Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Mol Basis Dis ; 1868(1): 166286, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34624498

ABSTRACT

Chronic stress leads to post-traumatic stress disorder (PTSD) and metabolic disorders including fatty liver. We hypothesized that stress-induced molecular mechanisms alter energy metabolism, thereby promoting hepatic lipid accumulation even after a stress-free recovery period. In this context, we investigated fibroblast growth factor-21 (FGF21) as protective for energy and glucose homeostasis. FGF21 knockout mice (B6.129S6(SJL)-Fgf21tm1.2Djm; FGF21KO) and control mice (C57BL6; WT) were subjected to chronic variable stress. Mice were examined directly after acute intervention (Cvs) and long-term after 3 months of recovery (3mCvs). In WT, Cvs reduced insulin sensitivity and hepatic lipid accumulation, whilst fatty acid uptake increased. FGF21KO mice responded to Cvs with improved glucose tolerance, insulin resistance but liver triglycerides and plasma lipids were unaltered. Hepatic gene expression was specifically altered by genotype and stress e.g. by PPARa and SREBP-1 regulated genes. The stress-induced alteration of hepatic metabolism persisted after stress recovery. In hepatocytes at 3mCvs, differential gene regulation and secreted proteins indicated a genotype specific progression of liver dysfunction. Overall, at 3mCvs FGF21 was involved in maintaining mitochondrial activity, attenuating de novo lipogenesis, increased fatty acid uptake and histone acetyltransferase activity. Glucocorticoid release and binding to the FGF21 promoter may contribute to prolonged FGF21 release and protection against hepatic lipid accumulation. In conclusion, we showed that stress favors fatty liver disease and FGF21 protected against hepatic lipid accumulation after previous chronic stress loading by i) restored physiological function, ii) modulated gene expression via DNA-modifying enzymes, and iii) maintained energy metabolism.


Subject(s)
Energy Metabolism/genetics , Fatty Liver/genetics , Fibroblast Growth Factors/genetics , Stress Disorders, Post-Traumatic/genetics , Animals , Fatty Liver/metabolism , Fatty Liver/pathology , Genotype , Glucose/metabolism , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Lipid Metabolism/genetics , Lipids/genetics , Liver/metabolism , Liver/pathology , Mice , Mice, Knockout , PPAR alpha/genetics , Sterol Regulatory Element Binding Protein 1/genetics , Stress Disorders, Post-Traumatic/metabolism , Stress Disorders, Post-Traumatic/pathology
2.
Mol Metab ; 42: 101062, 2020 12.
Article in English | MEDLINE | ID: mdl-32771698

ABSTRACT

OBJECTIVE: Physical exercise training is associated with increased glucose uptake in skeletal muscle and improved glycemic control. HDAC5, a class IIa histone deacetylase, has been shown to regulate transcription of the insulin-responsive glucose transporter GLUT4 in cultured muscle cells. In this study, we analyzed the contribution of HDAC5 to the transcriptional network in muscle and the beneficial effect of muscle contraction and regular exercise on glucose metabolism. METHODS: HDAC5 knockout mice (KO) and wild-type (WT) littermates were trained for 8 weeks on treadmills, metabolically phenotyped, and compared to sedentary controls. Hdac5-deficient skeletal muscle and cultured Hdac5-knockdown (KD) C2C12 myotubes were utilized for studies of gene expression and glucose metabolism. Chromatin immunoprecipitation (ChIP) studies were conducted to analyze Il6 promoter activity using H3K9ac and HDAC5 antibodies. RESULTS: Global transcriptome analysis of Hdac5 KO gastrocnemius muscle demonstrated activation of the IL-6 signaling pathway. Accordingly, knockdown of Hdac5 in C2C12 myotubes led to higher expression and secretion of IL-6 with enhanced insulin-stimulated activation of AKT that was reversed by Il6 knockdown. Moreover, Hdac5-deficient myotubes exhibited enhanced glucose uptake, glycogen synthesis, and elevated expression levels of the glucose transporter GLUT4. Transcription of Il6 was further enhanced by electrical pulse stimulation in Hdac5-deficient C2C12 myotubes. ChIP identified a ∼1 kb fragment of the Il6 promoter that interacts with HDAC5 and demonstrated increased activation-associated histone marker AcH3K9 in Hdac5-deficient muscle cells. Exercise intervention of HDAC5 KO mice resulted in improved systemic glucose tolerance as compared to WT controls. CONCLUSIONS: We identified HDAC5 as a negative epigenetic regulator of IL-6 synthesis and release in skeletal muscle. HDAC5 may exert beneficial effects through two different mechanisms, transcriptional control of genes required for glucose disposal and utilization, and HDAC5-dependent IL-6 signaling cross-talk to improve glucose uptake in muscle in response to exercise.


