1
|
Possa‐Paranhos IC, Butts J, Pyszka E, Nelson C, Congdon S, Cho D, Sweeney P. Medial hypothalamic MC3R signalling regulates energy rheostasis in adult mice. J Physiol 2025; 603:379-410. [PMID: 39718394 PMCID: PMC11737543 DOI: 10.1113/jp286699] [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: 04/08/2024] [Accepted: 11/11/2024] [Indexed: 12/25/2024] Open
Abstract
Although mammals resist both acute weight loss and weight gain, the neural circuitry mediating bi-directional defense against weight change is incompletely understood. Global constitutive deletion of the melanocortin-3-receptor (MC3R) impairs the behavioural response to both anorexic and orexigenic stimuli, with MC3R knockout mice demonstrating increased weight gain following anabolic challenges and increased weight loss following anorexic challenges (i.e. impaired energy rheostasis). However, the brain regions mediating this phenotype are not well understood. Here, we utilized MC3R floxed mice and viral injections of Cre-recombinase to selectively delete MC3R from the medial hypothalamus (MH) in adult mice. Behavioural assays were performed on these animals to test the role of MC3R in MH in the acute response to orexigenic and anorexic challenges. Complementary chemogenetic approaches were used in MC3R-Cre mice to localize and characterize the specific medial hypothalamic brain regions mediating the role of MC3R in energy homeostasis. Finally, we performed RNAscope in situ hybridization to map changes in the mRNA expression of MC3R, pro-opiomelanocortin and agouti-related peptide following energy rheostatic challenges, as well as to characterize the MC3R expressing cells in dorsal MH. Our results demonstrate that MC3R deletion in MH increases feeding and weight gain following high-fat diet feeding, and enhances the anorexic effects of semaglutide, in a sexually dimorphic manner. Furthermore, although the arcuate nucleus exerts an important role in MC3R-mediated effects on energy homeostasis, viral deletion in the dorsal MH also resulted in altered energy rheostasis, indicating that brain regions outside of the arcuate nucleus also contribute to the role of MC3R in energy rheostasis. Together, these results demonstrate that MC3R-mediated effects on energy rheostasis result from the loss of MC3R signalling in medial hypothalamic neurons and suggest an important role for dorsal-MH MC3R signalling in energy rheostasis. KEY POINTS: Melanocortin-3-receptor (MC3R) signalling regulates energy rheostasis in adult mice. Medial hypothalamus regulates energy rheostasis in adult mice. Energy rheostatic stimuli alter mRNA levels of agouti-related peptide, pro-opiomelanocortin and MC3R. Dorsal-medial hypothalamus (DMH) MC3R neurons increase locomotion and energy expenditure. MC3R cell types in DMH are sexually dimorphic.
Collapse
Affiliation(s)
| | - Jared Butts
- Department of Molecular and Integrative PhysiologyUniversity of Illinois Urbana‐ChampaignUrbanaILUSA
- University of Illinois Urbana‐Champaign Neuroscience ProgramUrbanaILUSA
| | - Emma Pyszka
- Department of Molecular and Integrative PhysiologyUniversity of Illinois Urbana‐ChampaignUrbanaILUSA
| | - Christina Nelson
- Department of Molecular and Integrative PhysiologyUniversity of Illinois Urbana‐ChampaignUrbanaILUSA
| | - Samuel Congdon
- Department of Molecular and Integrative PhysiologyUniversity of Illinois Urbana‐ChampaignUrbanaILUSA
| | - Dajin Cho
- Department of Molecular and Integrative PhysiologyUniversity of Illinois Urbana‐ChampaignUrbanaILUSA
- University of Illinois Urbana‐Champaign Neuroscience ProgramUrbanaILUSA
| | - Patrick Sweeney
- Department of Molecular and Integrative PhysiologyUniversity of Illinois Urbana‐ChampaignUrbanaILUSA
- University of Illinois Urbana‐Champaign Neuroscience ProgramUrbanaILUSA
| |
Collapse
|
2
|
Punger EM, Norris SLW, Stevens SC, Santos KH, Christy AC. Investigating the Effect of Enterally Administered Capromorelin on Body Weight in Mice (Mus musculus). Comp Med 2024; 74:327-335. [PMID: 39025662 PMCID: PMC11524401 DOI: 10.30802/aalas-cm-24-031] [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] [Received: 04/03/2024] [Revised: 04/24/2024] [Accepted: 06/13/2024] [Indexed: 07/20/2024]
Abstract
Significant weight loss in mice (Mus musculus) is a welfare concern and can alter physiology and behavior in ways that may confound research aims. In this study, factorial design was used to investigate the effect of enterally administered capromorelin on changes in mouse body weight overall and with various research-related interventions, such as administration of analgesics, anesthesia, or surgery. BALB/c mice (n = 61 [27 males/34 females] for analysis) were randomized into 8 intervention-treatment groups with 2 treatment allocations: capromorelin (10 mg/kg) or control, and 4 intervention allocations: no intervention; buprenorphine extended-release (XR) alone; buprenorphine XR, meloxicam, and anesthesia; or surgery under anesthesia with buprenorphine XR, meloxicam, and bupivacaine administered. Mice were habituated to handling, weighing, and voluntary consumption of condensed milk, which was used as the control solution and later a vehicle for capromorelin delivery, for 5 d (days 0 to 4). Then, mice received their interventions followed by 3 days of daily treatment or control administration (days 7 to 9). Body weights were measured daily (days 8 to 11 and day 14) to compare with baseline weights (days 0 to 4 and day 7) and evaluate for treatment and intervention effects on body weight. The interventions resulted in a decrease in group body weights 3 and 4 d after the interventions were conducted. Overall, body weights increased more in mice given capromorelin compared with control, and mice treated with capromorelin returned to, or exceeded, baseline weights faster. The weight loss was mitigated by capromorelin administration in all interventions except for the buprenorphine XR-only group. It is recommended to clinically consider enterally administered capromorelin to mitigate research-induced weight loss in mice.
Collapse
Affiliation(s)
- Elizabeth M Punger
- Veterinary Services Branch, Veterinary Medicine Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland; and
| | - Sarah L W Norris
- Research Support Branch, Regulated Research Administration Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland
| | - Stephen C Stevens
- Veterinary Services Branch, Veterinary Medicine Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland; and
| | - Kacee H Santos
- Veterinary Services Branch, Veterinary Medicine Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland; and
| | - Amanda C Christy
- Veterinary Services Branch, Veterinary Medicine Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland; and
| |
Collapse
|
3
|
Yılmaz D, Singh A, Wehrle E, Kuhn GA, Mathavan N, Müller R. Unveiling frailty: comprehensive and sex-specific characterization in prematurely aging PolgA mice. FRONTIERS IN AGING 2024; 5:1365716. [PMID: 39372332 PMCID: PMC11449839 DOI: 10.3389/fragi.2024.1365716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 09/09/2024] [Indexed: 10/08/2024]
Abstract
Frailty, a geriatric syndrome, is assessed using the frailty phenotype (FP) and frailty index (FI). While these approaches have been applied to aging mice, their effectiveness in prematurely aging mouse models such as PolgAD257A/D257A (PolgA) has not been completely explored. We demonstrated that frailty became evident in PolgA mice around 40 weeks, validated through body weight loss, reduced walking speed, decreased physical activity, and weaker grip strength. Moreover, we also identified sex differences in these mice with females exhibiting slightly more physical decline compared to males. Frailty prevalence in PolgA mice at 40 weeks parallels that observed in naturally aging mice at 27 months and aging humans at 65-70 years. These findings contribute to understanding frailty onset and sex-specific patterns in this prematurely aging mouse model, emphasizing the significance of the PolgA mouse model in investigating aging and related disorders.
Collapse
Affiliation(s)
- Dilara Yılmaz
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Amit Singh
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Esther Wehrle
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
- AO Research Institute Davos, Davos, Davos Platz, Switzerland
| | - Gisela A. Kuhn
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | | | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
4
|
Possa-Paranhos IC, Butts J, Pyszka E, Nelson C, Cho D, Sweeney P. Neuroanatomical dissection of the MC3R circuitry regulating energy rheostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.22.590573. [PMID: 38712101 PMCID: PMC11071362 DOI: 10.1101/2024.04.22.590573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Although mammals resist both acute weight loss and weight gain, the neural circuitry mediating bi-directional defense against weight change is incompletely understood. Global constitutive deletion of the melanocortin-3-receptor (MC3R) impairs the behavioral response to both anorexic and orexigenic stimuli, with MC3R knockout mice demonstrating increased weight gain following anabolic challenges and increased weight loss following anorexic challenges (i.e. impaired energy rheostasis). However, the brain regions mediating this phenotype remain incompletely understood. Here, we utilized MC3R floxed mice and viral injections of Cre-recombinase to selectively delete MC3R from medial hypothalamus (MH) in adult mice. Behavioral assays were performed on these animals to test the role of MC3R in MH in the acute response to orexigenic and anorexic challenges. Complementary chemogenetic approaches were used in MC3R-Cre mice to localize and characterize the specific medial hypothalamic brain regions mediating the role of MC3R in energy homeostasis. Finally, we performed RNAscope in situ hybridization to map changes in the mRNA expression of MC3R, POMC, and AgRP following energy rheostatic challenges. Our results demonstrate that MC3R deletion in MH increased feeding and weight gain following acute high fat diet feeding in males, and enhanced the anorexic effects of semaglutide, in a sexually dimorphic manner. Additionally, activation of DMH MC3R neurons increased energy expenditure and locomotion. Together, these results demonstrate that MC3R mediated effects on energy rheostasis result from the loss of MC3R signaling in the medial hypothalamus of adult animals and suggest an important role for DMH MC3R signaling in energy rheostasis.
Collapse
Affiliation(s)
| | - Jared Butts
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology
- University of Illinois Urbana-Champaign Neuroscience Program
| | - Emma Pyszka
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology
| | - Christina Nelson
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology
| | - Dajin Cho
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology
- University of Illinois Urbana-Champaign Neuroscience Program
| | - Patrick Sweeney
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology
- University of Illinois Urbana-Champaign Neuroscience Program
| |
Collapse
|
5
|
Krolenko EV, Kupriyanova OV, Nigmatullina LS, Grigoryeva TV, Roumiantsev SA, Shestopalov AV. Changes of the Concentration of Short-Chain Fatty Acids in the Intestines of Mice with Different Types of Obesity. Bull Exp Biol Med 2024; 176:347-353. [PMID: 38342814 DOI: 10.1007/s10517-024-06022-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Indexed: 02/13/2024]
Abstract
We studied the production of short-chain fatty acids (SCFA) by the intestinal microbiota in mice with obesity caused by a diet and a genetic defect in the leptin receptor gene. In mice, intestinal contents were examined and SCFA were quantitatively assayed by gas chromatography. SCFA concentration in the intestinal contents of mice with alimentary obesity model was significantly lower in the first phase of the experiment (day 14), and the change in their production in dynamics was fundamentally different from this process in the control group (standard diet). The dynamics of the concentration of these metabolites in the model of genetic obesity was similar to that in the control, but the production of SCFA was significantly reduced in mice with leptin resistance in the middle phase (day 60) of the experiment. These findings indicate that the production of SCFA is more influenced by the diet than by leptin resistance.
Collapse
Affiliation(s)
- E V Krolenko
- Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - O V Kupriyanova
- Institute of Fundamental Medicine and Biology, Kazan (Volga region) Federal University, Kazan, Republic of Tatarstan, Russia
| | - L S Nigmatullina
- Institute of Fundamental Medicine and Biology, Kazan (Volga region) Federal University, Kazan, Republic of Tatarstan, Russia
| | - T V Grigoryeva
- Institute of Fundamental Medicine and Biology, Kazan (Volga region) Federal University, Kazan, Republic of Tatarstan, Russia
| | - S A Roumiantsev
- Center of Digital and Translational Biomedicine, LLC "Center for Molecular Health", Moscow, Russia
- Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
- National Medical Research Center of Endocrinology, State Research Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - A V Shestopalov
- Center of Digital and Translational Biomedicine, LLC "Center for Molecular Health", Moscow, Russia
- Oncology, and Immunology, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Ministry of Health of the Russian Federation, Moscow, Russia
- Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
- National Medical Research Center of Endocrinology, State Research Center, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
6
|
Flier JS. Moderating "the great debate": The carbohydrate-insulin vs. the energy balance models of obesity. Cell Metab 2023; 35:737-741. [PMID: 37086719 DOI: 10.1016/j.cmet.2023.03.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/17/2023] [Accepted: 03/30/2023] [Indexed: 04/24/2023]
Abstract
The increased prevalence of obesity in recent decades is a topic of great scientific and medical interest, but despite many advances, the causes of this increase have not been adequately identified. In this context, two conflicting models for obesity-the carbohydrate-insulin model (CIM) and the energy balance model (EBM)-are being vigorously debated by distinct cohorts of experts in the field. The goal of this perspective is to assess this "conflict of models" from a neutral perspective. I conclude that although both models have produced useful insights, they differ fundamentally in what they seek to explain, and neither has yet provided a validated mechanistic account for the rising obesity prevalence in some but not all members of the population. Rather than engaging in such debates over competing models, the field should be more focused on establishing specific mechanistic insights in identified patient groups and, eventually, actionable interventions based on them.
