 Cells cytology, Th1 Cells immunology, Th2 Cells immunology, Th2 Cells metabolism, Corticotropin-Releasing Hormone physiology, Encephalomyelitis, Autoimmune, Experimental pathology, Inflammation Mediators physiology
- Abstract
Peripheral corticotropin-releasing hormone (CRH) is thought to have proinflammatory effects. We used the model of experimental autoimmune encephalomyelitis (EAE) to study the role of CRH in an immune-mediated disease. We showed that CRH-deficient mice are resistant to EAE, with a decrease in clinical score as well as decreased cellular infiltration in the CNS. Furthermore, Ag-specific responses of primed T cells as well as anti-CD3/anti-CD28 TCR costimulation were decreased in crh(-/-) mice with decreased production of Th1 cytokines and increased production of Th2 cytokines. Wild-type mice treated in vivo with a CRH antagonist showed a decrease in IFN-gamma production by primed T cells in vitro. This effect of CRH is independent of its ability to increase corticosterone production, because adrenalectomized wild-type mice had similar disease course and severity as control mice. We found that IkappaBalpha phosphorylation induced by TCR cross-linking was decreased in crh(-/-) T cells. We conclude that peripheral CRH exerts a proinflammatory effect in EAE with a selective increase in Th1-type responses. These findings have implications for the treatment of Th1-mediated diseases such as multiple sclerosis.
- Published
- 2005
- Full Text
- View/download PDF
30. NF-kappaB participates in the corticotropin-releasing, hormone-induced regulation of the pituitary proopiomelanocortin gene.
- Author
-
Karalis KP, Venihaki M, Zhao J, van Vlerken LE, and Chandras C
- Subjects
- Animals, Base Sequence, Cell Line, DNA Primers, Electrophoretic Mobility Shift Assay, Mice, NF-kappa B antagonists & inhibitors, Corticotropin-Releasing Hormone physiology, NF-kappa B physiology, Pituitary Gland metabolism, Pro-Opiomelanocortin genetics
- Abstract
Corticotropin-releasing hormone is a main regulator of mammalian stress response by stimulating pituitary proopiomelanocortin (POMC) gene expression, and thus adrenocorticotropic hormone (ACTH) secretion, which then causes glucocorticoid release from the adrenal. In a recent study in the pituitary corticotroph cell line AtT20, oxidative stress stimulated the activity of nuclear transcription factor B (NF-kappaB), whereas corticotropin-releasing hormone (CRH) inhibited both the constitutive and the oxidative stress-induced NF-kappaB DNA-binding activity. To further investigate the role of NF-kappaB on the CRH-induced pituitary POMC gene activation, AtT20 cells were transiently transfected with a POMC-luciferase construct mutated at an NF-kappaB binding site. After treatment with CRH, intracellular POMC-luciferase activity was significantly higher from the stimulation observed with transfection of the parental POMC-luciferase construct. In agreement with a previous report, CRH inhibited the constitutive NF-kappaB DNA-binding activity in AtT20 cells, as shown by electrophoretic mobility-shift assay, as soon as within 15 min of treatment. These effects of CRH were blocked by the CRH-R1 antagonist CP154,256. Our findings provide evidence that the regulation of corticotroph NF-kappaB activity by CRH is related to the activation of the pituitary POMC gene and, thus, may play an important role in stress response.