Subject(s)
Histone Deacetylases/metabolism , Insulin/metabolism , Interleukin-6/metabolism , Animals , Cell Line , Gene Expression/genetics , Glucose/metabolism , Histone Deacetylases/genetics , Interleukin-6/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Contraction/physiology , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Phosphorylation , Physical Conditioning, Animal/methods , Promoter Regions, Genetic/genetics , Signal Transduction/genetics
3.
J Clin Invest ; 130(11): 6093-6108, 2020 11 02.
Article in English | MEDLINE | ID: mdl-32780722

ABSTRACT

Recent genome-wide association studies (GWAS) identified DUSP8, encoding a dual-specificity phosphatase targeting mitogen-activated protein kinases, as a type 2 diabetes (T2D) risk gene. Here, we reveal that Dusp8 is a gatekeeper in the hypothalamic control of glucose homeostasis in mice and humans. Male, but not female, Dusp8 loss-of-function mice, either with global or corticotropin-releasing hormone neuron-specific deletion, had impaired systemic glucose tolerance and insulin sensitivity when exposed to high-fat diet (HFD). Mechanistically, we found impaired hypothalamic-pituitary-adrenal axis feedback, blunted sympathetic responsiveness, and chronically elevated corticosterone levels driven by hypothalamic hyperactivation of Jnk signaling. Accordingly, global Jnk1 ablation, AAV-mediated Dusp8 overexpression in the mediobasal hypothalamus, or metyrapone-induced chemical adrenalectomy rescued the impaired glucose homeostasis of obese male Dusp8-KO mice, respectively. The sex-specific role of murine Dusp8 in governing hypothalamic Jnk signaling, insulin sensitivity, and systemic glucose tolerance was consistent with functional MRI data in human volunteers that revealed an association of the DUSP8 rs2334499 risk variant with hypothalamic insulin resistance in men. Further, expression of DUSP8 was increased in the infundibular nucleus of T2D humans. In summary, our findings suggest the GWAS-identified gene Dusp8 as a novel hypothalamic factor that plays a functional role in the etiology of T2D.


Subject(s)
Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Type 2/enzymology , Dual-Specificity Phosphatases/metabolism , Hypothalamus/enzymology , Insulin Resistance , MAP Kinase Kinase 4/metabolism , Signal Transduction , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 2/genetics , Dual-Specificity Phosphatases/genetics , MAP Kinase Kinase 4/genetics , Mice , Mice, Knockout
4.
Phytomedicine ; 76: 153252, 2020 May 29.
Article in English | MEDLINE | ID: mdl-32505916

ABSTRACT

BACKGROUND: The occurrence of chronic wounds, account for significant suffering of diabetic people, together with increasing healthcare burden. The chronic wounds associated with diabetes do not undergo the normal healing process rather stagnate into chronic proinflammatory phase as well as declined fibroblast function and impaired cell migration. HYPOTHESIS: SIRT1, which is the most studied isoform of the sirtuin family in mammals, has now emerged as a crucial target for improving diabetic wound healing. It is an NAD+ dependent deacetylase, originally characterized to deacetylate histone proteins leading to heterochromatin formation and gene silencing. It is now known to regulate a number of cellular processes like cell proliferation, division, senescence, apoptosis, DNA repair, and metabolism. METHODOLOGY: The retrieval of potentially relevant studies was done by systematically searching of three databases (Google Scholar, Web of science and PubMed) in December 2019. The keywords used as search terms were related to SIRT1 and wound healing. The systematic search retrieved 649 papers that were potentially relevant and after selection procedure, 73 studies were included this review and discussed below. RESULTS: Many SIRT1 activating compounds (SACs) were found protective and improve diabetic wound healing through regulation of inflammation, cell migration, oxidative stress response and formation of granulation tissue at the wound site. CONCLUSIONS: However, contradictory reports describe the opposing role of SACs on the regulation of cell migration and cancer incidence. SACs are therefore subjected to intense research for understanding the mechanisms responsible for controlling cell migration and therefore possess prospective to enter the clinical arena in the foreseeable future.