Collapse
Affiliation(s)
- Jeffrey S Flier
- Department of Neurobiology and Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Pauli JR, da Silva ASR, de Freitas EC. An obesogenic diet mitigates brown adipose tissue-induced thermogenesis, shifting the metabolism towards triacylglycerol synthesis and favouring obesity development. J Physiol 2023; 601:1039-1040. [PMID: 36745510 DOI: 10.1113/jp284341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- José Rodrigo Pauli
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Adelino Sanchez Ramos da Silva
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil.,Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Ellen Cristini de Freitas
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil.,Department of Health Sciences, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
8
|
Gallop MR, Tobin SY, Chaix A. Finding balance: understanding the energetics of time-restricted feeding in mice. Obesity (Silver Spring) 2023; 31 Suppl 1:22-39. [PMID: 36513496 PMCID: PMC9877167 DOI: 10.1002/oby.23607] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/17/2022] [Accepted: 09/06/2022] [Indexed: 12/15/2022]
Abstract
Over the course of mammalian evolution, the ability to store energy likely conferred a survival advantage when food became scarce. A long-term increase in energy storage results from an imbalance between energy intake and energy expenditure, two tightly regulated parameters that generally balance out to maintain a fairly stable body weight. Understanding the molecular determinants of this feat likely holds the key to new therapeutic development to manage obesity and associated metabolic dysfunctions. Time-restricted feeding (TRF), a dietary intervention that limits feeding to the active phase, can prevent and treat obesity and metabolic dysfunction in rodents fed a high-fat diet, likely by exerting effects on energetic balance. Even when body weight is lower in mice on active-phase TRF, food intake is generally isocaloric as compared with ad libitum fed controls. This discrepancy between body weight and energy intake led to the hypothesis that energy expenditure is increased during TRF. However, at present, there is no consensus in the literature as to how TRF affects energy expenditure and energy balance as a whole, and the mechanisms behind metabolic adaptation under TRF are unknown. This review examines our current understanding of energy balance on TRF in rodents and provides a framework for future studies to evaluate the energetics of TRF and its molecular determinants.
Collapse
Affiliation(s)
- Molly R Gallop
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT
| | - Selene Y Tobin
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT
| | - Amandine Chaix
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT
| |
Collapse
|
9
|
Da Eira D, Jani S, Ceddia RB. An obesogenic diet impairs uncoupled substrate oxidation and promotes whitening of the brown adipose tissue in rats. J Physiol 2023; 601:69-82. [PMID: 36419345 DOI: 10.1113/jp283721] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/09/2022] [Indexed: 11/26/2022] Open
Abstract
Brown adipose tissue (BAT) is rich in mitochondria containing uncoupling protein 1 (UCP1), and dissipates energy through thermogenesis. However, even though BAT mass and its UCP1 content increase in rodents chronically fed a high-fat sucrose-enriched (HFS) diet, marked expansion of adiposity still occurs in these animals, suggesting insufficient BAT-mediated HFS diet-induced thermogenesis. Thus, the objective of this study was to investigate the metabolic and molecular mechanisms that regulate BAT thermogenesis in HFS-induced obesity. To accomplish this, rats were fed either a standard chow or HFS diet for 8 weeks. Subsequently, glucose and fatty acid metabolism and the molecular mechanisms underlying these processes were assessed in freshly isolated primary BAT adipocytes. Despite increasing BAT mass and its UCP1 content, the HFS diet reduced uncoupled glucose and palmitate oxidation in BAT adipocytes. It also markedly diminished tyrosine hydroxylase content and lipolysis in these cells. Conversely, glucose uptake, lactate production, glycerol incorporation into lipids, palmitate incorporation into triacylglycerol (TAG), phosphoenolpyruvate carboxykinase and glycerol kinase levels, and lipoprotein lipase and cluster of differentiation 36 gene expression were increased. In summary, a HFS diet enhanced glyceroneogenesis and shifted BAT metabolism toward TAG synthesis by impairing UCP1-mediated substrate oxidation and by enhancing fatty acid esterification in intact brown adipocytes. These adaptive metabolic responses to chronic HFS feeding attenuated BAT thermogenic capacity and favoured the development of obesity. KEY POINTS: Despite increasing brown adipose tissue (BAT) mass and levels of thermogenic proteins such as peroxisome proliferator-activated receptor γ coactivator 1α, carnitine palmitoyltransferase 1B and uncoupling protein 1 (UCP1), an obesogenic high-fat sucrose-enriched (HFS) diet attenuated uncoupled glucose and fatty acid oxidation in brown adipocytes. Brown adipocytes diverted glycerol and fatty acids toward triacylglycerol (TAG) synthesis by elevating the cellular machinery that promotes fatty acid uptake along with phosphoenolpyruvate carboxykinase and glycerol kinase levels. The HFS diet increased glucose uptake that supported lactate production and provided substrate for glyceroneogenesis and TAG synthesis in brown adipocytes. Impaired UCP-1-mediated thermogenic capacity and enhanced TAG storage in BAT adipocytes were consistent with reduced adipose triglyceride lipase and tyrosine hydroxylase levels in HFS diet-fed animals.
Collapse
Affiliation(s)
- Daniel Da Eira
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - Shailee Jani
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - Rolando B Ceddia
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| |
Collapse
|
10
|
Yanucil C, Kentrup D, Li X, Grabner A, Schramm K, Martinez EC, Li J, Campos I, Czaya B, Heitman K, Westbrook D, Wende AR, Sloan A, Roche JM, Fornoni A, Kapiloff MS, Faul C. FGF21-FGFR4 signaling in cardiac myocytes promotes concentric cardiac hypertrophy in mouse models of diabetes. Sci Rep 2022; 12:7326. [PMID: 35513431 PMCID: PMC9072546 DOI: 10.1038/s41598-022-11033-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 04/18/2022] [Indexed: 12/13/2022] Open
Abstract
Fibroblast growth factor (FGF) 21, a hormone that increases insulin sensitivity, has shown promise as a therapeutic agent to improve metabolic dysregulation. Here we report that FGF21 directly targets cardiac myocytes by binding β-klotho and FGF receptor (FGFR) 4. In combination with high glucose, FGF21 induces cardiac myocyte growth in width mediated by extracellular signal-regulated kinase 1/2 (ERK1/2) signaling. While short-term FGF21 elevation can be cardio-protective, we find that in type 2 diabetes (T2D) in mice, where serum FGF21 levels are elevated, FGFR4 activation induces concentric cardiac hypertrophy. As T2D patients are at risk for heart failure with preserved ejection fraction (HFpEF), we propose that induction of concentric hypertrophy by elevated FGF21-FGFR4 signaling may constitute a novel mechanism promoting T2D-associated HFpEF such that FGFR4 blockade might serve as a cardio-protective therapy in T2D. In addition, potential adverse cardiac effects of FGF21 mimetics currently in clinical trials should be investigated.
Collapse
Affiliation(s)
- Christopher Yanucil
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Tinsley Harrison Tower 611L, 1720 2nd Avenue South, Birmingham, AL, 35294, USA
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Dominik Kentrup
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Tinsley Harrison Tower 611L, 1720 2nd Avenue South, Birmingham, AL, 35294, USA
- Division of Nephrology and Hypertension, Center for Translational Metabolism and Health, Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, IL, USA
| | - Xueyi Li
- Departments of Ophthalmology and Medicine, Stanford Cardiovascular Institute, Stanford University, 1651 Page Mill Road, Mail Code 5356, Palo Alto, CA, USA
| | - Alexander Grabner
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Karla Schramm
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Eliana C Martinez
- Department of Pediatrics and Interdisciplinary Stem Cell Institute, Leonard M. Miller School of Medicine, University of Miami, FL, Miami, USA
| | - Jinliang Li
- Departments of Ophthalmology and Medicine, Stanford Cardiovascular Institute, Stanford University, 1651 Page Mill Road, Mail Code 5356, Palo Alto, CA, USA
- Department of Pediatrics and Interdisciplinary Stem Cell Institute, Leonard M. Miller School of Medicine, University of Miami, FL, Miami, USA
| | - Isaac Campos
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Tinsley Harrison Tower 611L, 1720 2nd Avenue South, Birmingham, AL, 35294, USA
| | - Brian Czaya
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Tinsley Harrison Tower 611L, 1720 2nd Avenue South, Birmingham, AL, 35294, USA
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Kylie Heitman
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Tinsley Harrison Tower 611L, 1720 2nd Avenue South, Birmingham, AL, 35294, USA
| | - David Westbrook
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Tinsley Harrison Tower 611L, 1720 2nd Avenue South, Birmingham, AL, 35294, USA
| | - Adam R Wende
- Division of Molecular & Cellular Pathology, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Alexis Sloan
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Johanna M Roche
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Alessia Fornoni
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Michael S Kapiloff
- Departments of Ophthalmology and Medicine, Stanford Cardiovascular Institute, Stanford University, 1651 Page Mill Road, Mail Code 5356, Palo Alto, CA, USA.
- Department of Pediatrics and Interdisciplinary Stem Cell Institute, Leonard M. Miller School of Medicine, University of Miami, FL, Miami, USA.
| | - Christian Faul
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Tinsley Harrison Tower 611L, 1720 2nd Avenue South, Birmingham, AL, 35294, USA.
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA.
| |
Collapse
|
11
|
Weldenegodguad M, Pokharel K, Niiranen L, Soppela P, Ammosov I, Honkatukia M, Lindeberg H, Peippo J, Reilas T, Mazzullo N, Mäkelä KA, Nyman T, Tervahauta A, Herzig KH, Stammler F, Kantanen J. Adipose gene expression profiles reveal insights into the adaptation of northern Eurasian semi-domestic reindeer (Rangifer tarandus). Commun Biol 2021; 4:1170. [PMID: 34620965 PMCID: PMC8497613 DOI: 10.1038/s42003-021-02703-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 09/21/2021] [Indexed: 02/08/2023] Open
Abstract
Reindeer (Rangifer tarandus) are semi-domesticated animals adapted to the challenging conditions of northern Eurasia. Adipose tissues play a crucial role in northern animals by altering gene expression in their tissues to regulate energy homoeostasis and thermogenic activity. Here, we perform transcriptome profiling by RNA sequencing of adipose tissues from three different anatomical depots: metacarpal (bone marrow), perirenal, and prescapular fat in Finnish and Even reindeer (in Sakha) during spring and winter. A total of 16,212 genes are expressed in our data. Gene expression profiles in metacarpal tissue are distinct from perirenal and prescapular adipose tissues. Notably, metacarpal adipose tissue appears to have a significant role in the regulation of the energy metabolism of reindeer in spring when their nutritional condition is poor after winter. During spring, genes associated with the immune system are upregulated in the perirenal and prescapular adipose tissue. Blood and tissue parameters reflecting general physiological and metabolic status show less seasonal variation in Even reindeer than in Finnish reindeer. This study identifies candidate genes potentially involved in immune response, fat deposition, and energy metabolism and provides new information on the mechanisms by which reindeer adapt to harsh arctic conditions.
