1
|
Zur Nedden S, Safari MS, Weber D, Kuenkel L, Garmsiri C, Lang L, Orset C, Freret T, Haelewyn B, Hotze M, Kwiatkowski M, Sarg B, Faserl K, Savic D, Skvortsova II, Krogsdam A, Carollo S, Trajanoski Z, Oberacher H, Zlotek D, Ostermaier F, Cameron A, Baier G, Baier-Bitterlich G. Protein kinase N1 deficiency results in upregulation of cerebral energy metabolism and is highly protective in in vivo and in vitro stroke models. Metabolism 2024; 161:156039. [PMID: 39332493 DOI: 10.1016/j.metabol.2024.156039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/06/2024] [Accepted: 09/21/2024] [Indexed: 09/29/2024]
Abstract
BACKGROUND AND AIM We recently identified protein kinase N1 (PKN1) as a master regulator of brain development. However, its function in the adult brain has not been clearly established. In this study, we assessed the cerebral energetic phenotype of wildtype (WT) and global Pkn1 knockout (Pkn1-/-) animals under physiological and pathophysiological conditions. METHODS Cerebral energy metabolism was analyzed by 13C6-glucose tracing in vivo and real time seahorse analysis of extracellular acidification rates as well as mitochondrial oxygen consumption rates (OCR) of brain slice punches in vitro. Isolated WT and Pkn1-/- brain mitochondria were tested for differences in OCR with different substrates. Metabolite levels were determined by mass spectrometric analysis in brain slices under control and energetic stress conditions, induced by oxygen-glucose deprivation and reperfusion, an in vitro model of ischemic stroke. Differences in enzyme activities were assessed by enzymatic assays, western blotting and bulk RNA sequencing. A middle cerebral artery occlusion stroke model was used to analyze lesion volumes and functional recovery in WT and Pkn1-/- mice. RESULTS Pkn1 deficiency resulted in a remarkable upregulation of cerebral energy metabolism, in vivo and in vitro. This was due to two separate mechanisms involving an enhanced glycolytic flux and higher pyruvate-induced mitochondrial OCR. Mechanistically we show that Pkn1-/- brain tissue exhibits an increased activity of the glycolysis rate-limiting enzyme phosphofructokinase. Additionally, glucose-1,6-bisphosphate levels, a metabolite that increases mitochondrial pyruvate uptake, were elevated upon Pkn1 deficiency. Consequently, Pkn1-/- brain slices had more ATP and a greater accumulation of ATP degradation metabolites during energetic stress. This translated into increased phosphorylation and activity of adenosine monophosphate (AMP)-activated protein kinase (AMPK) during in vitro stroke. Accordingly, Pkn1-/- brain slices showed a post-ischemic transcriptional upregulation of energy metabolism pathways and Pkn1 deficiency was strongly protective in in vitro and in vivo stroke models. While inhibition of mitochondrial pyruvate uptake only moderately affected the protective phenotype, inhibition of AMPK in Pkn1-/- slices increased post-ischemic cell death in vitro. CONCLUSION This is the first study to comprehensively demonstrate an essential and unique role of PKN1 in cerebral energy metabolism, regulating glycolysis and mitochondrial pyruvate-induced respiration. We further uncovered a highly protective phenotype of Pkn1 deficiency in both, in vitro and in vivo stroke models, validating inhibition of PKN1 as a promising new therapeutic target for the development of novel stroke therapies.
Collapse
Affiliation(s)
- Stephanie Zur Nedden
- Medical University of Innsbruck, CCB-Biocenter, Institute of Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria.
| | - Motahareh S Safari
- Medical University of Innsbruck, CCB-Biocenter, Institute of Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Dido Weber
- Medical University of Innsbruck, CCB-Biocenter, Institute of Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Louisa Kuenkel
- Medical University of Innsbruck, CCB-Biocenter, Institute of Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Carolin Garmsiri
- Medical University of Innsbruck, CCB-Biocenter, Institute of Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Luisa Lang
- Medical University of Innsbruck, CCB-Biocenter, Institute of Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Cyrille Orset
- Normandie University, UNICAEN, ESR3P, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Institute of Blood and Brain @ Caen-Normandie (BB@C), GIP Cyceron, Caen, France
| | - Tom Freret
- Normandie University, UNICAEN, ESR3P, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Institute of Blood and Brain @ Caen-Normandie (BB@C), GIP Cyceron, Caen, France
| | - Benoît Haelewyn
- Normandie University, UNICAEN, ESR3P, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Institute of Blood and Brain @ Caen-Normandie (BB@C), GIP Cyceron, Caen, France
| | - Madlen Hotze
- University of Innsbruck, Department of Biochemistry, Institute of Bioanalytic & Intermediary Metabolism, 6020 Innsbruck, Austria
| | - Marcel Kwiatkowski
- University of Innsbruck, Department of Biochemistry, Institute of Bioanalytic & Intermediary Metabolism, 6020 Innsbruck, Austria
| | - Bettina Sarg
- Medical University of Innsbruck, CCB-Biocenter, Institute of Medical Biochemistry, Protein Core Facility, 6020 Innsbruck, Austria
| | - Klaus Faserl
- Medical University of Innsbruck, CCB-Biocenter, Institute of Medical Biochemistry, Protein Core Facility, 6020 Innsbruck, Austria
| | - Dragana Savic
- Medical University of Innsbruck, Laboratory for Experimental and Translational Research on Radiation Oncology (EXTRO-Lab), Department of Therapeutic Radiology and Oncology, 6020 Innsbruck, Austria; Tyrolean Cancer Research Institute, Innsbruck A-6020, Austria
| | - Ira-Ida Skvortsova
- Medical University of Innsbruck, Laboratory for Experimental and Translational Research on Radiation Oncology (EXTRO-Lab), Department of Therapeutic Radiology and Oncology, 6020 Innsbruck, Austria; Tyrolean Cancer Research Institute, Innsbruck A-6020, Austria
| | - Anne Krogsdam
- Medical University of Innsbruck, CCB-Biocenter, Institute of Bioinformatics, 6020 Innsbruck, Austria
| | - Sandro Carollo
- Medical University of Innsbruck, CCB-Biocenter, Institute of Bioinformatics, 6020 Innsbruck, Austria
| | - Zlatko Trajanoski
- Medical University of Innsbruck, CCB-Biocenter, Institute of Bioinformatics, 6020 Innsbruck, Austria
| | - Herbert Oberacher
- Medical University of Innsbruck, Institute of Legal Medicine and Core Facility Metabolomics, 6020 Innsbruck, Austria
| | - Dominik Zlotek
- Medical University of Innsbruck, CCB-Biocenter, Institute of Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Florian Ostermaier
- Medical University of Innsbruck, CCB-Biocenter, Institute of Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Angus Cameron
- Kinase Biology Laboratory, John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Gottfried Baier
- Medical University of Innsbruck, Institute for Cell Genetics, 6020 Innsbruck, Austria
| | - Gabriele Baier-Bitterlich
- Medical University of Innsbruck, CCB-Biocenter, Institute of Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria
| |
Collapse
|
2
|
Roy SC, Sapkota S, Pasula MB, Briski KP. In Vivo Glucose Transporter-2 Regulation of Dorsomedial Versus Ventrolateral VMN Astrocyte Metabolic Sensor and Glycogen Metabolic Enzyme Gene Expression in Female Rat. Neurochem Res 2024; 49:3367-3382. [PMID: 39306597 DOI: 10.1007/s11064-024-04246-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 10/25/2024]
Abstract
Astrocyte glycogenolysis shapes ventromedial hypothalamic nucleus (VMN) regulation of glucostasis in vivo. Glucose transporter-2 (GLUT2), a plasma membrane glucose sensor, controls hypothalamic primary astrocyte culture glycogen metabolism in vitro. In vivo gene silencing tools and single-cell laser-catapult-microdissection/multiplex qPCR techniques were used here to examine whether GLUT2 governs dorsomedial (VMNdm) and/or ventrolateral (VMNvl) VMN astrocyte metabolic sensor and glycogen metabolic enzyme gene profiles. GLUT2 gene knockdown diminished astrocyte GLUT2 mRNA in both VMN divisions. Hypoglycemia caused GLUT2 siRNA-reversible up-regulation of this gene profile in the VMNdm, but down-regulated VMNvl astrocyte GLUT2 transcription. GLUT2 augmented baseline VMNdm and VMNvl astrocyte glucokinase (GCK) gene expression, but increased (VMNdm) or reduced (VMNvl) GCK transcription during hypoglycemia. GLUT2 imposed opposite control, namely stimulation versus inhibition of VMNdm or VMNvl astrocyte 5'-AMP-activated protein kinase-alpha 1 and -alpha 2 gene expression, respectively. GLUT2 stimulated astrocyte glycogen synthase (GS) gene expression in each VMN division. GLUT2 inhibited transcription of the AMP-sensitive glycogen phosphorylase (GP) isoform GP-brain type (GPbb) in each site, yet diminished (VMNdm) or augmented (VMNvl) astrocyte GP-muscle type (GPmm) mRNA. GLUT2 enhanced VMNdm and VMNvl glycogen accumulation during euglycemia, and curbed hypoglycemia-associated VMNdm glycogen depletion. Results show that VMN astrocytes exhibit opposite, division-specific GLUT2 transcriptional responsiveness to hypoglycemia. Data document divergent GLUT2 control of GCK, AMPK catalytic subunit, and GPmm gene profiles in VMNdm versus VMNvl astrocytes. Ongoing studies seek to determine how differential GLUT2 regulation of glucose and energy sensor function and glycogenolysis in each VMN location may affect local neuron responses to hypoglycemia.
Collapse
Affiliation(s)
- Sagor C Roy
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, 71201, USA
| | - Subash Sapkota
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, 71201, USA
| | - Madhu Babu Pasula
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, 71201, USA
| | - Karen P Briski
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, 71201, USA.
- UL System Foundation and Willis-Knighton Health Systems Professorship in Toxicology, College of Pharmacy, University of Louisiana at Monroe, Rm 356 Bienville Building, 1800 Bienville Drive, Monroe, LA, 71201, USA.
| |
Collapse
|
3
|
Sapkota S, Roy SC, Briski KP. Dorsomedial Ventromedial Hypothalamic Nucleus Growth Hormone-Releasing Hormone Neuron Steroidogenic Factor-1 Gene Targets in Female Rat. ASN Neuro 2024; 16:2403345. [PMID: 39401164 PMCID: PMC11792125 DOI: 10.1080/17590914.2024.2403345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/07/2024] [Indexed: 10/18/2024] Open
Abstract
The prospect that the ventromedial hypothalamic nucleus (VMN) transcription factor steroidogenic factor-1/NR5A1 (SF-1) may exert sex-dimorphic control of glucose counterregulation is unresolved. Recent studies in male rats show that SF-1 regulates transcription of co-expressed hypoglycemia-sensitive neurochemicals in dorsomedial VMN growth hormone-releasing hormone (Ghrh) neurons. Gene knockdown and laser-catapult-microdissection/single-cell multiplex qPCR techniques were used here in a female rat model to determine if SF-1 control of Ghrh neuron transmitter marker, energy sensor, and estrogen receptor (ER) variant mRNAs varies according to sex. Data show that in females, hypoglycemia elicits a gain of SF-1 inhibitory control of VMNdm Ghrh neuron Ghrh and Ghrh-receptor gene profiles and loss of augmentation of glutaminase transcription; SF-1 gene silencing diminished eu- and hypoglycemic patterns of neuronal nitric oxide gene transcription. SF-1 imposes divergent control of baseline and hypoglycemic glutamate decarboxylase65 (GAD)-1 (stimulatory) versus GAD2 (inhibitory) mRNAs in that sex. SF-1 stimulates baseline VMNdm Ghrh neuron PRKAA1/AMPKα1 and PRKAA2/AMPKα2 gene expression, yet causes opposite changes in these gene profiles during hypoglycemia. SF-1 exerts glucose-dependent control of ER-alpha and G-protein-coupled ER-1 transcription, but blunts ER-beta gene profiles during eu- and hypoglycemia. In females, SF-1 knockdown did not affect hypercorticosteronemia or hyperglucagonemia, but blunted hypoglycemic suppression of growth hormone secretion. Results show that SF-1 expression is critical for female rat VMNdm Ghrh neuron counterregulatory neurochemical, AMPK catalytic subunit, and ER gene transcription responses to hypoglycemia. Sex differences in direction of SF-1 control of distinctive gene profiles may result in observed disparities in SF-1 regulation of counterregulatory hormone secretion between sexes.
