1
|
Asgari R, Caceres-Valdiviezo M, Wu S, Hamel L, Humber BE, Agarwal SM, Fletcher PJ, Fulton S, Hahn MK, Pereira S. Regulation of energy balance by leptin as an adiposity signal and modulator of the reward system. Mol Metab 2024; 91:102078. [PMID: 39615837 DOI: 10.1016/j.molmet.2024.102078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/02/2024] [Accepted: 11/26/2024] [Indexed: 12/08/2024] Open
Abstract
BACKGROUND Leptin is an adipose tissue-derived hormone that plays a crucial role in body weight, appetite, and behaviour regulation. Leptin controls energy balance as an indicator of adiposity levels and as a modulator of the reward system, which is associated with liking palatable foods. Obesity is characterized by expanded adipose tissue mass and consequently, elevated concentrations of leptin in blood. Leptin's therapeutic potential for most forms of obesity is hampered by leptin resistance and a narrow dose-response window. SCOPE OF REVIEW This review describes the current knowledge of the brain regions and intracellular pathways through which leptin promotes negative energy balance and restrains neural circuits affecting food reward. We also describe mechanisms that hinder these biological responses in obesity and highlight potential therapeutic interventions. MAJOR CONCLUSIONS Additional research is necessary to understand how pathways engaged by leptin in different brain regions are interconnected in the control of energy balance.
Collapse
Affiliation(s)
| | - Maria Caceres-Valdiviezo
- Centre for Addiction and Mental Health, Toronto, ON, Canada; Laboratory of Omic Sciences, School of Medicine, Universidad de Especialidades Espíritu Santo, Samborondón, Ecuador
| | - Sally Wu
- Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Laurie Hamel
- Centre for Addiction and Mental Health, Toronto, ON, Canada
| | | | - Sri Mahavir Agarwal
- Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Banting & Best Diabetes Centre, University of Toronto, Toronto, ON, Canada
| | - Paul J Fletcher
- Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Department of Psychology, University of Toronto, Toronto, ON, Canada
| | - Stephanie Fulton
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal Diabetes Research Center, Montréal, QC, Canada; Department of Nutrition, Université de Montréal, QC, Canada
| | - Margaret K Hahn
- Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Banting & Best Diabetes Centre, University of Toronto, Toronto, ON, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada; Department of Pharmacology, University of Toronto, Toronto, ON, Canada.
| | - Sandra Pereira
- Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
2
|
Perez-Leighton C, Kerr B, Scherer PE, Baudrand R, Cortés V. The interplay between leptin, glucocorticoids, and GLP1 regulates food intake and feeding behaviour. Biol Rev Camb Philos Soc 2024; 99:653-674. [PMID: 38072002 DOI: 10.1111/brv.13039] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 11/21/2023] [Accepted: 12/01/2023] [Indexed: 05/09/2024]
Abstract
Nutritional, endocrine, and neurological signals converge in multiple brain centres to control feeding behaviour and food intake as part of the allostatic regulation of energy balance. Among the several neuroendocrine systems involved, the leptin, glucocorticoid, and glucagon-like peptide 1 (GLP1) systems have been extensively researched. Leptin is at the top hierarchical level since its complete absence is sufficient to trigger severe hyperphagia. Glucocorticoids are key regulators of the energy balance adaptation to stress and their sustained excess leads to excessive adiposity and metabolic perturbations. GLP1 participates in metabolic adaptation to food intake, regulating insulin secretion and satiety by parallel central and peripheral signalling systems. Herein, we review the brain and peripheral targets of these three hormone systems that integrate to regulate food intake, feeding behaviour, and metabolic homeostasis. We examine the functional relationships between leptin, glucocorticoids, and GLP1 at the central and peripheral levels, including the cross-regulation of their circulating levels and their cooperative or antagonistic actions at different brain centres. The pathophysiological roles of these neuroendocrine systems in dysregulated intake are explored in the two extremes of body adiposity - obesity and lipodystrophy - and eating behaviour disorders.
Collapse
Affiliation(s)
- Claudio Perez-Leighton
- Departmento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, 830024, Chile
| | - Bredford Kerr
- Centro de Biología Celular y Biomedicina-CEBICEM, Facultad de Medicina y Ciencia, Universidad San Sebastián, Carmen Sylva 2444, Providencia, Santiago, Chile
| | - Philipp E Scherer
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
| | - René Baudrand
- Departmento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, 830024, Chile
- Centro Translacional de Endocrinología (CETREN), Facultad de Medicina, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, 830024, Chile
| | - Víctor Cortés
- Departmento de Nutrición, Diabetes y Metabolismo, Facultad de Medicina, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, 830024, Chile
| |
Collapse
|
3
|
Heyward FD, Liu N, Jacobs C, Machado NLS, Ivison R, Uner A, Srinivasan H, Patel SJ, Gulko A, Sermersheim T, Tsai L, Rosen ED. AgRP neuron cis-regulatory analysis across hunger states reveals that IRF3 mediates leptin's acute effects. Nat Commun 2024; 15:4646. [PMID: 38821928 PMCID: PMC11143326 DOI: 10.1038/s41467-024-48885-y] [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: 12/08/2023] [Accepted: 05/14/2024] [Indexed: 06/02/2024] Open
Abstract
AgRP neurons in the arcuate nucleus of the hypothalamus (ARC) coordinate homeostatic changes in appetite associated with fluctuations in food availability and leptin signaling. Identifying the relevant transcriptional regulatory pathways in these neurons has been a priority, yet such attempts have been stymied due to their low abundance and the rich cellular diversity of the ARC. Here we generated AgRP neuron-specific transcriptomic and chromatin accessibility profiles from male mice during three distinct hunger states of satiety, fasting-induced hunger, and leptin-induced hunger suppression. Cis-regulatory analysis of these integrated datasets enabled the identification of 18 putative hunger-promoting and 29 putative hunger-suppressing transcriptional regulators in AgRP neurons, 16 of which were predicted to be transcriptional effectors of leptin. Within our dataset, Interferon regulatory factor 3 (IRF3) emerged as a leading candidate mediator of leptin-induced hunger-suppression. Measures of IRF3 activation in vitro and in vivo reveal an increase in IRF3 nuclear occupancy following leptin administration. Finally, gain- and loss-of-function experiments in vivo confirm the role of IRF3 in mediating the acute satiety-evoking effects of leptin in AgRP neurons. Thus, our findings identify IRF3 as a key mediator of the acute hunger-suppressing effects of leptin in AgRP neurons.
Collapse
Affiliation(s)
- Frankie D Heyward
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA.
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Nan Liu
- Cancer and Blood Disorders Center, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA, USA
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| | - Christopher Jacobs
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Natalia L S Machado
- Harvard Medical School, Boston, MA, USA
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Rachael Ivison
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Aykut Uner
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Harini Srinivasan
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Suraj J Patel
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Gastroenterology & Hepatology, UT Southwestern Medical Center, Dallas, TX, USA
- Center for Human Nutrition and Department of Internal Medicine, UT Southwestern Medical, Center, Dallas, TX, USA
| | - Anton Gulko
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Tyler Sermersheim
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Linus Tsai
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Evan D Rosen
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Gan HW, Cerbone M, Dattani MT. Appetite- and Weight-Regulating Neuroendocrine Circuitry in Hypothalamic Obesity. Endocr Rev 2024; 45:309-342. [PMID: 38019584 PMCID: PMC11074800 DOI: 10.1210/endrev/bnad033] [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: 02/04/2023] [Revised: 10/25/2023] [Accepted: 11/27/2023] [Indexed: 11/30/2023]
Abstract
Since hypothalamic obesity (HyOb) was first described over 120 years ago by Joseph Babinski and Alfred Fröhlich, advances in molecular genetic laboratory techniques have allowed us to elucidate various components of the intricate neurocircuitry governing appetite and weight regulation connecting the hypothalamus, pituitary gland, brainstem, adipose tissue, pancreas, and gastrointestinal tract. On a background of an increasing prevalence of population-level common obesity, the number of survivors of congenital (eg, septo-optic dysplasia, Prader-Willi syndrome) and acquired (eg, central nervous system tumors) hypothalamic disorders is increasing, thanks to earlier diagnosis and management as well as better oncological therapies. Although to date the discovery of several appetite-regulating peptides has led to the development of a range of targeted molecular therapies for monogenic obesity syndromes, outside of these disorders these discoveries have not translated into the development of efficacious treatments for other forms of HyOb. This review aims to summarize our current understanding of the neuroendocrine physiology of appetite and weight regulation, and explore our current understanding of the pathophysiology of HyOb.