Collapse
Affiliation(s)
- Melak Weldenegodguad
- grid.22642.300000 0004 4668 6757Natural Resources Institute Finland (Luke), Jokioinen, Finland ,grid.9668.10000 0001 0726 2490Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| | - Kisun Pokharel
- grid.22642.300000 0004 4668 6757Natural Resources Institute Finland (Luke), Jokioinen, Finland
| | - Laura Niiranen
- grid.10858.340000 0001 0941 4873Research Unit of Biomedicine, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Päivi Soppela
- grid.37430.330000 0001 0744 995XArctic Centre, University of Lapland, Rovaniemi, Finland
| | - Innokentyi Ammosov
- grid.495192.2Laboratory of Reindeer Husbandry and Traditional Industries, Yakut Scientific Research Institute of Agriculture, Yakutsk, The Sakha Republic (Yakutia) Russia
| | | | - Heli Lindeberg
- grid.22642.300000 0004 4668 6757Natural Resources Institute Finland (Luke), Maaninka, Finland
| | - Jaana Peippo
- grid.22642.300000 0004 4668 6757Natural Resources Institute Finland (Luke), Jokioinen, Finland ,NordGen—Nordic Genetic Resource Center, Ås, Norway
| | - Tiina Reilas
- grid.22642.300000 0004 4668 6757Natural Resources Institute Finland (Luke), Jokioinen, Finland
| | - Nuccio Mazzullo
- grid.37430.330000 0001 0744 995XArctic Centre, University of Lapland, Rovaniemi, Finland
| | - Kari A. Mäkelä
- grid.10858.340000 0001 0941 4873Research Unit of Biomedicine, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Tommi Nyman
- grid.454322.60000 0004 4910 9859Department of Ecosystems in the Barents Region, Norwegian Institute of Bioeconomy Research, Svanvik, Norway
| | - Arja Tervahauta
- grid.9668.10000 0001 0726 2490Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| | - Karl-Heinz Herzig
- grid.10858.340000 0001 0941 4873Research Unit of Biomedicine, Faculty of Medicine, University of Oulu, Oulu, Finland ,grid.10858.340000 0001 0941 4873Medical Research Center, Faculty of Medicine, University of Oulu, Oulu, Finland ,grid.412326.00000 0004 4685 4917Oulu University Hospital, Oulu, Finland ,grid.22254.330000 0001 2205 0971Institute of Pediatrics, Poznań University of Medical Sciences, Poznań, Poland
| | - Florian Stammler
- grid.37430.330000 0001 0744 995XArctic Centre, University of Lapland, Rovaniemi, Finland
| | - Juha Kantanen
- grid.22642.300000 0004 4668 6757Natural Resources Institute Finland (Luke), Jokioinen, Finland
| |
Collapse
|
12
|
Dapueto R, Rodriguez-Duarte J, Galliussi G, Kamaid A, Bresque M, Batthyány C, López GV, Escande C. A novel nitroalkene vitamin E analogue inhibits the NLRP3 inflammasome and protects against inflammation and glucose intolerance triggered by obesity. Redox Biol 2021; 39:101833. [PMID: 33352465 PMCID: PMC7750735 DOI: 10.1016/j.redox.2020.101833] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 12/15/2022] Open
Abstract
Chronic metabolic diseases, like obesity, type II diabetes and atherosclerosis often involve a low-grade and sterile systemic inflammatory state, in which activation of the pro-inflammatory transcription factor NF-kB and the NLRP3 inflammasome play a major role. It is well established that genetic inhibition of the NLRP3 inflammasome ameliorates acute and chronic inflammation. Indeed, accumulating experimental evidences in murine models and also in humans suggest that inhibition of the NLRP3 inflammasome might be a suitable approach to tackle the deleterious effects of chronic metabolic diseases. In this work, we explored our previously synthesized nitroalkene-Trolox™ derivative named NATx0, as a non-conventional anti-inflammatory strategy to treat chronic inflammatory diseases, such as obesity-induced glucose intolerance. We found that NATx0 inhibited NF-kB nuclear translocation and pro-inflammatory gene expression in macrophages in vitro. In addition, treatment with NATx0 prevented NLRP3 inflammasome activation after LPS/ATP stimulation in macrophages in vitro. When tested acutely in vivo, NATx0 inhibited neutrophil recruitment in zebrafish larvae, and also diminished IL-1β production after LPS challenge in mice. Finally, when NATx0 was administered chronically to diet-induced obese mice, it decreased muscle tissue inflammation and glucose intolerance, leading to improved glucose homeostasis. In conclusion, we propose that this novel nitroalkene-Trolox derivative is a suitable tool to tackle acute and chronic inflammation in vitro and in vivo mainly due to inhibition of NF-kB/NLRP3 activation.
Collapse
Affiliation(s)
- Rosina Dapueto
- Laboratory of Metabolic Diseases and Aging, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay; Laboratory of Vascular Biology and Drug Development, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Jorge Rodriguez-Duarte
- Laboratory of Vascular Biology and Drug Development, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Germán Galliussi
- Laboratory of Vascular Biology and Drug Development, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Andrés Kamaid
- Laboratory of Vascular Biology and Drug Development, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Mariana Bresque
- Laboratory of Metabolic Diseases and Aging, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Carlos Batthyány
- Laboratory of Vascular Biology and Drug Development, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay.
| | - Gloria V López
- Laboratory of Vascular Biology and Drug Development, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay; Departamento de Química Orgánica, Facultad de Química, Universidad de la República, Uruguay.
| | - Carlos Escande
- Laboratory of Metabolic Diseases and Aging, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay.
| |
Collapse
|
13
|
Ortín-Piqueras V, Freitag TL, Andersson LC, Lehtonen SH, Meri SK, Spillmann T, Frias R. Urinary Excretion of Iohexol as a Permeability Marker in a Mouse Model of Intestinal Inflammation: Time Course, Performance and Welfare Considerations. Animals (Basel) 2021; 11:ani11010079. [PMID: 33406796 PMCID: PMC7824797 DOI: 10.3390/ani11010079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/24/2020] [Accepted: 12/26/2020] [Indexed: 11/22/2022] Open
Abstract
Simple Summary In mammals, different diseases are associated with intestinal changes that may cause an increase in gut permeability. Intestinal permeability tests allow the evaluation of intestinal damage in humans, veterinary patients and laboratory animal models. When used in mouse models, these tests require that animals are singly housed in metabolic cages with a wire-grid floor to collect urine samples. This raises welfare concerns. Iohexol meets several criteria for an ideal intestinal permeability marker and has recently been used in several species. Here, we examined the performance of an intestinal permeability test using iohexol administered by mouth and following excretion over 24 h in urine. As a model, we chose immunodeficient mice with intestinal inflammation induced by adoptive transfer of effector/memory T cells. We collected urine samples at seven time points to profile the urinary excretion of iohexol, in addition to intestinal tissue samples for histological assessment. We conclude that a 6 h cumulative urine sample may be sufficient to evaluate small intestinal permeability in this mouse model and increased urinary excretion of iohexol is correlated with increased severity of duodenitis. The welfare of mice housed in metabolic cages could be improved by reducing the cage periods from 24 to 6 h. Abstract Intestinal permeability (IP) tests are used to assess intestinal damage in patients and research models. The probe iohexol has shown advantages compared to 51Cr-EDTA or absorbable/nonabsorbable sugars. During IP tests, animals are housed in metabolic cages (MCs) to collect urine. We examined the performance of an iohexol IP test in mice. Rag1-/- (C57BL/6) mice of both sexes were divided into controls or treatment groups, the latter receiving injections of effector/memory T cells to induce intestinal inflammation. After two, four and five weeks (W), a single dose of iohexol was orally administered. Urine was collected seven times over 24 h in MCs. Iohexol concentration was measured by ELISA. Intestinal histological damage was scored in duodenal sections. In control and treated mice of both sexes, urinary excretion of iohexol peaked at 4 h. From W2 to W4/W5, urinary iohexol excretion increased in treated mice of both sexes, consistent with development of duodenitis in this model. Positive correlations were observed between the urinary excretion of iohexol in W4/W5 and the histological severity of duodenitis in treated male mice. We conclude that a 6 h cumulative urine sample appears sufficient to evaluate small IP to iohexol in this mouse model, improving animal welfare by reducing cage periods.
Collapse
Affiliation(s)
- Victoria Ortín-Piqueras
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, FIN-00014 Helsinki, Finland;
- Comparative Medicine, Karolinska Institute, SE-171 77 Stockholm, Sweden;
- Correspondence:
| | - Tobias L Freitag
- Translational Immunology Research Program, University of Helsinki, FIN-00014 Helsinki, Finland; (T.L.F.); (S.K.M.)
| | - Leif C Andersson
- Department of Pathology, University of Helsinki, FIN-00014 Helsinki, Finland; (L.C.A.); (S.H.L.)
| | - Sanna H Lehtonen
- Department of Pathology, University of Helsinki, FIN-00014 Helsinki, Finland; (L.C.A.); (S.H.L.)
- Research Programme for Clinical and Molecular Medicine, University of Helsinki, FIN-00014 Helsinki, Finland
| | - Seppo K Meri
- Translational Immunology Research Program, University of Helsinki, FIN-00014 Helsinki, Finland; (T.L.F.); (S.K.M.)
| | - Thomas Spillmann
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, FIN-00014 Helsinki, Finland;
| | - Rafael Frias
- Comparative Medicine, Karolinska Institute, SE-171 77 Stockholm, Sweden;
| |
Collapse
|
14
|
Wang G. Testing the leanocentric locking-point theory by in silico partial lipectomy. QUANTITATIVE BIOLOGY 2021. [DOI: 10.15302/j-qb-021-0233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
15
|
McCarthy M, Brown T, Alarcon A, Williams C, Wu X, Abbott RD, Gimble J, Frazier T. Fat-On-A-Chip Models for Research and Discovery in Obesity and Its Metabolic Comorbidities. TISSUE ENGINEERING. PART B, REVIEWS 2020; 26:586-595. [PMID: 32216545 PMCID: PMC8196547 DOI: 10.1089/ten.teb.2019.0261] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 03/12/2020] [Indexed: 12/11/2022]
Abstract
The obesity epidemic and its associated comorbidities present a looming challenge to health care delivery throughout the world. Obesity is characterized as a sterile inflammatory process within adipose tissues leading to dysregulated secretion of bioactive adipokines such as adiponectin and leptin, as well as systemic metabolic dysfunction. The majority of current obesity research has focused primarily on preclinical animal models in vivo and two-dimensional cell culture models in vitro. Neither of these generalized approaches is optimal due to interspecies variability, insufficient accuracy with respect to predicting human outcomes, and failure to recapitulate the three-dimensional (3D) microenvironment. Consequently, there is a growing demand and need for more sophisticated microphysiological systems to reproduce more physiologically accurate human white and brown/beige adipose depots. To address this research need, human and murine cell lines and primary cultures are being combined with bioscaffolds to create functional 3D environments that are suitable for metabolically active adipose organoids in both static and perfusion bioreactor cultures. The development of these technologies will have considerable impact on the future pace of discovery for novel small molecules and biologics designed to prevent and treat metabolic syndrome and obesity in humans. Furthermore, when these adipose tissue models are integrated with other organ systems they will have applicability to obesity-related disorders such as diabetes, nonalcoholic fatty liver disease, and osteoarthritis. Impact statement The current review article summarizes the advances made within the organ-onchip field, as it pertains to adipose tissue models of obesity and obesity-related syndromes, such as diabetes, non-alcoholic fatty liver disease, and osteoarthritis. As humanized 3D adipose-derived constructs become more accessible to the research community, it is anticipated that they will accelerate and enhance the drug discovery pipeline for obesity, diabetes, and metabolic diseases by reducing the preclinical evaluation process and improving predictive accuracy. Such developments, applications, and usages of existing technologies can change the paradigm of personalized medicine and create substantial progress in our approach to modern medicine.
Collapse
Affiliation(s)
| | - Theodore Brown
- Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Andrea Alarcon
- LaCell LLC, New Orleans, Louisiana, USA
- Obatala Sciences, Inc., New Orleans, Louisiana, USA
| | | | - Xiying Wu
- LaCell LLC, New Orleans, Louisiana, USA
- Obatala Sciences, Inc., New Orleans, Louisiana, USA
| | - Rosalyn D. Abbott
- Materials Science and Engineering Department, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| | - Jeffrey Gimble
- LaCell LLC, New Orleans, Louisiana, USA
- Obatala Sciences, Inc., New Orleans, Louisiana, USA
| | - Trivia Frazier
- LaCell LLC, New Orleans, Louisiana, USA
- Obatala Sciences, Inc., New Orleans, Louisiana, USA
| |
Collapse
|
16
|
Wang G. Body Mass Dynamics Is Determined by the Metabolic Ohm's Law and Adipocyte-Autonomous Fat Mass Homeostasis. iScience 2020; 23:101176. [PMID: 32480131 PMCID: PMC7262567 DOI: 10.1016/j.isci.2020.101176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 04/05/2020] [Accepted: 05/14/2020] [Indexed: 11/06/2022] Open
Abstract
An ODE model integrating metabolic mechanisms with clinical data reveals an Ohm's law governing lifetime body mass dynamics, where fat and lean tissues are analogous to a parallel nonlinear capacitor and resistor, respectively. The law unexpectedly decouples weight stability (a cell-autonomous property of adipocytes) and weight change (a parabolic trajectory governed by Ohm's law). In middle age, insulin resistance causes fat accumulation to avoid excessive body shrinkage in old age. Moderate middle-age spread is thus natural, not an anomaly caused by hypothalamic defects, as proposed by lipostatic theory. These discoveries provide valuable insights into health care practices such as weight control and health assessment, explain certain observed phenomena, make testable predictions, and may help to resolve major conundrums in the field. The ODE model, which is more comprehensive than Ohm's law, is useful to study metabolism at the detailed microscopic levels.
Collapse
Affiliation(s)
- Guanyu Wang
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China; Guangdong Provincial Key Laboratory of Computational Science and Material Design, Shenzhen, Guangdong 518055, China; Guangdong Provincial Key Laboratory of Cell Microenviroment and Disease Research, Shenzhen, Guangdong 518055, China; Shenzhen Key Laboratory of Cell Microenviroment, Shenzhen, Guangdong 518055, China.
| |
Collapse
|
17
|
Chao AM, Wadden TA, Berkowitz RI, Blackburn G, Bolin P, Clark JM, Coday M, Curtis JM, Delahanty LM, Dutton GR, Evans M, Ewing LJ, Foreyt JP, Gay LJ, Gregg EW, Hazuda HP, Hill JO, Horton ES, Houston DK, Jakicic JM, Jeffery RW, Johnson KC, Kahn SE, Knowler WC, Kure A, Michalski KL, Montez MG, Neiberg RH, Patricio J, Peters A, Pi-Sunyer X, Pownall H, Reboussin D, Redmon B, Rejeski WJ, Steinburg H, Walker M, Williamson DA, Wing RR, Wyatt H, Yanovski SZ, Zhang P. Weight Change 2 Years After Termination of the Intensive Lifestyle Intervention in the Look AHEAD Study. Obesity (Silver Spring) 2020; 28:893-901. [PMID: 32320144 PMCID: PMC7437140 DOI: 10.1002/oby.22769] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 02/03/2020] [Indexed: 01/07/2023]
Abstract
OBJECTIVE This study evaluated weight changes after cessation of the 10-year intensive lifestyle intervention (ILI) in the Look AHEAD (Action for Health in Diabetes) study. It was hypothesized that ILI participants would be more likely to gain weight during the 2-year observational period following termination of weight-loss-maintenance counseling than would participants in the diabetes support and education (DSE) control group. METHODS Look AHEAD was a randomized controlled trial that compared the effects of ILI and DSE on cardiovascular morbidity and mortality in participants with overweight/obesity and type 2 diabetes. Look AHEAD was converted to an observational study in September 2012. RESULTS Two years after the end of the intervention (EOI), ILI and DSE participants lost a mean (SE) of 1.2 (0.2) kg and 1.8 (0.2) kg, respectively (P = 0.003). In addition, 31% of ILI and 23.9% of DSE participants gained ≥ 2% (P < 0.001) of EOI weight, whereas 36.3% and 45.9% of the respective groups lost ≥ 2% of EOI weight (P = 0.001). Two years after the EOI, ILI participants reported greater use of weight-control behaviors than DSE participants. CONCLUSIONS Both groups lost weight during the 2-year follow-up period, but more ILI than DSE participants gained ≥ 2% of EOI weight. Further understanding is needed of factors that affected long-term weight change in both groups.