Collapse
Affiliation(s)
- Subash Sapkota
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| | - Sagor C. Roy
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| | - Karen P. Briski
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| |
Collapse
|
4
|
Sapkota S, Roy SC, Shrestha R, Briski KP. Steroidogenic Factor-1 Regulation of Dorsomedial Ventromedial Hypothalamic Nucleus Ghrh Neuron Transmitter Marker and Estrogen Receptor Gene Expression in Male Rat. ASN Neuro 2024; 16:2368382. [PMID: 39024550 PMCID: PMC11262038 DOI: 10.1080/17590914.2024.2368382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2024] [Indexed: 07/20/2024] Open
Abstract
Ventromedial hypothalamic nucleus (VMN) growth hormone-releasing hormone (Ghrh) neurotransmission shapes counterregulatory hormone secretion. Dorsomedial VMN Ghrh neurons express the metabolic-sensitive transcription factor steroidogenic factor-1/NR5A1 (SF-1). In vivo SF-1 gene knockdown tools were used here to address the premise that in male rats, SF-1 may regulate basal and/or hypoglycemic patterns of Ghrh, co-transmitter biosynthetic enzyme, and estrogen receptor (ER) gene expression in these neurons. Single-cell multiplex qPCR analyses showed that SF-1 regulates basal profiles of mRNAs that encode Ghrh and protein markers for neurochemicals that suppress (γ-aminobutyric acid) or enhance (nitric oxide; glutamate) counterregulation. SF-1 siRNA pretreatment respectively exacerbated or blunted hypoglycemia-associated inhibition of glutamate decarboxylase67 (GAD67/GAD1) and -65 (GAD65/GAD2) transcripts. Hypoglycemia augmented or reduced nitric oxide synthase and glutaminase mRNAs, responses that were attenuated by SF-1 gene silencing. Ghrh and Ghrh receptor transcripts were correspondingly refractory to or increased by hypoglycemia, yet SF-1 knockdown decreased both gene profiles. Hypoglycemic inhibition of ER-alpha and G protein-coupled-ER gene expression was amplified by SF-1 siRNA pretreatment, whereas as ER-beta mRNA was amplified. SF-1 knockdown decreased (corticosterone) or elevated [glucagon, growth hormone (GH)] basal counterregulatory hormone profiles, but amplified hypoglycemic hypercorticosteronemia and -glucagonemia or prevented elevated GH release. Outcomes document SF-1 control of VMN Ghrh neuron counterregulatory neurotransmitter and ER gene transcription. SF-1 likely regulates Ghrh nerve cell receptivity to estradiol and release of distinctive neurochemicals during glucose homeostasis and systemic imbalance. VMN Ghrh neurons emerge as a likely substrate for SF-1 control of glucose counterregulation in the male rat.
Collapse
Affiliation(s)
- Subash Sapkota
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
| | - Sagor C Roy
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
| | - Rami Shrestha
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
| | - Karen P Briski
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
| |
Collapse
|
5
|
Xie L, Hao X, Xie J, Mo J, Yuan C, Chen W. Acetylated pelargonidin-3- O-glucoside alleviates hepatocyte lipid deposition through activating the AMPK-mediated lysosome-autophagy pathway and redox state. Food Funct 2024; 15:6929-6942. [PMID: 38659316 DOI: 10.1039/d4fo00185k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a worldwide public health issue, but a widely accepted therapy is still lacking until now. Anthocyanins are natural flavonoid compounds that possess various bioactivities, but their applications are limited due to their low bioavailability and stability. Acylated anthocyanins are reported to show higher stability, whereas their effects on NAFLD are still unclear. Herein, pelargonidin-3-O-(6''-acetyl)-glucoside (Ace Pg3G) was found to dose-dependently reduce intracellular lipid droplets and triglycerides, and improve cellular oxidative stress that accompanied lipid deposition. Besides, Ace Pg3G was proved to activate AMPK phosphorylation, thus stimulating AMPK-mediated lysosome-autophagy pathway to eliminate overloaded lipid. Further study unveiled that Ace Pg3G regulated genes related to lipid metabolism downstream of AMPK to inhibit lipid synthesis and accelerate lipid oxidation. Overall, this study provided the first evidence, to our best knowledge, that Ace Pg3G ameliorated free fatty acid-induced lipid deposition in hepatocytes through regulating AMPK-mediated autophagy pathways and redox state.
Collapse
Affiliation(s)
- Lianghua Xie
- Department of Traditional Chinese Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Xin Hao
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Jiahong Xie
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Jianling Mo
- Department of Traditional Chinese Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.
| | - Changzheng Yuan
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Wei Chen
- Department of Traditional Chinese Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
6
|
Sapkota S, Briski KP. Sex-Dimorphic Effects of Hypoglycemia on Metabolic Sensor mRNA Expression in Ventromedial Hypothalamic Nucleus-Dorsomedial Division (VMNdm) Growth Hormone-Releasing Hormone Neurons. ACS Chem Neurosci 2024; 15:2350-2358. [PMID: 38757688 DOI: 10.1021/acschemneuro.4c00206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024] Open
Abstract
Growth hormone-releasing hormone (Ghrh) neurons in the dorsomedial ventromedial hypothalamic nucleus (VMNdm) express the metabolic transcription factor steroidogenic factor-1 and hypoglycemia-sensitive neurochemicals of diverse chemical structures, transmission modes, and temporal signaling profiles. Ghrh imposes neuromodulatory control of coexpressed transmitters. Multiple metabolic sensory mechanisms are employed in the brain, including screening of the critical nutrient glucose or the energy currency ATP. Here, combinatory laser-catapult-microdissection/single-cell multiplex qPCR tools were used to investigate whether these neurons possess molecular machinery for monitoring cellular metabolic status and if these biomarkers exhibit sex-specific sensitivity to insulin-induced hypoglycemia. Data show that hypoglycemia up- (male) or downregulated (female) Ghrh neuron glucokinase (Gck) mRNA; Ghrh gene silencing decreased baseline and hypoglycemic patterns of Gck gene expression in each sex. Ghrh neuron glucokinase regulatory protein (Gckr) transcript levels were respectively diminished or augmented in hypoglycemic male vs female rats; this mRNA profile was decreased by Ghrh siRNA in both sexes. Gene transcripts encoding catalytic alpha subunits of the energy monitor 5-AMP-activated protein kinase (AMPK), i.e., Prkaa1 and 2, were increased by hypoglycemia in males, yet only the former mRNA was hypoglycemia-sensitive in females. Ghrh siRNA downregulated baseline and hypoglycemia-associated Prkaa subunit mRNAs in males but elicited divergent changes in Prkaa2 transcripts in eu- vs hypoglycemic females. Results provide unique evidence that VMNdm Ghrh neurons express the characterized metabolic sensor biomarkers glucokinase and AMPK and that the corresponding gene profiles exhibit distinctive sex-dimorphic transcriptional responses to hypoglycemia. Data further document Ghrh neuromodulation of baseline and hypoglycemic transcription patterns of these metabolic gene profiles.
Collapse
Affiliation(s)
- Subash Sapkota
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana 71201, United States
| | - Karen P Briski
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana 71201, United States
| |
Collapse
|
7
|
Carneiro L, Bernasconi R, Bernini A, Repond C, Pellerin L. Elevation of hypothalamic ketone bodies induces a decrease in energy expenditures and an increase risk of metabolic disorder. Mol Metab 2024; 83:101926. [PMID: 38553002 PMCID: PMC10999683 DOI: 10.1016/j.molmet.2024.101926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 03/18/2024] [Accepted: 03/22/2024] [Indexed: 04/04/2024] Open
Abstract
OBJECTIVE Ketone bodies (such as β-hydroxybutyrate or BHB) have been recently proposed as signals involved in brain regulation of energy homeostasis and obesity development. However, the precise role of ketone bodies sensing by the brain, and its impact on metabolic disorder development remains unclear. Nevertheless, partial deletion of the ubiquitous ketone bodies transporter MCT1 in mice (HE mice) results in diet-induced obesity resistance, while there is no alteration under normal chow diet. These results suggest that ketone bodies produced during the high fat diet would be important signals involved in obesity onset. METHODS In the present study we used a specific BHB infusion of the hypothalamus and analyzed the energy homeostasis of WT or HE mice fed a normal chow diet. RESULTS Our results indicate that high BHB levels sensed by the hypothalamus disrupt the brain regulation of energy homeostasis. This brain control dysregulation leads to peripheral alterations of energy expenditure mechanisms. CONCLUSIONS Altogether, the changes induced by high ketone bodies levels sensed by the brain increase the risk of obesity onset in mice.
Collapse
Affiliation(s)
- Lionel Carneiro
- Department of Physiology, University of Lausanne, 1005 Lausanne, Switzerland.
| | - Rocco Bernasconi
- Department of Physiology, University of Lausanne, 1005 Lausanne, Switzerland
| | - Adriano Bernini
- Department of Physiology, University of Lausanne, 1005 Lausanne, Switzerland
| | - Cendrine Repond
- Department of Physiology, University of Lausanne, 1005 Lausanne, Switzerland
| | - Luc Pellerin
- Department of Physiology, University of Lausanne, 1005 Lausanne, Switzerland; University and CHU of Poitiers, INSERM U1313, Poitiers, France.
| |
Collapse
|
8
|
Bheemanapally K, Briski KP. Differential G Protein-Coupled Estrogen Receptor-1 Regulation of Counter-Regulatory Transmitter Marker and 5'-AMP-Activated Protein Kinase Expression in Ventrolateral versus Dorsomedial Ventromedial Hypothalamic Nucleus. Neuroendocrinology 2023; 114:25-41. [PMID: 37699381 PMCID: PMC10843453 DOI: 10.1159/000533627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/14/2023] [Indexed: 09/14/2023]
Abstract
INTRODUCTION The ventromedial hypothalamic nucleus (VMN) is an estrogen receptor (ER)-rich structure that regulates glucostasis. The role of nuclear but not membrane G protein-coupled ER-1 (GPER) in that function has been studied. METHODS Gene silencing and laser-catapult microdissection/immunoblot tools were used to examine whether GPER regulates transmitter and energy sensor function in dorsomedial (VMNdm) and/or ventrolateral (VMNvl) VMN counter-regulatory nitrergic and γ-Aminobutyric acid (GABA) neurons. RESULTS Intra-VMN GPER siRNA administration to euglycemic animals did not affect VMNdm or -vl nitrergic neuron nitric oxide synthase (nNOS), but upregulated (VMNdm) or lacked influence on (VMNvl) GABA nerve cell glutamate decarboxylase65/67 (GAD) protein. Insulin-induced hypoglycemia (IIH) caused GPER knockdown-reversible augmentation of nNOS, 5'-AMP-activated protein kinase (AMPK), and phospho-AMPK proteins in nitrergic neurons in both divisions. IIH had dissimilar effects on VMNvl (unchanged) versus VMNdm (increased) GABAergic neuron GAD levels, yet GPER knockdown affected these profiles. GPER siRNA prevented hypoglycemic upregulation of VMNvl and -dm GABA neuron AMPK without altering pAMPK expression. CONCLUSIONS Outcomes infer that GPER exerts differential control of VMNdm versus -vl GABA transmission during glucostasis and is required for hypoglycemic upregulated nitrergic (VMNdm and -vl) and GABA (VMNdm) signaling. Glycogen metabolism is reported to regulate VMN nNOS and GAD proteins. Data show that GPER limits VMNvl glycogen phosphorylase (GP) protein expression and glycogen buildup during euglycemia but mediates hypoglycemic augmentation of VMNvl GP protein and glycogen content; VMNdm glycogen mass is refractory to GPER control. GPER regulation of VMNvl glycogen metabolism infers that this receptor may govern local counter-regulatory transmission in part by astrocyte metabolic coupling.