5.
Arch Pharm Res ; 43(2): 171-186, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31838653

ABSTRACT

Metabolic disorders are increasing at an alarming rate due to the stressful lifestyle and inappropriate diet schedule. The unorganized habits influence multiple epigenetic mechanisms like DNA methylation, histone post-translational modifications and miRNA expression. These epigenetic modifications are reversible in nature and regulate gene expression in response to external stimuli without altering the DNA sequence. Dietary herbs are enriched in various phytochemicals which additionally provide nutrition and health benefits; and are known to target these epigenetic gene regulatory mechanisms. They have been in use since human civilization for their health-promoting effects. Culinary spices and condiments which are generally used to enhance the taste of food are rich repositories of many phytochemicals which provides them their unique aroma. Considerable attention has been given to "Nutri-epigenetics" nowadays, with a focus on evaluating the potential of phytochemicals to regulate/neutralize various epigenetic modifications. This article aims at highlighting the epigenetic regulatory roles of phytochemicals present in condiments and spices with considerable health benefits.


Subject(s)
Biological Products/pharmacology , Epigenesis, Genetic/drug effects , Metabolic Diseases/drug therapy , Phytochemicals/pharmacology , Spices , Biological Products/chemistry , Epigenesis, Genetic/genetics , Humans , Metabolic Diseases/genetics , Phytochemicals/chemistry
7.
Nat Metab ; 1(2): 222-235, 2019 02.
Article in English | MEDLINE | ID: mdl-32694784

ABSTRACT

Heterogeneous populations of hypothalamic neurons orchestrate energy balance via the release of specific signatures of neuropeptides. However, how specific intracellular machinery controls peptidergic identities and function of individual hypothalamic neurons remains largely unknown. The transcription factor T-box 3 (Tbx3) is expressed in hypothalamic neurons sensing and governing energy status, whereas human TBX3 haploinsufficiency has been linked with obesity. Here, we demonstrate that loss of Tbx3 function in hypothalamic neurons causes weight gain and other metabolic disturbances by disrupting both the peptidergic identity and plasticity of Pomc/Cart and Agrp/Npy neurons. These alterations are observed after loss of Tbx3 in both immature hypothalamic neurons and terminally differentiated mouse neurons. We further establish the importance of Tbx3 for body weight regulation in Drosophila melanogaster and show that TBX3 is implicated in the differentiation of human embryonic stem cells into hypothalamic Pomc neurons. Our data indicate that Tbx3 directs the terminal specification of neurons as functional components of the melanocortin system and is required for maintaining their peptidergic identity. In summary, we report the discovery of a key mechanistic process underlying the functional heterogeneity of hypothalamic neurons governing body weight and systemic metabolism.


Subject(s)
Hypothalamus/metabolism , Melanocortins/metabolism , Neurons/metabolism , T-Box Domain Proteins/metabolism , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Animals , Body Weight , Energy Metabolism , Gene Expression Profiling , Green Fluorescent Proteins/genetics , Hypothalamus/cytology , Mice , Mice, Inbred C57BL , Pro-Opiomelanocortin/genetics , RNA, Messenger/genetics , T-Box Domain Proteins/genetics
8.
Diabetes ; 67(11): 2456-2465, 2018 11.
Article in English | MEDLINE | ID: mdl-30158241

ABSTRACT

Celastrol, a plant-derived constituent of traditional Chinese medicine, has been proposed to offer significant potential as an antiobesity drug. However, the molecular mechanism for this activity is unknown. We show that the weight-lowering effects of celastrol are driven by decreased food consumption. Although young Lep ob mice respond with a decrease in food intake and body weight, adult Lep db and Lep ob mice are unresponsive to celastrol, suggesting that functional leptin signaling in adult mice is required to elicit celastrol's catabolic actions. Protein tyrosine phosphatase 1 (PTP1B), a leptin negative-feedback regulator, has been previously reported to be one of celastrol's targets. However, we found that global PTP1B knockout (KO) and wild-type (WT) mice have comparable weight loss and hypophagia when treated with celastrol. Increased levels of uncoupling protein 1 (UCP1) in subcutaneous white and brown adipose tissue suggest celastrol-induced thermogenesis as a further mechanism. However, diet-induced obese UCP1 WT and KO mice have comparable weight loss upon celastrol treatment, and celastrol treatment has no effect on energy expenditure under ambient housing or thermoneutral conditions. Overall, our results suggest that celastrol-induced weight loss is hypophagia driven and age-dependently mediated by functional leptin signaling. Our data encourage reconsideration of therapeutic antiobesity strategies built on leptin sensitization.