Collapse
Affiliation(s)
- Hoong-Wei Gan
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Manuela Cerbone
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Mehul Tulsidas Dattani
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
5
|
De Luca SN, Kivivali L, Chong K, Kirby A, Lawther AJ, Nguyen JCD, Hale MW, Kent S. Calorie restriction partially attenuates sickness behavior induced by viral mimetic poly I:C. Behav Brain Res 2024; 457:114715. [PMID: 37838243 DOI: 10.1016/j.bbr.2023.114715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/28/2023] [Accepted: 10/11/2023] [Indexed: 10/16/2023]
Abstract
Calorie restriction (CR) has been shown to extend the mean and maximum lifespan in both preclinical and clinical settings. We have previously demonstrated that CR attenuates lipopolysaccharide (LPS)-induced fever and sickness behavior. CR also leads to reductions in pro-inflammatory and increases in anti-inflammatory profiles. LPS is a bacterial mimetic; however, few studies have explored this phenomenon utilizing a viral mimetic, such as polyinosinic:polycytidylic acid (poly I:C). Dose-dependently, poly I:C induced an increase in core body temperature (Tb), with the largest dose (5000 µg/kg) resulting in a 1.62 °C ( ± 0.23 °C) Tb increase at 7 h post-injection in ad libitum mice and was associated with reduced home-cage locomotor activity. We then investigated the effect of 50% CR for 28 days to attenuate fever and sickness behavior induced by a poly I:C (5000 µg/kg) viral immune challenge. CR resulted in the partial attenuation of fever and sickness behavior measures post-poly I:C. The freely fed, control mice demonstrated a 2.02 °C ( ± 0.22 °C) increase in Tb at 7 h post-injection compared to the CR poly I:C group which demonstrated an increase in Tb of 0.94 °C ( ± 0.27 °C). Locomotor patterns post-injection were different, CR mice displayed a reduction in activity during the light phase, and the control group displayed a reduction during the dark phase. CR moderately attenuated the neuroinflammatory response with a reduction in microglial density in the ventromedial nucleus of the hypothalamus. The fever and sickness behavior attenuation seen after CR may be driven by similar anti-inflammatory processes as after LPS; however, further investigation is required.
Collapse
Affiliation(s)
- Simone N De Luca
- School of Psychology & Public Health, La Trobe University, Melbourne, VIC, Australia; Centre for Respiratory Science & Health, School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Leah Kivivali
- School of Psychology & Public Health, La Trobe University, Melbourne, VIC, Australia
| | - Ken Chong
- School of Psychology & Public Health, La Trobe University, Melbourne, VIC, Australia
| | - Alice Kirby
- School of Psychology & Public Health, La Trobe University, Melbourne, VIC, Australia
| | - Adam J Lawther
- School of Psychology & Public Health, La Trobe University, Melbourne, VIC, Australia
| | - Jason C D Nguyen
- School of Psychology & Public Health, La Trobe University, Melbourne, VIC, Australia.
| | - Matthew W Hale
- School of Psychology & Public Health, La Trobe University, Melbourne, VIC, Australia
| | - Stephen Kent
- School of Psychology & Public Health, La Trobe University, Melbourne, VIC, Australia
| |
Collapse
|
6
|
Barrios-Nolasco A, Domínguez-López A, Miliar-García A, Cornejo-Garrido J, Jaramillo-Flores ME. Anti-Inflammatory Effect of Ethanolic Extract from Tabebuia rosea (Bertol.) DC., Quercetin, and Anti-Obesity Drugs in Adipose Tissue in Wistar Rats with Diet-Induced Obesity. Molecules 2023; 28:molecules28093801. [PMID: 37175211 PMCID: PMC10180162 DOI: 10.3390/molecules28093801] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/18/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Obesity is characterized by the excessive accumulation of fat, which triggers a low-grade chronic inflammatory process. Currently, the search for compounds with anti-obesogenic effects that help reduce body weight, as well as associated comorbidities, continues. Among this group of compounds are plant extracts and flavonoids with a great diversity of action mechanisms associated with their beneficial effects, such as anti-inflammatory effects and/or as signaling molecules. In the bark of Tabebuia rosea tree, there are different classes of metabolites with anti-inflammatory properties, such as quercetin. Therefore, the present work studied the effect of the ethanolic extract of T. rosea and quercetin on the mRNA of inflammation markers in obesity compared to the drugs currently used. Total RNA was extracted from epididymal adipose tissue of high-fat diet-induced obese Wistar rats treated with orlistat, phentermine, T. rosea extract, and quercetin. The rats treated with T. rosea and quercetin showed 36 and 31% reductions in body weight compared to the obese control, and they likewise inhibited pro-inflammatory molecules: Il6, Il1b, Il18, Lep, Hif1a, and Nfkb1 without modifying the expression of Socs1 and Socs3. Additionally, only T. rosea overexpressed Lipe. Both T. rosea and quercetin led to a reduction in the expression of pro-inflammatory genes, modifying signaling pathways, which led to the regulation of the obesity-inflammation state.
Collapse
Affiliation(s)
- Alejandro Barrios-Nolasco
- Laboratorio de Biología Celular y Productos Naturales, Escuela Nacional de Medicina y Homeopatía (ENMH), Instituto Politécnico Nacional, Guillermo Massieu Helguera 239, Col. La Escalera, Alcaldía Gustavo A. Madero, Ciudad de Mexico 07320, Mexico
| | - Aarón Domínguez-López
- Laboratorio de Biología Molecular, Escuela Superior de Medicina (ESM), Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Col. Casco de Santo Tomas, Alcaldía Miguel Hidalgo, Ciudad de Mexico 11340, Mexico
| | - Angel Miliar-García
- Laboratorio de Biología Molecular, Escuela Superior de Medicina (ESM), Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Col. Casco de Santo Tomas, Alcaldía Miguel Hidalgo, Ciudad de Mexico 11340, Mexico
| | - Jorge Cornejo-Garrido
- Laboratorio de Biología Celular y Productos Naturales, Escuela Nacional de Medicina y Homeopatía (ENMH), Instituto Politécnico Nacional, Guillermo Massieu Helguera 239, Col. La Escalera, Alcaldía Gustavo A. Madero, Ciudad de Mexico 07320, Mexico
| | - María Eugenia Jaramillo-Flores
- Laboratorio de Polímeros, Department de Ingeniería Bioquímica, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional, Wilfrido Massieu s/n esq. Manuel I. Stampa. Col. Unidad Profesional Adolfo López Mateos, Alcaldía Gustavo A. Madero, Ciudad de Mexico 07738, Mexico
| |
Collapse
|
7
|
Hristov M, Lazarov L. Inhibition of nitric oxide synthase or cystathionine gamma-lyase abolishes leptin-induced fever in male rats. J Therm Biol 2023; 112:103443. [PMID: 36796898 DOI: 10.1016/j.jtherbio.2022.103443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022]
Abstract
Leptin is an adipokine that regulates energy balance and immune function. Peripheral leptin administration elicits prostaglandin E₂-dependent fever in rats. The gasotransmitters nitric oxide (NO) and hydrogen sulfide (H₂S) are also involved in lipopolysaccharide (LPS)-induced fever response. However, there is no data in the literature indicating if these gasotransmitters have a role in leptin-induced fever response. Here, we investigate the inhibition of NO and H₂S enzymes neuronal nitric oxide synthase (nNOS), inducible nitric oxide synthase (iNOS), and cystathionine γ-lyase (CSE) in leptin-induced fever response, respectively. Selective nNOS inhibitor 7-nitroindazole (7-NI), selective iNOS inhibitor aminoguanidine (AG), and CSE inhibitor dl-propargylglycine (PAG) were administered intraperitoneally (ip). Body temperature (Tb), food intake, and body mass were recorded in fasted male rats. Leptin (0.5 mg/kg ip) induced a significant increase in Tb, whereas AG (50 mg/kg ip), 7-NI (10 mg/kg ip), or PAG (50 mg/kg ip) caused no changes in Tb. AG, 7-NI, or PAG abolished leptin increase in Tb. Our results highlight the potential involvement of iNOS, nNOS, and CSE in leptin-induced febrile response without affecting anorexic response to leptin in fasted male rats 24 h after leptin injection. Interestingly, all the inhibitors alone had the same anorexic effect induced by leptin. These findings have implications for understanding the role of NO and H₂S in leptin-induced febrile response.