Collapse
Affiliation(s)
- Khaggeswar Bheemanapally
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
| | - Karen P Briski
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
| |
Collapse
|
9
|
Napit PR, Ali MH, Mahmood ASMH, Ibrahim MMH, Briski KP. Sex-dimorphic hindbrain lactate regulation of ventromedial hypothalamic nucleus glucoregulatory neuron 5'-AMP-activated protein kinase activity and transmitter marker protein expression. Neuropeptides 2023; 99:102324. [PMID: 36791640 PMCID: PMC10175150 DOI: 10.1016/j.npep.2023.102324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 01/16/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023]
Abstract
BACKGROUND The oxidizable glycolytic end-product L-lactate is a gauge of nerve cell metabolic fuel stability that metabolic-sensory hindbrain A2 noradrenergic neurons impart to the brain glucose-regulatory network. Current research investigated the premise that hindbrain lactate deficiency exerts sex-specific control of energy sensor and transmitter marker protein responses to hypoglycemia in ventromedial hypothalamic nucleus (VMN) glucose-regulatory nitrergic and γ-aminobutyric acid (GABA) neurons. METHODS Nitric oxide synthase (nNOS)- or glutamate decarboxylase65/67 (GAD)-immunoreactive neurons were laser-catapult-microdissected from male and female rat VMN after subcutaneous insulin injection and caudal fourth ventricular L-lactate or vehicle infusion for Western blot protein analysis. RESULTS Hindbrain lactate repletion reversed hypoglycemia-associated augmentation (males) or inhibition (females) of nitrergic neuron nNOS expression, and prevented up-regulation of phosphorylated AMPK 5'-AMP-activated protein kinase (pAMPK) expression in those neurons. Hypoglycemic suppression of GABAergic neuron GAD protein was averted by exogenous lactate over the rostro-caudal length of the male VMN and in the middle region of the female VMN. Lactate normalized GABA neuron pAMPK profiles in hypoglycemic male (caudal VMN) and female (all VMN segments) rats. Hypoglycemic patterns of norepinephrine (NE) signaling were lactate-dependent throughout the male VMN, but confined to the rostral and middle female VMN. CONCLUSIONS Results document, in each sex, regional VMN glucose-regulatory transmitter responses to hypoglycemia that are controlled by hindbrain lactate status. Hindbrain metabolic-sensory regulation of hypoglycemia-correlated nitric oxide or GABA release may entail AMPK-dependent mechanisms in specific VMN rostro-caudal segments in each sex. Additional effort is required to examine the role of hindbrain lactoprivic-sensitive VMN neurotransmitters in lactate-mediated attenuation of hypoglycemic hyperglucagonemia and hypercorticosteronemia in male and female rats.
Collapse
Affiliation(s)
- Prabhat R Napit
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, United States
| | - Md Haider Ali
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, United States
| | - A S M Hasan Mahmood
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, United States
| | - Mostafa M H Ibrahim
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, United States
| | - Karen P Briski
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, United States.
| |
Collapse
|
10
|
Briski KP, Napit PR, Alhamyani A, Leprince J, Mahmood AH. Sex-Dimorphic Octadecaneuropeptide (ODN) Regulation of Ventromedial Hypothalamic Nucleus Glucoregulatory Neuron Function and Counterregulatory Hormone Secretion. ASN Neuro 2023; 15:17590914231167230. [PMID: 37194319 PMCID: PMC10196551 DOI: 10.1177/17590914231167230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 03/12/2023] [Accepted: 03/16/2023] [Indexed: 05/18/2023] Open
Abstract
Central endozepinergic signaling is implicated in glucose homeostasis. Ventromedial hypothalamic nucleus (VMN) metabolic monitoring governs glucose counter-regulation. VMN glucose-stimulatory nitric oxide (NO) and glucose-inhibitory γ-aminobutyric acid (GABA) neurons express the energy gauge 5'-AMP-activated protein kinase (AMPK). Current research addresses the premise that the astrocyte glio-peptide octadecaneuropeptide (ODN) imposes sex-dimorphic control of metabolic sensor activity and neurotransmitter signaling in these neurons. The ODN G-protein coupled-receptor antagonist cyclo(1-8)[DLeu5]OP (LV-1075) was administered intracerebroventricularly (icv) to euglycemic rats of each sex; additional groups were pretreated icv with the ODN isoactive surrogate ODN11-18 (OP) before insulin-induced hypoglycemia. Western blotting of laser-catapult-microdissected VMN NO and GABA neurons showed that hypoglycemia caused OP-reversible augmentation of phospho-, e.g., activated AMPK and nitric oxide synthase (nNOS) expression in rostral (female) or middle (male) VMN segments or ODN-dependent suppression of nNOS in male caudal VMN. OP prevented hypoglycemic down-regulation of glutamate decarboxylase profiles in female rat rostral VMN, without affecting AMPK activity. LV-1075 treatment of male, not female rats elevated plasma glucagon and corticosterone concentrations. Moreover, OP attenuated hypoglycemia-associated augmentation of these hormones in males only. Results identify, for each sex, regional VMN metabolic transmitter signals that are subject to endozepinergic regulation. Directional shifts and gain-or-loss of ODN control during eu- versus hypoglycemia infer that VMN neuron receptivity to or post-receptor processing of this stimulus may be modulated by energy state. In male, counter-regulatory hormone secretion may be governed principally by ODN-sensitive neural pathways, whereas this endocrine outflow may be controlled by parallel, redundant ODN-dependent and -independent mechanisms in female.
Collapse
Affiliation(s)
- Karen P. Briski
- School of Basic Pharmaceutical and
Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA,
USA
| | - Prabhat R. Napit
- School of Basic Pharmaceutical and
Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA,
USA
| | - Abdulrahman Alhamyani
- School of Basic Pharmaceutical and
Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA,
USA
| | - Jérôme Leprince
- Neuronal and Neuroendocrine Differentiation
and Communication Laboratory, Normandy University, INSERM U1239, PRIMACEN, Rouen,
France
| | - A.S.M. Hasan Mahmood
- School of Basic Pharmaceutical and
Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA,
USA
| |
Collapse
|
11
|
Roy SC, Sapkota S, Pasula MB, Bheemanapally K, Briski KP. Diazepam Binding Inhibitor Control of Eu- and Hypoglycemic Patterns of Ventromedial Hypothalamic Nucleus Glucose-Regulatory Signaling. ASN Neuro 2023; 15:17590914231214116. [PMID: 38031405 PMCID: PMC10687944 DOI: 10.1177/17590914231214116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/20/2023] [Accepted: 10/28/2023] [Indexed: 12/01/2023] Open
Abstract
Pharmacological stimulation/antagonism of astrocyte glio-peptide octadecaneuropeptide signaling alters ventromedial hypothalamic nucleus (VMN) counterregulatory γ-aminobutyric acid (GABA) and nitric oxide transmission. The current research used newly developed capillary zone electrophoresis-mass spectrometry methods to investigate hypoglycemia effects on VMN octadecaneuropeptide content, along with gene knockdown tools to determine if octadecaneuropeptide signaling regulates these transmitters during eu- and/or hypoglycemia. Hypoglycemia caused dissimilar adjustments in the octadecaneuropeptide precursor, i.e., diazepam-binding-inhibitor and octadecaneuropeptide levels in dorsomedial versus ventrolateral VMN. Intra-VMN diazepam-binding-inhibitor siRNA administration decreased baseline 67 and 65 kDa glutamate decarboxylase mRNA levels in GABAergic neurons laser-microdissected from each location, but only affected hypoglycemic transcript expression in ventrolateral VMN. This knockdown therapy imposed dissimilar effects on eu- and hypoglycemic glucokinase and 5'-AMP-activated protein kinase-alpha1 (AMPKα1) and -alpha2 (AMPKα2) gene profiles in dorsomedial versus ventrolateral GABAergic neurons. Diazepam-binding-inhibitor gene silencing up-regulated baseline (dorsomedial) or hypoglycemic (ventrolateral) nitrergic neuron neuronal nitric oxide synthase mRNA profiles. Baseline nitrergic cell glucokinase mRNA was up- (ventrolateral) or down- (dorsomedial) regulated by diazepam-binding-inhibitor siRNA, but knockdown enhanced hypoglycemic profiles in both sites. Nitrergic nerve cell AMPKα1 and -α2 transcripts exhibited division-specific responses to this genetic manipulation during eu- and hypoglycemia. Results document the utility of capillary zone electrophoresis-mass spectrometric tools for quantification of ODN in small-volume brain tissue samples. Data show that hypoglycemia has dissimilar effects on ODN signaling in the two major neuroanatomical divisions of the VMN and that this glio-peptide imposes differential control of glucose-regulatory neurotransmission in the VMNdm versus VMNvl during eu- and hypoglycemia.
Collapse
Affiliation(s)
- Sagor C. Roy
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| | - Subash Sapkota
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| | - Madhu Babu Pasula
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| | - Khaggeswar Bheemanapally
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| | - Karen P. Briski
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| |
Collapse
|
12
|
Haghshenas L, Nabi-Afjadi M, Zalpoor H, Bakhtiyari M, Marotta F. Energy Restriction on Cellular and Molecular Mechanisms in Aging. EVIDENCE-BASED FUNCTIONAL FOODS FOR PREVENTION OF AGE-RELATED DISEASES 2023:297-323. [DOI: 10.1007/978-981-99-0534-8_16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
13
|
Wu Z, Rao S, Li J, Ding N, Chen J, Feng L, Ma S, Hu C, Dai H, Wen L, Jiang Q, Deng J, Deng M, Tan C. Dietary adenosine 5’-monophosphate supplementation increases food intake and remodels energy expenditure in mice. Food Nutr Res 2022; 66:7680. [PMID: 35844957 PMCID: PMC9250134 DOI: 10.29219/fnr.v66.7680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 06/30/2021] [Accepted: 10/04/2021] [Indexed: 11/20/2022] Open
Abstract
Background Methods Results Conclusions
Collapse
Affiliation(s)
- Zifang Wu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Sujuan Rao
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Jiaying Li
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Ning Ding
- Guangzhou Customs Technology Center, 510623, China
| | - Jianzhao Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Li Feng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Shuo Ma
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Chengjun Hu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
- Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Haonan Dai
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Lijun Wen
- Guangdong Hinabiotech Co., Ltd., Guangzhou, China
| | - Qingyan Jiang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Jinping Deng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
- Ming Deng,
| | - Ming Deng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
- Ming Deng,
| | - Chengquan Tan
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
- Chengquan Tan, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.
| |
Collapse
|
14
|
Alhamyani A, Napit PR, Bheemanapally K, Sylvester PW, Briski KP. Singular versus combinatory glucose-sensitive signal control of metabolic sensor protein profiles in hypothalamic astrocyte cultures from each sex. Transl Neurosci 2022; 13:408-420. [PMID: 36518559 PMCID: PMC9719392 DOI: 10.1515/tnsci-2022-0259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/08/2022] [Accepted: 10/21/2022] [Indexed: 01/05/2025] Open
Abstract
Brain metabolic-sensory targets for modulatory glucose-sensitive endocrine and neurochemical signals remain unidentified. A hypothalamic astrocyte primary culture model was here used to investigate whether glucocorticoid receptor (GR) and noradrenergic signals regulate astrocyte glucose (glucose transporter-2 [GLUT2], glucokinase) and/or energy (5'-AMP-activated protein kinase [AMPK]) sensor reactivity to glucoprivation by sex. Glucose-supplied astrocytes of each sex showed increased GLUT2 expression after incubation with the GR agonist dexamethasone (DEX) or norepinephrine (NE); DEX plus NE (DEX/NE) augmented GLUT2 in the female, but not in male. Glucoprivation did not alter GLUT2 expression, but eliminated NE regulation of this protein in both sexes. Male and female astrocyte glucokinase profiles were refractory to all drug treatments, but were down-regulated by glucoprivation. Glucoprivation altered AMPK expression in male only, and caused divergent sex-specific changes in activated, i.e., phosphoAMPK (pAMPK) levels. DEX or DEX/NE inhibited (male) or stimulated (female) AMPK and pAMPK proteins in both glucose-supplied and -deprived astrocytes. In male, NE coincidently up-regulated AMPK and inhibited pAMPK profiles in glucose-supplied astrocytes; these effects were abolished by glucoprivation. In female, AMPK profiles were unaffected by NE irrespective of glucose status, whereas pAMPK expression was up-regulated by NE only during glucoprivation. Present outcomes document, for each sex, effects of glucose status on hypothalamic astrocyte glucokinase, AMPK, and pAMPK protein expression and on noradrenergic control of these profiles. Data also show that DEX and NE regulation of GLUT2 is sex-monomorphic, but both stimuli impose divergent sex-specific effects on AMPK and pAMPK. Further effort is warranted to characterize mechanisms responsible for sex-dimorphic GR and noradrenergic governance of hypothalamic astrocyte energy sensory function.