Subject(s)
Eating/drug effects , Obesity/metabolism , Plant Extracts/pharmacology , Triterpenes/pharmacology , Uncoupling Protein 1/metabolism , Weight Loss/drug effects , Animals , Diet, High-Fat , Energy Metabolism/drug effects , Mice, Knockout , Obesity/genetics , Pentacyclic Triterpenes , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Uncoupling Protein 1/genetics
9.
Mol Metab ; 16: 126-138, 2018 10.
Article in English | MEDLINE | ID: mdl-29980484

ABSTRACT

OBJECTIVE: Post-traumatic stress disorder (PTSD) increases type 2 diabetes risk, yet the underlying mechanisms are unclear. We investigated how early-life exposure to chronic stress affects long-term insulin sensitivity. METHODS: C57Bl/6J mice were exposed to chronic variable stress for 15 days (Cvs) and then recovered for three months without stress (Cvs3m). RESULTS: Cvs mice showed markedly increased plasma corticosterone and hepatic insulin resistance. Cvs3m mice exhibited improved whole-body insulin sensitivity along with enhanced adipose glucose uptake and skeletal muscle mitochondrial function and fatty acid oxidation. Plasma FGF21 levels were substantially increased and associated with expression of genes involved in fatty acid oxidation and formation of brown-like adipocytes. In humans, serum FGF21 levels were associated with stress coping long time after the exposure. CONCLUSIONS: Early-life exposure to chronic stress leads to long term improvements in insulin sensitivity, oxidative metabolism and adipose tissue remodeling. FGF21 contributes to a physiological memory mechanism to maintain metabolic homeostasis.


Subject(s)
Fibroblast Growth Factors/metabolism , Stress, Psychological/metabolism , Adipocytes/metabolism , Animals , Diabetes Mellitus, Experimental/metabolism , Disease Models, Animal , Energy Metabolism , Glucose/metabolism , Homeostasis , Insulin/metabolism , Insulin Resistance , Lipid Metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Oxidation-Reduction , Stress Disorders, Post-Traumatic/metabolism
10.
J Neuroinflammation ; 15(1): 35, 2018 Feb 08.
Article in English | MEDLINE | ID: mdl-29422055

ABSTRACT

ᅟ: Astrocytosis is a reactive process involving cellular, molecular, and functional changes to facilitate neuronal survival, myelin preservation, blood brain barrier function and protective glial scar formation upon brain insult. The overall pro- or anti-inflammatory impact of reactive astrocytes appears to be driven in a context- and disease-driven manner by modulation of astrocytic Ca2+ homeostasis and activation of Ca2+/calmodulin-activated serine/threonine phosphatase calcineurin. Here, we aimed to assess whether calcineurin is dispensable for astrocytosis in the hypothalamus driven by prolonged high fat diet (HFD) feeding. Global deletion of calcineurin A beta (gene name: Ppp3cb) led to a decrease of glial fibrillary acidic protein (GFAP)-positive cells in the ventromedial hypothalamus (VMH), dorsomedial hypothalamus (DMH), and arcuate nucleus (ARC) of mice exposed chronically to HFD. The concomitant decrease in Iba1-positive microglia in the VMH further suggests a modest impact of Ppp3cb deletion on microgliosis. Pharmacological inhibition of calcineurin activity by Fk506 had no impact on IBA1-positive microglia in hypothalami of mice acutely exposed to HFD for 1 week. However, Fk506-treated mice displayed a decrease in GFAP levels in the ARC. In vivo effects could not be replicated in cell culture, where calcineurin inhibition by Fk506 had no effect on astrocytic morphology, astrocytic cell death, GFAP, and vimentin protein levels or microglia numbers in primary hypothalamic astrocytes and microglia co-cultures. Further, adenoviral overexpression of calcineurin subunit Ppp3r1 in primary glia culture did not lead to an increase in GFAP fluorescence intensity. Overall, our results point to a prominent role of calcineurin in mediating hypothalamic astrocytosis as response to acute and chronic HFD exposure. Moreover, discrepant findings in vivo and in cell culture indicate the necessity of studying astrocytes in their "natural" environment, i.e., preserving an intact hypothalamic microenvironment with neurons and non-neuronal cells in close proximity.


Subject(s)
Calcineurin/deficiency , Diet, High-Fat/adverse effects , Gliosis/metabolism , Gliosis/prevention & control , Hypothalamus/metabolism , Animals , Astrocytes/metabolism , Cell Survival/physiology , Cells, Cultured , Gliosis/pathology , Hypothalamus/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
11.
Neuroscience ; 357: 241-254, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28627418