Collapse
Affiliation(s)
- Ariana M Chao
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Thomas A Wadden
- Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Robert I Berkowitz
- Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - George Blackburn
- Division of Nutrition, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Paula Bolin
- Southwestern American Indian Center, National Institute of Diabetes and Digestive and Kidney Diseases and St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Jeanne M Clark
- Division of General Internal Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Mace Coday
- Departments of Preventive Medicine and Psychiatry, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Jeffrey M Curtis
- Southwestern American Indian Center, National Institute of Diabetes and Digestive and Kidney Diseases and St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Linda M Delahanty
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Gareth R Dutton
- Division of Preventive Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Mary Evans
- Division of Digestive Diseases and Nutrition, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Linda J Ewing
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - John P Foreyt
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Linda J Gay
- Department of Psychiatry, The Miriam Hospital, Brown Medical School, Providence, Rhode Island, USA
| | - Edward W Gregg
- Division of Diabetes Translation, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Helen P Hazuda
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - James O Hill
- Anschutz Health and Wellness Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Edward S Horton
- Department of Integrative Physiology and Metabolism, Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Denise K Houston
- Department of Internal Medicine - Geriatrics, Wake Forest University, Winston-Salem, North Carolina, USA
| | - John M Jakicic
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Robert W Jeffery
- Divisions of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Karen C Johnson
- Departments of Preventive Medicine and Psychiatry, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Steven E Kahn
- Division of Metabolism, Endocrinology and Nutrition, US Department of Veterans Affairs Puget Sound Health Care System, University of Washington, Seattle, Washington, USA
| | - William C Knowler
- Southwestern American Indian Center, National Institute of Diabetes and Digestive and Kidney Diseases and St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Anne Kure
- Division of Metabolism, Endocrinology and Nutrition, US Department of Veterans Affairs Puget Sound Health Care System, University of Washington, Seattle, Washington, USA
| | - Katherine L Michalski
- Division of General Internal Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Maria G Montez
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Rebecca H Neiberg
- Department of Internal Medicine - Geriatrics, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Jennifer Patricio
- Department of Medicine, St. Luke's Roosevelt Hospital Center, Columbia University, New York, New York, USA
| | - Anne Peters
- Division of Endocrinology, University of Southern California, Los Angeles, California, USA
| | - Xavier Pi-Sunyer
- Department of Medicine, St. Luke's Roosevelt Hospital Center, Columbia University, New York, New York, USA
| | - Henry Pownall
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - David Reboussin
- Department of Internal Medicine - Geriatrics, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Bruce Redmon
- Divisions of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - W Jack Rejeski
- Department of Internal Medicine - Geriatrics, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Helmut Steinburg
- Departments of Preventive Medicine and Psychiatry, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Martha Walker
- Division of Endocrinology, University of Southern California, Los Angeles, California, USA
| | | | - Rena R Wing
- Department of Psychiatry, The Miriam Hospital, Brown Medical School, Providence, Rhode Island, USA
| | - Holly Wyatt
- Anschutz Health and Wellness Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Susan Z Yanovski
- Division of Digestive Diseases and Nutrition, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Ping Zhang
- Division of Diabetes Translation, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
18
|
Castegna A, Gissi R, Menga A, Montopoli M, Favia M, Viola A, Canton M. Pharmacological targets of metabolism in disease: Opportunities from macrophages. Pharmacol Ther 2020; 210:107521. [PMID: 32151665 DOI: 10.1016/j.pharmthera.2020.107521] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/28/2020] [Indexed: 12/12/2022]
Abstract
From advances in the knowledge of the immune system, it is emerging that the specialized functions displayed by macrophages during the course of an immune response are supported by specific and dynamically-connected metabolic programs. The study of immunometabolism is demonstrating that metabolic adaptations play a critical role in modulating inflammation and, conversely, inflammation deeply influences the acquisition of specific metabolic settings.This strict connection has been proven to be crucial for the execution of defined immune functional programs and it is now under investigation with respect to several human disorders, such as diabetes, sepsis, cancer, and autoimmunity. The abnormal remodelling of the metabolic pathways in macrophages is now emerging as both marker of disease and potential target of therapeutic intervention. By focusing on key pathological conditions, namely obesity and diabetes, rheumatoid arthritis, atherosclerosis and cancer, we will review the metabolic targets suitable for therapeutic intervention in macrophages. In addition, we will discuss the major obstacles and challenges related to the development of therapeutic strategies for a pharmacological targeting of macrophage's metabolism.
Collapse
Affiliation(s)
- Alessandra Castegna
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy; IBIOM-CNR, Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy; Fondazione Città della Speranza, Istituto di Ricerca Pediatrica, Padua, Italy.
| | - Rosanna Gissi
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Alessio Menga
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy; Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Monica Montopoli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy; Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
| | - Maria Favia
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Antonella Viola
- Department of Biomedical Sciences, University of Padua, Italy; Fondazione Città della Speranza, Istituto di Ricerca Pediatrica, Padua, Italy
| | - Marcella Canton
- Department of Biomedical Sciences, University of Padua, Italy; Fondazione Città della Speranza, Istituto di Ricerca Pediatrica, Padua, Italy.
| |
Collapse
|
19
|
Da Ré C, Souza JM, Fróes F, Taday J, dos Santos JP, Rodrigues L, Sesterheim P, Gonçalves CA, Leite MC. Neuroinflammation induced by lipopolysaccharide leads to memory impairment and alterations in hippocampal leptin signaling. Behav Brain Res 2020; 379:112360. [PMID: 31734263 DOI: 10.1016/j.bbr.2019.112360] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 10/21/2019] [Accepted: 11/13/2019] [Indexed: 12/29/2022]
|
20
|
Zecharia D, Rauch M, Sharabi-Nov A, Tamir S, Gutman R. Postnatal administration of leptin antagonist mitigates susceptibility to obesity under high-fat diet in male αMUPA mice. Am J Physiol Endocrinol Metab 2019; 317:E783-E793. [PMID: 31454257 DOI: 10.1152/ajpendo.00099.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Perturbations in postnatal leptin signaling have been associated with altered susceptibility to diet-induced obesity (DIO) under high-fat-diet (HFD), albeit with contradicting evidence. Previous studies have shown that alpha murine urokinase-type plasminogen activator (αMUPA) mice have a higher and longer postnatal leptin surge compared with their wild types (WTs) as well as lower body weight and food intake under regular diet (RD). Here we explored αMUPA's propensity for DIO and the effect of attenuating postnatal leptin signaling with leptin antagonist (LA) on energy homeostasis under both RD and HFD. Four-day-old αMUPA pups were treated on alternate days until postnatal day 18 with either vehicle or LA (10 or 20 mg·day-1·kg-1) and weaned into RD or HFD. Compared with RD-fed αMUPA males, HFD-fed αMUPA males showed higher energy intake, even when corrected for body weight difference, and became hyperinsulinemic and obese. Additionally, HFD-fed αMUPA males gained body weight at a higher rate than their WTs mainly because of strain differences in energy expenditure. LA administration did not affect strain differences under RD but attenuated αMUPA's hyperinsulinemia and DIO under HFD, most likely by mediating energy expenditure. Together with our previous findings, these results suggest that αMUPA's leptin surge underlies its higher susceptibility to obesity under HFD, highlighting the role of leptin-related developmental processes in inducing obesity in a postweaning obesogenic environment, at least in αMUPA males. This study therefore supports the use of αMUPA mice for elucidating developmental mechanisms of obesity and the efficacy of early-life manipulations via leptin surge axis in attenuating DIO.
Collapse
Affiliation(s)
- Danielle Zecharia
- Laboratory of Integrative Physiology, MIGAL-Galilee Research Institute. Kiryat Shmona, Israel
- Department of Biotechnology, Faculty of Sciences and Technology, Tel-Hai College, Upper Galilee, Israel
| | - Maayan Rauch
- Laboratory of Integrative Physiology, MIGAL-Galilee Research Institute. Kiryat Shmona, Israel
| | - Adi Sharabi-Nov
- Research Wing, Ziv Medical Center, Zefat, Israel
- Department of Nutritional Sciences, Faculty of Sciences and Technology, Tel-Hai College, Upper Galilee, Israel
| | - Snait Tamir
- Department of Nutritional Sciences, Faculty of Sciences and Technology, Tel-Hai College, Upper Galilee, Israel
- Laboratory of Human Health and Nutrition Sciences, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel
| | - Roee Gutman
- Laboratory of Integrative Physiology, MIGAL-Galilee Research Institute. Kiryat Shmona, Israel
- Department of Animal Sciences, Faculty of Sciences and Technology, Tel-Hai College, Upper Galilee, Israel
| |
Collapse
|
21
|
Sinden DS, Holman CD, Bare CJ, Sun X, Gade AR, Cohen DE, Pitt GS. Knockout of the X-linked Fgf13 in the hypothalamic paraventricular nucleus impairs sympathetic output to brown fat and causes obesity. FASEB J 2019; 33:11579-11594. [PMID: 31339804 PMCID: PMC6994920 DOI: 10.1096/fj.201901178r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/01/2019] [Indexed: 12/15/2022]
Abstract
Fibroblast growth factor (FGF)13, a nonsecreted, X-linked, FGF homologous factor, is differentially expressed in adipocytes in response to diet, yet Fgf13's role in metabolism has not been explored. Heterozygous Fgf13 knockouts fed normal chow and housed at 22°C showed hyperactivity accompanying reduced core temperature and obesity when housed at 30°C. Those heterozygous knockouts showed defects in thermogenesis even at 30°C and an inability to protect core temperature. Surprisingly, we detected trivial FGF13 in adipose of wild-type mice fed normal chow and no obesity in adipose-specific heterozygous knockouts housed at 30°C, and we detected an intact brown fat response through exogenous β3 agonist stimulation, suggesting a defect in sympathetic drive to brown adipose tissue. In contrast, hypothalamic-specific ablation of Fgf13 recapitulated weight gain at 30°C. Norepinephrine turnover in brown fat was reduced at both housing temperatures. Thus, our data suggest that impaired CNS regulation of sympathetic activation of brown fat underlies obesity and thermogenesis in Fgf13 heterozygous knockouts fed normal chow.-Sinden, D. S., Holman, C. D., Bare, C. J., Sun, X., Gade, A. R., Cohen, D. E., Pitt, G. S. Knockout of the X-linked Fgf13 in the hypothalamic paraventricular nucleus impairs sympathetic output to brown fat and causes obesity.
Collapse
Affiliation(s)
- Daniel S. Sinden
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
- Cardiovascular Research Institute, Weill Cornell Medical College, New York, New York, USA
| | - Corey D. Holman
- Division of Gastroenterology and Hepatology, Weill Cornell Medical College, New York, New York, USA
| | - Curtis J. Bare
- Division of Gastroenterology and Hepatology, Weill Cornell Medical College, New York, New York, USA
| | - Xiaolu Sun
- Cardiovascular Research Institute, Weill Cornell Medical College, New York, New York, USA
| | - Aravind R. Gade
- Cardiovascular Research Institute, Weill Cornell Medical College, New York, New York, USA
| | - David E. Cohen
- Division of Gastroenterology and Hepatology, Weill Cornell Medical College, New York, New York, USA
| | - Geoffrey S. Pitt
- Cardiovascular Research Institute, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
22
|
Control-theory models of body-weight regulation and body-weight-regulatory appetite. Appetite 2019; 144:104440. [PMID: 31494154 DOI: 10.1016/j.appet.2019.104440] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 08/08/2019] [Accepted: 09/02/2019] [Indexed: 12/24/2022]
Abstract
Human body weight (BW), or some variable related to it, is physiologically regulated. That is, negative feedback from changes in BW elicits compensatory influences on appetite, which may be called BW-regulatory appetite, and a component of energy expenditure (EE) called adaptive thermogenesis (AdEE). BW-regulatory appetite is of general significance because it appears to be related to a variety of aspects of human appetite beyond just energy intake. BW regulation, BW-regulatory appetite and AdEE are frequently discussed using concepts derived from control theory, which is the mathematical description of dynamic systems involving negative feedback. The aim of this review is to critically assess these discussions. Two general types of negative-feedback control have been invoked to describe BW regulation, set-point control and simple negative-feedback control, often called settling-point control in the BW literature. The distinguishing feature of set-point systems is the existence of an externally controlled target level of regulation, the set point. The performance of almost any negative-feedback regulatory system, however, can be modeled on the basis of feedback gain without including a set point. In both set-point and simple negative-feedback models of BW regulation, the precision of regulation is usually determined mainly by feedback gain, which refers to the transformations of feedback into compensatory changes in BW-regulatory appetite and AdEE. Stable BW most probably represents equilibria shaped by feedback gain and tonic open-loop challenges, especially obesogenic environments. Data indicate that simple negative-feedback control accurately models human BW regulation and that the set-point concept is superfluous unless its neuroendocrine representation is found in the brain. Additional research aimed at testing control-theory models in humans and non-human animals is warranted.