Collapse
Affiliation(s)
- Abdulrahman Alhamyani
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Rm 356 Bienville Building, 1800 Bienville Drive, Monroe, LA 71201, United States; Pharmaceuticals Chemistry Department, Faculty of Clinical Pharmacy, Al Baha University, Al Baha city, 65779, Saudi Arabia
| | - Prabhat R. Napit
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Rm 356 Bienville Building, 1800 Bienville Drive, Monroe, LA 71201, United States; Pharmaceuticals Chemistry Department, Faculty of Clinical Pharmacy, Al Baha University, Al Baha city, 65779, Saudi Arabia
| | - Khaggeswar Bheemanapally
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Rm 356 Bienville Building, 1800 Bienville Drive, Monroe, LA 71201, United States; Pharmaceuticals Chemistry Department, Faculty of Clinical Pharmacy, Al Baha University, Al Baha city, 65779, Saudi Arabia
| | - Paul W. Sylvester
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Rm 356 Bienville Building, 1800 Bienville Drive, Monroe, LA 71201, United States; Pharmaceuticals Chemistry Department, Faculty of Clinical Pharmacy, Al Baha University, Al Baha city, 65779, Saudi Arabia
| | - Karen P. Briski
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Rm 356 Bienville Building, 1800 Bienville Drive, Monroe, LA 71201, United States; Pharmaceuticals Chemistry Department, Faculty of Clinical Pharmacy, Al Baha University, Al Baha city, 65779, Saudi Arabia
| |
Collapse
|
15
|
Uddin MM, Ibrahim MMH, Briski KP. Glycogen Phosphorylase Isoform Regulation of Ventromedial Hypothalamic Nucleus Gluco-Regulatory Neuron 5'-AMP-Activated Protein Kinase and Transmitter Marker Protein Expression. ASN Neuro 2021; 13:17590914211035020. [PMID: 34596459 PMCID: PMC8495507 DOI: 10.1177/17590914211035020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Brain glycogen is remodeled during metabolic homeostasis and provides oxidizable
L-lactate equivalents. Brain glycogen phosphorylase (GP)-brain (GPbb;
AMP-sensitive) and -muscle (GPmm; norepinephrine-sensitive) type isoforms
facilitate stimulus-specific control of glycogen disassembly. Here, a whole
animal model involving stereotactic-targeted delivery of GPmm or GPbb siRNA to
the ventromedial hypothalamic nucleus (VMN) was used to investigate the premise
that these variants impose differential control of gluco-regulatory
transmission. Intra-VMN GPmm or GPbb siRNA administration inhibited glutamate
decarboxylate65/67 (GAD), a protein marker for the
gluco-inhibitory transmitter γ--aminobutyric acid (GABA), in the caudal VMN.
GPbb knockdown, respectively overturned or exacerbated hypoglycemia-associated
GAD suppression in rostral and caudal VMN. GPmm siRNA caused a segment-specific
reversal of hypoglycemic augmentation of the gluco-stimulatory transmitter
indicator, neuronal nitric oxide synthase (nNOS). In both cell types, GP siRNA
down-regulated 5′-AMP-activated protein kinase (AMPK) during euglycemia, but
hypoglycemic suppression of AMPK was reversed by GPmm targeting. GP knockdown
elevated baseline GABA neuron phosphoAMPK (pAMKP) content, and amplified
hypoglycemic augmentation of pAMPK expression in each neuron type. GPbb
knockdown increased corticosterone secretion in eu- and hypoglycemic rats.
Outcomes validate efficacy of GP siRNA delivery for manipulation of glycogen
breakdown in discrete brain structures in vivo, and document VMN GPbb control of
local GPmm expression. Results document GPmm and/or -bb regulation of GABAergic
and nitrergic transmission in discrete rostro-caudal VMN segments. Contrary
effects of glycogenolysis on metabolic-sensory AMPK protein during eu- versus
hypoglycemia may reflect energy state-specific astrocyte signaling. Amplifying
effects of GPbb knockdown on hypoglycemic stimulation of pAMPK infer that
glycogen mobilization by GPbb limits neuronal energy instability during
hypoglycemia.
Collapse
Affiliation(s)
- Md Main Uddin
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, 15512University of Louisiana Monroe, Monroe, LA, USA
| | - Mostafa M H Ibrahim
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, 15512University of Louisiana Monroe, Monroe, LA, USA
| | - Karen P Briski
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, 15512University of Louisiana Monroe, Monroe, LA, USA
| |
Collapse
|
16
|
Bheemanapally K, Ibrahim MMH, Alshamrani A, Briski KP. Ventromedial hypothalamic nucleus glycogen regulation of metabolic-sensory neuron AMPK and neurotransmitter expression: role of lactate. Am J Physiol Regul Integr Comp Physiol 2021; 320:R791-R799. [PMID: 33825506 PMCID: PMC8285616 DOI: 10.1152/ajpregu.00292.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 03/18/2021] [Accepted: 03/31/2021] [Indexed: 12/23/2022]
Abstract
Astrocyte glycogen is dynamically remodeled during metabolic stability and provides oxidizable l-lactate equivalents during neuroglucopenia. Current research investigated the hypothesis that ventromedial hypothalamic nucleus (VMN) glycogen metabolism controls glucostimulatory nitric oxide (NO) and/or glucoinhibitory gamma-aminobutyric acid (GABA) neuron 5'-AMP-activated protein kinase (AMPK) and transmitter marker, e.g., neuronal nitric oxide synthase (nNOS), and glutamate decarboxylase65/67 (GAD) protein expression. Adult ovariectomized estradiol-implanted female rats were injected into the VMN with the glycogen phosphorylase inhibitor 1,4-dideoxy-1,4-imino-d-arabinitol (DAB) before vehicle or l-lactate infusion. Western blot analysis of laser-catapult-microdissected nitrergic and GABAergic neurons showed that DAB caused lactate-reversible upregulation of nNOS and GAD proteins. DAB suppressed or increased total AMPK content of NO and GABA neurons, respectively, by lactate-independent mechanisms, but lactate prevented drug enhancement of pAMPK expression in nitrergic neurons. Inhibition of VMN glycogen disassembly caused divergent changes in counter-regulatory hormone, e.g. corticosterone (increased) and glucagon (decreased) secretion. Outcomes show that VMN glycogen metabolism controls local glucoregulatory transmission by means of lactate signal volume. Results implicate glycogen-derived lactate deficiency as a physiological stimulus of corticosterone release. Concurrent normalization of nitrergic neuron nNOS and pAMPK protein and corticosterone secretory response to DAB by lactate infers that the hypothalamic-pituitary-adrenal axis may be activated by VMN NO-mediated signals of cellular energy imbalance.
Collapse
Affiliation(s)
- Khaggeswar Bheemanapally
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana
| | - Mostafa M H Ibrahim
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana
| | - Ayed Alshamrani
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana
| | - Karen P Briski
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana
| |
Collapse
|
17
|
Uddin MM, Briski KP. Neuroestradiol regulation of ventromedial hypothalamic nucleus 5'-AMP-activated protein kinase activity and counterregulatory hormone secretion in hypoglycemic male versus female rats. AIMS Neurosci 2021; 8:133-147. [PMID: 33490375 PMCID: PMC7815480 DOI: 10.3934/neuroscience.2021006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/10/2020] [Indexed: 02/02/2023] Open
Abstract
Hypoglycemia activates the ultra-sensitive energy gauge 5′-AMP-activated protein kinase (AMPK) in ventromedial hypothalamic nucleus (VMN) gluco-regulatory neurons. The VMN is exemplified by high levels of expression of the enzyme aromatase, which converts testosterone to estradiol. This study examined the hypothesis that neuroestradiol imposes sex-dimorphic control of VMN AMPK activity during eu- and/or hypoglycemia. VMN tissue corresponding to distinct rostro-caudal segments was obtained by micropunch dissection from testes-intact male and estradiol-replaced ovariectomized female rats that were infused intracerebroventricularly with the aromatase inhibitor letrozole (Lz) before subcutaneous insulin (INS) injection. In euglycemic rats, Lz treatment elevated (male) or decreased (female) middle VMN phosphoAMPK content, with concurrent effects on total AMPK expression. Lz prevented hypoglycemic up-regulation of the mean pAMPK/AMPK ratio in rostral and middle segments of the male VMN, and significantly inhibited this proportion throughout the VMN of hypoglycemic female rats. Lz prevented glucagon secretion in hypoglycemic rats of each sex, and abolished hypoglycemic hypercorticosteronemia in males. Results show that neuroestradiol regulation of VMN AMPK activity during euglycemia is region-specific and gender-divergent, e.g. inhibitory in males versus stimulatory in females. Intra-VMN distribution of hypoglycemia-activated AMPK varies between sexes, but in each sex, locally-generated estradiol is critical for sensor reactivity to this stimulus. Coincident Lz attenuation of VMN AMPK and counter-regulatory hormone responses to hypoglycemia infers a possible cause-and-effect association. Further effort is needed to elucidate the cellular and molecular mechanisms that underlie sex-dimorphic neuroestradiol control of VMN total AMPK and phosphoAMPK expression during distinct metabolic states.
Collapse
Affiliation(s)
- Md Main Uddin
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA 71201, USA
| | - Karen P Briski
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA 71201, USA
| |
Collapse
|
18
|
Sex-dimorphic Rostro-caudal Patterns of 5'-AMP-activated Protein Kinase Activation and Glucoregulatory Transmitter Marker Protein Expression in the Ventrolateral Ventromedial Hypothalamic Nucleus (VMNvl) in Hypoglycemic Male and Female Rats: Impact of Estradiol. J Mol Neurosci 2020; 71:1082-1094. [PMID: 33231812 DOI: 10.1007/s12031-020-01730-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/07/2020] [Indexed: 12/20/2022]
Abstract
The ventromedial hypothalamic nucleus-ventrolateral part (VMNvl) is an estradiol-sensitive structure that controls sex-specific behavior. Electrical reactivity of VMNvl neurons to hypoglycemia infers that cellular energy stability is monitored there. Current research investigated the hypothesis that estradiol elicits sex-dimorphic patterns of VMNvl metabolic sensor activation and gluco-regulatory neurotransmission during hypoglycemia. Rostral-, middle-, and caudal-VMNvl tissue was separately micropunch-dissected from letrozole (Lz)- or vehicle-injected male and estradiol- or vehicle-implanted ovariectomized (OVX) female rats for Western blot analysis of total and phosphorylated 5'-AMP-activated protein kinase (AMPK) protein expression and gluco-stimulatory [neuronal nitric oxide synthase (nNOS); steroidogenic factor-1 (SF1) or -inhibitory (glutamate decarboxylase65/67 (GAD)] transmitter marker proteins after sc insulin (INS) or vehicle injection. In both sexes, hypoglycemic up-regulation of phosphoAMPK was estradiol-dependent in rostral and middle, but not caudal VMNvl. AMPK activity remained elevated after recovery from hypoglycemia over the rostro-caudal VMNvl in female, but only in the rostral segment in male. In each sex, hypoglycemia correspondingly augmented or suppressed nNOS profiles in rostral and middle versus caudal VMNvl; these segmental responses persisted longer in female. Rostral and middle segment SF1 protein was inhibited by estradiol-independent mechanisms in hypoglycemic males, but increased by estradiol-reliant mechanisms in female. After INS injection, GAD expression was inhibited in the male rostral VMNvl without estradiol involvement, but this hormone was required for broader suppression of this profile in the female. Neuroanatomical variability of VMNvl metabolic transmitter reactivity to hypoglycemia underscores the existence of functionally different subgroups in that structure. The regional distribution and estradiol sensitivity of hypoglycemia-sensitive VMNvl neurons of each neurochemical phenotype evidently vary between sexes.
Collapse
|
19
|
El-Tarabany MS, Saleh AA, El-Araby IE, El-Magd MA. Association of LEPR polymorphisms with egg production and growth performance in female Japanese quails. Anim Biotechnol 2020; 33:599-611. [PMID: 32865111 DOI: 10.1080/10495398.2020.1812617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This study aimed to screen intron 8 of the leptin receptor (LEPR) gene for polymorphisms in female Japanese quails. Two adjacent novel SNPs (A277G and A304G) were detected using PCR-SSCP and sequencing. These SNPs produced three haplotypes (AA/AA, AG/AG, and GG/GG) that were significantly (p ≤ 0.05) associated with growth and egg production traits. GG/GG haplotype-quails had significantly (p ≤ 0.05) lower egg production, feed intake, growth performance, lipid profile, serum levels of sex hormones (estradiol, progesterone, FSH, LH), and ovarian expressions of survivin, FSHR, and IGF1 than other quails. However, GG/GG quails had significantly (p ≤ 0.05) higher serum levels of LEP and mRNA levels of LEPR, LEP, and caspase 3 in the hypothalamus and ovaries. These higher levels of LEP/LEPR could not only reduce feed intake and body weight gain but also could induce apoptosis of ovarian cells (as indicated by lower survivin and IGF1 and higher caspase3 expression) which could inhibit the development of the follicles and the release of sex hormones with a subsequent decrease in egg production in GG/GG quails. Therefore, with these results, we suggest selecting Japanese quails with AA/AA and AG/AG haplotypes to improve the reproduction and growth performance of this flock.
