1
|
Friedman MI, Sørensen TIA, Taubes G, Lund J, Ludwig DS. Trapped fat: Obesity pathogenesis as an intrinsic disorder in metabolic fuel partitioning. Obes Rev 2024; 25:e13795. [PMID: 38961319 DOI: 10.1111/obr.13795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 05/24/2024] [Accepted: 06/13/2024] [Indexed: 07/05/2024]
Abstract
Our understanding of the pathophysiology of obesity remains at best incomplete despite a century of research. During this time, two alternative perspectives have helped shape thinking about the etiology of the disorder. The currently prevailing view holds that excessive fat accumulation results because energy intake exceeds energy expenditure, with excessive food consumption being the primary cause of the imbalance. The other perspective attributes the initiating cause of obesity to intrinsic metabolic defects that shift fuel partitioning from pathways for mobilization and oxidation to those for synthesis and storage. The resulting reduction in fuel oxidation and trapping of energy in adipose tissue drives a compensatory increase in energy intake and, under some conditions, a decrease in expenditure. This theory of obesity pathogenesis has historically garnered relatively less attention despite its pedigree. Here, we present an updated comprehensive formulation of the fuel partitioning theory, focused on evidence gathered over the last 80 years from major animal models of obesity showing a redirection of fuel fluxes from oxidation to storage and accumulation of excess body fat with energy intake equal to or even less than that of lean animals. The aim is to inform current discussions about the etiology of obesity and by so doing, help lay new foundations for the design of more efficacious approaches to obesity research, treatment and prevention.
Collapse
Affiliation(s)
| | - Thorkild I A Sørensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
- Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
- Center for Childhood Health, Copenhagen, Denmark
| | | | - Jens Lund
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - David S Ludwig
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Denmark
| |
Collapse
|
2
|
Burak MF, Stanley TL, Lawson EA, Campbell SL, Lynch L, Hasty AH, Domingos AI, Dixit VD, Hotamışlıgil GS, Sheedy FJ, Dixon AE, Brinkley TE, Hill JA, Donath MY, Grinspoon SK. Adiposity, immunity, and inflammation: interrelationships in health and disease: a report from 24th Annual Harvard Nutrition Obesity Symposium, June 2023. Am J Clin Nutr 2024; 120:257-268. [PMID: 38705359 PMCID: PMC11347817 DOI: 10.1016/j.ajcnut.2024.04.029] [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: 02/07/2024] [Revised: 04/09/2024] [Accepted: 04/29/2024] [Indexed: 05/07/2024] Open
Abstract
The rapidly evolving field of immunometabolism explores how changes in local immune environments may affect key metabolic and cellular processes, including that of adipose tissue. Importantly, these changes may contribute to low-grade systemic inflammation. In turn, chronic low-grade inflammation affecting adipose tissue may exacerbate the outcome of metabolic diseases. Novel advances in our understanding of immunometabolic processes may critically lead to interventions to reduce disease severity and progression. An important example in this regard relates to obesity, which has a multifaceted effect on immunity, activating the proinflammatory pathways such as the inflammasome and disrupting cellular homeostasis. This multifaceted effect of obesity can be investigated through study of downstream conditions using cellular and systemic investigative techniques. To further explore this field, the National Institutes of Health P30 Nutrition Obesity Research Center at Harvard, in partnership with Harvard Medical School, assembled experts to present at its 24th Annual Symposium entitled "Adiposity, Immunity, and Inflammation: Interrelationships in Health and Disease" on 7 June, 2023. This manuscript seeks to synthesize and present key findings from the symposium, highlighting new research and novel disease-specific advances in the field. Better understanding the interaction between metabolism and immunity offers promising preventative and treatment therapies for obesity-related immunometabolic diseases.
Collapse
Affiliation(s)
- Mehmet Furkan Burak
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States; Department of Molecular Metabolism and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA, United States.
| | - Takara L Stanley
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States; Division of Pediatric Endocrinology, Massachusetts General Hospital for Children and Harvard Medical School, Boston, MA, United States
| | - Elizabeth A Lawson
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Sophia L Campbell
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Lydia Lynch
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, VA Tennessee Valley Healthcare System, Nashville, TN, United States
| | - Ana I Domingos
- Department of Physiology, Anatomy & Genetics, Oxford University, Oxford, United Kingdom
| | - Vishwa D Dixit
- Department of Pathology, Department of Comparative Medicine, Department of Immunobiology, Yale School of Medicine, and Yale Center for Research on Aging, New Haven, CT, United States
| | - Gökhan S Hotamışlıgil
- Department of Molecular Metabolism and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Broad Institute of Harvard and MIT, Cambridge, MA, United States
| | - Frederick J Sheedy
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| | - Anne E Dixon
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| | - Tina E Brinkley
- Department of Internal Medicine, Section of Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Joseph A Hill
- Division of Cardiology, Department of Internal Medicine, Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Marc Y Donath
- Department of Biomedicine, University of Basel, Basel, Switzerland; Clinic of Endocrinology, Diabetes & Metabolism, University Hospital Basel, Basel, Switzerland
| | - Steven K Grinspoon
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
3
|
Rathod YD, Abdelgawad R, Hübner CA, Di Fulvio M. Slc12a2 loss in insulin-secreting β-cells links development of overweight and metabolic dysregulation to impaired satiation control of feeding. Am J Physiol Endocrinol Metab 2023; 325:E581-E594. [PMID: 37819196 PMCID: PMC10864024 DOI: 10.1152/ajpendo.00197.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/02/2023] [Accepted: 10/02/2023] [Indexed: 10/13/2023]
Abstract
Male mice lacking the Na+-K+-2Cl- cotransporter Slc12a2 (Nkcc1) specifically in insulin-secreting β-cells (Slc12a2βKO) have reduced β-cell mass and mild β-cell secretory dysfunction associated with overweight, glucose intolerance, insulin resistance, and metabolic abnormalities. Here, we confirmed and extended previous results to female Slc12a2βKO mice, which developed a similar metabolic syndrome-like phenotype as males, albeit milder. Notably, male and female Slc12a2βKO mice developed overweight without consuming excess calories. Analysis of the feeding microstructure revealed that young lean Slc12a2βKO male mice ate meals of higher caloric content and at a relatively lower frequency than normal mice, particularly during the night. In addition, overweight Slc12a2βKO mice consumed significantly larger meals than lean mice. Therefore, the reduced satiation control of feeding precedes the onset of overweight and is worsened in older Slc12a2βKO mice. However, the time spent between meals remained intact in lean and overweight Slc12a2βKO mice, indicating conserved satiety responses to ad libitum feeding. Nevertheless, satiety was intensified during and after refeeding only in overweight males. In lean females, satiety responses to refeeding were delayed relative to age- and body weight-matched control mice but normalized in overweight mice. Since meal size did not change during refeeding, these data suggested that the satiety control of eating after fasting is impaired in lean Slc12a2βKO mice before the onset of overweight and independently of their reduced satiation responses. Therefore, our results support the novel hypothesis that reduced satiation precedes the onset of overweight and the development of metabolic dysregulation.NEW & NOTEWORTHY Obesity, defined as excess fat accumulation, increases the absolute risk for metabolic diseases. Although obesity is usually attributed to increased food intake, we demonstrate that body weight gain can be hastened without consuming excess calories. In fact, impaired meal termination control, i.e., satiation, is detectable before the development of overweight in an animal model that develops a metabolic syndrome-like phenotype.
Collapse
Affiliation(s)
- Yakshkumar Dilipbhai Rathod
- Department of Pharmacology and Toxicology, School of Medicine Dayton, Wright State University, Ohio, United States
| | - Rana Abdelgawad
- Department of Pharmacology and Toxicology, School of Medicine Dayton, Wright State University, Ohio, United States
| | - Christian A Hübner
- Institut für Humangenetik Am Klinikum 1, Universitätsklinikum Jena, Jena, Germany
| | - Mauricio Di Fulvio
- Department of Pharmacology and Toxicology, School of Medicine Dayton, Wright State University, Ohio, United States
| |
Collapse
|
4
|
Chen H, Zhang H, Jia T, Wang Z, Zhu W. Roles of leptin on energy balance and thermoregulation in Eothenomys miletus. Front Physiol 2022; 13:1054107. [PMID: 36589465 PMCID: PMC9800980 DOI: 10.3389/fphys.2022.1054107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
Leptin is a hormone mainly synthesized and secreted by white adipose tissue (WAT), which regulates various physiological processes. To investigate the role of leptin in energy balance and thermoregulation in Eothenomys miletus, voles were randomly divided into leptin-injected and PBS-injected groups and placed at 25°C ± 1°C with a photoperiod of 12 L:12 D. They were housed under laboratory conditions for 28 days and compared in terms of body mass, food intake, water intake, core body temperature, interscapular skin temperature, resting metabolic rate (RMR), nonshivering thermogenesis (NST), liver and brown adipose tissue (BAT) thermogenic activity, and serum hormone levels. The results showed that leptin injection decreased body mass, body fat, food intake, and water intake. But it had no significant effect on carcass protein. Leptin injection increased core body temperature, interscapular skin temperature, resting metabolic rate, non-shivering thermogenesis, mitochondrial protein content and cytochrome C oxidase (COX) activity in liver and brown adipose tissue, uncoupling protein 1 (UCP1) content and thyroxin 5'-deiodinase (T45'-DII) activity in brown adipose tissue significantly. Serum leptin, triiodothyronine (T3), thyrotropin-releasing hormone (TRH) and corticotropin-releasing hormone (CRH) concentrations were also increased significantly. Correlation analysis showed that serum leptin levels were positively correlated with core body temperature, body mass loss, uncoupling protein 1 content, thyroxin 5'-deiodinase activity, nonshivering thermogenesis, and negatively correlated with food intake; thyroxin 5'-deiodinase and triiodothyronine levels were positively correlated, suggesting that thyroxin 5'-deiodinase may play an important role in leptin-induced thermogenesis in brown adipose tissue. In conclusion, our study shows that exogenous leptin is involved in the regulation of energy metabolism and thermoregulation in E. miletus, and thyroid hormone may play an important role in the process of leptin regulating energy balance in E. miletus.
Collapse
Affiliation(s)
- Huibao Chen
- Key Laboratory of Ecological Adaptive Evolution and Conservation on Animals-plants in Southwest Mountain Ecosystem of Yunnan Province Higher Institutes College, School of Life Sciences, Yunnan Normal University, Kunming, China
| | - Hao Zhang
- Key Laboratory of Ecological Adaptive Evolution and Conservation on Animals-plants in Southwest Mountain Ecosystem of Yunnan Province Higher Institutes College, School of Life Sciences, Yunnan Normal University, Kunming, China
| | - Ting Jia
- Yunnan College of Business Management, Kunming, China
| | - Zhengkun Wang
- Key Laboratory of Ecological Adaptive Evolution and Conservation on Animals-plants in Southwest Mountain Ecosystem of Yunnan Province Higher Institutes College, School of Life Sciences, Yunnan Normal University, Kunming, China
| | - Wanlong Zhu
- Key Laboratory of Ecological Adaptive Evolution and Conservation on Animals-plants in Southwest Mountain Ecosystem of Yunnan Province Higher Institutes College, School of Life Sciences, Yunnan Normal University, Kunming, China
- Engineering Research Center of Sustainable Development and Utilization of Biomass Energy Ministry of Education, Kunming, China
- Key Laboratory of Yunnan Province for Biomass Energy and Environment Biotechnology, Kunming, China
| |
Collapse
|
5
|
dos Santos KC, Olofsson C, Cunha JPMCM, Roberts F, Catrina S, Fex M, Ekberg NR, Spégel P. The impact of macronutrient composition on metabolic regulation: An Islet-Centric view. Acta Physiol (Oxf) 2022; 236:e13884. [PMID: 36056607 PMCID: PMC9787959 DOI: 10.1111/apha.13884] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 01/29/2023]
Abstract
AIM The influence of dietary carbohydrates and fats on weight gain is inconclusively understood. We studied the acute impact of these nutrients on the overall metabolic state utilizing the insulin:glucagon ratio (IGR). METHODS Following in vitro glucose and palmitate treatment, insulin and glucagon secretion from islets isolated from C57Bl/6J mice was measured. Our human in vivo study included 21 normoglycaemia (mean age 51.9 ± 16.5 years, BMI 23.9 ± 3.5 kg/m2 , and HbA1c 36.9 ± 3.3 mmol/mol) and 20 type 2 diabetes (T2D) diagnosed individuals (duration 12 ± 7 years, mean age 63.6 ± 4.5 years, BMI 29.1 ± 2.4 kg/m2 , and HbA1c 52.3 ± 9.5 mmol/mol). Individuals consumed a carbohydrate-rich or fat-rich meal (600 kcal) in a cross-over design. Plasma insulin and glucagon levels were measured at -30, -5, and 0 min, and every 30 min until 240 min after meal ingestion. RESULTS The IGR measured from mouse islets was determined solely by glucose levels. The palmitate-stimulated hormone secretion was largely glucose independent in the analysed mouse islets. The acute meal tolerance test demonstrated that insulin and glucagon secretion is dependent on glycaemic status and meal composition, whereas the IGR was dependent upon meal composition. The relative reduction in IGR elicited by the fat-rich meal was more pronounced in obese individuals. This effect was blunted in T2D individuals with elevated HbA1c levels. CONCLUSION The metabolic state in normoglycaemic individuals and T2D-diagnosed individuals is regulated by glucose. We demonstrate that consumption of a low carbohydrate diet, eliciting a catabolic state, may be beneficial for weight loss, particularly in obese individuals.