- Published
- 2004
- Full Text
- View/download PDF
31. Corticotropin-releasing hormone deficiency results in impaired splenocyte response to lipopolysaccharide.
- Author
-
Venihaki M, Zhao J, and Karalis KP
- Subjects
- Animals, Cell Division genetics, Cell Division immunology, Cells, Cultured, Corticotropin-Releasing Hormone biosynthesis, Corticotropin-Releasing Hormone pharmacology, Cytokines biosynthesis, Cytokines genetics, Cytokines metabolism, Down-Regulation genetics, Down-Regulation immunology, Interleukin-1 metabolism, Lymphocyte Activation genetics, Lymphocyte Subsets immunology, Lymphocyte Subsets metabolism, Lymphocyte Subsets pathology, Mice, Mice, Inbred C57BL, Mice, Knockout, NF-kappa B antagonists & inhibitors, NF-kappa B metabolism, RNA, Messenger antagonists & inhibitors, RNA, Messenger biosynthesis, Spleen drug effects, Spleen pathology, Tumor Necrosis Factor-alpha metabolism, Corticotropin-Releasing Hormone deficiency, Corticotropin-Releasing Hormone physiology, Lipopolysaccharides pharmacology, Spleen immunology, Spleen metabolism
- Abstract
Corticotropin-releasing hormone (Crh), a major mediator of the stress response, has been shown to exert both stimulatory and inhibitory effects on the regulation of the immune system, in vivo. In our present study, we used the Crh-/- mice to investigate the effect of Crh deficiency on leukocyte function in vitro. Our results show that following LPS treatment, TNF-alpha and IL-1beta expression was significantly compromised in Crh-/- splenocytes, an effect most likely mediated by the lower levels of NF-kappaB DNA binding activity measured in the same cells. Furthermore, we show here that the proliferation rate of Crh-/- splenocytes in response to LPS was decreased compared to Crh+/+ splenocytes. Taken together, our findings show that the presence of endogenous Crh is necessary for the normal function of leukocytes, in vitro.
- Published
- 2003
- Full Text
- View/download PDF
32. Regulation of nuclear factor-kappaB by corticotropin-releasing hormone in mouse thymocytes.
- Author
-
Zhao J and Karalis KP
- Subjects
- Animals, Base Sequence, Cells, Cultured, Cyclic AMP-Dependent Protein Kinases metabolism, DNA-Binding Proteins metabolism, Dexamethasone pharmacology, Female, I-kappa B Proteins metabolism, Kinetics, Mice, Mice, Inbred C57BL, NF-KappaB Inhibitor alpha, NF-kappa B antagonists & inhibitors, Oligodeoxyribonucleotides chemistry, Oligodeoxyribonucleotides metabolism, Protein Binding, Protein Kinase C metabolism, T-Lymphocytes drug effects, Corticotropin-Releasing Hormone pharmacology, NF-kappa B metabolism, T-Lymphocytes physiology
- Abstract
CRH, a major mediator of the stress response, has been shown to exert potent immunomodulatory effects in vivo, through mechanisms that have not been elucidated yet. To determine the molecular pathways mediating the proinflammatory effects of peripheral CRH, we studied its role in the activation of nuclear factor-kappaB (NF-kappaB), a transcription factor crucial for the regulation of a variety of inflammatory mediator genes. Our studies demonstrate that, in mouse thymocytes, CRH induces the NF-kappaB DNA-binding activity in a time- and dose-dependent manner, with parallel degradation of its inhibitor protein inhibitor of NF-kappaB. The effect of CRH is not inhibited by dexamethasone and is mediated by the protein kinase A and protein kinase C signaling pathways. In vivo, we show that CRH-deficient mice respond to lipopolysaccharide administration by reduced activation of thymus NF-kappaB, despite their significantly elevated proinflammatory cytokine and their low corticosterone levels. These findings suggest a putative molecular pathway mediating the proinflammatory effects of peripheral CRH through induction of the NF-kappaB DNA binding activity.
- Published
- 2002
- Full Text
- View/download PDF
33. Corticotropin-releasing hormone regulates IL-6 expression during inflammation.
- Author
-
Venihaki M, Dikkes P, Carrigan A, and Karalis KP
- Subjects
- Adrenalectomy, Adrenocorticotropic Hormone antagonists & inhibitors, Adrenocorticotropic Hormone blood, Animals, Corticosterone blood, Corticotropin-Releasing Hormone deficiency, Corticotropin-Releasing Hormone genetics, Gene Expression Regulation, Hypothalamo-Hypophyseal System physiology, Inflammation immunology, Inflammation pathology, Inflammation physiopathology, Interleukin-6 blood, Irritants toxicity, Mice, Mice, Inbred C57BL, Mice, Knockout, Pituitary-Adrenal System physiology, Turpentine toxicity, Corticotropin-Releasing Hormone physiology, Inflammation etiology, Interleukin-6 genetics
- Abstract
Stimulation of the hypothalamic-pituitary-adrenal (HPA) axis by proinflammatory cytokines results in increased release of glucocorticoid that restrains further development of the inflammatory process. IL-6 has been suggested to stimulate the HPA axis during immune activation independent of the input of hypothalamic corticotropin-releasing hormone (CRH). We used the corticotropin-releasing hormone-deficient (Crh(-/-)) mouse to elucidate the effect of CRH deficiency on IL-6 expression and IL-6-induced HPA axis activation during turpentine-induced inflammation. We demonstrate that during inflammation CRH is required for a normal adrenocorticotropin hormone (ACTH) increase but not for adrenal corticosterone rise. The paradoxical increase of plasma IL-6 associated with CRH deficiency suggests that IL-6 release during inflammation is CRH-dependent. We also demonstrate that adrenal IL-6 expression is CRH-dependent, as its basal and inflammation-induced expression is blocked by CRH deficiency. Our findings suggest that during inflammation, IL-6 most likely compensates for the effects of CRH deficiency on food intake. Finally, we confirm that the HPA axis response is defective in Crh(-/-)/IL-6(-/-) mice. These findings, along with the regulation of IL-6 by CRH, support the importance of the interaction between the immune system and the HPA axis in the pathophysiology of inflammatory diseases.