ABSTRACT

Elevated levels of oxidative stress and neuronal inflammation in the hypothalamus or ventral midbrain, respectively, represent common denominators for obesity and Parkinson's Disease (PD). However, little is known about defense mechanisms that protect neurons in these regions from oxidative damage. Here, we aimed to assess whether murine Gpx4, a crucial antioxidant enzyme that protects neurons from membrane damage and ferroptosis, is critical for the protection from neuronal inflammation in two distinct pathophysiologic diseases, namely metabolic dysfunction in diet-induced obesity or PD. Gpx4 was deleted from either AgRP or POMC neurons in the hypothalamus, essential for metabolic homeostasis, or from dopaminergic neurons in the ventral midbrain, governing behaviors such as anxiety or voluntary movement. To induce a pro-inflammatory environment, AgRP and POMC neuron-specific Gpx4 knockout mice were subjected to high-fat high-sucrose (HFHS) diet. To exacerbate oxidative stress in dopaminergic neurons of the ventral midbrain, we systemically co-deleted the PD-related gene DJ-1. Gpx4 was dispensable for the maintenance of cellular health and function of POMC neurons, even in mice exposed to obesogenic conditions. In contrast, HFHS-fed mice with Gpx4 deletion from AgRP neurons displayed increased body adiposity. Gpx4 expression and activity were diminished in the hypothalamus of HFHS-fed mice compared to standard diet-fed controls. Gpx4 deletion from dopaminergic neurons induced anxiety behavior, and diminished spontaneous locomotor activity when DJ-1 was co-deleted. Overall, these data suggest a physiological role for Gpx4 in balancing metabolic control signals and inflammation in AgRP but not POMC neurons. Moreover, Gpx4 appears to constitute an important rheostat against neuronal dysfunction and PD-like symptoms in dopaminergic circuitry within the ventral midbrain.


Subject(s)
Anxiety/enzymology , Body Weight/physiology , Glutathione Peroxidase/deficiency , Motor Activity/physiology , Obesity/enzymology , Parkinsonian Disorders/enzymology , Adiposity/physiology , Animals , Anxiety/immunology , Anxiety/pathology , Behavior, Animal/physiology , Diet, High-Fat , Dietary Sucrose , Dopaminergic Neurons/enzymology , Dopaminergic Neurons/immunology , Dopaminergic Neurons/pathology , Female , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Hypothalamus/enzymology , Hypothalamus/immunology , Hypothalamus/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Obesity/pathology , Oxidative Stress/physiology , Parkinsonian Disorders/immunology , Parkinsonian Disorders/pathology , Phospholipid Hydroperoxide Glutathione Peroxidase , Protein Deglycase DJ-1/genetics , Protein Deglycase DJ-1/metabolism , Sex Characteristics , Glutathione Peroxidase GPX1
12.
Nat Commun ; 7: 10782, 2016 Feb 29.
Article in English | MEDLINE | ID: mdl-26923837

ABSTRACT

Hypothalamic leptin signalling has a key role in food intake and energy-balance control and is often impaired in obese individuals. Here we identify histone deacetylase 5 (HDAC5) as a regulator of leptin signalling and organismal energy balance. Global HDAC5 KO mice have increased food intake and greater diet-induced obesity when fed high-fat diet. Pharmacological and genetic inhibition of HDAC5 activity in the mediobasal hypothalamus increases food intake and modulates pathways implicated in leptin signalling. We show HDAC5 directly regulates STAT3 localization and transcriptional activity via reciprocal STAT3 deacetylation at Lys685 and phosphorylation at Tyr705. In vivo, leptin sensitivity is substantially impaired in HDAC5 loss-of-function mice. Hypothalamic HDAC5 overexpression improves leptin action and partially protects against HFD-induced leptin resistance and obesity. Overall, our data suggest that hypothalamic HDAC5 activity is a regulator of leptin signalling that adapts food intake and body weight to our dietary environment.


Subject(s)
Hypothalamus/metabolism , Leptin/metabolism , Animals , Blood Glucose , Cell Line , Gene Expression Regulation , Gene Knockdown Techniques , Glucose Tolerance Test , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Infusions, Intraventricular , Insulin Resistance , Laser Capture Microdissection , Leptin/genetics , Male , Melanocyte-Stimulating Hormones/pharmacology , Mice , Mice, Inbred Strains , Mice, Knockout , Neurons/physiology , Rats , Rats, Wistar
13.
Cell Metab ; 22(5): 838-50, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26411342

ABSTRACT

Canonical protein phosphatase 3/calcineurin signaling is central to numerous physiological processes. Here we provide evidence that calcineurin plays a pivotal role in controlling systemic energy and body weight homeostasis. Knockdown of calcineurin in Drosophila melanogaster led to a decrease in body weight and energy stores, and increased energy expenditure. In mice, global deficiency of catalytic subunit Ppp3cb, and tissue-specific ablation of regulatory subunit Ppp3r1 from skeletal muscle, but not adipose tissue or liver, led to protection from high-fat-diet-induced obesity and comorbid sequelæ. Ser637 hyperphosphorylation of dynamin-related protein 1 (Drp1) in skeletal muscle of calcineurin-deficient mice was associated with mitochondrial elongation into power-cable-shaped filaments and increased mitochondrial respiration, but also with attenuated exercise performance. Our data suggest that calcineurin acts as highly conserved pivot for the adaptive metabolic responses to environmental changes such as high-fat, high-sugar diets or exercise.