Collapse
Affiliation(s)
- Klinsmann Carolo dos Santos
- Centre for Analysis and Synthesis, Department of ChemistryLund UniversityLundSweden,Unit of Molecular Metabolism, Department of Clinical Sciences in MalmöLund UniversityMalmöSweden
| | - Camilla Olofsson
- Department of Molecular Medicine and Surgery, Karolinska University HospitalKarolinska InstituteStockholmSweden
| | | | - Fiona Roberts
- Unit of Molecular Metabolism, Department of Clinical Sciences in MalmöLund UniversityMalmöSweden
| | - Sergiu‐Bogdan Catrina
- Department of Molecular Medicine and Surgery, Karolinska University HospitalKarolinska InstituteStockholmSweden,Centrum for DiabetesAcademic Specialist CentrumStockholmSweden
| | - Malin Fex
- Unit of Molecular Metabolism, Department of Clinical Sciences in MalmöLund UniversityMalmöSweden
| | - Neda Rajamand Ekberg
- Department of Molecular Medicine and Surgery, Karolinska University HospitalKarolinska InstituteStockholmSweden,Centrum for DiabetesAcademic Specialist CentrumStockholmSweden
| | - Peter Spégel
- Centre for Analysis and Synthesis, Department of ChemistryLund UniversityLundSweden
| |
Collapse
|
6
|
Martinez-Sanchez N, Sweeney O, Sidarta-Oliveira D, Caron A, Stanley SA, Domingos AI. The sympathetic nervous system in the 21st century: Neuroimmune interactions in metabolic homeostasis and obesity. Neuron 2022; 110:3597-3626. [PMID: 36327900 PMCID: PMC9986959 DOI: 10.1016/j.neuron.2022.10.017] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 08/23/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
The sympathetic nervous system maintains metabolic homeostasis by orchestrating the activity of organs such as the pancreas, liver, and white and brown adipose tissues. From the first renderings by Thomas Willis to contemporary techniques for visualization, tracing, and functional probing of axonal arborizations within organs, our understanding of the sympathetic nervous system has started to grow beyond classical models. In the present review, we outline the evolution of these findings and provide updated neuroanatomical maps of sympathetic innervation. We offer an autonomic framework for the neuroendocrine loop of leptin action, and we discuss the role of immune cells in regulating sympathetic terminals and metabolism. We highlight potential anti-obesity therapeutic approaches that emerge from the modern appreciation of SNS as a neural network vis a vis the historical fear of sympathomimetic pharmacology, while shifting focus from post- to pre-synaptic targeting. Finally, we critically appraise the field and where it needs to go.
Collapse
Affiliation(s)
| | - Owen Sweeney
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Davi Sidarta-Oliveira
- Physician-Scientist Graduate Program, Obesity and Comorbidities Research Center, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Alexandre Caron
- Faculty of Pharmacy, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Sarah A Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ana I Domingos
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK.
| |
Collapse
|
7
|
Glenny EM, Coleman MF, Giles ED, Wellberg EA, Hursting SD. Designing Relevant Preclinical Rodent Models for Studying Links Between Nutrition, Obesity, Metabolism, and Cancer. Annu Rev Nutr 2021; 41:253-282. [PMID: 34357792 PMCID: PMC8900211 DOI: 10.1146/annurev-nutr-120420-032437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Diet and nutrition are intricately related to cancer prevention, growth, and treatment response. Preclinical rodent models are a cornerstone to biomedical research and remain instrumental in our understanding of the relationship between cancer and diet and in the development of effective therapeutics. However, the success rate of translating promising findings from the bench to the bedside is suboptimal. Well-designed rodent models will be crucial to improving the impact basic science has on clinical treatment options. This review discusses essential experimental factors to consider when designing a preclinical cancer model with an emphasis on incorporatingthese models into studies interrogating diet, nutrition, and metabolism. The aims of this review are to (a) provide insight into relevant considerations when designing cancer models for obesity, nutrition, and metabolism research; (b) identify common pitfalls when selecting a rodent model; and (c) discuss strengths and limitations of available preclinical models.
Collapse
Affiliation(s)
- Elaine M Glenny
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA;
| | - Michael F Coleman
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA;
| | - Erin D Giles
- Department of Nutrition, Texas A&M University, College Station, Texas 77843, USA
| | - Elizabeth A Wellberg
- Department of Pathology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma 73104, USA
| | - Stephen D Hursting
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA;
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina 28081, USA
| |
Collapse
|
8
|
O'Brien CJO, Haberman ER, Domingos AI. A Tale of Three Systems: Toward a Neuroimmunoendocrine Model of Obesity. Annu Rev Cell Dev Biol 2021; 37:549-573. [PMID: 34613819 PMCID: PMC7614880 DOI: 10.1146/annurev-cellbio-120319-114106] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The prevalence of obesity is on the rise. What was once considered a simple disease of energy imbalance is now recognized as a complex condition perpetuated by neuro- and immunopathologies. In this review, we summarize the current knowledge of the neuroimmunoendocrine mechanisms underlying obesity. We examine the pleiotropic effects of leptin action in addition to its established role in the modulation of appetite, and we discuss the neural circuitry mediating leptin action and how this is altered with obesity, both centrally (leptin resistance) and in adipose tissues (sympathetic neuropathy). Finally, we dissect the numerous causal and consequential roles of adipose tissue macrophages in obesity and highlight recent key studies demonstrating their direct role in organismal energy homeostasis.
Collapse
Affiliation(s)
- Conan J O O'Brien
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, United Kingdom;
| | - Emma R Haberman
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, United Kingdom;
| | - Ana I Domingos
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, United Kingdom;
| |
Collapse
|
9
|
Isoda M, Ebihara K, Sawayama N, Murakami A, Ebihara C, Shibuya K, Takei A, Takei S, Wakabayashi T, Yamamuro D, Takahashi M, Nagashima S, Ishibashi S. Leptin sensitizing effect of 1,3-butanediol and its potential mechanism. Sci Rep 2021; 11:17691. [PMID: 34489483 PMCID: PMC8421515 DOI: 10.1038/s41598-021-96460-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 08/09/2021] [Indexed: 12/20/2022] Open
Abstract
Leptin is an adipocyte-derived hormone that regulates appetite and energy expenditure via the hypothalamus. Since the majority of obese subjects are leptin resistant, leptin sensitizers, rather than leptin itself, are expected to be anti-obesity drugs. Endoplasmic reticulum (ER) stress in the hypothalamus plays a key role in the pathogenesis of leptin resistance. ATP-deficient cells are vulnerable to ER stress and ATP treatment protects cells against ER stress. Thus, we investigated the therapeutic effects of oral 1,3-butanediol (BD) administration, which increases plasma β-hydroxybutyrate and hypothalamic ATP concentrations, in diet induced obese (DIO) mice with leptin resistance. BD treatment effectively decreased food intake and body weight in DIO mice. In contrast, BD treatment had no effect in leptin deficient ob/ob mice. Co-administration experiment demonstrated that BD treatment sensitizes leptin action in both DIO and ob/ob mice. We also demonstrated that BD treatment attenuates ER stress and leptin resistance at the hypothalamus level. This is the first report to confirm the leptin sensitizing effect of BD treatment in leptin resistant DIO mice. The present study provides collateral evidence suggesting that the effect of BD treatment is mediated by the elevation of hypothalamic ATP concentration. Ketone bodies and hypothalamic ATP are the potential target for the treatment of obesity and its complications.
Collapse
Affiliation(s)
- Masayo Isoda
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University School of Medicine, Yakushiji 3311-1, Shimotsuke, Tochigi, 329-0498, Japan
| | - Ken Ebihara
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University School of Medicine, Yakushiji 3311-1, Shimotsuke, Tochigi, 329-0498, Japan.
| | - Nagisa Sawayama
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University School of Medicine, Yakushiji 3311-1, Shimotsuke, Tochigi, 329-0498, Japan
| | - Akiko Murakami
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University School of Medicine, Yakushiji 3311-1, Shimotsuke, Tochigi, 329-0498, Japan
| | - Chihiro Ebihara
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University School of Medicine, Yakushiji 3311-1, Shimotsuke, Tochigi, 329-0498, Japan
| | - Koji Shibuya
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University School of Medicine, Yakushiji 3311-1, Shimotsuke, Tochigi, 329-0498, Japan
| | - Akihito Takei
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University School of Medicine, Yakushiji 3311-1, Shimotsuke, Tochigi, 329-0498, Japan
| | - Shoko Takei
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University School of Medicine, Yakushiji 3311-1, Shimotsuke, Tochigi, 329-0498, Japan
| | - Tetsuji Wakabayashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University School of Medicine, Yakushiji 3311-1, Shimotsuke, Tochigi, 329-0498, Japan
| | - Daisuke Yamamuro
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University School of Medicine, Yakushiji 3311-1, Shimotsuke, Tochigi, 329-0498, Japan
| | - Manabu Takahashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University School of Medicine, Yakushiji 3311-1, Shimotsuke, Tochigi, 329-0498, Japan
| | - Shuichi Nagashima
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University School of Medicine, Yakushiji 3311-1, Shimotsuke, Tochigi, 329-0498, Japan
| | - Shun Ishibashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University School of Medicine, Yakushiji 3311-1, Shimotsuke, Tochigi, 329-0498, Japan
| |
Collapse
|
10
|
Deshpande SSS, Nemani H, Balasinor NH. High fat diet-induced- and genetically inherited- obesity differential alters DNA demethylation pathways in the germline of adult male rats. Reprod Biol 2021; 21:100532. [PMID: 34246869 DOI: 10.1016/j.repbio.2021.100532] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 06/11/2021] [Accepted: 06/29/2021] [Indexed: 11/17/2022]
Abstract
Obesity is a multifactorial condition with predominantly genetic and environmental causes and is an emerging risk factor for male infertility/subfertility. Epigenetic mechanisms are vulnerable to genetic and environmental changes. Our earlier studies have shown differential effects of genetically inherited (GIO) - and diet-induced- obesity (DIO) on DNA methylation in male germline. Contrary to DNA methylation is DNA demethylation, which also regulates the gene expression by activating transcription. The present study aimed to delineate the effects of obesity on the DNA demethylation pathway using two rat models: GIO (WNIN/Ob) and DIO (high-fat diet). We observed differential alterations in enzymes involved in DNA demethylation by oxidation (Tet1-3) pathway in testis in both groups. An increase in Tets in DIO group and a decrease in GIO group were noted. Analysis of oxidation pathway intermediates (5-hmC, 5-fC, and 5-caC) did not show any effect on testis in DIO group but an increase in 5-hmC and decrease in 5-caC levels in GIO group was observed. Analysis of transcript levels of enzymes related to deamination pathway in testis showed an increase (Gadd45a, Aicda, and Tdg) in DIO group and a decrease (Gadd45a, Aicda, and Tdg) in GIO group. Also, 5-hmC levels were differentially altered in the spermatozoa of both groups without any changes in Tet enzyme levels. These findings highlight differences in effects of GIO and DIO on DNA demethylation mechanisms in male germline, which could be due to differences in endocrine and metabolic profile as well as white fat distribution observed earlier in two groups.
Collapse
Affiliation(s)
- Sharvari S S Deshpande
- Department of Neuroendocrinology, ICMR-National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400012, India
| | - Harishankar Nemani
- National Institute of Nutrition Animal Facility, ICMR-National Institute of Nutrition, Jamai-Osmania PO, Hyderabad, 500 007, India
| | - Nafisa H Balasinor
- Department of Neuroendocrinology, ICMR-National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400012, India.
| |
Collapse
|
11
|
Deshpande SS, Nemani H, Arumugam G, Ravichandran A, Balasinor NH. High-fat diet-induced and genetically inherited obesity differentially alters DNA methylation profile in the germline of adult male rats. Clin Epigenetics 2020; 12:179. [PMID: 33213487 PMCID: PMC7678167 DOI: 10.1186/s13148-020-00974-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 11/10/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Paternal obesity has been associated with reduced live birth rates. It could lead to inheritance of metabolic disturbances to the offspring through epigenetic mechanisms. However, obesity is a multifactorial disorder with genetic or environmental causes. Earlier we had demonstrated differential effects of high-fat diet-induced obesity (DIO) and genetically inherited obesity (GIO) on metabolic, hormonal profile, male fertility, and spermatogenesis using two rat models. The present study aimed to understand the effect of DIO and GIO on DNA methylation in male germline, and its subsequent effects on the resorbed (post-implantation embryo loss) and normal embryos. First, we assessed the DNA methylation enzymatic machinery in the testis by Real-Time PCR, followed global DNA methylation levels in spermatozoa and testicular cells by ELISA and flow cytometry, respectively. Further, we performed Methylation Sequencing in spermatozoa for both the groups. Sequencing data in spermatozoa from both the groups were validated using Pyrosequencing. Expression of the differentially methylated genes was assessed in the resorbed and normal embryos sired by the DIO group using Real-Time PCR for functional validation. RESULTS We noted a significant decrease in Dnmt transcript and global DNA methylation levels in the DIO group and an increase in the GIO group. Sequencing analysis showed 16,966 and 9113 differentially methylated regions in the spermatozoa of the DIO and GIO groups, respectively. Upon pathway analysis, we observed genes enriched in pathways involved in embryo growth and development namely Wnt, Hedgehog, TGF-beta, and Notch in spermatozoa for both the groups, the methylation status of which partially correlated with the gene expression pattern in resorbed and normal embryos sired by the DIO group. CONCLUSION Our study reports the mechanism by which diet-induced and genetically inherited obesity causes differential effects on the DNA methylation in the male germline that could be due to a difference in the white adipose tissue accumulation. These differences could either lead to embryo loss or transmit obesity-related traits to the offspring in adult life.