Collapse
Affiliation(s)
- Milen Hristov
- Department of Pharmacology and Toxicology, Faculty of Medicine, Medical University of Sofia, 2 "Zdrave" St., 1431, Sofia, Bulgaria.
| | - Lyudmil Lazarov
- Department of Pharmacology and Toxicology, Faculty of Medicine, Medical University of Sofia, 2 "Zdrave" St., 1431, Sofia, Bulgaria
| |
Collapse
|
8
|
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
|
9
|
Namgoung S, Chang Y, Woo CY, Kim Y, Kang J, Kwon R, Lim GY, Choi HR, Kim KH, Kim H, Hong YS, Zhao D, Cho J, Guallar E, Park HY, Ryu S. Metabolically healthy and unhealthy obesity and risk of vasomotor symptoms in premenopausal women: cross-sectional and cohort studies. BJOG 2022; 129:1926-1934. [PMID: 35596933 PMCID: PMC9541406 DOI: 10.1111/1471-0528.17224] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 12/18/2022]
Abstract
Objective To examine the relationship between metabolically healthy and unhealthy obesity phenotypes and risk of vasomotor symptoms (VMS) in premenopausal women. Design Prospective cohort study. Setting Middle‐aged women in a cohort based on regular health screening examinations. Population Premenopausal Korean women aged 42–52 years were recruited and were followed up for a median of 4.2 years. The cross‐sectional and cohort studies comprised 4672 women and 2590 women without VMS at baseline, respectively. Methods Adiposity measures included body mass index (BMI), waist circumference and percentage body fat. Being metabolically healthy was defined as not having any metabolic syndrome components or a homeostasis model assessment of insulin resistance of 2.5 or more. Main outcomes measures VMS (hot flushes and night sweats) assessed using the questionnaire. Results All adiposity measures were positively associated with an increased risk of VMS in both cross‐sectional and longitudinal studies. The multivariable‐adjusted prevalence ratio (95% confidence interval [CI]) for VMS comparing percentage body fat of 35% or more with the reference was 1.47 (95% CI 1.14–1.90) in metabolically healthy women, and the corresponding prevalence ratio was 2.32 (95% CI 1.42–3.78) in metabolically unhealthy women (Pinteraction = 0.334). The multivariable‐adjusted hazard ratio for incident VMS comparing percentage body fat of 35% or more with the reference was 1.34 (95% CI 1.00–1.79) in metabolically healthy women, whereas the corresponding hazard ratio was 3.61 (95% CI 1.81–7.20) in metabolically unhealthy women (Pinteraction = 0.036). The association between BMI, waist circumference and VMS did not significantly differ by metabolic health status. Conclusions Maintaining normal weight and being metabolically healthy may help to prevent VMS in premenopausal women. Tweetable abstract Avoiding obesity and a metabolically unhealthy status may help reduce vasomotor symptoms in premenopausal women. Avoiding obesity and a metabolically unhealthy status may help reduce vasomotor symptoms in premenopausal women.
Collapse
Affiliation(s)
- Sunju Namgoung
- Centre for Cohort Studies, Total Healthcare Centre, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of The Environmental Health Centre, Wonju Severance Christian Hospital, Yonsei University School of Medicine, Wonju, Korea
| | - Yoosoo Chang
- Centre for Cohort Studies, Total Healthcare Centre, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Occupational and Environmental Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Chae-Yeon Woo
- Centre for Cohort Studies, Total Healthcare Centre, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yejin Kim
- Centre for Cohort Studies, Total Healthcare Centre, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jeonggyu Kang
- Centre for Cohort Studies, Total Healthcare Centre, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ria Kwon
- Centre for Cohort Studies, Total Healthcare Centre, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea.,Institute of Medical Research, Sungkyunkwan University, School of Medicine, Suwon, Republic of Korea
| | - Ga-Young Lim
- Centre for Cohort Studies, Total Healthcare Centre, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea.,Institute of Medical Research, Sungkyunkwan University, School of Medicine, Suwon, Republic of Korea
| | - Hye Rin Choi
- Centre for Cohort Studies, Total Healthcare Centre, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea.,Institute of Medical Research, Sungkyunkwan University, School of Medicine, Suwon, Republic of Korea
| | - Kye-Hyun Kim
- Department of Obstetrics and Gynaecology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hoon Kim
- Department of Obstetrics and Gynaecology, Seoul National University College of Medicine, Seoul, Korea
| | - Yun Soo Hong
- Departments of Epidemiology and Medicine, and Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Di Zhao
- Departments of Epidemiology and Medicine, and Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Juhee Cho
- Departments of Epidemiology and Medicine, and Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA.,Department of Clinical Research Design & Evaluation, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Eliseo Guallar
- Departments of Epidemiology and Medicine, and Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Hyun-Young Park
- Department of Precision Medicine, National Institute of Health, Korea Disease Control and Prevention Agency (KDCA), Cheongju, Korea
| | - Seungho Ryu
- Centre for Cohort Studies, Total Healthcare Centre, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Occupational and Environmental Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
10
|
Niu X, Ding Y, Chen S, Gooneratne R, Ju X. Effect of Immune Stress on Growth Performance and Immune Functions of Livestock: Mechanisms and Prevention. Animals (Basel) 2022; 12:ani12070909. [PMID: 35405897 PMCID: PMC8996973 DOI: 10.3390/ani12070909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/19/2022] [Accepted: 03/26/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Immune stress is an important stressor in domestic animals that leads to decreased feed intake, slow growth, and reduced disease resistance of pigs and poultry. Especially in high-density animal feeding conditions, the risk factor of immune stress is extremely high, as they are easily harmed by pathogens, and frequent vaccinations are required to enhance the immunity function of the animals. This review mainly describes the causes, mechanisms of immune stress and its prevention and treatment measures. This provides a theoretical basis for further research and development of safe and efficient prevention and control measures for immune stress in animals. Abstract Immune stress markedly affects the immune function and growth performance of livestock, including poultry, resulting in financial loss to farmers. It can lead to decreased feed intake, reduced growth, and intestinal disorders. Studies have shown that pathogen-induced immune stress is mostly related to TLR4-related inflammatory signal pathway activation, excessive inflammatory cytokine release, oxidative stress, hormonal disorders, cell apoptosis, and intestinal microbial disorders. This paper reviews the occurrence of immune stress in livestock, its impact on immune function and growth performance, and strategies for immune stress prevention.
Collapse
Affiliation(s)
- Xueting Niu
- Department of Veterinary Medicine, Guangdong Ocean University, Zhanjiang 524088, China; (X.N.); (Y.D.); (S.C.)
- Marine Medical Research and Development Centre, Shenzhen Institute of Guangdong Ocean University, Shenzhen 518018, China
| | - Yuexia Ding
- Department of Veterinary Medicine, Guangdong Ocean University, Zhanjiang 524088, China; (X.N.); (Y.D.); (S.C.)
| | - Shengwei Chen
- Department of Veterinary Medicine, Guangdong Ocean University, Zhanjiang 524088, China; (X.N.); (Y.D.); (S.C.)