Subject(s)
Calcineurin/genetics , Intracellular Signaling Peptides and Proteins/genetics , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Obesity/metabolism , Animals , Body Weight , Calcineurin/metabolism , Calcium-Binding Proteins , Diet, High-Fat , Dynamins/metabolism , Energy Metabolism/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mitochondria/metabolism , Mitochondria/pathology , Muscle Proteins/metabolism , Muscle, Skeletal/pathology , Obesity/genetics , Obesity/pathology , Signal Transduction
14.
Endocrinology ; 156(5): 1714-23, 2015 May.
Article in English | MEDLINE | ID: mdl-25751638

ABSTRACT

Ghrelin is a circulating hormone that targets the central nervous system to regulate feeding and adiposity. The best-characterized neural system that mediates the effects of ghrelin on energy balance involves the activation of neuropeptide Y/agouti-related peptide neurons, expressed exclusively in the arcuate nucleus of the hypothalamus. However, ghrelin receptors are expressed in other neuronal populations involved in the control of energy balance. We combined laser capture microdissection of several nuclei of the central nervous system expressing the ghrelin receptor (GH secretagoge receptor) with microarray gene expression analysis to identify additional neuronal systems involved in the control of central nervous system-ghrelin action. We identified tachykinin-1 (Tac1) as a gene negatively regulated by ghrelin in the hypothalamus. Furthermore, we identified neuropeptide k as the TAC1-derived peptide with more prominent activity, inducing negative energy balance when delivered directly into the brain. Conversely, loss of Tac1 expression enhances the effectiveness of ghrelin promoting fat mass gain both in male and in female mice and increases the susceptibility to diet-induced obesity in ovariectomized mice. Taken together, our data demonstrate a role TAC1 in the control energy balance by regulating the levels of adiposity in response to ghrelin administration and to changes in the status of the gonadal function.


Subject(s)
Adiposity , Brain/metabolism , Energy Metabolism/genetics , Feeding Behavior/physiology , Ghrelin/metabolism , Obesity/genetics , Receptors, Ghrelin/metabolism , Tachykinins/genetics , Animals , Diet, High-Fat , Energy Metabolism/drug effects , Feeding Behavior/drug effects , Female , Gene Expression Profiling , Male , Mice , Obesity/metabolism , Polymerase Chain Reaction , RNA, Messenger/metabolism , Rats , Tachykinins/metabolism , Tachykinins/pharmacology
15.
PLoS One ; 8(4): e61822, 2013.
Article in English | MEDLINE | ID: mdl-23630616

ABSTRACT

Type 2 Diabetes is a global health burden and based on current estimates will become an even larger problem in the future. Developing new strategies to prevent and treat diabetes is a scientific challenge of high priority. The stomach hormone ghrelin has been associated with playing a role in the regulation of glucose homeostasis. However, its precise mechanism and impact on whole glucose metabolism remains to be elucidated. This study aims to clarify the role of the two ghrelin isoforms acyl- and desacyl ghrelin in regulating glucose homeostasis. Therefore ghrelin activating enzyme Ghrelin-O-acyltransferase (GOAT) was ablated in leptin-deficient ob/ob mice to study whether specific acyl ghrelin deficiency or desacyl ghrelin abundance modifies glucose tolerance on a massively obese background. As targeted deletion of acyl ghrelin does not improve glucose homeostasis in our GOAT-ob/ob mouse model we conclude that neither acyl ghrelin nor the increased ratio of desacyl/acyl ghrelin is crucial for controlling glucose homeostasis in the here presented model of massive obesity induced by leptin deficiency.


Subject(s)
Acyltransferases/genetics , Ghrelin/blood , Glucose Intolerance/enzymology , Leptin/deficiency , Protein Processing, Post-Translational , Acylation , Acyltransferases/deficiency , Adiposity , Animals , Body Weight , Female , Gene Knockout Techniques , Ghrelin/metabolism , Glucose/metabolism , Glucose Intolerance/blood , Homeostasis , Male , Membrane Proteins , Mice , Mice, Knockout , Mice, Obese , Obesity/blood , Obesity/enzymology , Phenotype
16.
J Clin Invest ; 123(1): 469-78, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23257354