Collapse
Affiliation(s)
- Sharvari S. Deshpande
- Department of Neuroendocrinology, ICMR-National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai 400012 India
| | - Harishankar Nemani
- National Institute of Nutrition Animal Facility, ICMR-National Institute of Nutrition, Jamai-Osmania PO, Hyderabad 500 007 India
| | - Gandhimathi Arumugam
- Genome Informatics Department, Genotypic Technologies Pvt. Ltd., #2/13, Balaji Complex, Poojari Layout, 80 Feet Road, R.M.V. 2nd stage, Bengaluru, India
| | - Avinash Ravichandran
- Genome Informatics Department, Genotypic Technologies Pvt. Ltd., #2/13, Balaji Complex, Poojari Layout, 80 Feet Road, R.M.V. 2nd stage, Bengaluru, India
| | - Nafisa H. Balasinor
- Department of Neuroendocrinology, ICMR-National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai 400012 India
| |
Collapse
|
12
|
Huang W, Queen NJ, McMurphy TB, Ali S, Cao L. Adipose PTEN regulates adult adipose tissue homeostasis and redistribution via a PTEN-leptin-sympathetic loop. Mol Metab 2019; 30:48-60. [PMID: 31767180 PMCID: PMC6812328 DOI: 10.1016/j.molmet.2019.09.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 09/20/2019] [Accepted: 09/22/2019] [Indexed: 01/19/2023] Open
Abstract
OBJECTIVE Despite the large body of work describing the tumor suppressor functions of Phosphatase and tensin homologue deleted on chromosome ten (PTEN), its roles in adipose homeostasis of adult animals are not yet fully understood. Here, we sought to determine the role of PTEN in whole-body adipose homeostasis. METHODS We genetically manipulated PTEN in specific fat depots through recombinant adeno-associated viral vector (rAAV)-based gene transfer of Cre recombinase to adult PTENflox mice. Additionally, we used a denervation agent, 6OHDA, to assess the role of sympathetic signaling in PTEN-related adipose remodeling. Furthermore, we chemically manipulated AKT signaling via a pan-AKT inhibitor, MK-2206, to assess the role of AKT in PTEN-related adipose remodeling. Finally, to understand the role of leptin and central signaling on peripheral tissues, we knocked down hypothalamic leptin receptor with a microRNA delivered by a rAAV vector. RESULTS Knockdown PTEN in individual fat depot resulted in massive expansion of the affected fat depot through activation of AKT signaling associated with suppression of lipolysis and induction of leptin. This hypertrophic expansion of the affected fat depot led to upregulation of PTEN level, higher lipolysis, and induction of white fat browning in other fat depots, and the compensatory reduced fat mass to maintain a set point of whole-body adiposity. Administration of AKT inhibitor MK-2206 prevented the adipose PTEN knockdown-associated effects. 6OHDA-mediated denervation demonstrated that sympathetic innervation was required for the PTEN knockdown-induced adipose redistribution. Knockdown hypothalamic leptin receptor attenuated the adipose redistribution induced by PTEN deficiency in individual fat depot. CONCLUSIONS Our results demonstrate the essential role of PTEN in adipose homeostasis, including mass and distribution in adulthood, and reveal an "adipose PTEN-leptin-sympathetic nervous system" feedback loop to maintain a set point of adipose PTEN and whole-body adiposity.
Collapse
Affiliation(s)
- Wei Huang
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA; The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Nicholas J Queen
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA; The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Travis B McMurphy
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA; The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Seemaab Ali
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA; The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Lei Cao
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA; The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA.
| |
Collapse
|
13
|
Yamada R, Odamaki S, Araki M, Watanabe T, Matsuo K, Uchida K, Kato T, Ozaki-Masuzawa Y, Takenaka A. Dietary protein restriction increases hepatic leptin receptor mRNA and plasma soluble leptin receptor in male rodents. PLoS One 2019; 14:e0219603. [PMID: 31306448 PMCID: PMC6629078 DOI: 10.1371/journal.pone.0219603] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 06/27/2019] [Indexed: 12/25/2022] Open
Abstract
Leptin is an adipokine that regulates adipose tissue mass through membrane-anchored leptin receptor (Ob-R). Extracellular domain of Ob-R in plasma is called soluble leptin receptor (sOb-R), and is the main leptin-binding protein. Based on a previous DNA microarray analysis that showed induction of hepatic Ob-R mRNA in low-protein diet-fed mice, this study aimed to clarify the effect of dietary protein restriction on hepatic Ob-R mRNA and plasma sOb-R levels. First, the effect of protein restriction on hepatic Ob-R mRNA level was examined together with fasting and food restriction using male rats as common experimental model for nutritional research. Hepatic Ob-R mRNA level was increased by feeding low-protein diet for 7 d, although not significantly influenced by 12-h fasting and sixty percent restriction in food consumption. Then, effect of protein restriction on liver Ob-R and plasma sOb-R was investigated using male mice because specific sOb-R ELISA was more available for mice. Hepatic Ob-R mRNA level was also increased in protein restricted-mice although it did not increase in hypothalamus. Hepatic Ob-R protein was decreased, whereas plasma sOb-R was increased by protein restriction. Because the concentration of sOb-R increased without changing plasma leptin concentration, free leptin in plasma was significantly reduced. The direct effect of amino acid deprivation on Ob-R mRNA level was not observed in rat hepatoma cells H4IIE cultured in amino acid deprived medium. In conclusion, dietary protein restriction increased hepatic Ob-R mRNA, resulting in increased plasma sOb-R concentration, which in turn, reduces plasma free leptin level and may modulate leptin activity.
Collapse
Affiliation(s)
- Riho Yamada
- Department of Agricultural Chemistry, School of Agriculture, Meiji University, Kawasaki, Kanagawa, Japan
| | - Shizuka Odamaki
- Department of Agricultural Chemistry, School of Agriculture, Meiji University, Kawasaki, Kanagawa, Japan
| | - Masaya Araki
- Department of Agricultural Chemistry, School of Agriculture, Meiji University, Kawasaki, Kanagawa, Japan
| | - Tasuku Watanabe
- Department of Agricultural Chemistry, School of Agriculture, Meiji University, Kawasaki, Kanagawa, Japan
| | - Keigo Matsuo
- Department of Agricultural Chemistry, School of Agriculture, Meiji University, Kawasaki, Kanagawa, Japan
| | - Kaito Uchida
- Department of Agricultural Chemistry, School of Agriculture, Meiji University, Kawasaki, Kanagawa, Japan
| | - Taku Kato
- Department of Agricultural Chemistry, School of Agriculture, Meiji University, Kawasaki, Kanagawa, Japan
| | - Yori Ozaki-Masuzawa
- Department of Agricultural Chemistry, School of Agriculture, Meiji University, Kawasaki, Kanagawa, Japan
| | - Asako Takenaka
- Department of Agricultural Chemistry, School of Agriculture, Meiji University, Kawasaki, Kanagawa, Japan
- * E-mail:
| |
Collapse
|
14
|
Deshpande SS, Nemani H, Pothani S, Khambata K, Kumar A, Kallamadi PR, Balasinor NH. Genetically Inherited Obesity and High-Fat Diet-Induced Obesity Differentially Alter Spermatogenesis in Adult Male Rats. Endocrinology 2019; 160:220-234. [PMID: 30496384 DOI: 10.1210/en.2018-00569] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 11/21/2018] [Indexed: 02/03/2023]
Abstract
Obesity is a multifactorial disorder with predominantly genetic and/or environmental causes. Our aim was to delineate effects of genetically inherited and high-fat diet-induced obesity on fertility and spermatogenesis using two Wistar rat models: genetically inherited obese (GIO) WNIN/Ob rats and diet-induced obese (DIO) rats, which received a high-fat diet. The terminal body weights were similar in both groups, but there was a significant difference in metabolic and hormone profiles between the groups. Fertility assessment revealed a significant decrease in the litter size due to increased pre- and postimplantation loss in the DIO group, whereas the rats in the GIO group were infertile due to lack of libido. Significantly decreased sperm counts were observed in the GIO group compared with the DIO group. Enumeration of testicular cells on the basis of ploidy and cell type-specific expression markers, to study the effect of obesity on spermatogenesis, demonstrated that the GIO and DIO states affected mitosis: spermatogonia and S-phase population were increased. However, distinctive effects were observed on meiosis and spermiogenesis in both the groups. Differential effects of GIO and DIO on fertility and spermatogenesis could be due to the significant difference in white adipose tissue accumulation between the groups and not due to high body weights. The differential effects of obesity suggest male obesity-induced infertility observed in humans could be a combination of genetic and environmental factors.
Collapse
Affiliation(s)
- Sharvari S Deshpande
- Department of Neuroendocrinology, National Institute for Research in Reproductive Health, Mumbai, India
| | - Harishankar Nemani
- National Centre for Laboratory Animal Sciences, National Institute of Nutrition, Hyderabad, India
| | - Suresh Pothani
- National Centre for Laboratory Animal Sciences, National Institute of Nutrition, Hyderabad, India
| | - Kushaan Khambata
- Department of Neuroendocrinology, National Institute for Research in Reproductive Health, Mumbai, India
| | - Anita Kumar
- Department of Neuroendocrinology, National Institute for Research in Reproductive Health, Mumbai, India
| | - Prathap Reddy Kallamadi
- National Centre for Laboratory Animal Sciences, National Institute of Nutrition, Hyderabad, India
| | - Nafisa H Balasinor
- Department of Neuroendocrinology, National Institute for Research in Reproductive Health, Mumbai, India
| |
Collapse
|
15
|
Ballak DB, Li S, Cavalli G, Stahl JL, Tengesdal IW, van Diepen JA, Klück V, Swartzwelter B, Azam T, Tack CJ, Stienstra R, Mandrup-Poulsen T, Seals DR, Dinarello CA. Interleukin-37 treatment of mice with metabolic syndrome improves insulin sensitivity and reduces pro-inflammatory cytokine production in adipose tissue. J Biol Chem 2018; 293:14224-14236. [PMID: 30006351 PMCID: PMC6139546 DOI: 10.1074/jbc.ra118.003698] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/06/2018] [Indexed: 12/22/2022] Open
Abstract
Obesity and the metabolic syndrome are characterized by chronic, low-grade inflammation mainly originating from expanding adipose tissue and resulting in inhibition of insulin signaling and disruption of glycemic control. Transgenic mice expressing human interleukin 37 (IL-37), an anti-inflammatory cytokine of the IL-1 family, are protected against metabolic syndrome when fed a high-fat diet (HFD) containing 45% fat. Here, we examined whether treatment with recombinant IL-37 ameliorates established insulin resistance and obesity-induced inflammation. WT mice were fed a HFD for 22 weeks and then treated daily with IL-37 (1 μg/mouse) during the last 2 weeks. Compared with vehicle only-treated mice, IL-37-treated mice exhibited reduced insulin in the plasma and had significant improvements in glucose tolerance and in insulin content of the islets. The IL-37 treatment also increased the levels of circulating IL-1 receptor antagonist. Cultured adipose tissues revealed that IL-37 treatment significantly decreases spontaneous secretions of IL-1β, tumor necrosis factor α (TNFα), and CXC motif chemokine ligand 1 (CXCL-1). We also fed mice a 60% fat diet with concomitant daily IL-37 for 2 weeks and observed decreased secretion of IL-1β, TNFα, and IL-6 and reduced intracellular levels of IL-1α in the liver and adipose tissue, along with improved plasma glucose clearance. Compared with vehicle treatment, these IL-37-treated mice had no apparent weight gain. In human adipose tissue cultures, the presence of 50 pm IL-37 reduced spontaneous release of TNFα and 50% of lipopolysaccharide-induced TNFα. These findings indicate that IL-37's anti-inflammatory effects can ameliorate established metabolic disturbances during obesity.