Collapse
Affiliation(s)
- Mahmoud S El-Tarabany
- Department of Animal Wealth Development, Animal Breeding and Production, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Ayman A Saleh
- Department of Animal Wealth Development, Veterinary Genetics & Genetic Engineering, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Iman E El-Araby
- Department of Animal Wealth Development, Veterinary Genetics & Genetic Engineering, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Mohammed A El-Magd
- Department of Anatomy & Embryology, Faculty of Veterinary Medicine, Kafrelsheikh University, Zagazig, Egypt
| |
Collapse
|
20
|
Evaluation of the Relationship between Adipose Metabolism Patterns and Secretion of Appetite-Related Endocrines on Chicken. Animals (Basel) 2020; 10:ani10081282. [PMID: 32727133 PMCID: PMC7460314 DOI: 10.3390/ani10081282] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 12/22/2022] Open
Abstract
Simple Summary The weight of an animal conforms to a certain growth pattern. Among others, feed, environment, and body composition, in addition to genetics, affect the animal’s feed consumption and body weight. Under normal circumstances, the body weight of an animal is mainly affected by feed intake, and body composition may significantly influence feed intake. Therefore, this report sets out the effects of fat accumulation on lipid metabolism and appetite, and finally introduces the effects of feeding patterns on animal feed intake. Abstract In addition to the influence of genes, the quality of poultry products is mainly controlled by the rearing environment or feed composition during rearing, and has to meet human use and economical needs. As the only source of energy for poultry, feed considerably affects the metabolic pattern of poultry and further affects the regulation of appetite-related endocrine secretion in poultry. Under normal circumstances, the accumulation of lipid in adipose reduces feed intake in poultry and increases the rate of adipose metabolism. When the adipose content in cells decreases, endocrines that promote food intake are secreted and increase nutrient concentrations in serum and cells. By regulating the balance between appetite and adipose metabolism, the poultry’s growth and posture can maintain a balanced state. In addition, increasing fiber composition in feed can effectively increase poultry welfare, body weight, lean composition and antioxidant levels in poultry. According to this, the concept that proper fiber content should be added to feed should be considered for better economic benefits, poultry welfare and meat productivity.
Collapse
|
21
|
Chen X, Yu Y, Zheng P, Jin T, He M, Zheng M, Song X, Jones A, Huang XF. Olanzapine increases AMPK-NPY orexigenic signaling by disrupting H1R-GHSR1a interaction in the hypothalamic neurons of mice. Psychoneuroendocrinology 2020; 114:104594. [PMID: 32007669 DOI: 10.1016/j.psyneuen.2020.104594] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 12/22/2019] [Accepted: 01/17/2020] [Indexed: 12/11/2022]
Abstract
Second generation antipsychotics, particularly olanzapine, induce severe obesity, which is associated with their antagonistic effect on the histamine H1 receptor (H1R). We have previously demonstrated that oral administration of olanzapine increases the concentration of neuropeptide Y (NPY) in the hypothalamus of rats, accompanied by hyperphagia and weight gain. However, it is unclear if the increased NPY after olanzapine administration is due to its direct effect on hypothalamic neurons and its H1R antagonistic property. In the present study, we showed that with an inverted U-shape dose-response curve, olanzapine increased NPY expression in the NPY-GFP hypothalamic neurons; however, this was not the case in the hypothalamic neurons of H1R knockout mice. Olanzapine inhibited the interaction of H1R and GHSR1a (ghrelin receptor) in the primary mouse hypothalamic neurons and NPY-GFP neurons examined by confocal fluorescence resonance energy transfer (FRET) technology. Furthermore, an H1R agonist, FMPH inhibited olanzapine activation of GHSR1a downstream signaling pAMPK and transcription factors of NPY (pFOXO1 and pCREB) in the hypothalamic NPY-GFP cell. However, an olanzapine analogue (E-Olan) with lower affinity to H1R presented negligible enhancement of pCREB within the nucleus of NPY neurons. These findings suggest that the H1R antagonist property of olanzapine inhibits the interaction of H1R and GHSR1a, activates GHSR1a downstream signaling pAMPK-FOXO1/pCREB and increases hypothalamic NPY: this could be one of the important molecular mechanisms of H1R antagonism of olanzapine-induced obesity in antipsychotic management of psychiatric disorders.
Collapse
Affiliation(s)
- Xiaoqi Chen
- Department of Endocrinology and Rheumatology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Illawarra Health and Medical Research Institute and Molecular Horizons, School of Medicine, University of Wollongong, NSW, 2522, Australia
| | - Yinghua Yu
- Department of Pathogen Biology and Immunology, Xuzhou Medical University and Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou, Jiangsu 221004, China.
| | - Peng Zheng
- Illawarra Health and Medical Research Institute and Molecular Horizons, School of Medicine, University of Wollongong, NSW, 2522, Australia
| | - Tiantian Jin
- Illawarra Health and Medical Research Institute and Molecular Horizons, School of Medicine, University of Wollongong, NSW, 2522, Australia
| | - Meng He
- School of Chemistry, Wuhan University of Technology, Wuhan, China
| | - Mingxuan Zheng
- Department of Pathogen Biology and Immunology, Xuzhou Medical University and Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou, Jiangsu 221004, China
| | - Xueqin Song
- School of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Alison Jones
- Illawarra Health and Medical Research Institute and Molecular Horizons, School of Medicine, University of Wollongong, NSW, 2522, Australia
| | - Xu-Feng Huang
- Illawarra Health and Medical Research Institute and Molecular Horizons, School of Medicine, University of Wollongong, NSW, 2522, Australia.
| |
Collapse
|
22
|
Hu X, Wang Y, Sheikhahmadi A, Li X, Buyse J, Lin H, Song Z. Effects of dietary energy level on appetite and central adenosine monophosphate-activated protein kinase (AMPK) in broilers. J Anim Sci 2019; 97:4488-4495. [PMID: 31586423 PMCID: PMC6827410 DOI: 10.1093/jas/skz312] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/03/2019] [Indexed: 12/23/2022] Open
Abstract
Adenosine monophosphate-activated protein kinase (AMPK) acts as a sensor of cellular energy changes and is involved in the control of food intake. A total of 216 1-d-old broilers were randomly allotted into 3 treatments with 6 replicates per treatment and 12 broilers in each cage. The dietary treatments included 1) high-energy (HE) diet (3,500 kcal/kg), 2) normal-energy (NE) diet (3,200 kcal/kg), and 3) low-energy (LE) diet (2,900 kcal/kg). The present study was conducted to investigate the effects of dietary energy level on appetite and the central AMPK signal pathway. The results showed that a HE diet increased average daily gain (ADG), whereas a LE diet had the opposite effect (P < 0.05, N = 6). The average daily feed intake (ADFI) of the chickens fed the LE diet was significantly higher than that of the control (P < 0.05, N = 6). Overall, the feed conversion rate gradually decreased with increasing dietary energy level (P < 0.05, N = 6). Moreover, the chickens fed the LE and HE diets demonstrated markedly improved urea content compared with the control group (P < 0.0001, N = 8). The triglyceride (TG) content in the LE group was obviously higher than that in the HE group but showed no change compared with the control (P = 0.0678, N = 8). The abdominal fat rate gradually increased with increased dietary energy level (P = 0.0927, N = 8). The HE group showed downregulated gene expression levels of liver kinase B1 (LKB1), neuropeptide Y (NPY), cholecystokinin (CCK), and glucocorticoid receptor (GR) in the hypothalamus compared with the control group (P < 0.05, N = 8). However, LE treatment significantly increased the mRNA level of AMP-activated protein kinase α2 (AMPKα2) compared with other groups (P = 0.0110, N = 8). In conclusion, a HE diet inhibited appetite and central AMPK signaling. In contrast, a LE diet activated central AMPK and appetite. Overall, the central AMPK signal pathway and appetite were modulated in accordance with the energy level in the diet to regulate nutritional status and maintain energy homeostasis in birds.
Collapse
Affiliation(s)
- Xiyi Hu
- Department of Animal Science, Shandong Agricultural University, Taian, Shandong, China
| | - Yufeng Wang
- Division Animal and Human Health Engineering, Department of Biosystems, KU Leuven, Kasteelpark Arenberg, Leuven, Belgium
| | - Ardashir Sheikhahmadi
- Department of Animal Science, Faculty of Agriculture, University of Kurdistan, Sanandaj, Iran
| | - Xianlei Li
- Department of Animal Science, Shandong Agricultural University, Taian, Shandong, China
| | - Johan Buyse
- Division Animal and Human Health Engineering, Department of Biosystems, KU Leuven, Kasteelpark Arenberg, Leuven, Belgium
| | - Hai Lin
- Department of Animal Science, Shandong Agricultural University, Taian, Shandong, China
| | - Zhigang Song
- Department of Animal Science, Shandong Agricultural University, Taian, Shandong, China
| |
Collapse
|
23
|
Liver kinase B1 induces browning phenotype in 3 T3-L1 adipocytes. Gene 2019; 682:33-41. [PMID: 30296566 DOI: 10.1016/j.gene.2018.10.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 09/25/2018] [Accepted: 10/04/2018] [Indexed: 12/11/2022]
|
24
|
Quines CB, Rosa SG, Velasquez D, Prado VC, Neto JS, Nogueira CW. (p-ClPhSe)2 stabilizes metabolic function in a rat model of neuroendocrine obesity induced by monosodium glutamate. Food Chem Toxicol 2018; 118:168-180. [PMID: 29738801 DOI: 10.1016/j.fct.2018.05.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 04/30/2018] [Accepted: 05/04/2018] [Indexed: 12/13/2022]
|
25
|
|
26
|
Weng Y, Lin J, Liu H, Wu H, Yan Z, Zhao J. AMPK activation by Tanshinone IIA protects neuronal cells from oxygen-glucose deprivation. Oncotarget 2017; 9:4511-4521. [PMID: 29435120 PMCID: PMC5796991 DOI: 10.18632/oncotarget.23391] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 12/01/2017] [Indexed: 12/25/2022] Open
Abstract
The current study tested the potential neuroprotective function of Tanshinone IIA (ThIIA) in neuronal cells with oxygen-glucose deprivation (ODG) and re-oxygenation (OGDR). In SH-SY5Y neuronal cells and primary murine cortical neurons, ThIIA pre-treatment attenuated OGDR-induced viability reduction and apoptosis. Further, OGDR-induced mitochondrial depolarization, reactive oxygen species production, lipid peroxidation and DNA damages in neuronal cells were significantly attenuated by ThIIA. ThIIA activated AMP-activated protein kinase (AMPK) signaling, which was essential for neuroprotection against OGDR. AMPKα1 knockdown or complete knockout in SH-SY5Y cells abolished ThIIA-induced AMPK activation and neuroprotection against OGDR. Further studies found that ThIIA up-regulated microRNA-135b to downregulate the AMPK phosphatase Ppm1e. Notably, knockdown of Ppm1e by targeted shRNA or forced microRNA-135b expression also activated AMPK and protected SH-SY5Y cells from OGDR. Together, AMPK activation by ThIIA protects neuronal cells from OGDR. microRNA-135b-mediated silence of Ppm1e could be the key mechanism of AMPK activation by ThIIA.
Collapse
Affiliation(s)
- Yingfeng Weng
- Department of Neurology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jixian Lin
- Department of Neurology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hui Liu
- Department of Neurology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hui Wu
- Department of Neurology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhimin Yan
- Department of Neurology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Zhao
- Department of Neurology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
27
|
Affiliation(s)
- Komal Saraswat
- Department of Biochemistry, University of Allahabad, Allahabad, India
| | | |
Collapse
|
28
|
Energy balance, body composition, sedentariness and appetite regulation: pathways to obesity. Clin Sci (Lond) 2017; 130:1615-28. [PMID: 27503946 DOI: 10.1042/cs20160006] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 06/03/2016] [Indexed: 12/18/2022]
Abstract
Energy balance is not a simple algebraic sum of energy expenditure and energy intake as often depicted in communications. Energy balance is a dynamic process and there exist reciprocal effects between food intake and energy expenditure. An important distinction is that of metabolic and behavioural components of energy expenditure. These components not only contribute to the energy budget directly, but also by influencing the energy intake side of the equation. It has recently been demonstrated that resting metabolic rate (RMR) is a potential driver of energy intake, and evidence is accumulating on the influence of physical activity (behavioural energy expenditure) on mechanisms of satiety and appetite control. These effects are associated with changes in leptin and insulin sensitivity, and in the plasma levels of gastrointestinal (GI) peptides such as glucagon-like peptide-1 (GLP-1), ghrelin and cholecystokinin (CCK). The influence of fat-free mass on energy expenditure and as a driver of energy intake directs attention to molecules emanating from skeletal tissue as potential appetite signals. Sedentariness (physical inactivity) is positively associated with adiposity and is proposed to be a source of overconsumption and appetite dysregulation. The molecular signals underlying these effects are not known but represent a target for research.