- Marine Medical Research and Development Centre, Shenzhen Institute of Guangdong Ocean University, Shenzhen 518018, China
| | - Ravi Gooneratne
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln 7647, New Zealand;
| | - Xianghong Ju
- Department of Veterinary Medicine, Guangdong Ocean University, Zhanjiang 524088, China; (X.N.); (Y.D.); (S.C.)
- Marine Medical Research and Development Centre, Shenzhen Institute of Guangdong Ocean University, Shenzhen 518018, China
- Correspondence:
| |
Collapse
|
11
|
Gonzalez A, Cheung WW, Perens EA, Oliveira EA, Gertler A, Mak RH. A Leptin Receptor Antagonist Attenuates Adipose Tissue Browning and Muscle Wasting in Infantile Nephropathic Cystinosis-Associated Cachexia. Cells 2021; 10:1954. [PMID: 34440723 PMCID: PMC8393983 DOI: 10.3390/cells10081954] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/19/2021] [Accepted: 07/24/2021] [Indexed: 12/12/2022] Open
Abstract
Mice lacking the functional cystinosin gene (Ctns-/-), a model of infantile nephropathic cystinosis (INC), exhibit the cachexia phenotype with adipose tissue browning and muscle wasting. Elevated leptin signaling is an important cause of chronic kidney disease-associated cachexia. The pegylated leptin receptor antagonist (PLA) binds to but does not activate the leptin receptor. We tested the efficacy of this PLA in Ctns-/- mice. We treated 12-month-old Ctns-/- mice and control mice with PLA (7 mg/kg/day, IP) or saline as a vehicle for 28 days. PLA normalized food intake and weight gain, increased fat and lean mass, decreased metabolic rate and improved muscle function. It also attenuated perturbations of energy homeostasis in adipose tissue and muscle in Ctns-/- mice. PLA attenuated adipose tissue browning in Ctns-/- mice. PLA increased gastrocnemius weight and fiber size as well as attenuated muscle fat infiltration in Ctns-/- mice. This was accompanied by correcting the increased expression of muscle wasting signaling while promoting the decreased expression of myogenesis in gastrocnemius of Ctns-/- mice. PLA attenuated aberrant expressed muscle genes that have been associated with muscle atrophy, increased energy expenditure and lipolysis in Ctns-/- mice. Leptin antagonism may represent a viable therapeutic strategy for adipose tissue browning and muscle wasting in INC.
Collapse
MESH Headings
- Adipose Tissue, Brown/drug effects
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, Brown/pathology
- Adipose Tissue, White/drug effects
- Adipose Tissue, White/metabolism
- Adipose Tissue, White/pathology
- Amino Acid Transport Systems, Neutral/genetics
- Amino Acid Transport Systems, Neutral/metabolism
- Animals
- Body Composition/drug effects
- Cachexia/etiology
- Cachexia/metabolism
- Cachexia/pathology
- Cachexia/prevention & control
- Cystinosis/complications
- Cystinosis/drug therapy
- Cystinosis/metabolism
- Cystinosis/pathology
- Disease Models, Animal
- Hormone Antagonists/pharmacology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Atrophy/etiology
- Muscular Atrophy/metabolism
- Muscular Atrophy/pathology
- Muscular Atrophy/prevention & control
- Receptors, Leptin/antagonists & inhibitors
- Receptors, Leptin/metabolism
- Signal Transduction
- Mice
Collapse
Affiliation(s)
- Alex Gonzalez
- Division of Pediatric Nephrology, Rady Children’s Hospital, University of California, San Diego, CA 92093-0831, USA; (A.G.); (W.W.C.); (E.A.P.); (E.A.O.)
| | - Wai W. Cheung
- Division of Pediatric Nephrology, Rady Children’s Hospital, University of California, San Diego, CA 92093-0831, USA; (A.G.); (W.W.C.); (E.A.P.); (E.A.O.)
| | - Elliot A. Perens
- Division of Pediatric Nephrology, Rady Children’s Hospital, University of California, San Diego, CA 92093-0831, USA; (A.G.); (W.W.C.); (E.A.P.); (E.A.O.)
| | - Eduardo A. Oliveira
- Division of Pediatric Nephrology, Rady Children’s Hospital, University of California, San Diego, CA 92093-0831, USA; (A.G.); (W.W.C.); (E.A.P.); (E.A.O.)
- Health Sciences Postgraduate Program, School of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte 30130-100, MG, Brazil
| | - Arieh Gertler
- Institute of Biochemistry, Food Science and Nutrition, Hebrew University of Jerusalem, Rehovot 7610001, Israel;
| | - Robert H. Mak
- Division of Pediatric Nephrology, Rady Children’s Hospital, University of California, San Diego, CA 92093-0831, USA; (A.G.); (W.W.C.); (E.A.P.); (E.A.O.)
| |
Collapse
|
12
|
Barton JR, Snook AE, Waldman SA. From leptin to lasers: the past and present of mouse models of obesity. Expert Opin Drug Discov 2021; 16:777-790. [PMID: 33472452 PMCID: PMC8243785 DOI: 10.1080/17460441.2021.1877654] [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: 09/11/2020] [Accepted: 01/14/2021] [Indexed: 10/22/2022]
Abstract
Introduction: Obesity is a prevalent condition that accounts for significant morbidity and mortality across the globe. Despite substantial effort, most obesity pharmacotherapies have proven unsafe or ineffective. The use of obese mouse models provides unique insight into the hormones and mechanisms that regulate appetite and metabolism. Paramount among these models are the 'obese' and 'diabetic' mice that revealed the powerful satiety hormone leptin, revolutionizing obesity research.Areas Covered: In this article, the authors discuss work on leptin therapy, and the clinical response to leptin in humans. The authors describe the use of modern mouse genetics to study targetable mechanisms for genetic forms of human obesity. Additionally, they describe mouse models of neuromodulation and their utility in unraveling neural circuits that govern appetite and metabolism.Expert opinion: Combining past and present models of obesity is required for the development of safe, effective, and impactful obesity therapy. Current research in obesity can benefit from repositories of genetically engineered mouse models to discover interactions between appetitive systems and circuits. Combining leptin therapy with other satiety signals comprising the gut-brain axis is a promising approach to induce significant enduring weight loss.
Collapse
Affiliation(s)
- Joshua R. Barton
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Adam E. Snook
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Scott A. Waldman
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
13
|
O'Connor KM, Ashoori M, Dias ML, Dempsey EM, O'Halloran KD, McDonald FB. Influence of innate immune activation on endocrine and metabolic pathways in infancy. Am J Physiol Endocrinol Metab 2021; 321:E24-E46. [PMID: 33900849 DOI: 10.1152/ajpendo.00542.2020] [Citation(s) in RCA: 2] [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] [Indexed: 12/12/2022]
Abstract
Prematurity is the leading cause of neonatal morbidity and mortality worldwide. Premature infants often require extended hospital stays, with increased risk of developing infection compared with term infants. A picture is emerging of wide-ranging deleterious consequences resulting from innate immune system activation in the newborn infant. Those who survive infection have been exposed to a stimulus that can impose long-lasting alterations into later life. In this review, we discuss sepsis-driven alterations in integrated neuroendocrine and metabolic pathways and highlight current knowledge gaps in respect of neonatal sepsis. We review established biomarkers for sepsis and extend the discussion to examine emerging findings from human and animal models of neonatal sepsis that propose novel biomarkers for early identification of sepsis. Future research in this area is required to establish a greater understanding of the distinct neonatal signature of early and late-stage infection, to improve diagnosis, curtail inappropriate antibiotic use, and promote precision medicine through a biomarker-guided empirical and adjunctive treatment approach for neonatal sepsis. There is an unmet clinical need to decrease sepsis-induced morbidity in neonates, to limit and prevent adverse consequences in later life and decrease mortality.