Collapse
Affiliation(s)
- Dov B. Ballak
- From the Department of Medicine, University of Colorado Denver, Aurora, Colorado 80045, ,the Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado 80309
| | - Suzhao Li
- From the Department of Medicine, University of Colorado Denver, Aurora, Colorado 80045
| | - Giulio Cavalli
- From the Department of Medicine, University of Colorado Denver, Aurora, Colorado 80045
| | - Jonathan L. Stahl
- the Department of Biomedical Sciences, University of Copenhagen, 1165 Copenhagen, Denmark
| | - Isak W. Tengesdal
- From the Department of Medicine, University of Colorado Denver, Aurora, Colorado 80045
| | - Janna A. van Diepen
- the Department of Medicine, Radboud University Medical Center, 6525 Nijmegen, The Netherlands, and
| | - Viola Klück
- From the Department of Medicine, University of Colorado Denver, Aurora, Colorado 80045
| | - Benjamin Swartzwelter
- From the Department of Medicine, University of Colorado Denver, Aurora, Colorado 80045
| | - Tania Azam
- From the Department of Medicine, University of Colorado Denver, Aurora, Colorado 80045
| | - Cees J. Tack
- the Department of Medicine, Radboud University Medical Center, 6525 Nijmegen, The Netherlands, and
| | - Rinke Stienstra
- the Department of Medicine, Radboud University Medical Center, 6525 Nijmegen, The Netherlands, and ,the Division of Human Nutrition, Wageningen University, 6525 Wageningen, The Netherlands
| | - Thomas Mandrup-Poulsen
- the Department of Biomedical Sciences, University of Copenhagen, 1165 Copenhagen, Denmark
| | - Douglas R. Seals
- the Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado 80309
| | - Charles A. Dinarello
- From the Department of Medicine, University of Colorado Denver, Aurora, Colorado 80045, ,the Department of Medicine, Radboud University Medical Center, 6525 Nijmegen, The Netherlands, and , To whom correspondence should be addressed:
Dept. of Medicine, University of Colorado Denver, Aurora, Colorado 80045. Tel.:
303-724-6174; Fax:
303-724-6178; E-mail:
| |
Collapse
|
16
|
Ferguson D, Blenden M, Hutson I, Du Y, Harris CA. Mouse Embryonic Fibroblasts Protect ob/ob Mice From Obesity and Metabolic Complications. Endocrinology 2018; 159:3275-3286. [PMID: 30085057 PMCID: PMC6109302 DOI: 10.1210/en.2018-00561] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 07/25/2018] [Indexed: 12/22/2022]
Abstract
The global obesity epidemic is fueling alarming rates of diabetes, associated with increased risk of cardiovascular disease and cancer. Leptin is a hormone secreted by adipose tissue that is a key regulator of body weight (BW) and energy expenditure. Leptin-deficient humans and mice are obese, diabetic, and infertile and have hepatic steatosis. Although leptin replacement therapy can alleviate the pathologies seen in leptin-deficient patients and mouse models, treatment is costly and requires daily injections. Because adipocytes are the source of leptin secretion, we investigated whether mouse embryonic fibroblasts (MEFs), capable of forming adipocytes, could be injected into ob/ob mice and prevent the metabolic phenotype seen in these leptin-deficient mice. We performed a single subcutaneous injection of MEFs into leptin-deficient ob/ob mice. The MEF injection formed a single fat pad that is histologically similar to white adipose tissue. The ob/ob mice receiving MEFs (obRs) had significantly lower BW compared with nontreated ob/ob mice, primarily because of decreased adipose tissue mass. Additionally, obR mice had significantly less liver steatosis and greater glucose tolerance and insulin sensitivity. obR mice also manifested lower food intake and greater energy expenditure than ob/ob mice, providing a mechanism underlying their metabolic improvement. Furthermore, obRs have sustained metabolic protection and restoration of fertility. Collectively, our studies show the importance of functional adipocytes in preventing metabolic abnormalities seen in leptin deficiency and point to the possibility of cell-based therapies for the treatment of leptin-deficient states.
Collapse
Affiliation(s)
- Daniel Ferguson
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, Missouri
| | - Mitchell Blenden
- Department of Medical Education, College of Medicine, University of Central Florida, Orlando, Florida
| | - Irina Hutson
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, Missouri
| | - Yingqiu Du
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, Missouri
| | - Charles A Harris
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, Missouri
- Department of Medicine, Veterans Affairs St. Louis Healthcare System, John Cochran Division, St. Louis, Missouri
| |
Collapse
|
17
|
Comparative Proteome Analysis Reveals Lipid Metabolism-Related Protein Networks in Response to Rump Fat Mobilization. Int J Mol Sci 2018; 19:ijms19092556. [PMID: 30154394 PMCID: PMC6164786 DOI: 10.3390/ijms19092556] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 08/17/2018] [Accepted: 08/17/2018] [Indexed: 01/09/2023] Open
Abstract
Altay is a typical fat-tailed sheep breed displaying the unique ability to rapidly mobilize fat, which is vital for maintaining a normal metabolism that facilitates its survival in lengthy winter conditions. However, the physiological, biochemical, and molecular mechanisms underlying fat mobilization remain to be elucidated. In this study, the monitoring of rump fat adipocyte sizes disclosed a positive correlation between cell size and fat deposition ability. In addition, we subjected sheep to persistent starvation to imitate the conditions that trigger rump fat mobilization and screened 112 differentially expressed proteins using the isobaric peptide labeling approach. Notably, increased secretion of leptin and adiponectin activated the key fat mobilization signaling pathways under persistent starvation conditions. Furthermore, the upregulation of resistin (RETN), heat-shock protein 72 (HSP72), and complement factor D (CFD) promoted lipolysis, whereas the downregulation of cell death-inducing DFFA-like effector C (CIDEC) inhibited lipid droplet fusion, and the increase in HSP72 and apolipoprotein AI (Apo-AI) levels activated the body’s stress mechanisms. The synergistic actions of the above hormones, genes, and signaling pathways form a molecular network that functions in improving the adaptability of Altay sheep to extreme environments. Our findings provide a reference for elucidating the complex molecular mechanisms underlying rump fat mobilization.
Collapse
|
18
|
Perry RJ, Wang Y, Cline GW, Rabin-Court A, Song JD, Dufour S, Zhang XM, Petersen KF, Shulman GI. Leptin Mediates a Glucose-Fatty Acid Cycle to Maintain Glucose Homeostasis in Starvation. Cell 2018; 172:234-248.e17. [PMID: 29307489 PMCID: PMC5766366 DOI: 10.1016/j.cell.2017.12.001] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 09/09/2017] [Accepted: 11/29/2017] [Indexed: 02/02/2023]
Abstract
The transition from the fed to the fasted state necessitates a shift from carbohydrate to fat metabolism that is thought to be mostly orchestrated by reductions in plasma insulin concentrations. Here, we show in awake rats that insulinopenia per se does not cause this transition but that both hypoleptinemia and insulinopenia are necessary. Furthermore, we show that hypoleptinemia mediates a glucose-fatty acid cycle through activation of the hypothalamic-pituitary-adrenal axis, resulting in increased white adipose tissue (WAT) lipolysis rates and increased hepatic acetyl-coenzyme A (CoA) content, which are essential to maintain gluconeogenesis during starvation. We also show that in prolonged starvation, substrate limitation due to reduced rates of glucose-alanine cycling lowers rates of hepatic mitochondrial anaplerosis, oxidation, and gluconeogenesis. Taken together, these data identify a leptin-mediated glucose-fatty acid cycle that integrates responses of the muscle, WAT, and liver to promote a shift from carbohydrate to fat oxidation and maintain glucose homeostasis during starvation.
Collapse
Affiliation(s)
- Rachel J Perry
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Yongliang Wang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Gary W Cline
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Aviva Rabin-Court
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Joongyu D Song
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Sylvie Dufour
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Xian Man Zhang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Kitt Falk Petersen
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Gerald I Shulman
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA; Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW This perspective is motivated by the need to question dogma that does not work: that the problem is insulin resistance (IR). We highlight the need to investigate potential environmental obesogens and toxins. RECENT FINDINGS The prequel to severe metabolic disease includes three interacting components that are abnormal: (a) IR, (b) elevated lipids and (c) elevated basal insulin (HI). HI is more common than IR and is a significant independent predictor of diabetes. We hypothesize that (1) the initiating defect is HI that increases nutrient consumption and hyperlipidemia (HL); (2) the cause of HI may include food additives, environmental obesogens or toxins that have entered our food supply since 1980; and (3) HI is sustained by HL derived from increased adipose mass and leads to IR. We suggest that HI and HL are early indicators of metabolic dysfunction and treating and reversing these abnormalities may prevent the development of more serious metabolic disease.
Collapse
Affiliation(s)
- Karel A. Erion
- 0000 0004 0367 5222grid.475010.7Obesity Research Center, Department of Medicine, Boston University School of Medicine, 650 Albany St, Boston, MA 02118 USA
- 0000 0000 9632 6718grid.19006.3eDivision of Endocrinology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA USA
| | - Barbara E. Corkey
- 0000 0004 0367 5222grid.475010.7Obesity Research Center, Department of Medicine, Boston University School of Medicine, 650 Albany St, Boston, MA 02118 USA
| |
Collapse
|
20
|
Takanashi M, Taira Y, Okazaki S, Takase S, Kimura T, Li CC, Xu PF, Noda A, Sakata I, Kumagai H, Ikeda Y, Iizuka Y, Yahagi N, Shimano H, Osuga JI, Ishibashi S, Kadowaki T, Okazaki H. Role of Hormone-sensitive Lipase in Leptin-Promoted Fat Loss and Glucose Lowering. J Atheroscler Thromb 2017; 24:1105-1116. [PMID: 28413180 PMCID: PMC5684476 DOI: 10.5551/jat.39552] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aim: Myriad biological effects of leptin may lead to broad therapeutic applications for various metabolic diseases, including diabetes and its complications; however, in contrast to its anorexic effect, the molecular mechanisms underlying adipopenic and glucose-lowering effects of leptin have not been fully understood. Here we aim to clarify the role of hormone-sensitive lipase (HSL) in leptin's action. Methods: Wild-type (WT) and HSL-deficient (HSLKO) mice were made hyperleptinemic by two commonly-used methods: adenovirus-mediated overexpression of leptin and continuous subcutaneous infusion of leptin by osmotic pumps. The amount of food intake, body weights, organ weights, and parameters of glucose and lipid metabolism were measured. Results: Hyperleptinemia equally suppressed the food intake in WT and HSLKO mice. On the other hand, leptin-mediated fat loss and glucose-lowering were significantly blunted in the absence of HSL when leptin was overexpressed by recombinant adenovirus carrying leptin. By osmotic pumps, the fat-losing and glucose-lowering effects of leptin were milder due to lower levels of hyperleptinemia; although the difference between WT and HSLKO mice did not reach statistical significance, HSLKO mice had a tendency to retain more fat than WT mice in the face of hyperleptinemia. Conclusions: We clarify for the first time the role of HSL in leptin's effect using a genetic model: leptin-promoted fat loss and glucose-lowering are at least in part mediated via HSL-mediated lipolysis. Further studies to define the pathophysiological role of adipocyte lipases in leptin action may lead to a new therapeutic approach to circumvent leptin resistance.
Collapse
Affiliation(s)
- Mikio Takanashi
- Departments of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Yoshino Taira
- Departments of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Sachiko Okazaki
- Departments of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Satoru Takase
- Departments of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Takeshi Kimura
- Departments of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Cheng Cheng Li
- Departments of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Peng Fei Xu
- Departments of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Akari Noda
- Departments of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Ichiro Sakata
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University
| | - Hidetoshi Kumagai
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| | - Yuichi Ikeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| | - Yoko Iizuka
- Departments of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Naoya Yahagi
- Departments of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Hitoshi Shimano
- Departments of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Jun-Ichi Osuga
- Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University
| | - Shun Ishibashi
- Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University
| | - Takashi Kadowaki
- Departments of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Hiroaki Okazaki
- Departments of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| |
Collapse
|
21
|
Pandit R, Beerens S, Adan RAH. Role of leptin in energy expenditure: the hypothalamic perspective. Am J Physiol Regul Integr Comp Physiol 2017; 312:R938-R947. [PMID: 28356295 DOI: 10.1152/ajpregu.00045.2016] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 03/27/2017] [Accepted: 03/28/2017] [Indexed: 01/08/2023]
Abstract
The adipocyte-derived hormone leptin is a peripheral signal that informs the brain about the metabolic status of an organism. Although traditionally viewed as an appetite-suppressing hormone, studies in the past decade have highlighted the role of leptin in energy expenditure. Leptin has been shown to increase energy expenditure in particular through its effects on the cardiovascular system and brown adipose tissue (BAT) thermogenesis via the hypothalamus. The current review summarizes the role of leptin signaling in various hypothalamic nuclei and its effects on the sympathetic nervous system to influence blood pressure, heart rate, and BAT thermogenesis. Specifically, the role of leptin signaling on three different hypothalamic nuclei, the dorsomedial hypothalamus, the ventromedial hypothalamus, and the arcuate nucleus, is reviewed. It is known that all of these brain regions influence the sympathetic nervous system activity and thereby regulate BAT thermogenesis and the cardiovascular system. Thus the current work focuses on how leptin signaling in specific neuronal populations within these hypothalamic nuclei influences certain aspects of energy expenditure.