ABSTRACT

The scaffold protein p62 (sequestosome 1; SQSTM1) is an emerging key molecular link among the metabolic, immune, and proliferative processes of the cell. Here, we report that adipocyte-specific, but not CNS-, liver-, muscle-, or myeloid-specific p62-deficient mice are obese and exhibit a decreased metabolic rate caused by impaired nonshivering thermogenesis. Our results show that p62 regulates energy metabolism via control of mitochondrial function in brown adipose tissue (BAT). Accordingly, adipocyte-specific p62 deficiency led to impaired mitochondrial function, causing BAT to become unresponsive to ß-adrenergic stimuli. Ablation of p62 leads to decreased activation of p38 targets, affecting signaling molecules that control mitochondrial function, such as ATF2, CREB, PGC1α, DIO2, NRF1, CYTC, COX2, ATP5ß, and UCP1. p62 ablation in HIB1B and BAT primary cells demonstrated that p62 controls thermogenesis in a cell-autonomous manner, independently of brown adipocyte development or differentiation. Together, our data identify p62 as a novel regulator of mitochondrial function and brown fat thermogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Heat-Shock Proteins/metabolism , Mitochondria/metabolism , Thermogenesis/physiology , Adaptor Proteins, Signal Transducing/genetics , Adipocytes, Brown/cytology , Adipose Tissue, Brown/cytology , Animals , Cells, Cultured , Heat-Shock Proteins/genetics , MAP Kinase Signaling System/physiology , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Organ Specificity/genetics , Sequestosome-1 Protein , Transcription Factors/genetics , Transcription Factors/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Nat Med ; 18(12): 1847-56, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23142820

ABSTRACT

We report the development of a new combinatorial approach that allows for peptide-mediated selective tissue targeting of nuclear hormone pharmacology while eliminating adverse effects in other tissues. Specifically, we report the development of a glucagon-like peptide-1 (GLP-1)-estrogen conjugate that has superior sex-independent efficacy over either of the individual hormones alone to correct obesity, hyperglycemia and dyslipidemia in mice. The therapeutic benefits are driven by pleiotropic dual hormone action to improve energy, glucose and lipid metabolism, as shown by loss-of-function models and genetic action profiling. Notably, the peptide-based targeting strategy also prevents hallmark side effects of estrogen in male and female mice, such as reproductive endocrine toxicity and oncogenicity. Collectively, selective activation of estrogen receptors in GLP-1-targeted tissues produces unprecedented efficacy to enhance the metabolic benefits of GLP-1 agonism. This example of targeting the metabolic syndrome represents the discovery of a new class of therapeutics that enables synergistic co-agonism through peptide-based selective delivery of small molecules. Although our observations with the GLP-1-estrogen conjugate justify translational studies for diabetes and obesity, the multitude of other possible combinations of peptides and small molecules may offer equal promise for other diseases.


Subject(s)
Estrogens/pharmacology , Glucagon-Like Peptide 1/pharmacology , Metabolic Syndrome/drug therapy , Receptors, Estrogen/metabolism , Analysis of Variance , Animals , Binding, Competitive , Body Composition/physiology , Chromatography, High Pressure Liquid , Drug Combinations , Drug Discovery , Estrogens/metabolism , Estrogens/therapeutic use , Female , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/therapeutic use , Glucose Tolerance Test , Humans , MCF-7 Cells , Magnetic Resonance Imaging , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Estrogen/genetics , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays
18.
J Pept Sci ; 18(6): 383-93, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22565812

ABSTRACT

The identification of leptin as a mediator of body weight regulation provided much initial excitement for the treatment of obesity. Unfortunately, leptin monotherapy is insufficient in reversing obesity in rodents or humans. Recent findings suggest that amylin is able to restore leptin sensitivity and when used in combination with leptin enhances body weight loss in obese rodents and humans. However, as the uniqueness of this combination therapy remains unclear, we assessed whether co-administration of leptin with other weight loss-inducing hormones equally restores leptin responsiveness in diet-induced obese (DIO) mice. Accordingly, we report here the design and characterization of a series of site-specifically enhanced leptin analogs of high potency and sustained action that, when administered in combination with exendin-4 or fibroblast growth factor 21 (FGF21), restores leptin responsiveness in DIO mice after an initial body weight loss of 30%. Using either combination, body weight loss was enhanced compared with either exendin-4 or FGF21 monotherapy, and leptin alone was sufficient to maintain the reduced body weight. In contrast, leptin monotherapy proved ineffective when identical weight loss was induced by caloric restriction alone over a comparable time. Accordingly, we find that a hypothalamic counter-regulatory response to weight loss, assessed using changes in hypothalamic agouti related peptide (AgRP) levels, is triggered by caloric restriction, but blunted by treatment with exendin-4. We conclude that leptin re-sensitization requires pharmacotherapy but does not appear to be restricted to a unique signaling pathway. Our findings provide preclinical evidence that high activity, long-acting leptin analogs are additively efficacious when used in combination with other weight-lowering agents.