Collapse
Affiliation(s)
- K M O'Connor
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - M Ashoori
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
- Irish Centre for Maternal and Child Health Research (INFANT), University College Cork, Cork, Ireland
| | - M L Dias
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - E M Dempsey
- Irish Centre for Maternal and Child Health Research (INFANT), University College Cork, Cork, Ireland
- Department of Paediatrics and Child Health, School of Medicine, College of Medicine and Health, Cork University Hospital, Wilton, Cork, Ireland
| | - K D O'Halloran
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
- Irish Centre for Maternal and Child Health Research (INFANT), University College Cork, Cork, Ireland
| | - F B McDonald
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
- Irish Centre for Maternal and Child Health Research (INFANT), University College Cork, Cork, Ireland
| |
Collapse
|
14
|
Brown WE, Bradford BJ. Invited review: Mechanisms of hypophagia during disease. J Dairy Sci 2021; 104:9418-9436. [PMID: 34099296 DOI: 10.3168/jds.2021-20217] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/24/2021] [Indexed: 12/26/2022]
Abstract
Suppression of appetite, or hypophagia, is among the most recognizable effects of disease in livestock, with the potential to impair growth, reproduction, and lactation. The continued evolution of the field of immunology has led to a greater understanding of the immune and endocrine signaling networks underlying this conserved response to disease. Inflammatory mediators, especially including the cytokines tumor necrosis factor-α and interleukin-1β, are likely pivotal to disease-induced hypophagia, based on findings in both rodents and cattle. However, the specific mechanisms linking a cytokine surge to decreased feeding behavior are more difficult to pin down and likely include direct effects on appetite centers in the brain, alteration of gastric motility, and modulation of other endocrine factors that influence appetite and satiety. These insights into the mechanisms for disease-induced hypophagia have great relevance for management of neonatal calves, mature cows transitioning to lactation, and cows experiencing mastitis; however, it is not necessarily the case that increasing feed intake by any means possible will improve health outcomes for diseased cattle. We explore conflicting effects of hypophagia on immune responses, which may be impaired by the lack of specific substrates, versus apparent benefits for controlling the growth of some pathogens. Anti-inflammatory strategies have shown promise for promoting recovery of feed intake following some conditions but not others. Finally, we explore the potential for early disease detection through automated monitoring of feeding behavior and consider which strategies may be implemented to respond to early hypophagia.
Collapse
Affiliation(s)
- W E Brown
- Department of Animal Sciences & Industry, Kansas State University, Manhattan 66506
| | - B J Bradford
- Department of Animal Science, Michigan State University, East Lansing 48824.
| |
Collapse
|
15
|
Minimal effects from a single exposure to sevoflurane in adult male and female Sprague-Dawley rats. Neurotoxicol Teratol 2021; 84:106955. [PMID: 33465422 DOI: 10.1016/j.ntt.2021.106955] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 12/16/2020] [Accepted: 01/11/2021] [Indexed: 02/03/2023]
Abstract
Many people undergo procedures requiring general anesthesia each day and adverse cognitive effects have been reported in response to that anesthesia. Postoperative Cognitive Dysfunction (POCD) may occur in as many as 80% of adults during the first post-surgical week and can have lasting effects. Here, the cognitive and motor effects of sevoflurane exposure in Sprague-Dawley rats was examined along with body weights, blood oxygen saturation, heart rate, and body temperature. Male and female rats were exposed to 2.5% sevoflurane or medical grade air for one hour at postnatal day 115. Beginning the following day, rats began a series of behavioral tests examining locomotor activity, motor coordination, novel object recognition, and spatial learning and memory in a water maze. Blood oxygen saturation, heart rate, and body temperature were not affected by the sevoflurane exposure. A slight effect on locomotor activity was detected, but no effects on motor coordination, novel object recognition, or spatial learning and memory were observed. Brain weights following behavioral testing did not differ. The results reported here along with existing literature suggest sevoflurane is largely without effects on later cognition in adult rodents when exposure is of a relatively short duration and at a relatively low concentration.
Collapse
|
16
|
Abstract
The landmark discoveries of leptin and adiponectin firmly established adipose tissue as a sophisticated and highly active endocrine organ, opening a new era of investigating adipose-mediated tissue crosstalk. Both obesity-associated hyperleptinemia and hypoadiponectinemia are important biomarkers to predict cardiovascular outcomes, suggesting a crucial role for adiponectin and leptin in obesity-associated cardiovascular disorders. Normal physiological levels of adiponectin and leptin are indeed essential to maintain proper cardiovascular function. Insufficient adiponectin and leptin signaling results in cardiovascular dysfunction. However, a paradox of high levels of both leptin and adiponectin is emerging in the pathogenesis of cardiovascular disorders. Here, we (1) summarize the recent progress in the field of adiponectin and leptin and its association with cardiovascular disorders, (2) further discuss the underlying mechanisms for this new paradox of leptin and adiponectin action, and (3) explore the possible application of partial leptin reduction, in addition to increasing the adiponectin/leptin ratio as a means to prevent or reverse cardiovascular disorders.
Collapse
Affiliation(s)
- Shangang Zhao
- Touchstone Diabetes Center, Department of Internal Medicine (S.Z., C.M.K., P.E.S.), The University of Texas Southwestern Medical Center, Dallas
| | - Christine M Kusminski
- Touchstone Diabetes Center, Department of Internal Medicine (S.Z., C.M.K., P.E.S.), The University of Texas Southwestern Medical Center, Dallas
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine (S.Z., C.M.K., P.E.S.), The University of Texas Southwestern Medical Center, Dallas.,Department of Cell Biology (P.E.S.), The University of Texas Southwestern Medical Center, Dallas
| |
Collapse
|
17
|
Oztas B, Sahin D, Kir H, Kuskay S, Ates N. Effects of leptin, ghrelin and neuropeptide y on spike-wave discharge activity and certain biochemical parameters in WAG/Rij rats with genetic absence epilepsy. J Neuroimmunol 2020; 351:577454. [PMID: 33333420 DOI: 10.1016/j.jneuroim.2020.577454] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 11/26/2020] [Accepted: 12/06/2020] [Indexed: 12/14/2022]
Abstract
This study aimed to evaluate the effects of leptin, ghrelin and neuropeptide-Y on the development of nonconvulsive seizure activity and their role on combating oxidative stress and cytokines produced by the systemic immune response in the WAG/Rij rat model for genetic absence epilepsy. Current study showed that all three peptides aggravated spike wave discharges activity and affected the oxidative stress in WAG/Rij rats without any significant changes in the levels of IL-1β, IL-6 and TNF-α except leptin that only induced an increment in the concentration of IL-1β. Our results support the modulatory role of these endogenous peptides on absence epilepsy.
Collapse
Affiliation(s)
- Berrin Oztas
- Kocaeli University, Faculty of Medicine, Department of Biochemistry, Kocaeli, Turkey
| | - Deniz Sahin
- Kocaeli University, Faculty of Medicine, Department of Physiology, Kocaeli, Turkey.
| | - Hale Kir
- Kocaeli University, Faculty of Medicine, Department of Biochemistry, Kocaeli, Turkey
| | - Sevinc Kuskay
- Kocaeli University, Faculty of Medicine, Department of Biochemistry, Kocaeli, Turkey
| | - Nurbay Ates
- Kocaeli University, Faculty of Medicine, Department of Physiology, Kocaeli, Turkey
| |
Collapse
|
18
|
Delezie J, Gill JF, Santos G, Karrer-Cardel B, Handschin C. PGC-1β-expressing POMC neurons mediate the effect of leptin on thermoregulation in the mouse. Sci Rep 2020; 10:16888. [PMID: 33060645 PMCID: PMC7567876 DOI: 10.1038/s41598-020-73794-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023] Open
Abstract
The arcuate nucleus (ARC) of the hypothalamus is a key regulator of food intake, brown adipose tissue (BAT) thermogenesis, and locomotor activity. Whole-body deficiency of the transcriptional coactivator peroxisome proliferator-activated receptor γ (PPARγ) coactivator-1β (PGC-1β) disrupts mouse circadian locomotor activity and BAT-regulated thermogenesis, in association with altered gene expression at the central level. We examined whether PGC-1β expression in the ARC is required for proper energy balance and locomotor behavior by generating mice lacking the PGC-1β gene specifically in pro-opiomelanocortin (POMC) neurons. POMC neuron-specific deletion of PGC-1β did not impact locomotor behavior, food intake, body composition, energy fuel utilization and metabolic rate in fed, 24-h fasted and 24-h refed conditions. In contrast, in the fed state, deletion of PGC-1β in POMC cells elevated core body temperature during the nighttime period. Importantly, this higher body temperature is not associated with changes in BAT function and gene expression. Conversely, we provide evidence that mice lacking PGC-1β in POMC neurons are more sensitive to the effect of leptin on heat dissipation. Our data indicate that PGC-1β-expressing POMC neurons are part of a circuit controlling body temperature homeostasis and that PGC-1β function in these neurons is involved in the thermoregulatory effect of leptin.