Collapse
Affiliation(s)
- R Pandit
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - S Beerens
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - R A H Adan
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
22
|
Carcangiu V, Giannetto C, Luridiana S, Fazio F, Mura MC, Parmeggiani A, Piccione G. Lactation influences the serum level of leptin and growth hormone during the daily bathyphase in ewes. BIOL RHYTHM RES 2016. [DOI: 10.1080/09291016.2016.1223808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
23
|
Mela V, Vargas A, Meza C, Kachani M, Wagner EJ. Modulatory influences of estradiol and other anorexigenic hormones on metabotropic, Gi/o-coupled receptor function in the hypothalamic control of energy homeostasis. J Steroid Biochem Mol Biol 2016; 160:15-26. [PMID: 26232394 PMCID: PMC4732935 DOI: 10.1016/j.jsbmb.2015.07.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 07/22/2015] [Accepted: 07/26/2015] [Indexed: 11/22/2022]
Abstract
The appetite suppressant actions of estradiol are due to its ability to attenuate orexigenic signals and potentiate anorexigenic signals. The work from my laboratory has shown that male guinea pigs are more sensitive to the hyperphagic and hypothermic effects of cannabinoids than their female counterparts. Cannabinoid sensitivity is further dampened by the activational effects of estradiol. This occurs via the hypothalamic feeding circuitry, where estradiol rapidly attenuates the cannabinoid CB1 receptor-mediated presynaptic inhibition of glutamatergic input onto anorexigenic proopiomelanocortin (POMC) neurons in the arcuate nucleus. This disruption is blocked by the estrogen receptor antagonist ICI 182,780, and associated with increased expression of phosphatidylinositol-3-kinase (PI3K). Moreover, the ability of estradiol to reduce both the cannabinoid-induced hyperphagia and glutamate release onto POMC neurons is abrogated by the PI3K inhibitor PI 828. The peptide orphanin FQ/nociceptin (OFQ/N) activates opioid receptor-like (ORL)1 receptors to hyperpolarize and inhibit POMC neurons via the activation of postsynaptic G protein-gated, inwardly-rectifying (GIRK) channels. We have demonstrated that the fasting-induced hyperphagia observed in ORL1-null mice is blunted compared to wild type controls. In addition, the ORL1 receptor-mediated activation of GIRK channels in POMC neurons from ovariectomized female rats is markedly impaired by estradiol. The estrogenic attenuation of presynaptic CB1 and postsynaptic ORL1 receptor function may be part of a more generalized mechanism through which anorexigenic hormones suppress orexigenic signaling. Indeed, we have found that leptin robustly suppresses the OFQ/N-induced activation of GIRK channels in POMC neurons. Furthermore, its ability to augment excitatory input onto POMC neurons is blocked by PI 828. Thus, estradiol and other hormones like leptin reduce energy intake at least partly by activating PI3K to disrupt the pleiotropic functions of Gi/o-coupled receptors that inhibit anorexigenic POMC neurons.
Collapse
Affiliation(s)
- Virginia Mela
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States
| | - Amanda Vargas
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States
| | - Cecilia Meza
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States
| | - Malika Kachani
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766, United States
| | - Edward J Wagner
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States.
| |
Collapse
|
24
|
Stern JH, Rutkowski JM, Scherer PE. Adiponectin, Leptin, and Fatty Acids in the Maintenance of Metabolic Homeostasis through Adipose Tissue Crosstalk. Cell Metab 2016; 23:770-84. [PMID: 27166942 PMCID: PMC4864949 DOI: 10.1016/j.cmet.2016.04.011] [Citation(s) in RCA: 724] [Impact Index Per Article: 80.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Metabolism research has made tremendous progress over the last several decades in establishing the adipocyte as a central rheostat in the regulation of systemic nutrient and energy homeostasis. Operating at multiple levels of control, the adipocyte communicates with organ systems to adjust gene expression, glucoregulatory hormone exocytosis, enzymatic reactions, and nutrient flux to equilibrate the metabolic demands of a positive or negative energy balance. The identification of these mechanisms has great potential to identify novel targets for the treatment of diabetes and related metabolic disorders. Herein, we review the central role of the adipocyte in the maintenance of metabolic homeostasis, highlighting three critical mediators: adiponectin, leptin, and fatty acids.
Collapse
Affiliation(s)
- Jennifer H Stern
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Joseph M Rutkowski
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
25
|
Ehrhardt RA, Foskolos A, Giesy SL, Wesolowski SR, Krumm CS, Butler WR, Quirk SM, Waldron MR, Boisclair YR. Increased plasma leptin attenuates adaptive metabolism in early lactating dairy cows. J Endocrinol 2016; 229:145-57. [PMID: 26957637 DOI: 10.1530/joe-16-0031] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 03/03/2016] [Indexed: 12/31/2022]
Abstract
Mammals meet the increased nutritional demands of lactation through a combination of increased feed intake and a collection of adaptations known as adaptive metabolism (e.g., glucose sparing via insulin resistance, mobilization of endogenous reserves, and increased metabolic efficiency via reduced thyroid hormones). In the modern dairy cow, adaptive metabolism predominates over increased feed intake at the onset of lactation and develops concurrently with a reduction in plasma leptin. To address the role of leptin in the adaptive metabolism of early lactation, we asked which adaptations could be countered by a constant 96-h intravenous infusion of human leptin (hLeptin) starting on day 8 of lactation. Compared to saline infusion (Control), hLeptin did not alter energy intake or milk energy output but caused a modest increase in body weight loss. hLeptin reduced plasma glucose by 9% and hepatic glycogen content by 73%, and these effects were associated with a 17% increase in glucose disposal during an insulin tolerance test. hLeptin attenuated the accumulation of triglyceride in the liver by 28% in the absence of effects on plasma levels of the anti-lipolytic hormone insulin or plasma levels of free fatty acids, a marker of lipid mobilization from adipose tissue. Finally, hLeptin increased the plasma concentrations of T4 and T3 by nearly 50% without affecting other neurally regulated hormones (i.e., cortisol and luteinizing hormone (LH)). Overall these data implicate the periparturient reduction in plasma leptin as one of the signals promoting conservation of glucose and energy at the onset of lactation in the energy-deficient dairy cow.
Collapse
Affiliation(s)
- Richard A Ehrhardt
- Departments of Animal Science and Large Animal Clinical SciencesMichigan State University, East Lansing, Michigan, USA
| | - Andreas Foskolos
- Institute of Biological, Environmental and Rural SciencesAberystwyth University, Aberystwyth, UK
| | - Sarah L Giesy
- Department of Animal ScienceCornell University, Ithaca, New York, USA
| | | | | | - W Ronald Butler
- Department of Animal ScienceCornell University, Ithaca, New York, USA
| | - Susan M Quirk
- Department of Animal ScienceCornell University, Ithaca, New York, USA
| | - Matthew R Waldron
- Department of Animal ScienceCornell University, Ithaca, New York, USA
| | - Yves R Boisclair
- Department of Animal ScienceCornell University, Ithaca, New York, USA
| |
Collapse
|
26
|
Bolze F, Morath V, Bast A, Rink N, Schlapschy M, Mocek S, Skerra A, Klingenspor M. Long-Acting PASylated Leptin Ameliorates Obesity by Promoting Satiety and Preventing Hypometabolism in Leptin-Deficient Lep(ob/ob) Mice. Endocrinology 2016; 157:233-44. [PMID: 26492472 DOI: 10.1210/en.2015-1519] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Body weight loss of Lep(ob/ob) mice in response to leptin is larger than expected from the reduction in energy intake alone, suggesting a thermogenic action of unknown magnitude. We exploited the superior pharmacological properties of a novel long-acting leptin prepared via PASylation to study the contribution of its anorexigenic and thermogenic effects. PASylation, the genetic fusion of leptin with a conformationally disordered polypeptide comprising 600 Pro/Ala/Ser (PAS) residues, provides a superior way to increase the hydrodynamic volume of the fusion protein, thus retarding kidney filtration and extending plasma half-life. Here a single PAS(600)-leptin injection (300 pmol/g) resulted in a maximal weight reduction of 21% 6 days after application. The negative energy balance of 300 kJ/(4 d) was driven by a decrease in energy intake, whereas energy expenditure remained stable. Mice that were food restricted to the same extent showed an energy deficit of only 220 kJ/(4 d) owing to recurring torpor bouts. Therefore, the anorexigenic effect of PAS(600)-leptin contributes 75% to weight loss, whereas the thermogenic action accounts for 25% by preventing hypometabolism. In a second experiment, just four injections of PAS(600)-leptin (100 pmol/g) administered in 5- to 6-day intervals rectified the Lep(ob/ob) phenotype. In total, 16 nmol of PAS(600)-leptin per mouse triggered a weight loss of 43% within 20 days and normalized hypothermia and glucose homeostasis as well as hepatic steatosis. The beneficial properties of PAS(600)-leptin are substantiated by a comparison with previous studies in which approximately 400 nmol (∼25-fold) unmodified leptin was mandatory to achieve similar improvements.
Collapse
Affiliation(s)
- Florian Bolze
- Lehrstuhl für Molekulare Ernährungsmedizin (F.B., A.B., N.R., S.M., M.K.), Else Kröner-Fresenius Center and ZIEL-Research Center for Nutrition and Food Science, and Munich Center for Integrated Protein Science and Lehrstuhl für Biologische Chemie (V.M., M.S., A.S.), Technische Universität München, 85350 Freising-Weihenstephan, Germany; and XL-protein GmbH (A.S.), 85354 Freising, Germany
| | - Volker Morath
- Lehrstuhl für Molekulare Ernährungsmedizin (F.B., A.B., N.R., S.M., M.K.), Else Kröner-Fresenius Center and ZIEL-Research Center for Nutrition and Food Science, and Munich Center for Integrated Protein Science and Lehrstuhl für Biologische Chemie (V.M., M.S., A.S.), Technische Universität München, 85350 Freising-Weihenstephan, Germany; and XL-protein GmbH (A.S.), 85354 Freising, Germany
| | - Andrea Bast
- Lehrstuhl für Molekulare Ernährungsmedizin (F.B., A.B., N.R., S.M., M.K.), Else Kröner-Fresenius Center and ZIEL-Research Center for Nutrition and Food Science, and Munich Center for Integrated Protein Science and Lehrstuhl für Biologische Chemie (V.M., M.S., A.S.), Technische Universität München, 85350 Freising-Weihenstephan, Germany; and XL-protein GmbH (A.S.), 85354 Freising, Germany
| | - Nadine Rink
- Lehrstuhl für Molekulare Ernährungsmedizin (F.B., A.B., N.R., S.M., M.K.), Else Kröner-Fresenius Center and ZIEL-Research Center for Nutrition and Food Science, and Munich Center for Integrated Protein Science and Lehrstuhl für Biologische Chemie (V.M., M.S., A.S.), Technische Universität München, 85350 Freising-Weihenstephan, Germany; and XL-protein GmbH (A.S.), 85354 Freising, Germany
| | - Martin Schlapschy
- Lehrstuhl für Molekulare Ernährungsmedizin (F.B., A.B., N.R., S.M., M.K.), Else Kröner-Fresenius Center and ZIEL-Research Center for Nutrition and Food Science, and Munich Center for Integrated Protein Science and Lehrstuhl für Biologische Chemie (V.M., M.S., A.S.), Technische Universität München, 85350 Freising-Weihenstephan, Germany; and XL-protein GmbH (A.S.), 85354 Freising, Germany
| | - Sabine Mocek
- Lehrstuhl für Molekulare Ernährungsmedizin (F.B., A.B., N.R., S.M., M.K.), Else Kröner-Fresenius Center and ZIEL-Research Center for Nutrition and Food Science, and Munich Center for Integrated Protein Science and Lehrstuhl für Biologische Chemie (V.M., M.S., A.S.), Technische Universität München, 85350 Freising-Weihenstephan, Germany; and XL-protein GmbH (A.S.), 85354 Freising, Germany
| | - Arne Skerra
- Lehrstuhl für Molekulare Ernährungsmedizin (F.B., A.B., N.R., S.M., M.K.), Else Kröner-Fresenius Center and ZIEL-Research Center for Nutrition and Food Science, and Munich Center for Integrated Protein Science and Lehrstuhl für Biologische Chemie (V.M., M.S., A.S.), Technische Universität München, 85350 Freising-Weihenstephan, Germany; and XL-protein GmbH (A.S.), 85354 Freising, Germany
| | - Martin Klingenspor
- Lehrstuhl für Molekulare Ernährungsmedizin (F.B., A.B., N.R., S.M., M.K.), Else Kröner-Fresenius Center and ZIEL-Research Center for Nutrition and Food Science, and Munich Center for Integrated Protein Science and Lehrstuhl für Biologische Chemie (V.M., M.S., A.S.), Technische Universität München, 85350 Freising-Weihenstephan, Germany; and XL-protein GmbH (A.S.), 85354 Freising, Germany
| |
Collapse
|
27
|
Turner RT, Dube M, Branscum AJ, Wong CP, Olson DA, Zhong X, Kweh MF, Larkin IV, Wronski TJ, Rosen CJ, Kalra SP, Iwaniec UT. Hypothalamic leptin gene therapy reduces body weight without accelerating age-related bone loss. J Endocrinol 2015; 227:129-41. [PMID: 26487675 PMCID: PMC4917201 DOI: 10.1530/joe-15-0280] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/14/2015] [Indexed: 02/04/2023]
Abstract
Excessive weight gain in adults is associated with a variety of negative health outcomes. Unfortunately, dieting, exercise, and pharmacological interventions have had limited long-term success in weight control and can result in detrimental side effects, including accelerating age-related cancellous bone loss. We investigated the efficacy of using hypothalamic leptin gene therapy as an alternative method for reducing weight in skeletally-mature (9 months old) female rats and determined the impact of leptin-induced weight loss on bone mass, density, and microarchitecture, and serum biomarkers of bone turnover (CTx and osteocalcin). Rats were implanted with cannulae in the 3rd ventricle of the hypothalamus and injected with either recombinant adeno-associated virus encoding the gene for rat leptin (rAAV-Leptin, n=7) or a control vector encoding green fluorescent protein (rAAV-GFP, n=10) and sacrificed 18 weeks later. A baseline control group (n=7) was sacrificed at vector administration. rAAV-Leptin-treated rats lost weight (-4±2%) while rAAV-GFP-treated rats gained weight (14±2%) during the study. At study termination, rAAV-Leptin-treated rats weighed 17% less than rAAV-GFP-treated rats and had lower abdominal white adipose tissue weight (-80%), serum leptin (-77%), and serum IGF1 (-34%). Cancellous bone volume fraction in distal femur metaphysis and epiphysis, and in lumbar vertebra tended to be lower (P<0.1) in rAAV-GFP-treated rats (13.5 months old) compared to baseline control rats (9 months old). Significant differences in cancellous bone or biomarkers of bone turnover were not detected between rAAV-Leptin and rAAV-GFP rats. In summary, rAAV-Leptin-treated rats maintained a lower body weight compared to baseline and rAAV-GFP-treated rats with minimal effects on bone mass, density, microarchitecture, or biochemical markers of bone turnover.