Collapse
|
29
|
Song Y, Oh GH, Kim MB, Hwang JK. Fucosterol inhibits adipogenesis through the activation of AMPK and Wnt/β-catenin signaling pathways. Food Sci Biotechnol 2017; 26:489-494. [PMID: 30263569 DOI: 10.1007/s10068-017-0067-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 12/31/2016] [Accepted: 01/04/2017] [Indexed: 12/20/2022] Open
Abstract
Fucosterol is a sterol constituent primarily derived from brown algae. Recently, the antiadipogenic effect of fucosterol has been reported; however, its molecular mechanism remains to be studied. Fucosterol effectively upregulated the phosphorylations of both adenosine monophosphate (AMP)-activated protein kinase (AMPK) and acetyl-CoA carboxylase (ACC), and downregulated the expression levels of lipogenesis-related factors. Moreover, fucosterol activated the major components of the Wnt/β-catenin signaling pathway, including β-catenin, disheveled 2 (DVL2), and cyclin D1 (CCND1), whereas it inactivated glycogen synthase kinase 3β (p-GSK3β) by stimulating its phosphorylation. In the presence or absence of fucosterol, the adipogenic transcriptional factors [peroxisome proliferator activated-receptor γ (PPARγ), CCAAT/enhancer binding protein α (C/EBPα), and sterol regulatory element binding protein-1c (SREBP-1c)] were upregulated by the inhibition of AMPK by compound C or the knockdown of β-catenin by siRNA. Overall, these data demonstrate that fucosterol prevents adipogenesis by mediating both AMPK- and Wnt/β-catenin-signaling pathways.
Collapse
Affiliation(s)
- Youngwoo Song
- 1Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03772 Korea
| | - Ga Hui Oh
- 2Department of Biomaterials Science and Engineering, Yonsei University, Seoul, 03772 Korea
| | - Mi-Bo Kim
- 1Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03772 Korea
| | - Jae-Kwan Hwang
- 1Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03772 Korea.,2Department of Biomaterials Science and Engineering, Yonsei University, Seoul, 03772 Korea
| |
Collapse
|
30
|
Autophagy Induced by Areca Nut Extract Contributes to Decreasing Cisplatin Toxicity in Oral Squamous Cell Carcinoma Cells: Roles of Reactive Oxygen Species/AMPK Signaling. Int J Mol Sci 2017; 18:ijms18030524. [PMID: 28257034 PMCID: PMC5372540 DOI: 10.3390/ijms18030524] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 02/14/2017] [Accepted: 02/22/2017] [Indexed: 02/07/2023] Open
Abstract
Chewing areca nut is closely associated with oral squamous cell carcinoma (OSCC). The current study aimed to investigate potential associations between areca nut extract (ANE) and cisplatin toxicity in OSCC cells. OSCC cells (Cal-27 and Scc-9) viability and apoptosis were analyzed after treatment with ANE and/or cisplatin. The expressions of proteins associated with autophagy and the AMP-activated protein kinase (AMPK) signaling network were evaluated. We revealed that advanced OSCC patients with areca nut chewing habits presented higher LC3 expression and poorer prognosis. Reactive oxygen species (ROS)-mediated autophagy was induced after pro-longed treatment of ANE (six days, 3 μg). Cisplatin toxicity (IC50, 48 h) was decreased in OSCC cells after ANE treatment (six days, 3 μg). Cisplatin toxicity could be enhanced by reversed autophagy by pretreatment of 3-methyladenine (3-MA), N-acetyl-l-cysteine (NAC), or Compound C. Cleaved-Poly-(ADP-ribose) polymerase (cl-PARP) and cleaved-caspase 3 (cl-caspase 3) were downregulated in ANE-treated OSCC cells in the presence of cisplatin, which was also reversed by NAC and Compound C. Collectively, ANE could decrease cisplatin toxicity of OSCC by inducing autophagy, which involves the ROS and AMPK/mTOR signaling pathway.
Collapse
|
31
|
Dagon Y, Mantzoros CS, Kim YB. AMPK↔Sirt1: From a signaling network to a combination drug. Metabolism 2016; 65:1692-1694. [PMID: 27558832 DOI: 10.1016/j.metabol.2016.07.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 07/25/2016] [Indexed: 01/09/2023]
Affiliation(s)
- Yossi Dagon
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
32
|
Pimentel GD, Contreras C, López M. Fatty Acids and Hypothalamic Dysfunction in Obesity. HANDBOOK OF LIPIDS IN HUMAN FUNCTION 2016:557-582. [DOI: 10.1016/b978-1-63067-036-8.00021-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
33
|
Wang D, Xu C, Wang T, Li H, Li Y, Ren J, Tian Y, Li Z, Jiao Y, Kang X, Liu X. Discovery and functional characterization of leptin and its receptors in Japanese quail (Coturnix japonica). Gen Comp Endocrinol 2016; 225:1-12. [PMID: 26342967 DOI: 10.1016/j.ygcen.2015.09.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 08/07/2015] [Accepted: 09/01/2015] [Indexed: 12/31/2022]
Abstract
Leptin is an important endocrine regulation factor of food intake and energy homeostasis in mammals; however, the existence of a poultry leptin gene (LEP) is still debated. Here, for the first time, we report the cloning of a partial exon 3 sequence of LEP (qLEP) and four different leptin receptor splicing variants, including a long receptor (qLEPRl) and three soluble receptors (qLEPR-a, qLEPR-b and qLEPR-c) in Japanese quail (Coturnix japonica). The qLEP gene had high GC content (64%), which is similar to other reported avian leptin genes. The encoded qLEP protein possessed the conserved pair of cysteine residues that are required to form a lasso knot for full biological activity, but shared relatively low identities with LEPs of other vertebrates. The translated qLEPRl protein contained 1143 amino acids and shared high amino acid sequence identity with a chicken homolog (89% identity). qLEPRl also contained all the motifs, domains, and basic tyrosine residues that are conserved in the LEPRl proteins of other vertebrates. qRT-PCR analysis showed that LEP and the four LEPR variants were expressed extensively in all tissues examined; the expression levels of LEP were relatively high in hypothalamus, skeletal muscle, and pancreas, while the expression levels of the LEPRs were highest in the pituitary. Compared with the expression levels of juvenile qLEP and total qLEPR (including all LEPR variants), the expression levels of mature qLEP and total qLEPR were up-regulated in the hypothalamus and pituitary, and down-regulated in the ovary. The expressions of LEP/LEPR increased when fasting and decreased when refeeding in the brain and peripheral tissues of juvenile quail, which suggested that the LEP/LEPR system modulated food intake and energy expenditure, although, unlike in mammals, LEP may actually act to inhibit food intake during fasting, at least in juvenile quail. The results indicate that qLEP and qLEPR have unique expression patterns and that the encoded proteins play important roles in the regulation of reproduction and energy status in Japanese quail.
Collapse
Affiliation(s)
- Dandan Wang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Chunlin Xu
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Taian Wang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Hong Li
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Yanmin Li
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Junxiao Ren
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Yadong Tian
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China; Henan Innovative Engineering Research Center of Poultry Germplasm Resource, Henan Agricultural University, Zhengzhou 450002, China; International Joint Research Laboratory for Poultry Breeding of Henan, Henan Agricultural University, Zhengzhou 450002, China
| | - Zhuanjian Li
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China; Henan Innovative Engineering Research Center of Poultry Germplasm Resource, Henan Agricultural University, Zhengzhou 450002, China; International Joint Research Laboratory for Poultry Breeding of Henan, Henan Agricultural University, Zhengzhou 450002, China
| | - Yuping Jiao
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China; Institute of Animal Husbandry and Veterinary Medicine, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Xiangtao Kang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China; Henan Innovative Engineering Research Center of Poultry Germplasm Resource, Henan Agricultural University, Zhengzhou 450002, China; International Joint Research Laboratory for Poultry Breeding of Henan, Henan Agricultural University, Zhengzhou 450002, China.
| | - Xiaojun Liu
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China; Henan Innovative Engineering Research Center of Poultry Germplasm Resource, Henan Agricultural University, Zhengzhou 450002, China; International Joint Research Laboratory for Poultry Breeding of Henan, Henan Agricultural University, Zhengzhou 450002, China.
| |
Collapse
|
34
|
Ezeoke CC, Morley JE. Pathophysiology of anorexia in the cancer cachexia syndrome. J Cachexia Sarcopenia Muscle 2015; 6:287-302. [PMID: 26675762 PMCID: PMC4670736 DOI: 10.1002/jcsm.12059] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 06/11/2015] [Accepted: 06/22/2015] [Indexed: 12/20/2022] Open
Abstract
Anorexia is commonly present in persons with cancer and a major component of cancer cachexia. There are multiple causes of anorexia in cancer. Peripherally, these can be due to (i) substances released from or by the tumour, e.g. pro-inflammatory cytokines, lactate, and parathormone-related peptide; (ii) tumours causing dysphagia or altering gut function; (iii) tumours altering nutrients, e.g. zinc deficiency; (iv) tumours causing hypoxia; (v) increased peripheral tryptophan leading to increased central serotonin; or (vi) alterations of release of peripheral hormones that alter feeding, e.g. peptide tyrosine tyrosine and ghrelin. Central effects include depression and pain, decreasing the desire to eat. Within the central nervous system, tumours create multiple alterations in neurotransmitters, neuropeptides, and prostaglandins that modulate feeding. Many of these neurotransmitters appear to produce their anorectic effects through the adenosine monophosphate kinase/methylmalonyl coenzyme A/fatty acid system in the hypothalamus. Dynamin is a guanosine triphosphatase that is responsible for internalization of melanocortin 4 receptors and prostaglandin receptors. Dynamin is up-regulated in a mouse model of cancer anorexia. A number of drugs, e.g. megestrol acetate, cannabinoids, and ghrelin agonists, have been shown to have some ability to be orexigenic in cancer patients.
Collapse
Affiliation(s)
- Chukwuemeka Charles Ezeoke
- United States Navy Medical Corps and PGY-2, Internal Medicine Residency, Saint Louis University HospitalSt. Louis, MO, USA
| | - John E Morley
- Division of Geriatrics, Saint Louis University School of Medicine1402 S. Grand Blvd., M238, St. Louis, MO, 63104, USA
- Division of Endocrinology, Saint Louis University School of MedicineSt. Louis, MO, USA
| |
Collapse
|
35
|
Weisová P, Pfeiffer S, Prehn JHM. AMP‐Activated Protein Kinase (AMPK) as a Cellular Energy Sensor and Therapeutic Target for Neuroprotection. THE FUNCTIONS, DISEASE‐RELATED DYSFUNCTIONS, AND THERAPEUTIC TARGETING OF NEURONAL MITOCHONDRIA 2015:130-145. [DOI: 10.1002/9781119017127.ch5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
36
|
Moreto F, Kano HT, Torezan GA, de Oliveira EP, Manda RM, Teixeira O, Michelin E, Correa CR, Burini RC. Changes in malondialdehyde and C-reactive protein concentrations after lifestyle modification are related to different metabolic syndrome-associated pathophysiological processes. Diabetes Metab Syndr 2015; 9:218-222. [PMID: 25956753 DOI: 10.1016/j.dsx.2015.04.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
AIMS Metabolic syndrome (MetS) is often accompanied by pro-oxidative and pro-inflammatory processes. Lifestyle modification (LiSM) may act as primary treatment for these processes. This study aimed to elucidate influencing factors on changes of malondialdehyde (MDA) and C-reactive protein (CRP) concentrations after a LiSM intervention. METHODS Sixty subjects (53 yrs, 84% women) clinically approved to attend a 20 weeks LiSM-program were submitted to weekly nutritional counseling and physical activities combining aerobic (3 times/week) and resistance (2 times/week) exercises. Before and after intervention they were assessed for anthropometric, clinical, cardiorespiratory fitness test (CRF) and laboratory markers. Statistical analyses performed were multiple regression analysis and backward stepwise with p<0.05 and R(2) as influence index. RESULTS LiSM was responsible for elevations in CRF, healthy eating index (HEI), total plasma antioxidant capacity (TAP) and HDL-C along with reductions in waist circumference measures and MetS (47-40%) prevalence. MDA and CRP did not change after LiSM, however, we observed that MDA concentrations were positively influenced (R(2)=0.35) by fasting blood glucose (β=0.64) and HOMA-IR (β=0.58) whereas CRP concentrations were by plasma gamma-glutamyltransferase activity (β=0.54; R(2)=0.29). CONCLUSIONS Pro-oxidant and pro-inflammatory states of MetS can be attenuated after lifestyle modification if glucose metabolism homeostasis were recovered and if liver inflammation were reduced, respectively.