Collapse
Affiliation(s)
- Julien Delezie
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056, Basel, Switzerland
| | - Jonathan F Gill
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056, Basel, Switzerland
| | - Gesa Santos
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056, Basel, Switzerland
| | | | - Christoph Handschin
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056, Basel, Switzerland.
| |
Collapse
|
19
|
Shi Z, Pelletier NE, Wong J, Li B, Sdrulla AD, Madden CJ, Marks DL, Brooks VL. Leptin increases sympathetic nerve activity via induction of its own receptor in the paraventricular nucleus. eLife 2020; 9:e55357. [PMID: 32538782 PMCID: PMC7316512 DOI: 10.7554/elife.55357] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 06/12/2020] [Indexed: 12/11/2022] Open
Abstract
Whether leptin acts in the paraventricular nucleus (PVN) to increase sympathetic nerve activity (SNA) is unclear, since PVN leptin receptors (LepR) are sparse. We show in rats that PVN leptin slowly increases SNA to muscle and brown adipose tissue, because it induces the expression of its own receptor and synergizes with local glutamatergic neurons. PVN LepR are not expressed in astroglia and rarely in microglia; instead, glutamatergic neurons express LepR, some of which project to a key presympathetic hub, the rostral ventrolateral medulla (RVLM). In PVN slices from mice expressing GCaMP6, leptin excites glutamatergic neurons. LepR are expressed mainly in thyrotropin-releasing hormone (TRH) neurons, some of which project to the RVLM. Injections of TRH into the RVLM and dorsomedial hypothalamus increase SNA, highlighting these nuclei as likely targets. We suggest that this neuropathway becomes important in obesity, in which elevated leptin maintains the hypothalamic pituitary thyroid axis, despite leptin resistance.
Collapse
Affiliation(s)
- Zhigang Shi
- Department of Physiology and PharmacologyPortlandUnited States
| | | | - Jennifer Wong
- Department of Physiology and PharmacologyPortlandUnited States
| | - Baoxin Li
- Department of Physiology and PharmacologyPortlandUnited States
| | - Andrei D Sdrulla
- Department of Anesthesiology and Perioperative MedicinePortlandUnited States
| | | | - Daniel L Marks
- Department of Pediatrics, Pape Family Pediatric Research Institute, Brenden-Colson Center for Pancreatic Care Oregon Health & Science UniversityPortlandUnited States
| | | |
Collapse
|
20
|
Huang Z, Chen J, Li B, Zeng B, Chou CH, Zheng X, Xie J, Li H, Hao Y, Chen G, Pei F, Shen B, Kraus VB, Wei H, Zhou X, Cheng L. Faecal microbiota transplantation from metabolically compromised human donors accelerates osteoarthritis in mice. Ann Rheum Dis 2020; 79:646-656. [PMID: 32205337 PMCID: PMC7384301 DOI: 10.1136/annrheumdis-2019-216471] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 02/17/2020] [Accepted: 03/03/2020] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Emerging evidence suggests that the microbiome plays an important role in the pathogenesis of osteoarthritis (OA). We aimed to test the two-hit model of OA pathogenesis and potentiation in which one 'hit' is provided by an adverse gut microbiome that activates innate immunity; the other 'hit' is underlying joint damage. METHODS Medical history, faecal and blood samples were collected from human healthy controls (OA-METS-, n=4), knee OA without metabolic syndrome (OA+METS-, n=7) and knee OA with metabolic syndrome (OA+METS+, n=9). Each group of human faecal samples, whose microbial composition was identified by 16S rRNA sequencing, was pooled and transplanted into germ-free mice 2 weeks prior to meniscal/ligamentous injury (MLI) (n≥6 per group). Eight weeks after MLI, mice were evaluated for histological OA severity and synovitis, systemic inflammation and gut permeability. RESULTS Histological OA severity following MLI was minimal in germ-free mice. Compared with the other groups, transplantation with the OA+METS+ microbiome was associated with higher mean systemic concentrations of inflammatory biomarkers (interleukin-1β, interleukin-6 and macrophage inflammatory protein-1α), higher gut permeability and worse OA severity. A greater abundance of Fusobacterium and Faecalibaterium and lesser abundance of Ruminococcaceae in transplanted mice were consistently correlated with OA severity and systemic biomarkers concentrations. CONCLUSION The study clearly establishes a direct gut microbiome-OA connection that sets the stage for a new means of exploring OA pathogenesis and potentially new OA therapeutics. Alterations of Fusobacterium, Faecalibaterium and Ruminococcaceae suggest a role of these particular microbes in exacerbating OA.
Collapse
Affiliation(s)
- ZeYu Huang
- State Key Laboratory of Oral Diseases and National Clinical Research for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Orthopaedic Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Jing Chen
- State Key Laboratory of Oral Diseases and National Clinical Research for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - BoLei Li
- State Key Laboratory of Oral Diseases and National Clinical Research for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Benhua Zeng
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Ching-Heng Chou
- Duke Molecular Physiology Institute, Duke University School of Medicine, Duke University, Durham, North Carolina, USA
| | - Xin Zheng
- State Key Laboratory of Oral Diseases and National Clinical Research for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - JingWei Xie
- Department of Orthopaedic Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Hao Li
- State Key Laboratory of Oral Diseases and National Clinical Research for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yu Hao
- State Key Laboratory of Oral Diseases and National Clinical Research for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Guo Chen
- Department of Orthopaedic Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - FuXing Pei
- Department of Orthopaedic Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Bin Shen
- Department of Orthopaedic Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Virginia B Kraus
- Duke Molecular Physiology Institute, Duke University School of Medicine, Duke University, Durham, North Carolina, USA
- Division of Rheumatology, Department of Medicine, Duke University School of Medicine, Duke University, Durham, North Carolina, USA
| | - Hong Wei
- Central Laboratory, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases and National Clinical Research for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases and National Clinical Research for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
21
|
Zhao S, Li N, Zhu Y, Straub L, Zhang Z, Wang MY, Zhu Q, Kusminski CM, Elmquist JK, Scherer PE. Partial leptin deficiency confers resistance to diet-induced obesity in mice. Mol Metab 2020; 37:100995. [PMID: 32289482 PMCID: PMC7229277 DOI: 10.1016/j.molmet.2020.100995] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/30/2020] [Accepted: 04/06/2020] [Indexed: 12/20/2022] Open
Abstract
Objective Hyperleptinemia per se is sufficient to promote leptin resistance in the obese state. Leptin sensitivity can be restored by reducing circulating leptin levels within a physiologically healthy range and is a viable antiobesity and antidiabetic strategy. However, a previous study suggests that partial leptin deficiency favors diet-induced obesity and related metabolic disorders in mice, arguing that a lower leptin level may indeed promote diet-induced obesity and its associated metabolic disorders. Here, we aim to elucidate what the impact of partial leptin deficiency is on fat mass and insulin sensitivity. Methods We used two different mouse models of partial leptin deficiency: an adipocyte-specific congenital heterozygous leptin knockout mouse line (LepHZ) and the well-established whole body heterozygous leptin knockout mouse (OBHZ). The metabolic studies of OBHZ and LepHZ mice were performed both on normal carbohydrate-rich chow diet and on a high-fat diet (HFD). Male and female mice were included in the study to account for sex-specific differences. Body weight, food intake, glucose tolerance, and insulin tolerance were tested. Histology of adipose tissue and liver tissue allowed insights into adipose tissue inflammation and hepatic triglyceride content. Immunohistochemistry was paired with RT-PCR analysis for expression levels of inflammatory markers. Results Both OBHZ and LepHZ mice displayed reduced circulating leptin levels on the chow diet and HFD. On chow diet, male OBHZ and LepHZ mice showed elevated fat mass and body weight, while their glucose tolerance and insulin sensitivity remained unchanged. However, the inability in partially leptin-deficient mice to fully induce circulating leptin during the development of diet-induced obesity results in reduced food intake and leaner mice with lower body weight compared to their littermate controls. Importantly, a strong reduction of adipose tissue inflammation is observed along with improvements in insulin sensitivity and enhanced glucose tolerance. Additionally, partial leptin deficiency protects the mice from fatty liver and liver fibrosis. Chronically HFD-fed OBHZ and LepHZ mice remain more sensitive to exogenous leptin injection, as reflected by their reduced food intake upon an acute leptin treatment. Conclusion In response to HFD feeding, the inability to upregulate leptin levels due to partial leptin deficiency protects mice from diet-induced obesity and metabolic dysregulation. Thus, in an obesogenic environment, maintaining lower leptin levels is highly beneficial for both obesity and diabetes management. Chronic leptin reduction represents a viable preventive strategy whose efficacy awaits clinical testing. Partial leptin deficiency protects from diet-induced obesity. Reduced leptin protects from diet-induced obesity independent of sex. Reduction of circulating leptin levels inhibits HFD-induced adipose tissue inflammation. Partial leptin deficiency confers resistance to HFD-induced liver fibrosis. Partial leptin deficiency enhances leptin sensitivity.