Collapse
Affiliation(s)
- Russell T Turner
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Michael Dube
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Adam J Branscum
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Carmen P Wong
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Dawn A Olson
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Xiaoying Zhong
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Mercedes F Kweh
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Iske V Larkin
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Thomas J Wronski
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Clifford J Rosen
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Satya P Kalra
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Urszula T Iwaniec
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| |
Collapse
|
28
|
Intracerebroventricular administration of leptin increase physical activity but has no effect on thermogenesis in cold-acclimated rats. Sci Rep 2015; 5:11189. [PMID: 26053156 PMCID: PMC4459185 DOI: 10.1038/srep11189] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 04/17/2015] [Indexed: 12/03/2022] Open
Abstract
Most small homotherms display low leptin level in response to chronic cold exposure. Cold-induced hypoleptinemia was proved to induce hyperphagia. However, it is still not clear whether hypoleptinemia regulates energy expenditure in cold condition. We try to answer this question in chronic cold-acclimated rats. Results showed that 5-day intracerebroventricular(ICV) infusion of leptin (5 μg/day) had no effects on basal and adaptive thermogenesis and uncoupling protein 1 expression. Physical activity was increased by leptin treatment. We further determined whether ghrelin could reverse the increasing effect of leptin on physical activity. Coadministration of ghrelin (1.2 μg/day) completely reversed the effect of leptin on physical activity. Collectively, this study indicated the regulation of leptin on energy expenditure during cold acclimation may be mainly mediated by physical activity but not by thermogenesis. Our study outlined behavioral role of leptin during the adaptation to cold, which adds some new knowledge to promote our understanding of cold-induced metabolic adaptation.
Collapse
|
29
|
Meredith ME, Salameh TS, Banks WA. Intranasal Delivery of Proteins and Peptides in the Treatment of Neurodegenerative Diseases. AAPS JOURNAL 2015; 17:780-7. [PMID: 25801717 DOI: 10.1208/s12248-015-9719-7] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 01/12/2015] [Indexed: 12/28/2022]
Abstract
The blood-brain barrier (BBB) is a major impediment to the therapeutic delivery of peptides and proteins to the brain. Intranasal delivery often provides a non-invasive means to bypass the BBB. Advantages of using intranasal delivery include minimizing exposure to peripheral organs and tissues, thus reducing systemic side effects. It also allows substances that typically have rapid degradation in the blood time to exert their effect. Intranasal delivery provides the ability to target proteins and peptides to specific regions of the brain when administered with substrates like cyclodextrins. In this review, we examined the use of intranasal delivery of various proteins and peptides that have implications in the treatment of neurodegenerative diseases, focusing especially on albumin, exendin/GLP-1, GALP, insulin, leptin, and PACAP. We have described their rationale for use, distribution in the brain after intranasal injection, how intranasal administration differed from other modes of delivery, and their use in clinical trials, if applicable. Intranasal delivery of drugs, peptides, and other proteins could be very useful in the future for the prevention or treatment of brain related diseases.
Collapse
|
30
|
Kaiyala KJ, Ogimoto K, Nelson JT, Schwartz MW, Morton GJ. Leptin signaling is required for adaptive changes in food intake, but not energy expenditure, in response to different thermal conditions. PLoS One 2015; 10:e0119391. [PMID: 25756181 PMCID: PMC4355297 DOI: 10.1371/journal.pone.0119391] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 01/30/2015] [Indexed: 12/18/2022] Open
Abstract
Survival of free-living animals depends on the ability to maintain core body temperature in the face of rapid and dramatic changes in their thermal environment. If food intake is not adjusted to meet the changing energy demands associated with changes of ambient temperature, a serious challenge to body energy stores can occur. To more fully understand the coupling of thermoregulation to energy homeostasis in normal animals and to investigate the role of the adipose hormone leptin to this process, comprehensive measures of energy homeostasis and core temperature were obtained in leptin-deficient ob/ob mice and their wild-type (WT) littermate controls when housed under cool (14°C), usual (22°C) or ∼ thermoneutral (30°C) conditions. Our findings extend previous evidence that WT mice robustly defend normothermia in response to either a lowering (14°C) or an increase (30°C) of ambient temperature without changes in body weight or body composition. In contrast, leptin-deficient, ob/ob mice fail to defend normothermia at ambient temperatures lower than thermoneutrality and exhibit marked losses of both body fat and lean mass when exposed to cooler environments (14°C). Our findings further demonstrate a strong inverse relationship between ambient temperature and energy expenditure in WT mice, a relationship that is preserved in ob/ob mice. However, thermal conductance analysis indicates defective heat retention in ob/ob mice, irrespective of temperature. While a negative relationship between ambient temperature and energy intake also exists in WT mice, this relationship is disrupted in ob/ob mice. Thus, to meet the thermoregulatory demands of different ambient temperatures, leptin signaling is required for adaptive changes in both energy intake and thermal conductance. A better understanding of the mechanisms coupling thermoregulation to energy homeostasis may lead to the development of new approaches for the treatment of obesity.
Collapse
Affiliation(s)
- Karl J. Kaiyala
- Department of Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA, United States of America
| | - Kayoko Ogimoto
- Diabetes and Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Jarrell T. Nelson
- Diabetes and Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Michael W. Schwartz
- Diabetes and Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Gregory J. Morton
- Diabetes and Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, WA, United States of America
- * E-mail:
| |
Collapse
|
31
|
Xu L. Leptin action in the midbrain: From reward to stress. J Chem Neuroanat 2014; 61-62:256-65. [DOI: 10.1016/j.jchemneu.2014.06.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 06/13/2014] [Accepted: 06/25/2014] [Indexed: 12/11/2022]
|
32
|
The adipokine Retnla modulates cholesterol homeostasis in hyperlipidemic mice. Nat Commun 2014; 5:4410. [PMID: 25022542 DOI: 10.1038/ncomms5410] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Accepted: 06/16/2014] [Indexed: 01/05/2023] Open
Abstract
Hyperlipidemia is a well-recognized risk factor for atherosclerosis and can be regulated by adipokines. Expression of the adipokine resistin-like molecule alpha (Retnla) is regulated by food intake; whether Retnla has a role in the pathogenesis of hyperlipidemia and atherosclerosis is unknown. Here we report that Retnla has a cholesterol-lowering effect and protects against atherosclerosis in low-density lipoprotein receptor-deficient mice. On a high-fat diet, Retnla deficiency promotes hypercholesterolaemia and atherosclerosis, whereas Retnla overexpression reverses these effects and improves the serum lipoprotein profile, with decreased cholesterol in the very low-density lipoprotein fraction concomitant with reduced serum apolipoprotein B levels. We show that Retnla upregulates cholesterol-7-α-hydroxylase, a key hepatic enzyme in the cholesterol catabolic pathway, through induction of its transcriptional activator liver receptor homologue-1, leading to increased excretion of cholesterol in the form of bile acids. These findings define Retnla as a novel therapeutic target for treating hypercholesterolaemia and atherosclerosis.
Collapse
|
33
|
Abstract
The fat‐derived hormone, leptin, is well known to regulate body weight. However, there is now substantial evidence that leptin also plays a primary role in the regulation of glucose homeostasis, independent of actions on food intake, energy expenditure or body weight. As such, leptin might have clinical utility in treating hyperglycemia, particularly in conditions of leptin deficiency, such as lipodystrophy and diabetes mellitus. The mechanisms through which leptin modulates glucose metabolism have not been fully elucidated. Leptin receptors are widely expressed in peripheral tissues, including the endocrine pancreas, liver, skeletal muscle and adipose, and both direct and indirect leptin action on these tissues contributes to the control of glucose homeostasis. Here we review the role of leptin in glucose homeostasis, along with our present understanding of the mechanisms involved. (J Diabetes Invest, doi: 10.1111/j.2040‐1124.2012.00203.x, 2012)
Collapse
Affiliation(s)
- Heather C Denroche
- Department of Cellular and Physiological Sciences, The Life Sciences Institute
| | - Frank K Huynh
- Department of Cellular and Physiological Sciences, The Life Sciences Institute
| | - Timothy J Kieffer
- Department of Cellular and Physiological Sciences, The Life Sciences Institute ; Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
34
|
Wu F, Song Y, Li F, He X, Ma J, Feng T, Guan B, Wang L, Li S, Liu X, Liu Y, Mao M, Liu J, Bai S, Song C. Wen-dan decoction improves negative emotions in sleep-deprived rats by regulating orexin-a and leptin expression. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2014; 2014:872547. [PMID: 24864160 PMCID: PMC4016855 DOI: 10.1155/2014/872547] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 03/12/2014] [Accepted: 03/15/2014] [Indexed: 11/18/2022]
Abstract
Wen-Dan Decoction (WDD), a formula of traditional Chinese medicine, has been clinically used for treating insomnia for approximately 800 years. However, the therapeutic mechanisms of WDD remain unclear. Orexin-A plays a key role in the sleep-wake cycle, while leptin function is opposite to orexin-A. Thus, orexin-A and leptin may be important factors in sleep disorders. In this study, 48 rats were divided into control, model, WDD-treated, and diazepam-treated groups. The model of insomnia was produced by sleep deprivation (SD) for 14 days. The expressions of orexin-A, leptin, and their receptors in blood serum, prefrontal cortex, and hypothalamus were detected by enzyme-linked immunosorbent assay, immunohistochemistry, and real time PCR. Open field tests showed that SD increased both crossing movement (Cm) and rearing-movement (Rm) times. Orexin-A and leptin levels in blood serum increased after SD but decreased in brain compared to the control group. mRNA expressions of orexin receptor 1 and leptin receptor after SD were decreased in the prefrontal cortex but were increased in hypothalamus. WDD treatment normalized the behavior and upregulated orexin-A, leptin, orexin receptor 1 and leptin receptor in brain. The findings suggest that WDD treatment may regulate SD-induced negative emotions by regulating orexin-A and leptin expression.
Collapse
Affiliation(s)
- Fengzhi Wu
- School of Preclinical Medicine, Beijing University of Chinese Medicine, No. 11 Bei Sanhuan Donglu, Chaoyang District, Beijing 100029, China
| | - Yuehan Song
- School of Preclinical Medicine, Beijing University of Chinese Medicine, No. 11 Bei Sanhuan Donglu, Chaoyang District, Beijing 100029, China
| | - Feng Li
- School of Preclinical Medicine, Beijing University of Chinese Medicine, No. 11 Bei Sanhuan Donglu, Chaoyang District, Beijing 100029, China
| | - Xin He
- School of Preclinical Medicine, Beijing University of Chinese Medicine, No. 11 Bei Sanhuan Donglu, Chaoyang District, Beijing 100029, China
| | - Jie Ma
- School of Preclinical Medicine, Beijing University of Chinese Medicine, No. 11 Bei Sanhuan Donglu, Chaoyang District, Beijing 100029, China
| | - Ting Feng
- Traditional Chinese Medical Hospital of Ji'nan, Shandong 250012, China
| | - Binghe Guan
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Liye Wang
- School of Preclinical Medicine, Beijing University of Chinese Medicine, No. 11 Bei Sanhuan Donglu, Chaoyang District, Beijing 100029, China
| | - Sinai Li
- School of Preclinical Medicine, Beijing University of Chinese Medicine, No. 11 Bei Sanhuan Donglu, Chaoyang District, Beijing 100029, China
| | - Xiaolan Liu
- School of Preclinical Medicine, Beijing University of Chinese Medicine, No. 11 Bei Sanhuan Donglu, Chaoyang District, Beijing 100029, China
| | - Yan Liu
- School of Preclinical Medicine, Beijing University of Chinese Medicine, No. 11 Bei Sanhuan Donglu, Chaoyang District, Beijing 100029, China
| | - Meng Mao
- School of Preclinical Medicine, Beijing University of Chinese Medicine, No. 11 Bei Sanhuan Donglu, Chaoyang District, Beijing 100029, China
| | - Jing Liu
- School of Preclinical Medicine, Beijing University of Chinese Medicine, No. 11 Bei Sanhuan Donglu, Chaoyang District, Beijing 100029, China
| | - Shijing Bai
- School of Preclinical Medicine, Beijing University of Chinese Medicine, No. 11 Bei Sanhuan Donglu, Chaoyang District, Beijing 100029, China
| | - Cai Song
- Graduate Institute of Neural and Cognitive Sciences, China Medical University and CMU Hospital, Taichung 40402, Taiwan
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| |
Collapse
|
35
|
Suzuki Y, Nakahara K, Maruyama K, Okame R, Ensho T, Inoue Y, Murakami N. Changes in mRNA expression of arcuate nucleus appetite-regulating peptides during lactation in rats. J Mol Endocrinol 2014; 52:97-109. [PMID: 24299740 PMCID: PMC3907180 DOI: 10.1530/jme-13-0015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The contribution of hypothalamic appetite-regulating peptides to further hyperphagia accompanying the course of lactation in rats was investigated by using PCR array and real-time PCR. Furthermore, changes in the mRNA expression for appetite-regulating peptides in the hypothalamic arcuate nucleus (ARC) were analyzed at all stages of pregnancy and lactation, and also after weaning. Food intake was significantly higher during pregnancy, lactation, and after weaning than during non-lactation periods. During lactation, ARC expression of mRNAs for agouti-related protein (AgRP) and peptide YY was increased, whereas that of mRNAs for proopiomelanocortin (POMC) and cholecystokinin (CCK) was decreased, in comparison with non-lactation periods. The increase in AgRP mRNA expression during lactation was especially marked. The plasma level of leptin was significantly decreased during the course of lactation, whereas that of acyl-ghrelin was unchanged. In addition, food intake was negatively correlated with the plasma leptin level during lactation. This study has clarified synchronous changes in the expression of many appetite-regulating peptides in ARC of rats during lactation. Our results suggest that hyperphagia during lactation in rats is caused by decreases in POMC and CCK expression and increases in AgRP expression in ARC, the latter being most notable. Together with the decrease in the blood leptin level, such changes in mRNA expression may explain the further hyperphagia accompanying the course of lactation.