Collapse
|
23
|
Wensveen FM, Šestan M, Turk Wensveen T, Polić B. 'Beauty and the beast' in infection: How immune-endocrine interactions regulate systemic metabolism in the context of infection. Eur J Immunol 2019; 49:982-995. [PMID: 31106860 DOI: 10.1002/eji.201847895] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/28/2019] [Accepted: 05/17/2019] [Indexed: 02/07/2023]
Abstract
The immune and endocrine systems ensure two vital functions in the body. The immune system protects us from lethal pathogens, whereas the endocrine system ensures proper metabolic function of peripheral organs by regulating systemic homeostasis. These two systems were long thought to operate independently. The immune system uses cytokines and immune receptors, whereas the endocrine system uses hormones to regulate metabolism. However, recent findings show that the immune and endocrine systems closely interact, especially regarding regulation of glucose metabolism. In response to pathogen encounter, cytokines modify responsiveness of peripheral organs to endocrine signals, resulting in altered levels of blood hormones such as insulin, which promotes the ability of the body to fight infection. Here we provide an overview of recent literature describing various mechanisms, which the immune system utilizes to modify endocrine regulation of systemic metabolism. Moreover, we will describe how these immune-endocrine interactions derail in the context of obesity. From a clinical perspective we will elaborate how infection and obesity aggravate the development of metabolic diseases such as diabetes mellitus type 2 in humans. In summary, this review provides a comprehensive overview of immune-induced changes in systemic metabolism following infection, with a focus on regulation of glucose metabolism.
Collapse
Affiliation(s)
- Felix M Wensveen
- Department of Histology and Embryology, University of Rijeka School of Medicine, Rijeka, Croatia
| | - Marko Šestan
- Department of Histology and Embryology, University of Rijeka School of Medicine, Rijeka, Croatia
| | - Tamara Turk Wensveen
- Department of Endocrinology, Diabetes and Metabolic Diseases, Clinical hospital center Rijeka, Rijeka, Croatia
| | - Bojan Polić
- Department of Histology and Embryology, University of Rijeka School of Medicine, Rijeka, Croatia
| |
Collapse
|
24
|
Sendžikaitė G, Hanna CW, Stewart-Morgan KR, Ivanova E, Kelsey G. A DNMT3A PWWP mutation leads to methylation of bivalent chromatin and growth retardation in mice. Nat Commun 2019; 10:1884. [PMID: 31015495 PMCID: PMC6478690 DOI: 10.1038/s41467-019-09713-w] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 03/26/2019] [Indexed: 11/08/2022] Open
Abstract
DNA methyltransferases (DNMTs) deposit DNA methylation, which regulates gene expression and is essential for mammalian development. Histone post-translational modifications modulate the recruitment and activity of DNMTs. The PWWP domains of DNMT3A and DNMT3B are posited to interact with histone 3 lysine 36 trimethylation (H3K36me3); however, the functionality of this interaction for DNMT3A remains untested in vivo. Here we present a mouse model carrying a D329A point mutation in the DNMT3A PWWP domain. The mutation causes dominant postnatal growth retardation. At the molecular level, it results in progressive DNA hypermethylation across domains marked by H3K27me3 and bivalent chromatin, and de-repression of developmental regulatory genes in adult hypothalamus. Evaluation of non-CpG methylation, a marker of de novo methylation, further demonstrates the altered recruitment and activity of DNMT3AD329A at bivalent domains. This work provides key molecular insights into the function of the DNMT3A-PWWP domain and role of DNMT3A in regulating postnatal growth.
Collapse
Affiliation(s)
| | - Courtney W Hanna
- Epigenetics Programme, Babraham Institute, Cambridge, CB22 3AT, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, CB2 3EG, UK
| | - Kathleen R Stewart-Morgan
- Epigenetics Programme, Babraham Institute, Cambridge, CB22 3AT, UK
- Biotech Research & Innovation Centre, 2200, Copenhagen, Denmark
| | - Elena Ivanova
- Epigenetics Programme, Babraham Institute, Cambridge, CB22 3AT, UK
| | - Gavin Kelsey
- Epigenetics Programme, Babraham Institute, Cambridge, CB22 3AT, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, CB2 3EG, UK.
| |
Collapse
|
25
|
Effects of polyphenol (carob) supplementation on body composition and aerobic capacity in taekwondo athletes. Physiol Behav 2019; 205:22-28. [PMID: 30853622 DOI: 10.1016/j.physbeh.2019.03.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 03/06/2019] [Accepted: 03/06/2019] [Indexed: 01/08/2023]
Abstract
Herbal products and supplements use by athletes has increased over the past decade. One such item being polyphenols. These are reported to reduce weight and modify body composition, which could aid athletes in many sports. Therefore, the purpose of the study was to determine the effect of 6 weeks supplementation with carob, a naturally occurring polyphenol, on body composition and aerobic capacity in youth taekwondo athletes. Twenty-three taekwondo athletes (21.9 ± 1.2 years; 1.64 ± 0.03 m; 67.4 ± 17.3 kg;BMI: 22.8 ± 5.5 kg/m2) participated in a short-term (6-week) double-blind randomized design parallel fully controlled training study (pre-to-post measurements): Supplemented group (SG), n = 11;placebo group (PG), n = 12. Body composition, aerobic capacity, heart rate and RPE were analyzed before and after 6 weeks of carob rich polyphenol ingestion. Significantly greater decreases in weight were observed for SG and PG (-2.82% and - 0.51%respectively) with differences between groups (p < 0.001). No significant differences were reported in percentage body fat and muscular volume between groups. Our results revealed an improvement of aerobic performance score and RPE with differences between groups. A cute polyphenol supplementation seemed to be effective in reducing body weight and improving aerobic performance in athletes.
Collapse
|
26
|
Daghestani MH, Daghestani MH, Daghistani MH, Bjørklund G, Chirumbolo S, Warsy A. The influence of the rs1137101 genotypes of leptin receptor gene on the demographic and metabolic profile of normal Saudi females and those suffering from polycystic ovarian syndrome. BMC WOMENS HEALTH 2019; 19:10. [PMID: 30635060 PMCID: PMC6329086 DOI: 10.1186/s12905-018-0706-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 12/28/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND Polycystic ovarian syndrome (PCOS) is of frequent occurrence in Saudi females and is often associated with obesity, insulin resistance, hypogonadotropic hypogonadism, and infertility. Since these features are also associated with leptin receptor (LEP-R) deficiency, several studies have attempted to link LEP-R gene polymorphisms to PCOS. METHODS The purpose of this study is to assess the possible association of LEP-R gene polymorphism (rs1137101) with the main obesity-linked metabolic parameters in Saudi female patients affected by PCOS. A cohort of 122 Saudi female subjects, attending the outpatient's clinics at Makkah, Saudi Arabia and diagnosed with PCOS was investigated. Metabolic parameters in serum samples, including lipidogram, glucose, leptin, ghrelin and insulin and obesity markers (BMI, W/H ratio, HOMA) were assayed and compared with values from 130 healthy female volunteers (controls). The genotyping of rs1137101 polymorphism in the leptin receptor gene by amplification (PCR) followed by DNA sequencing, was conducted in both groups (PCOS and controls). RESULTS Waist/hip ratio (W/H ratio), leptin serum levels and triglycerides appeared to be associated with PCOS but, aside from W/H ratio (AA s GG p = 0.009), this association also occurred for controls. No significant association in the leptin gene polymorphic locus rs1137101 with PCOS was seen in the results of the present study. In the control group, BMI, W/H ratio, leptin, Insulin, and HOMA-IR were significantly higher in the GG genotype compared to AA. CONCLUSION Despite previous suggestion about a relationship between rs1137101, serum leptin levels, and PCOS, our studies do not show any statistical association and further investigations; possibly by also evaluating obese patients should be needed to elucidate this issue better.
Collapse
Affiliation(s)
- Maha H Daghestani
- Department of Zoology, College of Science. Director of Central Laboratory, Female Center for Scientific & Medical Colleges, King Saud University, Riyadh, Saudi Arabia
| | - Mazin H Daghestani
- Department of Obstetrics and Gynecology, Umm Al-Qura University, P.O. Box 424, Makkah, 21955, Saudi Arabia
| | - Mamoon H Daghistani
- Department of Surgery, King Abdulaziz Medical City, National Guard Health Affairs, Jeddah, Kingdom of Saudi Arabia
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Mo i Rana, Norway
| | - Salvatore Chirumbolo
- Department of Neurological and Movement Sciences, University of Verona, Verona, Italy
| | - Arjumand Warsy
- Central Laboratory, Female Center for Scientific and Medical Colleges, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
27
|
Müller MJ, Krawczak M, Heymsfield S, Schutz Y, Dulloo A, Blundell J, Geisler C, Bosy-Westphal A. Thanks for opening an overdue discussion on GWAS of BMI: a reply to Prof. Speakman et al. Int J Obes (Lond) 2018; 43:217-218. [PMID: 30482932 DOI: 10.1038/s41366-018-0264-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/08/2018] [Accepted: 10/15/2018] [Indexed: 11/09/2022]
Affiliation(s)
- Manfred J Müller
- Institut für Humanernährung und Lebensmittelkunde, Christian-Albrechts-Universität zu Kiel, Kiel, Germany.
| | - Michael Krawczak
- Institut für Medizinische Informatik und Statistik, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | | | - Yves Schutz
- Institute de Physiology, University of Lausanne, Lausanne, Switzerland
| | - Abdul Dulloo
- Department of Medicine, Division of Physiology, University of Fribourg, Fribourg, Switzerland
| | - John Blundell
- Institute of Psychological Sciences, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Corinna Geisler
- Institut für Humanernährung und Lebensmittelkunde, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Anja Bosy-Westphal
- Institut für Humanernährung und Lebensmittelkunde, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| |
Collapse
|
28
|
Abstract
During the last decades, research on adipose tissues has spread in parallel with the extension of obesity. Several observations converged on the idea that adipose tissues are organized in a large organ with endocrine and plastic properties. Two parenchymal components: white (WATs) and brown adipose tissues (BATs) are contained in subcutaneous and visceral compartments. Although both have endocrine properties, their function differs: WAT store lipids to allow intervals between meals, BAT burns lipids for thermogenesis. In spite of these opposite functions, they share the ability for reciprocal reversible transdifferentiation to tackle special physiologic needs. Thus, chronic need for thermogenesis induces browning and chronic positive energy balance induce whitening. Lineage tracing and data from explant studies strongly suggest other remodeling properties of this organ. During pregnancy and lactation breast WAT transdifferentiates into milk-secreting glands, composed by cells with abundant cytoplasmic lipids (pink adipocytes) and in the postlactation period pink adipocytes transdifferentiate back into WAT and BAT. The plastic properties of mature adipocytes are supported also by a liposecretion process in vitro where adult cell in culture transdifferentiate to differentiated fibroblast-like elements able to give rise to different phenotypes (rainbow adipocytes). In addition, the inflammasome system is activated in stressed adipocytes from obese adipose tissue. These adipocytes die and debris are reabsorbed by macrophages inducing a chronic low-grade inflammation, potentially contributing to insulin resistance and T2 diabetes. Thus, the plastic properties of this organ could open new therapeutic perspectives in the obesity-related metabolic disease and in breast pathologies. © 2018 American Physiological Society. Compr Physiol 8:1357-1431, 2018.