Collapse
Affiliation(s)
- Shangang Zhao
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Na Li
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yi Zhu
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Leon Straub
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Zhuzhen Zhang
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - May-Yun Wang
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Qingzhang Zhu
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christine M Kusminski
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joel K Elmquist
- Division of Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
22
|
Abstract
Animals that lack the hormone leptin become grossly obese, purportedly for 2 reasons: increased food intake and decreased energy expenditure (thermogenesis). This review examines the experimental evidence for the thermogenesis component. Analysis of the data available led us to conclude that the reports indicating hypometabolism in the leptin-deficient ob/ob mice (as well as in the leptin-receptor-deficient db/db mice and fa/fa rats) derive from a misleading calculation artefact resulting from expression of energy expenditure per gram of body weight and not per intact organism. Correspondingly, the body weight-reducing effects of leptin are not augmented by enhanced thermogenesis. Congruent with this, there is no evidence that the ob/ob mouse demonstrates atrophied brown adipose tissue or diminished levels of total UCP1 mRNA or protein when the ob mutation is studied on the inbred C57BL/6 mouse background, but a reduced sympathetic nerve activity is observed. On the outbred "Aston" mouse background, brown adipose tissue atrophy is seen, but whether this is of quantitative significance for the development of obesity has not been demonstrated. We conclude that leptin is not a thermogenic hormone. Rather, leptin has effects on body temperature regulation, by opposing torpor bouts and by shifting thermoregulatory thresholds. The central pathways behind these effects are largely unexplored.
Collapse
Affiliation(s)
- Alexander W Fischer
- Department of Molecular Biosciences, The Wenner-Gren Institute, The Arrhenius Laboratories F3, Stockholm University, Stockholm, Sweden.,Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Barbara Cannon
- Department of Molecular Biosciences, The Wenner-Gren Institute, The Arrhenius Laboratories F3, Stockholm University, Stockholm, Sweden
| | - Jan Nedergaard
- Department of Molecular Biosciences, The Wenner-Gren Institute, The Arrhenius Laboratories F3, Stockholm University, Stockholm, Sweden
| |
Collapse
|
23
|
Ryu KJ, Park H, Park JS, Lee YW, Kim SY, Kim H, Jeong Y, Kim YJ, Yi KW, Shin JH, Hur JY, Kim T. Vasomotor Symptoms: More Than Temporary Menopausal Symptoms. J Menopausal Med 2020; 26:147-153. [PMID: 33423402 PMCID: PMC7797223 DOI: 10.6118/jmm.20030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 10/05/2020] [Indexed: 01/10/2023] Open
Abstract
Vasomotor symptoms (VMS), such as hot flashes and night sweating, are classic menopausal symptoms experienced by a majority of perimenopausal and postmenopausal women. VMS have received a great deal of attention due to their relationship with cardiometabolic risk. Further, accumulating evidence indicates that VMS are associated with an increased risk of several chronic diseases, including metabolic syndrome, type 2 diabetes mellitus, nonalcoholic fatty liver diseases, and osteoporosis in perimenopausal and postmenopausal women. These findings suggest VMS as biomarkers of impaired cardiometabolic conditions rather than just temporary symptoms in menopausal women, warranting further studies to confirm the casual relationship of VMS with these diseases and the exact underlying mechanism in this context.
Collapse
Affiliation(s)
- Ki Jin Ryu
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul, Korea
| | - Hyuntae Park
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul, Korea.
| | | | - Yeon Woo Lee
- Korea University College of Medicine, Seoul, Korea
| | | | - Hayun Kim
- Korea University College of Medicine, Seoul, Korea
| | - Youngmi Jeong
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul, Korea
| | - Yong Jin Kim
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul, Korea
| | - Kyong Wook Yi
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul, Korea
| | - Jung Ho Shin
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul, Korea
| | - Jun Young Hur
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul, Korea
| | - Tak Kim
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
24
|
Aguilar-Valles A, Rodrigue B, Matta-Camacho E. Maternal Immune Activation and the Development of Dopaminergic Neurotransmission of the Offspring: Relevance for Schizophrenia and Other Psychoses. Front Psychiatry 2020; 11:852. [PMID: 33061910 PMCID: PMC7475700 DOI: 10.3389/fpsyt.2020.00852] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/04/2020] [Indexed: 12/21/2022] Open
Abstract
Prenatal infections have been linked to the development of schizophrenia (SCZ) and other neurodevelopmental disorders in the offspring, and work in animal models indicates that this is to occur through the maternal inflammatory response triggered by infection. Several studies in animal models demonstrated that acute inflammatory episodes are sufficient to trigger brain alterations in the adult offspring, especially in the mesolimbic dopamine (DA) system, involved in the pathophysiology of SCZ and other disorders involving psychosis. In the current review, we synthesize the literature on the clinical studies implicating prenatal infectious events in the development of SCZ. Then, we summarize evidence from animal models of maternal immune activation (MIA) and the behavioral and molecular alterations relevant for the function of the DAergic system. Furthermore, we discuss the evidence supporting the involvement of maternal cytokines, such as interleukin 6 (IL-6) and leptin (a hormone with effects on inflammation) in mediating the effects of MIA on the fetal brain, leading to the long-lasting effects on the offspring. In particular, IL-6 has been involved in mediating the effects of MIA animal models in the offspring through actions on the placenta, induction of IL-17a, or triggering the decrease in non-heme iron (hypoferremia). Maternal infection is very likely interacting with additional genetic and environmental risk factors in the development of SCZ; systematically investigating how these interactions produce specific phenotypes is the next step in understanding the etiology of complex psychiatric disorders.