Subject(s)
Diet/adverse effects , Fibroblast Growth Factors/pharmacology , Leptin/analogs & derivatives , Leptin/pharmacology , Obesity/metabolism , Peptides/pharmacology , Venoms/pharmacology , Animals , Body Weight , Drug Combinations , Exenatide , Fibroblast Growth Factors/administration & dosage , Leptin/administration & dosage , Leptin/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Obese , Models, Molecular , Obesity/chemically induced , Obesity/drug therapy , Peptides/administration & dosage , Polyethylene Glycols/chemistry , Venoms/administration & dosage
19.
PLoS One ; 7(2): e32100, 2012.
Article in English | MEDLINE | ID: mdl-22363801

ABSTRACT

OBJECTIVE: Ghrelin acylation by ghrelin O-acyltransferase (GOAT) has recently been reported to be essential for the prevention of hypoglycemia during prolonged negative energy balance. Using a unique set of four different genetic loss-of-function models for the GOAT/ghrelin/growth hormone secretagogue receptor (GHSR) system, we thoroughly tested the hypothesis that lack-of-ghrelin activation or signaling would lead to hypoglycemia during caloric deprivation. METHODOLOGY: Male and female knockout (KO) mice for GOAT, ghrelin, GHSR, or both ghrelin and GHSR (dKO) were subjected to prolonged calorie restriction (40% of ad libitum chow intake). Body weight, fat mass, and glucose levels were recorded daily and compared to wildtype (WT) controls. Forty-eight hour blood glucose profiles were generated for each individual mouse when 2% or less body fat mass was reached. Blood samples were obtained for analysis of circulating levels of acyl- and desacyl-ghrelin, IGF-1, and insulin. PRINCIPAL FINDINGS: Chronic calorie restriction progressively decreased body weight and body fat mass in all mice regardless of genotype. When fat mass was depleted to 2% or less of body weight for 2 consecutive days, random hypoglycemic events occurred in some mice across all genotypes. There was no increase in the incidence of hypoglycemia in any of the four loss-of-function models for ghrelin signaling including GOAT KO mice. Furthermore, no differences in insulin or IGF-1 levels were observed between genotypes. CONCLUSION: The endogenous GOAT-ghrelin-GHSR system is not essential for the maintenance of euglycemia during prolonged calorie restriction.


Subject(s)
Acyltransferases/metabolism , Caloric Restriction , Ghrelin/metabolism , Hypoglycemia/prevention & control , Adiposity , Animals , Blood Glucose/metabolism , Body Weight , Female , Genotype , Ghrelin/blood , Hypoglycemia/blood , Insulin/blood , Insulin-Like Growth Factor I/metabolism , Male , Membrane Proteins , Mice , Mice, Knockout , Models, Animal
20.
Mol Metab ; 1(1-2): 95-100, 2012.
Article in English | MEDLINE | ID: mdl-24024123

ABSTRACT

Obesity, type 2 diabetes, and related diseases represent major health threats to modern society. Related pathophysiology of impaired neuronal function in hypothalamic control centers regulating metabolism and body weight has been dissected extensively and recent studies have started focusing on potential roles of astrocytes and microglia. The hypothalamic vascular system, however, which maintains the microenvironment necessary for appropriate neuronal function, has been largely understudied. We recently discovered that high fat/high sucrose diet exposure leads to increased hypothalamic presence of immunoglobulin G (IgG1). Investigating this phenomenon further, we have discovered a significant increase in blood vessel length and density in the arcuate nucleus (ARC) of the hypothalamus in mice fed a high fat/high sucrose diet, compared to matched controls fed standard chow diet. We also found a clearly increased presence of α-smooth muscle actin immunoreactive vessels, which are rarely present in the ARC and indicate an increase in the formation of new arterial vessels. Along the blood brain barrier, an increase of degenerated endothelial cells are observed. Moreover, such hypothalamic angiogenesis was not limited to rodent models. We also found an increase in the number of arterioles of the infundibular nucleus (the human equivalent of the mouse ARC) in patients with type 2 diabetes, suggesting angiogenesis occurs in the human hypothalamus of diabetics. Our discovery reveals novel hypothalamic pathophysiology, which is reminiscent of diabetic retinopathy and suggests a potential functional involvement of the hypothalamic vasculature in the later stage pathogenesis of metabolic syndrome.

SELECTION OF CITATIONS
SEARCH DETAIL
...