Collapse
Affiliation(s)
- Saverio Cinti
- Professor of Human Anatomy, Director, Center of Obesity, University of Ancona (Politecnica delle Marche), Ancona, Italy
| |
Collapse
|
29
|
Müller MJ, Geisler C, Heymsfield SB, Bosy-Westphal A. Recent advances in understanding body weight homeostasis in humans. F1000Res 2018; 7. [PMID: 30026913 PMCID: PMC6039924 DOI: 10.12688/f1000research.14151.1] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/29/2018] [Indexed: 12/19/2022] Open
Abstract
Presently, control of body weight is assumed to exist, but there is no consensus framework of body weight homeostasis. Three different models have been proposed, with a "set point" suggesting (i) a more or less tight and (ii) symmetric or asymmetric biological control of body weight resulting from feedback loops from peripheral organs and tissues (e.g. leptin secreted from adipose tissue) to a central control system within the hypothalamus. Alternatively, a "settling point" rather than a set point reflects metabolic adaptations to energy imbalance without any need for feedback control. Finally, the "dual intervention point" model combines both paradigms with two set points and a settling point between them. In humans, observational studies on large populations do not provide consistent evidence for a biological control of body weight, which, if it exists, may be overridden by the influences of the obesogenic environment and culture on personal behavior and experiences. To re-address the issue of body weight homeostasis, there is a need for targeted protocols based on sound concepts, e.g. lean rather than overweight subjects should be investigated before, during, and after weight loss and weight regain. In addition, improved methods and a multi-level-multi-systemic approach are needed to address the associations (i) between masses of individual body components and (ii) between masses and metabolic functions in the contexts of neurohumoral control and systemic effects. In the future, simplifications and the use of crude and non-biological phenotypes (i.e. body mass index and waist circumference) should be avoided. Since changes in body weight follow the mismatch between tightly controlled energy expenditure at loosely controlled energy intake, control (or even a set point) is more likely to be about energy expenditure rather than about body weight itself.
Collapse
Affiliation(s)
- Manfred J Müller
- Institute of Human Nutrition and Food Science, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Corinna Geisler
- Institute of Human Nutrition and Food Science, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | | | - Anja Bosy-Westphal
- Institute of Human Nutrition and Food Science, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| |
Collapse
|
30
|
Park HS, Cho JH, Kim KW, Chung WS, Song MY. Effects of Panax ginseng on Obesity in Animal Models: A Systematic Review and Meta-Analysis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2018; 2018:2719794. [PMID: 29861768 PMCID: PMC5976977 DOI: 10.1155/2018/2719794] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 02/05/2018] [Indexed: 01/11/2023]
Abstract
OBJECTIVE To determine the antiobesity effects of Panax ginseng in animals. METHODS We conducted a systematic search for all controlled trials (up to March 2017) that assessed the antiobesity effects of P. ginseng in animal obesity models in the PubMed, EMBASE, Cochrane library, Web of Science, and Scopus databases. The primary outcome was final body weight measured at the longest follow-up time after administration of the intervention. The secondary outcome was the lipid profile. We assessed methodological quality using the SYRCLE risk of bias tool, and RevMan 5.3 was used to perform a meta-analysis. Finally, a subgroup analysis of parameters including intervention duration, animal models, and type of ginseng was performed. RESULT We identified 16 studies that met the inclusion criteria. Data from the meta-analysis indicated that the intervention group had a significantly lower body weight than the control group (SMD: -1.50, 95% CI: -1.90 to -1.11, χ2: 78.14, P < 0.0001, I2 = 58%). Final body weight was lower in an animal obesity model induced by high-fat diet than in genetic models. Also the intervention group had a significantly higher serum HDL level and lower serum LDL, TG, and TC level than the control group. CONCLUSION Our meta-analysis indicated that oral administration of P. ginseng significantly inhibits weight gain and improves serum lipid profiles in animal obesity models. However, causes of obesity and type of ginseng may affect treatment effects.
Collapse
Affiliation(s)
- Hye-Sung Park
- Department of Korean Rehabilitation Medicine, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 130-701, Republic of Korea
| | - Jae-Heung Cho
- Department of Korean Rehabilitation Medicine, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 130-701, Republic of Korea
| | - Koh-Woon Kim
- Department of Korean Rehabilitation Medicine, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 130-701, Republic of Korea
| | - Won-Seok Chung
- Department of Korean Rehabilitation Medicine, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 130-701, Republic of Korea
| | - Mi-Yeon Song
- Department of Korean Rehabilitation Medicine, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 130-701, Republic of Korea
| |
Collapse
|
31
|
Fischer IP, Irmler M, Meyer CW, Sachs SJ, Neff F, Hrabě de Angelis M, Beckers J, Tschöp MH, Hofmann SM, Ussar S. A history of obesity leaves an inflammatory fingerprint in liver and adipose tissue. Int J Obes (Lond) 2018; 42:507-517. [PMID: 28901330 PMCID: PMC5880583 DOI: 10.1038/ijo.2017.224] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/28/2017] [Accepted: 09/04/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND/OBJECTIVES Dieting is a popular yet often ineffective way to lower body weight, as the majority of people regain most of their pre-dieting weights in a relatively short time. The underlying molecular mechanisms driving weight regain and the increased risk for metabolic disease are still incompletely understood. Here we investigate the molecular alterations inherited from a history of obesity. METHODS In our model, male high-fat diet (HFD)-fed obese C57BL/6J mice were switched to a low caloric chow diet, resulting in a decline of body weight to that of lean mice. We measured body composition, as well as metrics of glucose, insulin and lipid homeostasis. This was accompanied by histological and gene expression analysis of adipose tissue and liver to assess adipose tissue inflammation and hepatosteatosis. Moreover, acute hypothalamic response to (re-) exposure to HFD was assessed by qPCR. RESULTS & CONCLUSIONS Within 7 weeks after diet switch, most obesity-associated phenotypes, such as body mass, glucose intolerance and blood metabolite levels were reversed. However, hepatic inflammation, hepatic steatosis as well as hypertrophy and inflammation of perigonadal, but not subcutaneous, adipocytes persisted in formerly obese mice. Transcriptional profiling of liver and perigonadal fat revealed an upregulation of pathways associated with immune function and cellularity. Thus, we show that weight reduction leaves signs of inflammation in liver and perigonadal fat, indicating that persisting proinflammatory signals in liver and adipose tissue could contribute to an increased risk of formerly obese subjects to develop the metabolic syndrome upon recurring weight gain.
Collapse
Affiliation(s)
- I P Fischer
- JRG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Center Munich, Garching, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
| | - M Irmler
- Institute for Experimental Genetics, Helmholtz Zentrum München, München-Neuherberg, Germany
| | - C W Meyer
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
| | - S J Sachs
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Medizinische Klinik und Poliklinik IV der LMU, Munich, Germany
- Institute for Diabetes and Regeneration, Helmholtz Diabetes Center at Helmholtz Zentrum München, Garching, Germany
| | - F Neff
- Institute for Pathology, Helmholtz Zentrum München, München-Neuherberg, Germany
| | - M Hrabě de Angelis
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Institute for Experimental Genetics, Helmholtz Zentrum München, München-Neuherberg, Germany
- Technische Universität München, Lehrstuhl für Experimentelle Genetik, Freising, Germany
| | - J Beckers
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Institute for Experimental Genetics, Helmholtz Zentrum München, München-Neuherberg, Germany
- Technische Universität München, Lehrstuhl für Experimentelle Genetik, Freising, Germany
| | - M H Tschöp
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Garching, Germany
| | - S M Hofmann
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Medizinische Klinik und Poliklinik IV der LMU, Munich, Germany
- Institute for Diabetes and Regeneration, Helmholtz Diabetes Center at Helmholtz Zentrum München, Garching, Germany
| | - S Ussar
- JRG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Center Munich, Garching, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| |
Collapse
|
32
|
Haas V, Stengel A, Mähler A, Gerlach G, Lehmann C, Boschmann M, de Zwaan M, Herpertz S. Metabolic Barriers to Weight Gain in Patients With Anorexia Nervosa: A Young Adult Case Report. Front Psychiatry 2018; 9:199. [PMID: 29867616 PMCID: PMC5968865 DOI: 10.3389/fpsyt.2018.00199] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 04/30/2018] [Indexed: 11/18/2022] Open
Abstract
Background: Over-proportionally high energy requirements in some patients with anorexia nervosa (AN) have been reported, but their exact origin remains unclear. Objective: To objectively measure metabolic alterations in an AN patient with high energy requirements as judged by clinical observation. Materials and Methods: We present the case of a young woman with AN (index patient, IP; 19 years, admission BMI 13.9 kg/m2). After 3 months of treatment at BMI 17.4 kg/m2, we assessed her resting energy expenditure (REE), respiratory exchange ratio (RER), diet-induced thermogenesis (DIT), seated non-exercise physical activity (NEPA in Volt by infrared sensors), and exercise activity thermogenesis (EAT) in a metabolic chamber; body composition (bioimpedance analysis), energy intake (15d-food protocol), physical activity (accelerometry) and endocrine parameters. The IP was compared for REE, RER, DIT and seated NEPA to six AN patients (AN-C) and four healthy women (HC-1), and for EAT to another six healthy women (HC-2). Results: Our IP showed high REE (110% of predicted REE according to Harris & Benedict) and high seated NEPA (47% increase over AN-C, 40% over HC-1), whereas DIT (IP: 78 vs. HC-1: 145 ± 51 kJ/180 min) and EAT (IP: 157 vs. HC-2: 235 ± 30 kJ/30 min) were low, when compared with HC. The other AN patients showed a lower REE (AN: 87 ± 2% vs. HC: 97 ± 2% predicted) at increased DIT (AN: 187 ± 91 vs. HC: 145 ± 51 kJ/180 min) when compared with HC. RER of the IP was low (IP: 0.72 vs. 0.77 in AN-C; 0.77 in HC-1 and 0.80 in HC-2). Conclusions: Complex and variable disturbances of energy metabolism might exist in a subgroup of patients with AN during refeeding, which could lead to unexpectedly high energy requirements. Future studies need to confirm the existence, and investigate the characteristics and prevalence of this subgroup. Clinical trial Registry number: NCT02087280, https://www.clinicaltrials.gov/.
Collapse
Affiliation(s)
- Verena Haas
- Department of Child and Adolescent Psychiatry, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Andreas Stengel
- Department of Psychosomatic Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Anja Mähler
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Gabriele Gerlach
- Department of Psychosomatic Medicine and Psychotherapy, LWL-Universitätsklinikum, Ruhr-Universität Bochum, Bochum, Germany
| | - Celine Lehmann
- Department of Child and Adolescent Psychiatry, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Psychosomatic Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Boschmann
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Martina de Zwaan
- Department of Psychosomatic Medicine and Psychotherapy, Hannover Medical School, Hanover, Germany
| | - Stephan Herpertz
- Department of Psychosomatic Medicine and Psychotherapy, LWL-Universitätsklinikum, Ruhr-Universität Bochum, Bochum, Germany
| |
Collapse
|
33
|
Rubinstein M, Low MJ. Molecular and functional genetics of the proopiomelanocortin gene, food intake regulation and obesity. FEBS Lett 2017; 591:2593-2606. [PMID: 28771698 PMCID: PMC9975356 DOI: 10.1002/1873-3468.12776] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 07/31/2017] [Accepted: 07/31/2017] [Indexed: 12/20/2022]
Abstract
A specter is haunting the world, the specter of obesity. During the last decade, this pandemia has skyrocketed threatening children, adolescents and lower income families worldwide. Although driven by an increase in the consumption of ultraprocessed edibles of poor nutritional value, the obesogenic changes in contemporary human lifestyle affect people differently, revealing that some individuals are more prone to develop increased adiposity. During the last years, we performed a variety of genetic, evolutionary, biochemical and behavioral experiments that allowed us to understand how a group of neurons present in the arcuate nucleus of the hypothalamus regulate the expression of the proopiomelanocortin (Pomc) gene and induce satiety. We disentangled the neuronal transcriptional code of Pomc by identifying the cis-acting regulatory elements and primary transcription factors controlling hypothalamic Pomc expression and determined their functional importance in the regulation of food intake and adiposity. Altogether, our studies reviewed here shed light on the power and limitations of the mammalian central satiety pathways and may contribute to the development of individual and collective strategies to reduce the debilitating effects of the self-induced obesity pandemia.
Collapse
Affiliation(s)
- Marcelo Rubinstein
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina,Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Argentina,Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Malcolm J. Low
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA,Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
34
|
Andermann ML, Lowell BB. Toward a Wiring Diagram Understanding of Appetite Control. Neuron 2017; 95:757-778. [PMID: 28817798 DOI: 10.1016/j.neuron.2017.06.014] [Citation(s) in RCA: 373] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 06/02/2017] [Accepted: 06/07/2017] [Indexed: 01/26/2023]
Abstract
Prior mouse genetic research has set the stage for a deep understanding of appetite regulation. This goal is now being realized through the use of recent technological advances, such as the ability to map connectivity between neurons, manipulate neural activity in real time, and measure neural activity during behavior. Indeed, major progress has been made with regard to meal-related gut control of appetite, arcuate nucleus-based hypothalamic circuits linking energy state to the motivational drive, hunger, and, finally, limbic and cognitive processes that bring about hunger-mediated increases in reward value and perception of food. Unexpected findings are also being made; for example, the rapid regulation of homeostatic neurons by cues that predict future food consumption. The aim of this review is to cover the major underpinnings of appetite regulation, describe recent advances resulting from new technologies, and synthesize these findings into an updated view of appetite regulation.