Collapse
Affiliation(s)
- Fernando Moreto
- Sao Paulo State University, Botucatu School of Medicine, Botucatu, Brazil.
| | - Hugo T Kano
- Sao Paulo State University, Botucatu School of Medicine, Botucatu, Brazil.
| | - Gabriel A Torezan
- Sao Paulo State University, Botucatu School of Medicine, Botucatu, Brazil.
| | | | - Rodrigo M Manda
- Sao Paulo State University, Botucatu School of Medicine, Botucatu, Brazil.
| | - Okesley Teixeira
- Sao Paulo State University, Botucatu School of Medicine, Botucatu, Brazil.
| | - Edilaine Michelin
- Sao Paulo State University, Botucatu School of Medicine, Botucatu, Brazil.
| | - Camila R Correa
- Sao Paulo State University, Botucatu School of Medicine, Botucatu, Brazil.
| | - Roberto C Burini
- Sao Paulo State University, Botucatu School of Medicine, Botucatu, Brazil.
| |
Collapse
|
37
|
Abstract
AbstractEnergy restriction (ER; also known as caloric restriction) is the only nutritional intervention that has repeatedly been shown to increase lifespan in model organisms and may delay ageing in humans. In the present review we discuss current scientific literature on ER and its molecular, metabolic and hormonal effects. Moreover, criteria for the classification of substances that might induce positive ER-like changes without having to reduce energy intake are summarised. Additionally, the putative ER mimetics (ERM) 2-deoxy-d-glucose, metformin, rapamycin, resveratrol, spermidine and lipoic acid and their suggested molecular targets are discussed. While there are reports on these ERM candidates that describe lifespan extension in model organisms, data on longevity-inducing effects in higher organisms such as mice remain controversial or are missing. Furthermore, some of these candidates produce detrimental side effects such as immunosuppression or lactic acidosis, or have not been tested for safety in long-term studies. Up to now, there are no known ERM that could be recommended without limitations for use in humans.
Collapse
|
38
|
Mendes MCS, Pimentel GD, Costa FO, Carvalheira JBC. Molecular and neuroendocrine mechanisms of cancer cachexia. J Endocrinol 2015; 226:R29-R43. [PMID: 26112046 DOI: 10.1530/joe-15-0170] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/22/2015] [Indexed: 02/05/2023]
Abstract
Cancer and its morbidities, such as cancer cachexia, constitute a major public health problem. Although cancer cachexia has afflicted humanity for centuries, its underlying multifactorial and complex physiopathology has hindered the understanding of its mechanism. During the last few decades we have witnessed a dramatic increase in the understanding of cancer cachexia pathophysiology. Anorexia and muscle and adipose tissue wasting are the main features of cancer cachexia. These apparently independent symptoms have humoral factors secreted by the tumor as a common cause. Importantly, the hypothalamus has emerged as an organ that senses the peripheral signals emanating from the tumoral environment, and not only elicits anorexia but also contributes to the development of muscle and adipose tissue loss. Herein, we review the roles of factors secreted by the tumor and its effects on the hypothalamus, muscle and adipose tissue, as well as highlighting the key targets that are being exploited for cancer cachexia treatment.
Collapse
Affiliation(s)
- Maria Carolina S Mendes
- Department of Internal MedicineFaculty of Medical Sciences, State University of Campinas (UNICAMP), MA: 13083-970 Campinas, Sao Paulo, Brazil
| | - Gustavo D Pimentel
- Department of Internal MedicineFaculty of Medical Sciences, State University of Campinas (UNICAMP), MA: 13083-970 Campinas, Sao Paulo, Brazil
| | - Felipe O Costa
- Department of Internal MedicineFaculty of Medical Sciences, State University of Campinas (UNICAMP), MA: 13083-970 Campinas, Sao Paulo, Brazil
| | - José B C Carvalheira
- Department of Internal MedicineFaculty of Medical Sciences, State University of Campinas (UNICAMP), MA: 13083-970 Campinas, Sao Paulo, Brazil
| |
Collapse
|
39
|
Dagon Y, Mantzoros C, Kim YB. Exercising insulin sensitivity: AMPK turns on autophagy! Metabolism 2015; 64:655-7. [PMID: 25819737 PMCID: PMC4709017 DOI: 10.1016/j.metabol.2015.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 03/07/2015] [Indexed: 01/18/2023]
Affiliation(s)
- Yossi Dagon
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Christos Mantzoros
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Young-Bum Kim
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
40
|
Martínez de Morentin PB, Lage R, González-García I, Ruíz-Pino F, Martins L, Fernández-Mallo D, Gallego R, Fernø J, Señarís R, Saha AK, Tovar S, Diéguez C, Nogueiras R, Tena-Sempere M, López M. Pregnancy induces resistance to the anorectic effect of hypothalamic malonyl-CoA and the thermogenic effect of hypothalamic AMPK inhibition in female rats. Endocrinology 2015; 156:947-60. [PMID: 25535827 PMCID: PMC4330316 DOI: 10.1210/en.2014-1611] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 12/19/2014] [Indexed: 12/20/2022]
Abstract
During gestation, hyperphagia is necessary to cope with the metabolic demands of embryonic development. There were three main aims of this study: Firstly, to investigate the effect of pregnancy on hypothalamic fatty acid metabolism, a key pathway for the regulation of energy balance; secondly, to study whether pregnancy induces resistance to the anorectic effect of fatty acid synthase (FAS) inhibition and accumulation of malonyl-coenzyme A (CoA) in the hypothalamus; and, thirdly, to study whether changes in hypothalamic AMPK signaling are associated with brown adipose tissue (BAT) thermogenesis during pregnancy. Our data suggest that in pregnant rats, the hypothalamic fatty acid pathway shows an overall state that should lead to anorexia and elevated BAT thermogenesis: decreased activities of AMP-activated protein kinase (AMPK), FAS, and carnitine palmitoyltransferase 1, coupled with increased acetyl-CoA carboxylase function with subsequent elevation of malonyl-CoA levels. This profile seems dependent of estradiol levels but not prolactin or progesterone. Despite the apparent anorexic and thermogenic signaling in the hypothalamus, pregnant rats remain hyperphagic and display reduced temperature and BAT function. Actually, pregnant rats develop resistance to the anorectic effects of central FAS inhibition, which is associated with a reduction of proopiomelanocortin (POMC) expression and its transcription factors phospho-signal transducer and activator of transcription 3, and phospho-forkhead box O1. This evidence demonstrates that pregnancy induces a state of resistance to the anorectic and thermogenic actions of hypothalamic cellular signals of energy surplus, which, in parallel to the already known refractoriness to leptin effects, likely contributes to gestational hyperphagia and adiposity.
Collapse
Affiliation(s)
- Pablo B Martínez de Morentin
- Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS) (P.B.M.d.M., R.L., I.G.-G., L.M., D.F.M., R.S., S.T., C.D., R.N., M.L.), University of Santiago de Compostela (USC)-Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela 15782, Spain; Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de la Obesidad y Nutrición (CIBERobn) (P.B.M.d.M., R.L., I.G.-G., F.R.-P., L.M., D.F.M., S.T., C.D., R.N., M.T.-S., M.L.), Santiago de Compostela 15706, Spain; Department of Cell Biology, Physiology and Immunology (F.R.-P., M.T.-S.), University of Córdoba, Córdoba 14004, Spain; Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC)/Hospital Universitario Reina Sofía (F.R.-P., M.T.-S.), Córdoba 14004, Spain; Department of Morphological Sciences (R.G.), School of Medicine, University of Santiago de Compostela, Santiago de Compostela 15782, Spain; Department of Clinical Science (J.F.), K. G. Jebsen Center for Diabetes Research, University of Bergen, Bergen, N-5021, Norway; and Diabetes Research Unit, EBRC-827 (A.K.S.), Boston Medical Center, Boston, Massachusetts 02118
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Rapamycin and everolimus facilitate hepatitis E virus replication: revealing a basal defense mechanism of PI3K-PKB-mTOR pathway. J Hepatol 2014; 61:746-54. [PMID: 24859454 DOI: 10.1016/j.jhep.2014.05.026] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Revised: 05/08/2014] [Accepted: 05/09/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Humans are frequently exposed to hepatitis E virus (HEV). Nevertheless, the disease mainly affects pregnant women and immunocompromised individuals. Organ recipients receiving immunosuppressants, such as rapalogs, to prevent rejection have a high risk for developing chronic hepatitis following HEV infection. Rapalogs constitute potent inhibitors of mTOR including rapamycin and everolimus. As a master kinase, the mechanism-of-action of mTOR is not only associated with the immunosuppressive capacity of rapalogs but is also tightly regulated during pregnancy because of increased nutritional demands. METHODS We thus investigated the role of mTOR in HEV infection by using two state-of-the-art cell culture models: a subgenomic HEV containing luciferase reporter and a full-length HEV infectious cell culture system. RESULTS In both subgenomic and full-length HEV models, HEV infection was aggressively escalated by treatment of rapamycin or everolimus. Inhibition of mTOR was confirmed by Western blot showing the inhibition of its downstream target, S6 phosphorylation. Consistently, stable silencing of mTOR by lentiviral RNAi resulted in a significant increase in intracellular HEV RNA, suggesting an antiviral function of mTOR in HEV infection. By targeting a series of other up- and downstream elements of mTOR signaling, we further revealed an effective basal defense mechanism of the PI3K-PKB-mTOR pathway against HEV, which is through the phosphorylated eIF4E-binding protein 1 (4E-BP1), however independent of autophagy formation. CONCLUSIONS The discovery that PI3K-PKB-mTOR pathway limits HEV infection through 4E-BP1 and acts as a gate-keeper in human HEV target cells bears significant implications in managing immunosuppression in HEV-infected organ transplantation recipients.
Collapse
|
42
|
Lindholm-Perry AK, Kuehn LA, Oliver WT, Kern RJ, Cushman RA, Miles JR, McNeel AK, Freetly HC. DNA polymorphisms and transcript abundance ofPRKAG2and phosphorylated AMP-activated protein kinase in the rumen are associated with gain and feed intake in beef steers. Anim Genet 2014; 45:461-72. [DOI: 10.1111/age.12151] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2014] [Indexed: 02/02/2023]
Affiliation(s)
- A. K. Lindholm-Perry
- USDA; ARS; U.S. Meat Animal Research Center; PO Box 166 Clay Center NE 68933 USA
| | - L. A. Kuehn
- USDA; ARS; U.S. Meat Animal Research Center; PO Box 166 Clay Center NE 68933 USA
| | - W. T. Oliver
- USDA; ARS; U.S. Meat Animal Research Center; PO Box 166 Clay Center NE 68933 USA
| | - R. J. Kern
- USDA; ARS; U.S. Meat Animal Research Center; PO Box 166 Clay Center NE 68933 USA
- University of Wyoming; Department of Animal Science; 1000 E University Ave Laramie WY 82071 USA
| | - R. A. Cushman
- USDA; ARS; U.S. Meat Animal Research Center; PO Box 166 Clay Center NE 68933 USA
| | - J. R. Miles
- USDA; ARS; U.S. Meat Animal Research Center; PO Box 166 Clay Center NE 68933 USA
| | - A. K. McNeel
- USDA; ARS; U.S. Meat Animal Research Center; PO Box 166 Clay Center NE 68933 USA
| | - H. C. Freetly
- USDA; ARS; U.S. Meat Animal Research Center; PO Box 166 Clay Center NE 68933 USA
| |
Collapse
|
43
|
Thiamine deficiency induces anorexia by inhibiting hypothalamic AMPK. Neuroscience 2014; 267:102-13. [PMID: 24607345 DOI: 10.1016/j.neuroscience.2014.02.033] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 02/08/2014] [Accepted: 02/21/2014] [Indexed: 01/26/2023]
Abstract
Obesity and eating disorders are prevailing health concerns worldwide. It is important to understand the regulation of food intake and energy metabolism. Thiamine (vitamin B1) is an essential nutrient. Thiamine deficiency (TD) can cause a number of disorders in humans, such as Beriberi and Wernicke-Korsakoff syndrome. We demonstrated here that TD caused anorexia in C57BL/6 mice. After feeding a TD diet for 16days, the mice displayed a significant decrease in food intake and an increase in resting energy expenditure (REE), which resulted in a severe weight loss. At the 22nd day, the food intake was reduced by 69% and 74% for male and female mice, respectively in TD group. The REE increased by ninefolds in TD group. The loss of body weight (17-24%) was similar between male and female animals and mainly resulted from the reduction of fat mass (49% decrease). Re-supplementation of thiamine (benfotiamine) restored animal's appetite, leading to a total recovery of body weight. The hypothalamic adenosine monophosphate-activated protein kinase (AMPK) is a critical regulator of food intake. TD inhibited the phosphorylation of AMPK in the arcuate nucleus (ARN) and paraventricular nucleus (PVN) of the hypothalamus without affecting its expression. TD-induced inhibition of AMPK phosphorylation was reversed once thiamine was re-supplemented. In contrast, TD increased AMPK phosphorylation in the skeletal muscle and upregulated the uncoupling protein (UCP)-1 in brown adipose tissues which was consistent with increased basal energy expenditure. Re-administration of thiamine stabilized AMPK phosphorylation in the skeletal muscle as well as energy expenditure. Taken together, TD may induce anorexia by inhibiting hypothalamic AMPK activity. With a simultaneous increase in energy expenditure, TD caused an overall body weight loss. The results suggest that the status of thiamine levels in the body may affect food intake and body weight.