Collapse
Affiliation(s)
- Yoshihiro Suzuki
- Department of Veterinary Physiology, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Japan
| | | | | | | | | | | | | |
Collapse
|
36
|
Moreau J, Ciriello J. Effects of acute intermittent hypoxia on energy balance and hypothalamic feeding pathways. Neuroscience 2013; 253:350-60. [DOI: 10.1016/j.neuroscience.2013.09.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Revised: 09/03/2013] [Accepted: 09/04/2013] [Indexed: 01/20/2023]
|
37
|
Yamagata S, Kageyama K, Akimoto K, Watanuki Y, Suda T, Daimon M. Regulation of corticotropin-releasing factor and urocortin 2/3 mRNA by leptin in hypothalamic N39 cells. Peptides 2013; 50:1-7. [PMID: 24083959 DOI: 10.1016/j.peptides.2013.09.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 09/23/2013] [Accepted: 09/23/2013] [Indexed: 01/02/2023]
Abstract
Corticotropin-releasing factor (CRF) activates the pituitary-adrenal axis during stress, and shows anorectic effects via CRF type 1 receptors in the hypothalamus. Both urocortin (Ucn) 2 and Ucn3 also act as anorectic neuropeptides via CRF type 2 receptors. Leptin, a product of the obesity gene secreted mainly from adipose tissue, reduces food intake and increases energy expenditure. A possible interaction between leptin and CRF/Ucns has been suggested, as leptin can regulate expression and activation of CRF and Ucns in the hypothalamus. This study aimed to explore the possible function of leptin in the hypothalamus, and its effects in regulating CRF and Ucns. The study identified mRNA expression of the leptin receptor (Ob-R) and its subtypes, CRF, and Ucn2/3 in mouse hypothalamic N39 cells. Leptin stimulated signal transducer and activators of transcription type 3 (STAT3) phosphorylation, directly increased the mRNA levels of both CRF and Ucn2/3 in hypothalamic cells, and increased Ob-Rb mRNA levels. A Janus kinase inhibitor inhibited the leptin-mediated increase in STAT3 phosphorylation, and then the increases in CRF and Ucn2/3 mRNA levels. Leptin may contribute to a stress response or anorectic effect via the regulation of CRF and Ucn2/3 in the hypothalamus.
Collapse
Affiliation(s)
- Satoshi Yamagata
- Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | | | | | | | | | | |
Collapse
|
38
|
Sinchak K, Wagner EJ. Estradiol signaling in the regulation of reproduction and energy balance. Front Neuroendocrinol 2012; 33:342-63. [PMID: 22981653 PMCID: PMC3496056 DOI: 10.1016/j.yfrne.2012.08.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 08/18/2012] [Accepted: 08/22/2012] [Indexed: 12/14/2022]
Abstract
Our knowledge of membrane estrogenic signaling mechanisms and their interactions that regulate physiology and behavior has grown rapidly over the past three decades. The discovery of novel membrane estrogen receptors and their signaling mechanisms has started to reveal the complex timing and interactions of these various signaling mechanisms with classical genomic steroid actions within the nervous system to regulate physiology and behavior. The activation of the various estrogenic signaling mechanisms is site specific and differs across the estrous cycle acting through both classical genomic mechanisms and rapid membrane-initiated signaling to coordinate reproductive behavior and physiology. This review focuses on our current understanding of estrogenic signaling mechanisms to promote: (1) sexual receptivity within the arcuate nucleus of the hypothalamus, (2) estrogen positive feedback that stimulates de novo neuroprogesterone synthesis to trigger the luteinizing hormone surge important for ovulation and estrous cyclicity, and (3) alterations in energy balance.
Collapse
Affiliation(s)
- Kevin Sinchak
- Department of Biological Sciences, California State University, Long Beach, 1250 Bellflower Blvd., Long Beach, CA 90840-9502, United States.
| | | |
Collapse
|
39
|
Nikolic M, Boban M, Ljubicic N, Supanc V, Mirosevic G, Pezo Nikolic B, Krpan R, Posavec L, Zjacic-Rotkvic V, Bekavac-Beslin M, Gacina P. Morbidly obese are ghrelin and leptin hyporesponders with lesser intragastric balloon treatment efficiency : ghrelin and leptin changes in relation to obesity treatment. Obes Surg 2012; 21:1597-604. [PMID: 21494811 DOI: 10.1007/s11695-011-0414-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Ghrelin and leptin recently emerged as the most influential neuroendocrine factors in the pathophysiology of obesity. The said peptides act in reciprocity and are responsible for regulation of appetite and energy metabolism. Intragastric balloons acquired worldwide popularity for obesity treatment. However, the roles of ghrelin and leptin in intragastric balloon treatment were still not systematically studied. METHODS A prospective single-center study included 43 Caucasians treated with BioEnterics intragastric balloon, with age range of 18-60, and divided to non-morbid (body mass index cutoff 40 kg/m(2)) or morbid type of obesity, with 12 months follow-up. Serum hormonal samples were taken from fasting patients and kept frozen until analyses. RESULTS Significant differences were observed in anthropometrics and there were no differences between genders or comorbidities. The baseline weight for non-morbid vs. morbid was 104 kg (90-135) vs. 128.5 kg (104-197). Weight loss was statistically different between the studied groups during the study course with a median control weight at 6 months of 92 kg (72-121) vs. 107 kg (84-163), p < 0.001. Treatment was successful for 18 (94.7%) vs. 16 (66.7%) patients, p = 0.026. Ghrelin varied from 333.3 to 3,416.8 pg/ml and leptin from 1.7 to 61.2 ng/ml, with a statistically significant time-dependent relationship. A significant difference (p = 0.04) with emphasized ghrelin peak was found in the 3rd month of treatment for non-morbidly obese subjects. CONCLUSIONS The importance of ghrelin and leptin in treatment-induced changes was reaffirmed. Ghrelin hyper-response in non-morbidly obese subjects characterized greater short-term treatment efficiency and landmarked an inclination to weight regain. The results suggest a potential pattern of individualization between obese patients according to body mass index towards intragastric balloon or bariatric surgery. Further studies are needed in order to get better insights in the pathophysiologic mechanisms of obesity.
Collapse
Affiliation(s)
- Marko Nikolic
- Interventional Gastroenterology Unit, Department of Internal Medicine, University Hospital Centre "Sestre Milosrdnice", Vinogradska 29, Zagreb, 10000, Croatia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Schulz C, Paulus K, Jöhren O, Lehnert H. Intranasal leptin reduces appetite and induces weight loss in rats with diet-induced obesity (DIO). Endocrinology 2012; 153:143-53. [PMID: 22128019 DOI: 10.1210/en.2011-1586] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Resistance to brain-mediated effects of leptin is a characteristic feature of obesity, resulting from alterations in leptin receptor signaling in hypothalamic neurons and/or transport across the blood-brain-barrier. We have shown previously, that the latter can be circumvented by intranasal (i.n.) application of leptin in lean rats. This prompted us to test i.n. leptin in animals with diet-induced obesity (DIO) as a basis for future human administration. DIO was induced in male Wistar rats by feeding a cafeteria diet for 25 or 32 wk, respectively. Consecutively, these DIO animals (seven to eight per treatment) and standard diet rats (lean) (14-15 per treatment, matched for age and diet duration) were treated with 0.1, 0.2 mg/kg leptin, or control solution i.n. daily for 4 wk before onset of dark period. Energy intake and body weight were measured daily; blood glucose, serum insulin, and leptin were measured before and after treatment. Expression of hypothalamic neuropeptides was assessed by quantitative real-time PCR. We demonstrate, for the first time, that i.n. leptin reduces appetite and induces weight loss in DIO to the same extent as in lean rats. Our findings are supported accordingly by an altered expression pattern of anorexigenic and orexigenic neuropeptides in the hypothalamus, e.g. proopiomelanocortin, cocaine and amphetamine-related transcript, neuropeptide Y, agouti-related protein. It now appears clear that i.n. leptin is effectively acting in obese animals in the same fashion as in their lean counterparts. These findings now clearly warrant studies in humans and may open new perspectives in the treatment of obesity.
Collapse
Affiliation(s)
- Carla Schulz
- Department of Internal Medicine I, Luebeck University, Ratzeburger Allee 160, 23538 Luebeck, Germany.
| | | | | | | |
Collapse
|
41
|
Owyang C, Heldsinger A. Vagal control of satiety and hormonal regulation of appetite. J Neurogastroenterol Motil 2011; 17:338-48. [PMID: 22148102 PMCID: PMC3228973 DOI: 10.5056/jnm.2011.17.4.338] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 09/10/2011] [Accepted: 09/15/2011] [Indexed: 12/27/2022] Open
Abstract
The paradigm for the control of feeding behavior has changed significantly. In this review, we present evidence that the separation of function in which cholecystokinin (CCK) controls short-term food intake and leptin regulate long-term eating behavior and body weight become less clear. In addition to the hypothalamus, the vagus nerve is critically involved in the control of feeding by transmitting signals arising from the upper gut to the nucleus of the solitary tract. Among the peripheral mediators, CCK is the key peptide involved in generating the satiety signal via the vagus. Leptin receptors have also been identified in the vagus nerve. Studies in the rodents clearly indicate that leptin and CCK interact synergistically to induce short-term inhibition of food intake and long-term reduction of body weight. The synergistic interaction between vagal CCK-A receptor and leptin is mediated by the phosphorylation of signal transducer and activator of transcription3 (STAT3), which in turn, activates closure of K(+) channels, leading to membrane depolarization and neuronal firing. This involves the interaction between CCK/SRC/phosphoinositide 3-kinase cascades and leptin/Janus kinase-2/phosphoinositide 3-kinase/STAT3 signaling pathways. It is conceivable that malfunctioning of these signaling molecules may result in eating disorders.
Collapse
Affiliation(s)
- Chung Owyang
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA.
| | | |
Collapse
|
42
|
Zhao ZJ. Serum leptin, energy budget, and thermogenesis in striped hamsters exposed to consecutive decreases in ambient temperatures. Physiol Biochem Zool 2011; 84:560-72. [PMID: 22030849 DOI: 10.1086/662553] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Leptin has been found to be a direct participant in the regulation of both energy intake and energy expenditure in small mammals showing seasonal declines in body mass (M(b)) and fat mass, but its roles in an animal exhibiting seasonally increased thermogenesis and unchanged M(b) remain unclear. Serum leptin levels, energy budget, and thermogenesis were measured in striped hamsters exposed to consecutive decreases in ambient temperatures ranging from 23° to -23°C. Cold-exposed hamsters had significant increases in gross energy intake (GEI), the rate of basal metabolism, nonshivering thermogenesis, and activity of cytochrome c oxidase (COX) in brown adipose tissue (BAT), compared with control hamsters, indicating a cold-induced elevation of thermogenesis. Body mass and fat content were decreased in cold-exposed animals, and serum leptin levels were increased in hamsters exposed to temperatures of -8°C and below in inverse proportion to body fat content. Serum leptin levels were positively correlated with GEI and BAT COX activity in cold-exposed hamsters, but no such relationships were observed in control animals. These findings suggest that cold-exposed hamsters increase food consumption to meet the energy requirements for increased BAT thermogenesis. The increases in serum leptin levels are likely involved in increased thermogenesis in hamsters under cold stress. Cold-exposed hamsters may become leptin resistant, which is associated with impaired regulation of food intake. This new natural model of leptin resistance may also provide insight into the dynamic long-term control of energy homeostasis for animals that do not exhibit seasonal decline in M(b).
Collapse
Affiliation(s)
- Zhi-Jun Zhao
- School of Agricultural Science, Liaocheng University, Liaocheng, Shandong 252059, China.
| |
Collapse
|
43
|
Abstract
Rodent models of fatty liver disease are essential research tools that provide a window into disease pathogenesis and a testing ground for prevention and treatment. Models come in many varieties involving dietary and genetic manipulations, and sometimes both. High-energy diets that induce obesity do not uniformly cause fatty liver disease; this has prompted close scrutiny of specific macronutrients and nutrient combinations to determine which have the greatest potential for hepatotoxicity. At the same time, diets that do not cause obesity or the metabolic syndrome but do cause severe steatohepatitis have been exploited to study factors important to progressive liver injury, including cell death, oxidative stress, and immune activation. Rodents with a genetic predisposition to overeating offer yet another model in which to explore the evolution of fatty liver disease. In some animals that overeat, steatohepatitis can develop even without resorting to a high-energy diet. Importantly, these models and others have been used to document that aerobic exercise can prevent or reduce fatty liver disease. This review focuses primarily on lessons learned about steatohepatitis from manipulations of diet and eating behavior. Numerous additional insights about hepatic lipid metabolism, which have been gained from genetically engineered mice, are also mentioned.