Collapse
Affiliation(s)
- Mark L Andermann
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA.
| | - Bradford B Lowell
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
35
|
Bae CR, Hino J, Hosoda H, Arai Y, Son C, Makino H, Tokudome T, Tomita T, Kimura T, Nojiri T, Hosoda K, Miyazato M, Kangawa K. Overexpression of C-type Natriuretic Peptide in Endothelial Cells Protects against Insulin Resistance and Inflammation during Diet-induced Obesity. Sci Rep 2017; 7:9807. [PMID: 28852070 PMCID: PMC5574992 DOI: 10.1038/s41598-017-10240-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 08/02/2017] [Indexed: 02/08/2023] Open
Abstract
The endogenous peptide C-type natriuretic peptide (CNP) binds its receptor, guanylyl cyclase B (GCB), and is expressed by endothelial cells in diverse tissues. Because the endothelial cells of visceral adipose tissue have recently been reported to play a role in lipid metabolism and inflammation, we investigated the effects of CNP on features of obesity by using transgenic (Tg) mice in which CNP was placed under the control of the Tie2 promoter and was thus overexpressed in endothelial cells (E-CNP). Here we show that increased brown adipose tissue thermogenesis in E-CNP Tg mice increased energy expenditure, decreased mesenteric white adipose tissue (MesWAT) fat weight and adipocyte hypertrophy, and prevented the development of fatty liver. Furthermore, CNP overexpression improved glucose tolerance, decreased insulin resistance, and inhibited macrophage infiltration in MesWAT, thus suppressing pro-inflammation during high-fat diet–induced obesity. Our findings indicate an important role for the CNP produced by the endothelial cells in the regulation of MesWAT hypertrophy, insulin resistance, and inflammation during high-fat diet–induced obesity.
Collapse
Affiliation(s)
- Cho-Rong Bae
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Jun Hino
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan.
| | - Hiroshi Hosoda
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Yuji Arai
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Cheol Son
- Division of Endocrinology and Metabolism, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan.,Omics Research Center, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Hisashi Makino
- Division of Endocrinology and Metabolism, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Takeshi Tokudome
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Tsutomu Tomita
- Division of Endocrinology and Metabolism, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan.,Biobank, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Toru Kimura
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Takashi Nojiri
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Kiminori Hosoda
- Division of Endocrinology and Metabolism, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Mikiya Miyazato
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Kenji Kangawa
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan.
| |
Collapse
|
36
|
Gillman MW, Rifas-Shiman SL, Fernandez-Barres S, Kleinman K, Taveras EM, Oken E. Beverage Intake During Pregnancy and Childhood Adiposity. Pediatrics 2017; 140:peds.2017-0031. [PMID: 28689188 PMCID: PMC5527670 DOI: 10.1542/peds.2017-0031] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/12/2017] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES To examine associations of sugar sweetened beverages (SSBs) and other beverage intake during pregnancy with adiposity in midchildhood (median age of 7.7 years). METHODS We studied 1078 mother-child pairs in Project Viva, a prospective prebirth cohort study. Exposures were sugary and nonsugary beverage intake assessed in the first and second trimesters of pregnancy by using a food frequency questionnaire. Main outcome measures were offspring overall adiposity (BMI z score, fat mass index [FMI, kg/m2] from dual-energy radiograph absorptiometry, and sum of subscapular [SS] and triceps [TR] skinfold thicknesses) and central adiposity (SS:TR ratio and waist circumference). RESULTS In the second trimester, mean (SD) SSB intake was 0.6 (0.9) servings per day. Thirty-two percent of mothers were multiracial or people of color, 68% were college graduates, and 10% smoked during pregnancy. In midchildhood, mean (SD) BMI z score was 0.38 (1.00), and the FMI was 4.4 (1.9) kg/m2. In multivariable models adjusted for multiple maternal and child covariates, each additional serving per day of SSB was associated with higher BMI z scores (0.07 U; 95% confidence interval [CI]: -0.01 to 0.15), FMI (0.15 kg/m2; 95% CI: -0.01 to 0.30), SS + TR (0.85 mm; 95% CI: 0.06 to 1.64), and waist circumference (0.65 cm; 95% CI: 0.01 to 1.28). Stratified models suggested that the associations were due primarily to maternal, not child, SSB intake and to sugary soda rather than fruit drinks or juice. We did not find differences between boys and girls, nor did we observe the effects of sugar-free soda or of first-trimester beverage intake. CONCLUSIONS Higher SSB intake during the second trimester of pregnancy was associated with greater adiposity in midchildhood.
Collapse
Affiliation(s)
- Matthew W. Gillman
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts
| | - Sheryl L. Rifas-Shiman
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts
| | - Silvia Fernandez-Barres
- Nutrition and Mental Health Research Group, Universitat Rovira I Virgili, Reus, Spain;,ISGlobal, Center for Research in Environmental Epidemiology, Barcelona, Spain
| | - Ken Kleinman
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts, Amherst, Massachusetts; and
| | - Elsie M. Taveras
- Division of General Academic Pediatrics, Department of Pediatrics, Massachusetts General Hospital for Children, Boston, Massachusetts
| | - Emily Oken
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts
| |
Collapse
|
37
|
Farhat G, Drummond S, Al-Dujaili EAS. Polyphenols and Their Role in Obesity Management: A Systematic Review of Randomized Clinical Trials. Phytother Res 2017; 31:1005-1018. [PMID: 28493374 DOI: 10.1002/ptr.5830] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 04/06/2017] [Accepted: 04/17/2017] [Indexed: 12/21/2022]
Abstract
Polyphenols have been suggested to reduce body weight and modify body composition through different mechanisms. These effects have been extensively studied in animals and in vitro and to a lesser extent in humans. The aim of this review is to consider the association between polyphenols and body weight status by focusing on human intervention studies. We conducted a systematic literature search in MEDLINE (via EBSCOhost), ProQuest CENTRAL, and Cochrane CENTRAL without time restrictions. Randomized controlled trials assessing the effect of polyphenols on weight and/or body composition in the overweight and/or obese population were included. Nineteen studies met our inclusion criteria. Results suggest that further research is required before supporting a potential role of polyphenols in reducing weight in overweight and obese individuals (nine studies showed a significant decrease in weight by a mean of 1.47 ± 0.58 kg). Nevertheless, several studies indicated that polyphenols might be effective in preventing small increases in weight during periods of overfeeding rather than reducing weight as such. The outcomes noted do not yet support polyphenol supplementation as a complementary approach in weight loss diets. Further larger trials with a duration of 12 months or more are needed to elucidate the effect of polyphenols on body weight status. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Grace Farhat
- Department of Dietetics, Nutrition and Biological Sciences, Queen Margaret University, Musselburgh, East Lothian, EH21 6UU, UK
| | - Sandra Drummond
- Department of Dietetics, Nutrition and Biological Sciences, Queen Margaret University, Musselburgh, East Lothian, EH21 6UU, UK
| | | |
Collapse
|
38
|
Ivić V, Blažetić S, Labak I, Balog M, Vondrak L, Blažeković R, Vari SG, Heffer M. Ovariectomy and chronic stress lead toward leptin resistance in the satiety centers and insulin resistance in the hippocampus of Sprague-Dawley rats. Croat Med J 2017; 57:194-206. [PMID: 27106360 PMCID: PMC4856194 DOI: 10.3325/cmj.2016.57.194] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Aim To evaluate the changes in the expression level of gonadal steroid, insulin, and leptin receptors in the brain of adult Sprague-Dawley female rats due to ovariectomy and/or chronic stress. Methods Sixteen-week-old ovariectomized and non-ovariectomized female Sprague-Dawley rats were divided in two groups and exposed to three 10-day-sessions of sham or chronic stress. After the last stress-session the brains were collected and free-floating immunohistochemical staining was performed using androgen (AR), progesterone (PR), estrogen-β (ER-β), insulin (IR-α), and leptin receptor (ObR) antibodies. The level of receptors expression was analyzed in hypothalamic (HTH), cortical (CTX), dopaminergic (VTA/SNC), and hippocampal regions (HIPP). Results Ovariectomy downregulated AR in the hypothalamic satiety centers and hippocampus. It prevented or attenuated the stress-specific upregulation of AR in these regions. The main difference in stress response between non-ovariectomized and ovariectomized females was in PR level. Ovariectomized ones had increased PR level in the HTH, VTA, and HIPP. Combination of stressors pushed the hypothalamic satiety centers toward the rise of ObR and susceptibility to leptin resistance. When exposed to combined stressors, the HIPP, SNC and piriform cortex upregulated the expression of IR-α and the possibility to develop insulin resistance. Conclusion Ovariectomy exacerbates the effect of chronic stress by preventing gonadal receptor-specific stress response reflected in the upregulation of AR in the satiety and hippocampal regions, while stress after ovariectomy usually raises PR. The final outcome of inadequate stress response is reflected in the upregulation of ObR in the satiety centers and IR-α in the regions susceptible to early neurodegeneration. We discussed the possibility of stress induced metabolic changes under conditions of hormone deprivation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Marija Heffer
- Marija Heffer, Department of Medical Biology and Genetics, Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, Cara Hadrijana 10/E, HR-31000 Osijek, Croatia,
| |
Collapse
|
39
|
Abstract
Metabolic adaptation to weight changes relates to body weight control, obesity and malnutrition. Adaptive thermogenesis (AT) refers to changes in resting and non-resting energy expenditure (REE and nREE) which are independent from changes in fat-free mass (FFM) and FFM composition. AT differs in response to changes in energy balance. With negative energy balance, AT is directed towards energy sparing. It relates to a reset of biological defence of body weight and mainly refers to REE. After weight loss, AT of nREE adds to weight maintenance. During overfeeding, energy dissipation is explained by AT of the nREE component only. As to body weight regulation during weight loss, AT relates to two different set points with a settling between them. During early weight loss, the first set is related to depleted glycogen stores associated with the fall in insulin secretion where AT adds to meet brain's energy needs. During maintenance of reduced weight, the second set is related to low leptin levels keeping energy expenditure low to prevent triglyceride stores getting too low which is a risk for some basic biological functions (e.g., reproduction). Innovative topics of AT in humans are on its definition and assessment, its dynamics related to weight loss and its constitutional and neuro-endocrine determinants.
Collapse
Affiliation(s)
- Manfred J Müller
- Institute of Human Nutrition and Food Science, Faculty of Agricultural and Nutritional Sciences, University of Kiel, Düsternbrooker Weg 17, D-24105, Kiel, Germany.
| | - Janna Enderle
- Institute of Human Nutrition and Food Science, Faculty of Agricultural and Nutritional Sciences, University of Kiel, Düsternbrooker Weg 17, D-24105, Kiel, Germany
| | - Anja Bosy-Westphal
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| |
Collapse
|
40
|
Metabolic control of immune tolerance in health and autoimmunity. Semin Immunol 2016; 28:491-504. [PMID: 27720234 DOI: 10.1016/j.smim.2016.09.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 09/23/2016] [Accepted: 09/30/2016] [Indexed: 12/15/2022]
Abstract
The filed that links immunity and metabolism is rapidly expanding. The adipose tissue, by secreting a series of immune regulators called adipokines, represents the common mediator linking metabolic processes and immune system functions. The dysregulation of adipokine secretion, occurring in obese individuals or in conditions of malnutrition or dietary restriction, affects the activity of immune cells resulting in inflammatory autoimmune responses or increased susceptibility to infectious diseases. Alterations of cell metabolism that characterize several autoimmune diseases strongly support the idea that the immune tolerance is also regulated by metabolic pathways. The comprehension of the molecular mechanisms underlying these alterations may lead to the development of novel therapeutic strategies to control immune cell differentiation and function in conditions of autoimmunity.
Collapse
|
41
|
Rosenbaum M, Leibel RL. Models of energy homeostasis in response to maintenance of reduced body weight. Obesity (Silver Spring) 2016; 24:1620-9. [PMID: 27460711 PMCID: PMC4965234 DOI: 10.1002/oby.21559] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 04/04/2016] [Accepted: 04/06/2016] [Indexed: 01/07/2023]
Abstract
OBJECTIVE To test three proposed models for adaptive thermogenesis in compartments of energy expenditure following different degrees of weight loss. Specifically, (1) there is no adaptive thermogenesis [constant relationship of energy expenditure (EE) to metabolic mass]. (2) There is a fixed degree of adaptive thermogenesis once fat stores are below a "threshold." (3) The degree of adaptive thermogenesis is proportional to weight loss. METHODS The relationship between weight loss and EE was examined in 17 inpatient subjects with stable weight and obesity studied at usual weight and again following a 10% and a 20% weight loss. RESULTS Following initial weight loss (10%), resting (REE) and non-resting (NREE) EE were significantly below those predicted on the basis of the amount and composition of weight lost. Further reductions below predicted values of NREE but not REE occurred following an additional 10% weight loss. Changes in body weight, composition, and/or energy stores were significantly correlated with changes in EE. CONCLUSIONS All models are applicable to the decline in EE following weight loss. The disproportionate decline in REE is consistent with a threshold model (no change with further weight loss) while the disproportionate decline in NREE is largely reflective of the degree of weight loss.