Collapse
|
44
|
Olofsson B. The olfactory neuron AWC promotes avoidance of normally palatable food following chronic dietary restriction. ACTA ACUST UNITED AC 2014; 217:1790-8. [PMID: 24577446 PMCID: PMC4020945 DOI: 10.1242/jeb.099929] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Changes in metabolic state alter foraging behavior and food preference in animals. Here, I show that normally attractive food becomes repulsive to Caenorhabditis elegans if animals are chronically undernourished as a result of alimentary tract defects. This behavioral plasticity is achieved in two ways: increased food leaving and induction of aversive behavior towards food. A particularly strong food avoider is defective in the chitin synthase that makes the pharyngeal lining. Food avoidance induced by underfeeding is mediated by cGMP signaling in the olfactory neurons AWC and AWB, and the gustatory neurons ASJ and ASK. Food avoidance is enhanced by increased population density and is reduced if the animals are unable to correctly interpret their nutritional state as a result of defects in the AMP kinase or TOR/S6kinase pathways. The TGF-β/DBL-1 pathway suppresses food avoidance and the cellular basis for this is distinct from its role in aversive olfactory learning of harmful food. This study suggests that nutritional state feedback via nutrient sensors, population size and olfactory neurons guides food preference in C. elegans.
Collapse
Affiliation(s)
- Birgitta Olofsson
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
| |
Collapse
|
45
|
Genetic deletion of Rheb1 in the brain reduces food intake and causes hypoglycemia with altered peripheral metabolism. Int J Mol Sci 2014; 15:1499-510. [PMID: 24451134 PMCID: PMC3907882 DOI: 10.3390/ijms15011499] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Revised: 12/12/2013] [Accepted: 01/07/2014] [Indexed: 02/05/2023] Open
Abstract
Excessive food/energy intake is linked to obesity and metabolic disorders, such as diabetes. The hypothalamus in the brain plays a critical role in the control of food intake and peripheral metabolism. The signaling pathways in hypothalamic neurons that regulate food intake and peripheral metabolism need to be better understood for developing pharmacological interventions to manage eating behavior and obesity. Mammalian target of rapamycin (mTOR), a serine/threonine kinase, is a master regulator of cellular metabolism in different cell types. Pharmacological manipulations of mTOR complex 1 (mTORC1) activity in hypothalamic neurons alter food intake and body weight. Our previous study identified Rheb1 (Ras homolog enriched in brain 1) as an essential activator of mTORC1 activity in the brain. Here we examine whether central Rheb1 regulates food intake and peripheral metabolism through mTORC1 signaling. We find that genetic deletion of Rheb1 in the brain causes a reduction in mTORC1 activity and impairs normal food intake. As a result, Rheb1 knockout mice exhibit hypoglycemia and increased lipid mobilization in adipose tissue and ketogenesis in the liver. Our work highlights the importance of central Rheb1 signaling in euglycemia and energy homeostasis in animals.
Collapse
|
46
|
Miller JL, Linville TD, Dykens EM. Effects of metformin in children and adolescents with Prader-Willi syndrome and early-onset morbid obesity: a pilot study. J Pediatr Endocrinol Metab 2014; 27:23-9. [PMID: 23893676 PMCID: PMC3864175 DOI: 10.1515/jpem-2013-0116] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 06/03/2013] [Indexed: 12/14/2022]
Abstract
Prader-Willi syndrome (PWS) is one of the most commonly recognized causes of early-onset childhood obesity. Individuals with PWS have significant hyperphagia and decreased recognition of satiety. The exact etiology of the hyperphagia remains unknown and, therefore, untreatable. We conducted a pilot, open-label study of response to metformin in 21 children with PWS and six with early morbid obesity (EMO). Participants had significant insulin resistance and glucose intolerance on oral glucose tolerance testing (OGTT) and were started on metformin for these biochemical findings. We administered the Hyperphagia Questionnaire to parents of patients before and after starting metformin treatment. Both the PWS and EMO groups showed significant improvements in food-related distress, anxiety, and ability to be redirected away from food on the Hyperphagia Questionnaire. In the PWS group, improvements were predominantly seen in females. Within the PWS group, responders to metformin had higher 2-h glucose levels on OGTT (7.48 mmol/L vs. 4.235 mmol/L; p=0.003) and higher fasting insulin levels (116 pmol/L vs. 53.5 pmol/L; p=0.04). Additionally, parents of 5/13 individuals with PWS and 5/6 with EMO reported that their child was able to feel full while on metformin (for many this was the first time they had ever described a feeling of fullness). Metformin may improve sense of satiety and decrease anxiety about food in some individuals with PWS and EMO. Positive response to metformin may depend on the degree of hyperinsulinism and glucose intolerance. Nonetheless, the results of this pilot study bear further investigation.
Collapse
|
47
|
Romo-Vaquero M, Larrosa M, Yáñez-Gascón MJ, Issaly N, Flanagan J, Roller M, Tomás-Barberán FA, Espín JC, García-Conesa MT. A rosemary extract enriched in carnosic acid improves circulating adipocytokines and modulates key metabolic sensors in lean Zucker rats: Critical and contrasting differences in the obese genotype. Mol Nutr Food Res 2013; 58:942-53. [DOI: 10.1002/mnfr.201300524] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 10/02/2013] [Accepted: 10/03/2013] [Indexed: 12/12/2022]
Affiliation(s)
- María Romo-Vaquero
- Research Group on Quality; Safety, and Bioactivity of Plant Foods; Department of Food Science and Technology; CEBAS-CSIC; Murcia Spain
| | - Mar Larrosa
- Research Group on Quality; Safety, and Bioactivity of Plant Foods; Department of Food Science and Technology; CEBAS-CSIC; Murcia Spain
| | - María J. Yáñez-Gascón
- Research Group on Quality; Safety, and Bioactivity of Plant Foods; Department of Food Science and Technology; CEBAS-CSIC; Murcia Spain
| | | | | | | | - Francisco A. Tomás-Barberán
- Research Group on Quality; Safety, and Bioactivity of Plant Foods; Department of Food Science and Technology; CEBAS-CSIC; Murcia Spain
| | - Juan C. Espín
- Research Group on Quality; Safety, and Bioactivity of Plant Foods; Department of Food Science and Technology; CEBAS-CSIC; Murcia Spain
| | - María-Teresa García-Conesa
- Research Group on Quality; Safety, and Bioactivity of Plant Foods; Department of Food Science and Technology; CEBAS-CSIC; Murcia Spain
| |
Collapse
|
48
|
Implication of transcriptional repression in compound C-induced apoptosis in cancer cells. Cell Death Dis 2013; 4:e883. [PMID: 24157877 PMCID: PMC3920957 DOI: 10.1038/cddis.2013.419] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 09/14/2013] [Accepted: 09/17/2013] [Indexed: 12/25/2022]
Abstract
Compound C, a well-known inhibitor of AMP-activated protein kinase (AMPK), has been reported to induce apoptosis in some types of cells. However, the underlying mechanisms remain largely unclear. Using a DNA microarray analysis, we found that the expression of many genes was downregulated upon treatment with compound C. Importantly, compound C caused transcriptional repression with the induction of p53, a well-known marker of transcriptional stress response, in several cancer cell lines. Compound C did not induce the phosphorylation of p53 but dramatically increased the protein level of p53 similar to some other transcriptional inhibitors, including 5,6-dichloro-1-β-D-ribobenzimidazole (DRB). Consistent with previous reports, we found that compound C initiated apoptotic death of cancer cells in an AMPK-independent manner. Similar to DRB and actinomycin D (ActD), two classic transcription inhibitors, compound C not only resulted in the loss of Bcl-2 and Bcl-xl protein but also induced the phosphorylation of eukaryotic initiation factor-alpha (eIF2α) on Ser51. Hence, the phosphorylation of eIF2α might be a novel marker of transcriptional inhibition. It is noteworthy that compound C-mediated apoptosis of cancer cells is correlated with decreased expression of Bcl-2 and Bcl-xl and the phosphorylation of eIF2α on Ser51. Remarkably, compound C exhibits potent anticancer activities in vivo. Taken together, our data suggest that compound C may be an attractive candidate for anticancer drug development.
Collapse
|
49
|
McClung CA. How might circadian rhythms control mood? Let me count the ways.. Biol Psychiatry 2013; 74:242-9. [PMID: 23558300 PMCID: PMC3725187 DOI: 10.1016/j.biopsych.2013.02.019] [Citation(s) in RCA: 342] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 02/06/2013] [Accepted: 02/25/2013] [Indexed: 12/28/2022]
Abstract
Mood disorders are serious diseases that affect a large portion of the population. There have been many hypotheses put forth over the years to explain the development of major depression, bipolar disorder, and other mood disorders. These hypotheses include disruptions in monoamine transmission, hypothalamus-pituitary-adrenal axis function, immune function, neurogenesis, mitochondrial dysfunction, and neuropeptide signaling (to name a few). Nearly all people suffering from mood disorders have significant disruptions in circadian rhythms and the sleep/wake cycle. In fact, altered sleep patterns are one of the major diagnostic criteria for these disorders. Moreover, environmental disruptions to circadian rhythms, including shift work, travel across time zones, and irregular social schedules, tend to precipitate or exacerbate mood-related episodes. Recent studies have found that molecular clocks are found throughout the brain and body where they participate in the regulation of most physiological processes, including those thought to be involved in mood regulation. This review will summarize recent data that implicate the circadian system as a vital regulator of a variety of systems that are thought to play a role in the development of mood disorders.
Collapse
Affiliation(s)
- Colleen A McClung
- Department of Psychiatry and Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
| |
Collapse
|
50
|
Vitzel KF, Bikopoulos G, Hung S, Curi R, Ceddia RB. Loss of the anorexic response to systemic 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside administration despite reducing hypothalamic AMP-activated protein kinase phosphorylation in insulin-deficient rats. PLoS One 2013; 8:e71944. [PMID: 23967267 PMCID: PMC3743807 DOI: 10.1371/journal.pone.0071944] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 07/09/2013] [Indexed: 02/07/2023] Open
Abstract
This study tested whether chronic systemic administration of 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside (AICAR) could attenuate hyperphagia, reduce lean and fat mass losses, and improve whole-body energy homeostasis in insulin-deficient rats. Male Wistar rats were first rendered diabetic through streptozotocin (STZ) administration and then intraperitoneally injected with AICAR for 7 consecutive days. Food and water intake, ambulatory activity, and energy expenditure were assessed at the end of the AICAR-treatment period. Blood was collected for circulating leptin measurement and the hypothalami were extracted for the determination of suppressor of cytokine signaling 3 (SOCS3) content, as well as the content and phosphorylation of AMP-kinase (AMPK), acetyl-CoA carboxylase (ACC), and the signal transducer and activator of transcription 3 (STAT3). Rats were thoroughly dissected for adiposity and lean body mass (LBM) determinations. In non-diabetic rats, despite reducing adiposity, AICAR increased (∼1.7-fold) circulating leptin and reduced hypothalamic SOCS3 content and food intake by 67% and 25%, respectively. The anorexic effect of AICAR was lost in diabetic rats, even though hypothalamic AMPK and ACC phosphorylation markedly decreased in these animals. Importantly, hypothalamic SOCS3 and STAT3 levels remained elevated and reduced, respectively, after treatment of insulin-deficient rats with AICAR. Diabetic rats were lethargic and displayed marked losses of fat and LBM. AICAR treatment increased ambulatory activity and whole-body energy expenditure while also attenuating diabetes-induced fat and LBM losses. In conclusion, AICAR did not reverse hyperphagia, but it promoted anti-catabolic effects on skeletal muscle and fat, enhanced spontaneous physical activity, and improved the ability of rats to cope with the diabetes-induced dysfunctional alterations in glucose metabolism and whole-body energy homeostasis.
Collapse
Affiliation(s)
- Kaio F. Vitzel
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, SP, Brazil
| | - George Bikopoulos
- School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Steven Hung
- School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Rui Curi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, SP, Brazil
| | - Rolando B. Ceddia
- School of Kinesiology and Health Science, York University, Toronto, ON, Canada
- * E-mail:
| |
Collapse
|