- Published
- 2001
- Full Text
- View/download PDF
34. The physiology of corticotropin-releasing hormone deficiency in mice.
- Author
-
Muglia LJ, Jacobson L, Weninger SC, Karalis KP, Jeong K, and Majzoub JA
- Subjects
- Adrenocorticotropic Hormone biosynthesis, Adrenocorticotropic Hormone metabolism, Animals, Inflammation physiopathology, Mice, Mice, Knockout, Circadian Rhythm physiology, Corticotropin-Releasing Hormone deficiency, Corticotropin-Releasing Hormone physiology, Hypothalamo-Hypophyseal System physiology
- Abstract
A review of the generation and characterization of corticotropin-releasing hormone (CRH)-deficient mice is presented. The studies summarized demonstrate the central role of CRH in the pituitary-adrenal axis response to stress, circadian stimulation, and glucocorticoid withdrawal. Additionally, pro-inflammatory actions of CRH at sites of local inflammation are given further support. In contrast, behavioral effects during stress that had been ascribed to CRH action are not altered in CRH-deficient mice. The normal behavioral response to stress in CRH-deficient mice strongly suggests the importance of other, possibly as yet undiscovered, CRH-like molecules.
- Published
- 2001
- Full Text
- View/download PDF
35. Corticotropin-releasing hormone deficiency unmasks the proinflammatory effect of epinephrine.
- Author
-
Karalis KP, Kontopoulos E, Muglia LJ, and Majzoub JA
- Subjects
- Animals, Catecholamines physiology, Male, Mice, Mice, Knockout, Adrenal Glands physiology, Corticotropin-Releasing Hormone physiology, Epinephrine physiology, Inflammation physiopathology
- Abstract
Traditionally, the adrenal gland has been considered an important endocrine component of the pathway to inhibit acute inflammation via hypothalamic corticotropin-releasing hormone (CRH)-mediated secretion of glucocorticoid. Immunoreactive CRH found in inflamed tissues is a potent proinflammatory factor. Using genetic and pharmacological models of CRH deficiency, we now show that CRH deficiency unmasks a major proinflammatory effect of epinephrine secreted from the adrenal medulla. Together, epinephrine and peripheral CRH stimulate inflammation, and glucocorticoid acts as a counterbalancing force in this regard. Our findings suggest that stimulation of the acute inflammatory response should be included with the other "fight-or-flight" actions of epinephrine.
- Published
- 1999
- Full Text
- View/download PDF
36. Placental corticotropin-releasing hormone: function and regulation.
- Author
-
Majzoub JA and Karalis KP
- Subjects
- Animals, Corticotropin-Releasing Hormone metabolism, Embryonic and Fetal Development physiology, Female, Humans, Hypothalamo-Hypophyseal System physiology, Pituitary-Adrenal System physiology, Pregnancy, Primates physiology, Corticotropin-Releasing Hormone physiology, Placenta metabolism
- Abstract
Corticotropin-releasing hormone is a neuropeptide placentally expressed among mammals only in primates. Its expression increases as much as 100 times during the last 6 to 8 weeks of pregnancy and is paradoxically stimulated by glucocorticoids. Increasing evidence suggests that placental corticotropin-releasing hormone may have evolved in primates to stimulate fetal adrenocorticotropin release and adrenal steroidogenesis, thus satisfying the high demand for synthesis of dehydroepiandrosterone, the predominant source of placental estradiol. Concomitant stimulation by placental corticotropin-releasing hormone of fetal cortisol and dehydroepiandrosterone would couple the glucocorticoid effects on fetal organ maturation with the timing of parturition, an obvious benefit in postnatal survival.
- Published
- 1999
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.