Collapse
Affiliation(s)
- Michael Rosenbaum
- Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, New York, USA
- Department of Medicine, College of Physicians & Surgeons, Columbia University, New York, New York, USA
| | - Rudolph L Leibel
- Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, New York, USA
- Department of Medicine, College of Physicians & Surgeons, Columbia University, New York, New York, USA
| |
Collapse
|
42
|
Rasmussen EB, Robertson SH, Rodriguez LR. The utility of behavioral economics in expanding the free-feed model of obesity. Behav Processes 2016; 127:25-34. [PMID: 26923097 PMCID: PMC4868767 DOI: 10.1016/j.beproc.2016.02.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 02/18/2016] [Accepted: 02/22/2016] [Indexed: 01/19/2023]
Abstract
Animal models of obesity are numerous and diverse in terms of identifying specific neural and peripheral mechanisms related to obesity; however, they are limited when it comes to behavior. The standard behavioral measure of food intake in most animal models occurs in a free-feeding environment. While easy and cost-effective for the researcher, the free-feeding environment omits some of the most important features of obesity-related food consumption-namely, properties of food availability, such as effort and delay to obtaining food. Behavior economics expands behavioral measures of obesity animal models by identifying such behavioral mechanisms. First, economic demand analysis allows researchers to understand the role of effort in food procurement, and how physiological and neural mechanisms are related. Second, studies on delay discounting contribute to a growing literature that shows that sensitivity to delayed food- and food-related outcomes is likely a fundamental process of obesity. Together, these data expand the animal model in a manner that better characterizes how environmental factors influence food consumption.
Collapse
|
43
|
Bennett BJ, Hall KD, Hu FB, McCartney AL, Roberto C. Nutrition and the science of disease prevention: a systems approach to support metabolic health. Ann N Y Acad Sci 2015; 1352:1-12. [PMID: 26415028 DOI: 10.1111/nyas.12945] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 08/26/2015] [Indexed: 12/31/2022]
Abstract
Progress in nutritional science, genetics, computer science, and behavioral economics can be leveraged to address the challenge of noncommunicable disease. This report highlights the connection between nutrition and the complex science of preventing disease and discusses the promotion of optimal metabolic health, building on input from several complementary disciplines. The discussion focuses on (1) the basic science of optimal metabolic health, including data from gene-diet interactions, microbiome, and epidemiological research in nutrition, with the goal of defining better targets and interventions, and (2) how nutrition, from pharma to lifestyle, can build on systems science to address complex issues.
Collapse
Affiliation(s)
- Brian J Bennett
- Departments of Genetics and Nutrition, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina
| | - Kevin D Hall
- Integrative Physiology Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Frank B Hu
- Departments of Nutrition and Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Anne L McCartney
- Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Christina Roberto
- Departments of Social and Behavioral Sciences and Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
44
|
Wensveen FM, Valentić S, Šestan M, Wensveen TT, Polić B. Interactions between adipose tissue and the immune system in health and malnutrition. Semin Immunol 2015; 27:322-33. [PMID: 26603491 DOI: 10.1016/j.smim.2015.10.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 10/26/2015] [Accepted: 10/27/2015] [Indexed: 02/07/2023]
Abstract
Adipose tissue provides the body with a storage depot of nutrients that is drained during times of starvation and replenished when food sources are abundant. As such, it is the primary sensor for nutrient availability in the milieu of an organism, which it communicates to the body through the excretion of hormones. Adipose tissue regulates a multitude of body functions associated with metabolism, such as gluconeogenesis, feeding and nutrient uptake. The immune system forms a vital layer of protection against micro-organisms that try to gain access to the nutrients contained in the body. Because infections need to be resolved as quickly as possible, speed is favored over energy-efficiency in an immune response. Especially when immune cells are activated, they switch to fast, but energy-inefficient anaerobic respiration to fulfill their energetic needs. Despite the necessity for an effective immune system, it is not given free rein in its energy expenditure. Signals derived from adipose tissue limit immune cell numbers and activity under conditions of nutrient shortage, whereas they allow proper immune cell activity when food sources are sufficiently available. When excessive fat accumulation occurs, such as in diet-induced obesity, adipose tissue becomes the site of pathological immune cell activation, causing chronic low-grade systemic inflammation. Obesity is therefore associated with a number of disorders in which the immune system plays a central role, such as atherosclerosis and non-alcoholic steatohepatitis. In this review, we will discuss the way in which adipose tissue regulates activity of the immune system under healthy and pathological conditions.
Collapse
Affiliation(s)
- Felix M Wensveen
- Department of Histology & Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia; Department of Experimental Immunology, Amsterdam Medical Centre, Amsterdam, The Netherlands
| | - Sonja Valentić
- Department of Histology & Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Marko Šestan
- Department of Histology & Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | | | - Bojan Polić
- Department of Histology & Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.
| |
Collapse
|
45
|
Müller MJ, Enderle J, Pourhassan M, Braun W, Eggeling B, Lagerpusch M, Glüer CC, Kehayias JJ, Kiosz D, Bosy-Westphal A. Metabolic adaptation to caloric restriction and subsequent refeeding: the Minnesota Starvation Experiment revisited. Am J Clin Nutr 2015; 102:807-19. [PMID: 26399868 DOI: 10.3945/ajcn.115.109173] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 07/28/2015] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Adaptive thermogenesis (AT) is the fat-free mass (FFM)-independent reduction of resting energy expenditure (REE) to caloric restriction (CR). AT attenuates weight loss and favors weight regain. Its variance, dynamics, and control remain obscure. OBJECTIVES Our aims were to address the variance and kinetics of AT, its associations with body composition in the context of endocrine determinants, and its effect on weight regain. DESIGN Thirty-two nonobese men underwent sequential overfeeding (1 wk at +50% of energy needs), CR (3 wk at -50% of energy needs), and refeeding (2 wk at +50% of energy needs). AT and its determinants were measured together with body composition as assessed with the use of quantitative magnetic resonance, whole-body MRI, isotope dilution, and nitrogen and fluid balances. RESULTS Changes in body weight were +1.8 kg (overfeeding), -6.0 kg (CR), and +3.5 kg (refeeding). CR reduced fat mass and FFM by 114 and 159 g/d, respectively. Within FFM, skeletal muscle (-5%), liver (-13%), and kidneys (-8%) decreased. CR also led to reductions in REE (-266 kcal/d), respiratory quotient (-15%), heart rate (-14%), blood pressure (-7%), creatinine clearance (-12%), energy cost of walking (-22%), activity of the sympathetic nervous system (SNS) (-38%), and plasma leptin (-44%), insulin (-54%), adiponectin (-49%), 3,5,3'-tri-iodo-thyronine (T3) (-39%), and testosterone (-11%). AT was 108 kcal/d or 48% of the decrease in REE. Changes in FFM composition explained 36 kcal, which left 72 kcal/d for true AT. The decrease in AT became significant at ≤3 d of CR and was related to decreases in insulin secretion (r = 0.92, P < 0.001), heart rate (r = 0.60, P < 0.05), creatinine clearance (r = 0.79, P < 0.05), negative fluid balance (r = 0.51, P < 0.01), and the free water clearance rate (r = -0.90, P < 0.002). SNS activity and plasma leptin, ghrelin, and T3 and their changes with CR were not related to AT. CONCLUSION During early weight loss, AT is associated with a fall in insulin secretion and body fluid balance. This trial was registered at clinicaltrials.gov as NCT01737034.
Collapse
Affiliation(s)
| | | | | | - Wiebke Braun
- Institute of Human Nutrition and Food Science and
| | | | | | - Claus-Christian Glüer
- Clinic of Radiology and Neuroradiology, Biomedical Imaging, Molecular Imaging North Competence Centre, University Medical Center Schleswig Holstein, Christian-Albrechts University, Kiel, Germany
| | - Joseph J Kehayias
- the Body Composition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA; and
| | - Dieter Kiosz
- Institute of Human Nutrition and Food Science and
| | - Anja Bosy-Westphal
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| |
Collapse
|
46
|
Insensitivity of well-conditioned mature sheep to central administration of a leptin receptor antagonist. Animal 2015. [PMID: 26220331 DOI: 10.1017/s1751731115001159] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Ruminants remain productive during the energy insufficiency of late pregnancy or early lactation by evoking metabolic adaptations sparing available energy and nutrients (e.g. higher metabolic efficiency and induction of insulin resistance). A deficit in central leptin signaling triggers these adaptations in rodents but whether it does in ruminants remains unclear. To address this issue, five mature ewes were implanted with intracerebroventricular (ICV) cannula in the third ventricle. They were used in two experiments with an ovine leptin antagonist (OLA) when well-conditioned (average body condition score of 3.7 on a 5 point scale). The first experiment tested the ability of OLA to antagonize leptin under in vivo conditions. Ewes received continuous ICV infusion of artificial cerebrospinal fluid (aCSF), ovine leptin (4 µg/h) or the combination of ovine leptin (4 µg/h) and its mutant version OLA (40 µg/h) for 48 h. Dry matter intake (DMI) was measured every day and blood samples were collected on the last day of infusion. ICV infusion of leptin reduced DMI by 24% (P < 0.05), and this effect was completely abolished by OLA co-infusion. A second experiment tested whether a reduction in endogenous leptin signaling in the brain triggers metabolic adaptations. This involved continuous ICV infusions of aCSF or OLA alone (40 µg/h) for 4 consecutive days. The infusion of OLA did not alter voluntary DMI over the treatment period or on any individual day. OLA did not affect plasma variables indicative of insulin action (glucose, non-esterified fatty acids, insulin and the disposition of plasma glucose during an insulin tolerance test) or plasma cortisol, but tended to reduce plasma triiodothyronine and thyroxine (P < 0.07). Overall, these data show that a reduction of central leptin signaling has little impact on insulin action in well-conditioned mature sheep. They also raise the possibility that reduced central leptin signaling plays a role in controlling thyroid hormone production.
Collapse
|
47
|
Yu YH, Vasselli JR, Zhang Y, Mechanick JI, Korner J, Peterli R. Metabolic vs. hedonic obesity: a conceptual distinction and its clinical implications. Obes Rev 2015; 16:234-47. [PMID: 25588316 PMCID: PMC5053237 DOI: 10.1111/obr.12246] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 11/19/2014] [Accepted: 11/19/2014] [Indexed: 01/01/2023]
Abstract
Body weight is determined via both metabolic and hedonic mechanisms. Metabolic regulation of body weight centres around the 'body weight set point', which is programmed by energy balance circuitry in the hypothalamus and other specific brain regions. The metabolic body weight set point has a genetic basis, but exposure to an obesogenic environment may elicit allostatic responses and upward drift of the set point, leading to a higher maintained body weight. However, an elevated steady-state body weight may also be achieved without an alteration of the metabolic set point, via sustained hedonic over-eating, which is governed by the reward system of the brain and can override homeostatic metabolic signals. While hedonic signals are potent influences in determining food intake, metabolic regulation involves the active control of both food intake and energy expenditure. When overweight is due to elevation of the metabolic set point ('metabolic obesity'), energy expenditure theoretically falls onto the standard energy-mass regression line. In contrast, when a steady-state weight is above the metabolic set point due to hedonic over-eating ('hedonic obesity'), a persistent compensatory increase in energy expenditure per unit metabolic mass may be demonstrable. Recognition of the two types of obesity may lead to more effective treatment and prevention of obesity.
Collapse
Affiliation(s)
- Y-H Yu
- Weight Loss and Diabetes Center, Greenwich Hospital, Greenwich, CT, USA; Endocrinology Associates of Greenwich, Northeast Medical Group, Yale New-Haven Health System, Greenwich, CT, USA
| | | | | | | | | | | |
Collapse
|
48
|
Calorie restriction-mediated restoration of hypothalamic signal transducer and activator of transcription 3 (STAT3) phosphorylation is not effective for lowering the body weight set point in IRS-2 knockout obese mice. Diabetol Int 2015. [DOI: 10.1007/s13340-015-0205-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
49
|
Nystrom FH, Quon MJ. Man shall not live by bread alone. Nutrition 2014; 31:244-7. [PMID: 25466671 DOI: 10.1016/j.nut.2014.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 10/22/2014] [Indexed: 10/24/2022]
Affiliation(s)
- Fredrik H Nystrom
- Department of Medical and Health Sciences, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Michael J Quon
- Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
50
|
Abstract
Mammals regulate fat mass so that increases or reductions in adipose tissue mass activate responses that favor return to one's previous weight. A reduction in fat mass activates a system that increases food intake and reduces energy expenditure; conversely, overfeeding and rapid adipose tissue expansion reduces food intake and increases energy expenditure. With the identification of leptin nearly two decades ago, the central circuit that defends against reductions in body fat was revealed. However, the systems that defend against rapid expansion of fat mass remain largely unknown. Here we review the physiology of the overfed state and evidence for a distinct regulatory system, which unlike the leptin-mediated system, we propose primarily measures a functional aspect of adipose tissue and not total mass per se.
Collapse
Affiliation(s)
- Yann Ravussin
- Department of Medicine, Columbia University, 1150 St. Nicholas Ave, New York, NY 10032, USA; Naomi Berrie Diabetes Center, Columbia University, 1150 St. Nicholas Ave, New York, NY 10032, USA
| | - Rudolph L Leibel
- Naomi Berrie Diabetes Center, Columbia University, 1150 St. Nicholas Ave, New York, NY 10032, USA; Department of Pediatrics, Columbia University, 1150 St. Nicholas Ave, New York, NY 10032, USA
| | - Anthony W Ferrante
- Department of Medicine, Columbia University, 1150 St. Nicholas Ave, New York, NY 10032, USA; Naomi Berrie Diabetes Center, Columbia University, 1150 St. Nicholas Ave, New York, NY 10032, USA.
| |
Collapse
|