Collapse
Affiliation(s)
- Jacquelyn J Maher
- Liver Center and Department of Medicine, University of California, San Francisco San Francisco, California, USA.
| |
Collapse
|
44
|
Buchbinder S, Bärtsch U, Müller M, Zorn M, Nawroth PP, Schilling T. A novel missense mutation T101N in the melanocortin-4 receptor gene associated with obesity. GENETICS AND MOLECULAR RESEARCH 2011; 10:1042-9. [PMID: 21710454 DOI: 10.4238/vol10-2gmr948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Mutations in the melanocortin-4 receptor (MC4R) are associated with severe obesity, independent of their effect on cortisol or thyroid-stimulating hormone levels. We examined a morbidly obese male (BMI = 62 kg/m²) with a binge-eating disorder and eight family members for mutations in the MC4R gene and potential differences in leptin levels. Fifty healthy individuals served as controls. Sequence analysis revealed a novel heterozygous missense mutation (c.302 C>A, p.T101N) located in the second transmembrane domain of the receptor, which was not detected in controls. The Fisher exact test revealed an association between the T101N mutation and history of obesity (P < 0.05) in the family. The Kruskal-Wallis test showed an association between the mutation and the leptin/BMI ratio (P < 0.05), while there was no association between the T101N mutation and diabetes or arterial hypertension in the family. Although the available family was small, we could show a significant association between the heterozygous T101N mutation and obesity.
Collapse
Affiliation(s)
- S Buchbinder
- Department of Internal Medicine 1 and Clinical Chemistry, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
45
|
Donahoo WT, Stob NR, Ammon S, Levin N, Eckel RH. Leptin increases skeletal muscle lipoprotein lipase and postprandial lipid metabolism in mice. Metabolism 2011; 60:438-43. [PMID: 20494377 DOI: 10.1016/j.metabol.2010.03.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2009] [Revised: 11/23/2009] [Accepted: 03/23/2010] [Indexed: 11/16/2022]
Abstract
The ability of leptin to preserve lean tissue during weight loss may be in part due to differences in nutrient partitioning. Because lipoprotein lipase (LPL) plays a key role in partitioning lipid nutrients, this study was conducted to test the hypothesis that leptin would modify the tissue-specific regulation of LPL and result in increased lipid oxidation and decreased storage. The effects of daily intraperitoneal leptin injections (2 mg/kg body weight) over 2 weeks on LPL activity and postprandial lipid metabolism were tested in both wild-type (WT), leptin-deficient ob/ob obese mice and mice pair fed to the leptin-treated mice. On the experimental day, mice were given food by gavage, blood was drawn periodically, and adipose tissue and skeletal muscle were harvested for measurements of LPL activity at 240 minutes. After 2 weeks of leptin administration, skeletal muscle LPL (SMLPL) activity was increased in leptin-treated compared with pair-fed (P = .012) and WT (P = .002) mice. There was no effect of leptin or pair feeding on postprandial adipose tissue LPL activity. In ob/ob mice, leptin treatment normalized the decrease in postprandial free fatty acid concentration (P = .066). Leptin had no effect on either the area under the triglyceride (TG) excursion or the integrated area under the TG excursion in WT mice. In ob/ob mice, however, the TG excursion was lower in the leptin-treated than the pair-fed mice by area under the TG excursion (P = .012) and was lower than in the WT mice by integrated area under the TG excursion (P = .027). As expected, 2 weeks of leptin treatment decreased body weight in both the WT and ob/ob mice (-2.6% and -10.4%, respectively). Leptin treatment increased SMLPL, an effect that may have contributed to the leptin-induced weight loss. The leptin-induced decreased postprandial TG excursion in ob/ob mice suggests that leptin acts to augment clearance of postprandial TG-rich lipoprotein lipid and that this increase may in part be secondary to the increased activity of SMLPL. The trend for decreased postprandial free fatty acid may indicate that leptin decreases adipose tissue lipid stores without increasing lipolysis.
Collapse
Affiliation(s)
- William T Donahoo
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Denver, 12801 East 17th Avenue, Mail Stop 8106, Aurora, CO 80045, USA
| | | | | | | | | |
Collapse
|
46
|
Kim KY, Kim JY, Sung YY, Jung WH, Kim HY, Park JS, Cheon HG, Rhee SD. Inhibitory effect of leptin on rosiglitazone-induced differentiation of primary adipocytes prepared from TallyHO/Jng mice. Biochem Biophys Res Commun 2011; 406:584-9. [PMID: 21352814 DOI: 10.1016/j.bbrc.2011.02.095] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Accepted: 02/19/2011] [Indexed: 11/25/2022]
Abstract
The effects of leptin on rosiglitazone-induced adipocyte differentiation were investigated in the primary adipocytes prepared from subcutaneous fat of TallyHO/Jng (TallyHO) mouse, a recently developed model animal for type 2 diabetes mellitus (T2DM). The treatment of leptin inhibited the rosiglitazone-induced adipocyte differentiation with a decreased expression of peroxisome proliferator-activated receptor γ (PPARγ) a key adipogenic transcription factor, both in mRNA and protein levels. Leptin (10 nM) was sufficient to inhibit the adipocyte differentiation, which seemed to come from increased expression of leptin receptor genes in the fat of TallyHO mice. The inhibition of adipogenesis by leptin was restored by the treatment of inhibitors for extracellular-signal-regulated kinase (ERK) (PD98059) and signal transducer and activator of transcription-1 (STAT1) (fludarabine). Furthermore, in vivo intraperitoneal administration of PD98059 and fludarabine increased the PPARγ expression in the subcutaneous fat of TallyHO mice. These data suggest that leptin could inhibit the PPARγ expression and adipocyte differentiation in its physiological concentration in TallyHO mice.
Collapse
Affiliation(s)
- Ki Young Kim
- Medicinal Science Division, Korea Research Institute of Chemical Technology, 100 Jang-dong, Yuseong, 305-600 Daejon, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Dilger AC, Spurlock ME, Grant AL, Gerrard DE. Myostatin null mice respond differently to dietary-induced and genetic obesity. Anim Sci J 2011; 81:586-93. [PMID: 20887312 DOI: 10.1111/j.1740-0929.2010.00776.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Our objective was to determine sensitivity of myostatin null (MN) mice to obesity induction by dietary or genetic means. To induce dietary obesity, 3-week-old wild type (WT) and MN mice were fed diets with 60% calories (HF) or 10% calories from fat (LF) for 4 weeks. MN mice did gain body fat on the HF diet but to a lesser extent than WT mice. Body weight and fat content was similar in MN mice fed HF and LF diets. To induce genetic obesity, the MN mutation was incorporated into leptin db/db (DB) mice generating mice homozygous for each mutation (MNDB). Nine-week-old MNDB mice were obese, similar to DB mice. Body weight, body fat content, fat pad weight and adipocyte size were all increased in MNDB mice compared to MN and WT mice and were quite similar to DB mice. However, fasting blood glucose, an indicator of insulin resistance and diabetes, was reduced in MNDB mice compared to DB mice. These results indicate that MN mice gain less body fat than WT on a HF diet, but the MN mutation does not alter fat accumulation caused by DB mutation. Thus, MN mice are not always resistant to obesity development.
Collapse
Affiliation(s)
- Anna C Dilger
- Department of Animal Sciences, Purdue University, W Lafayette, Indiana, USA.
| | | | | | | |
Collapse
|
48
|
Mendoza J, Lopez-Lopez C, Revel FG, Jeanneau K, Delerue F, Prinssen E, Challet E, Moreau JL, Grundschober C. Dimorphic effects of leptin on the circadian and hypocretinergic systems of mice. J Neuroendocrinol 2011; 23:28-38. [PMID: 20874776 DOI: 10.1111/j.1365-2826.2010.02072.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The hormone leptin controls food intake and body weight through its receptor in the hypothalamus, and may modulate physiological functions such as reproduction, sleep or circadian timing. In the present study, the effects of leptin on the resetting of the circadian clock, the hypothalamic suprachiasmatic nucleus (SCN) and on the activity of the hypocretinergic system were examined in vivo, with comparative analysis between male and female mice. A single leptin injection (5 mg/kg) at both the onset and offset of the activity period did not alter locomotion of mice housed under a 12 : 12 h light/dark cycle and did not shift the circadian behavioral rhythm of mice housed in constant darkness. By contrast, leptin potentiated the phase-shifting effect of a 30-min light-pulse on behavioural rhythms during the late subjective night, although only in females. This was accompanied by a higher induction of the clock genes Per1 and Per2 in the SCN. A 2-week chronic exposure to a physiological dose of leptin (100 μg/kg per day) decreased locomotor activity, expression of hypocretin receptor 1 and 2, as well as the number of hypocretin-immunoreactive neurones only in female mice, whereas the number of c-fos-positive hypocretinergic neurones was reduced in both genders. These results highlight a dimorphic effect of leptin on the hypocretinergic system and on the response of the circadian clock to light. Leptin may thus modulate the sleep/wake cycle and circadian system beside its well-established action on food intake and regulation of body weight.
Collapse
Affiliation(s)
- J Mendoza
- Department of Neurobiology of Rhythms, Institute of Cellular and Integrative Neurosciences, Centre National de la Recherche Scientifique, UPR3212 University of Strasbourg, Strasbourg, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Tsutsumi A, Motoshima H, Kondo T, Kawasaki S, Matsumura T, Hanatani S, Igata M, Ishii N, Kinoshita H, Kawashima J, Taketa K, Furukawa N, Tsuruzoe K, Nishikawa T, Araki E. Caloric restriction decreases ER stress in liver and adipose tissue in ob/ob mice. Biochem Biophys Res Commun 2010; 404:339-44. [PMID: 21134353 DOI: 10.1016/j.bbrc.2010.11.120] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Accepted: 11/26/2010] [Indexed: 11/26/2022]
Abstract
Endoplasmic reticulum (ER) stress plays a crucial role in the development of insulin resistance and diabetes. Although caloric restriction (CR) improves obesity-related disorders, the effects of CR on ER stress in obesity remain unknown. To investigate how CR affects ER stress in obesity, ob/ob mice were assigned to either ad libitum (AL) (ob-AL) or CR (ob-CR) feeding (2 g food/day) for 1-4 weeks. The body weight (BW) of ob-CR mice decreased to the level of lean AL-fed littermates (lean-AL) within 2 weeks. BW of lean-AL and ob-CR mice was less than that of ob-AL mice. The ob-CR mice showed improved glucose tolerance and hepatic insulin action compared with ob-AL mice. Levels of ER stress markers such as phosphorylated PKR-like ER kinase (PERK) and eukaryotic translation initiation factor 2α and the mRNA expression of activating transcription factor 4 were significantly higher in the liver and epididymal fat from ob-AL mice compared with lean-AL mice. CR for 2 and 4 weeks significantly reduced all of these markers to less than 35% and 50%, respectively, of the levels in ob-AL mice. CR also significantly reduced the phosphorylation of insulin receptor substrate (IRS)-1 and c-Jun NH(2)-terminal kinase (JNK) in ob/ob mice. The CR-mediated decrease in PERK phosphorylation was similar to that induced by 4-phenyl butyric acid, which reduces ER stress in vivo. In conclusion, CR reduced ER stress and improved hepatic insulin action by suppressing JNK-mediated IRS-1 serine-phosphorylation in ob/ob mice.
Collapse
Affiliation(s)
- Atsuyuki Tsutsumi
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Sharma AN, Elased KM, Garrett TL, Lucot JB. Neurobehavioral deficits in db/db diabetic mice. Physiol Behav 2010; 101:381-8. [PMID: 20637218 DOI: 10.1016/j.physbeh.2010.07.002] [Citation(s) in RCA: 141] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Revised: 07/07/2010] [Accepted: 07/08/2010] [Indexed: 01/02/2023]
Abstract
Recent clinical studies indicate neurobehavioral disturbances in type-2 diabetics. However, there is paucity of preclinical research to support this concept. The validity of db/db mouse as an animal model to study type-2 diabetes and related complications is known. The present study was designed to investigate comprehensively the db/db mouse behavior as preclinical evidence of type-2 diabetes related major neurobehavioral complications. We tested juvenile (5-6weeks) and adult (10-11weeks) db/db mice for behavioral depression in forced swim test (FST), psychosis-like symptoms using pre-pulse inhibition (PPI) test, anxiety behavior employing elevated plus maze (EPM) test, locomotor behavior and thigmotaxis using open field test and working memory deficits in Y-maze test. Both juvenile and adult group db/db mice displayed behavioral despair with increased immobility time in FST. There was an age-dependent progression of psychosis-like symptoms with disrupted PPI in adult db/db mice. In the EPM test, db/db mice were less anxious as observed by increased percent open arms time and entries. They were also hypo-locomotive as evident by a decrease in their basic and fine movements. There was no impairment of working memory in the Y-maze test in db/db mice. This is the first report of depression, psychosis-like symptoms and anxiolytic behavior of db/db mouse strain. It is tempting to speculate that this mouse strain can serve as useful preclinical model to study type-2 diabetes related neurobehavioral complications.
Collapse
Affiliation(s)
- Ajaykumar N Sharma
- Wright State University, Boonshoft School of Medicine, Department of Pharmacology and Toxicology, Dayton, OH 45435, USA
| | | | | | | |
Collapse
|