Collapse
Affiliation(s)
| | - Brandon Rodrigue
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | | |
Collapse
|
25
|
Gregg BA, Parker PA, Waller KM, Schneider LG, Garcia M, Bradford B, Daniel JA, Whitlock BK. Effects of central and peripheral administration of an acute-phase protein, α-1-acid-glycoprotein, on feed intake and rectal temperature in sheep. J Anim Sci 2019; 97:4783-4791. [PMID: 31679022 PMCID: PMC6915238 DOI: 10.1093/jas/skz336] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 10/29/2019] [Indexed: 12/21/2022] Open
Abstract
In rodents, an acute-phase protein, α-1-acid-glycoprotein (AGP), was shown to provide a link between inflammation and suppression of feed intake by acting as a leptin receptor agonist. The objective of this study was to determine the effects of AGP on feed intake and rectal temperature in sheep. Ewes were ovariectomized, implanted with a cannula into a lateral ventricle of the brain, and kept indoors in individual pens. Feed intake and rectal temperature were determined for sheep in all experiments. In the first experiment, ewes (n = 4) received 1 of 4 treatments [0 (control), 0.012 (low), 0.06 (medium), or 0.30 (high) mg/kg BW AGP] into the lateral ventricle (ICV). All sheep received all treatments in a Latin square design balanced for carryover effects with 10 d between treatments. In the second experiment, ewes (n = 10) received 1 of 2 treatments (0 and 3 mg/kg BW of AGP) intravenously (IV) in a completely randomized design. In the third experiment, ewes (n = 19) received peripheral treatments (IV) of an antipyretic [0 (control) or 2.2 mg/kg BW flunixin meglumine (FLU)] 30 min before receiving central AGP [0 (control) or 0.3 mg/kg BW of AGP] in a completely randomized design. All data were analyzed using a mixed model analysis of variance and tested for effects of treatment, time, and the interaction of treatment and time. Cumulative 48-h feed intake after administration of treatments was also determined. In the first experiment, there was no effect of ICV treatment (P = 0.37) on feed intake rate or on cumulative feed intake (P = 0.31). There was an effect of ICV treatment (P = 0.002) on rectal temperatures, which were greater (P < 0.05) after the high dose of centrally administered AGP. In the second experiment, there was no effect of AGP administration IV on feed intake rate (P = 0.98), on cumulative feed intake (P = 0.41) or on rectal temperature (P = 0.71). In the third experiment, there was an effect of central AGP treatment (P < 0.0001) and an interaction of central AGP and time (P < 0.0001) on rectal temperature, whereas FLU had no effect (P = 0.93), demonstrating that AGP increased rectal temperatures regardless of antipyretic treatment. These results indicate that central AGP increases rectal temperature in sheep by pathways that do not involve prostaglandins. Further research is needed to determine whether AGP may be an important integrator of energy balance and inflammation.
Collapse
Affiliation(s)
- Brittany A Gregg
- College of Veterinary Medicine, University of Tennessee, Knoxville, TN
- Department of Animal Science, University of Tennessee, Knoxville, TN
| | - Paxton A Parker
- College of Veterinary Medicine, University of Tennessee, Knoxville, TN
| | - Kathryn M Waller
- College of Veterinary Medicine, University of Tennessee, Knoxville, TN
| | | | - Miriam Garcia
- Department of Animal Sciences and Industry, Kansas State University, Manhattan, KS
| | - Barry Bradford
- Department of Animal Sciences and Industry, Kansas State University, Manhattan, KS
| | - Joseph A Daniel
- School of Mathematical and Natural Sciences, Berry College, Mount Berry, GA
| | - Brian K Whitlock
- College of Veterinary Medicine, University of Tennessee, Knoxville, TN
- Department of Animal Science, University of Tennessee, Knoxville, TN
| |
Collapse
|
26
|
The Influence of Photoperiod on the Action of Exogenous Leptin on Gene Expression of Proinflammatory Cytokines and Their Receptors in the Thoracic Perivascular Adipose Tissue (PVAT) in Ewes. Mediators Inflamm 2019; 2019:7129476. [PMID: 31780867 PMCID: PMC6875191 DOI: 10.1155/2019/7129476] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 09/02/2019] [Accepted: 10/09/2019] [Indexed: 12/31/2022] Open
Abstract
Leptin resistance is either a condition induced by human obesity or a natural phenomenon associated with seasonality in ruminants. In the cardiovascular system, the leptin resistance state presence is a complex issue. Moreover, the perivascular adipose tissue (PVAT) appears to be crucial as a source of proinflammatory cytokines and as a site of interaction for leptin contributing to endothelium dysfunction and atherosclerosis progression. So the aim of this study was to examine the influence of the photoperiod on the action of exogenous leptin on gene expression of selected proinflammatory cytokines and their receptors in thoracic PVAT of ewe with or without prior lipopolysaccharide (LPS) stimulation. The experiment was conducted on 48 adult, female ewes divided into 4 group (n = 6 in each): control, with LPS intravenous (iv.) injection (400 ng/kg of BW), with leptin iv. injection (20 μg/kg BW), and with LPS and 30-minute-later leptin injection, during short-day (SD) and long-day (LD) seasons. Three hours after LPS/control treatment, animals were euthanized to collect the PVAT adherent to the aorta wall. The leptin injection enhanced IL1B gene expression only in the LD season; however, in both seasons leptin injection intensified LPS-induced increase in IL1B gene expression. IL1R2 gene expression was increased by leptin injection only in the SD season. Neither IL6 nor its receptor and signal transducer gene expressions were influenced by leptin administration. Leptin injection increased TNFA gene expression regardless of photoperiodic conditions. Only in the SD season did leptin treatment increase the gene expression of both TNFα receptors. To conclude, leptin may modulate the inflammatory reaction progress in PVAT. In ewe, the sensitivity of PVAT on leptin action is dependent upon the photoperiodic condition with stronger effects stated in the SD season.
Collapse
|
27
|
Musolino ST, Schartner EP, Hutchinson MR, Salem A. Minocycline attenuates 3,4-methylenedioxymethamphetamine-induced hyperthermia in the rat brain. Eur J Pharmacol 2019; 858:172495. [PMID: 31238065 DOI: 10.1016/j.ejphar.2019.172495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 06/21/2019] [Accepted: 06/21/2019] [Indexed: 10/26/2022]
Abstract
Hyperthermia is most dangerous clinical symptom of acute MDMA administration, and a key factor related to potentially life-threatening MDMA-induced complications. MDMA induces a consistently faster onset of brain hyperthermia when compared to a delayed and moderate hyperthermia in the body, and the most harmful effects of MDMA are related to its modulation of neural functions. The primary focus of this study was to investigate the effects of minocycline, a centrally acting tetracycline derivative on MDMA-induced brain hyperthermia at high ambient temperature. However, we also simultaneously recorded body temperature, heart rate, and locomotor activity changes, allowing us to gain a better understanding of the mechanisms underlying the MDMA-induced hyperthermic response. We also investigated the effects of MDMA at normal ambient temperature to provide further evidence as to the importance of environmental factors on the intensity of MDMA's temperature effects. At normal ambient temperature, MDMA (10 mg/kg, i.p.) induced a significant brain and body hypothermia for the first 90 min following drug administration, and significantly increased heart rate and locomotor activity compared to saline controls. At high ambient temperature however, MDMA (10 mg/kg, i.p.) induced a robust and extended brain and body hyperthermia, as well as significantly increased heart rate and locomotor activity. A 3-day minocycline (50 mg/kg, i.p.) pre-treatment significantly attenuated MDMA-induced increases in brain temperature, body temperature, heart rate, and locomotor activity. Our findings indicate that minocycline is more effective in attenuating the exacerbated MDMA-induced hyperthermic response in the brain compared to the body at high ambient temperature.
Collapse
Affiliation(s)
- Stefan T Musolino
- ARC Centre of Excellence for Nanoscale BioPhotonics and Institute for Photonics and Advanced Sensing, Adelaide, SA, 5005, Australia; Discipline of Pharmacology, Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia.
| | - Erik P Schartner
- ARC Centre of Excellence for Nanoscale BioPhotonics and Institute for Photonics and Advanced Sensing, Adelaide, SA, 5005, Australia; School of Physical Sciences, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Mark R Hutchinson
- ARC Centre of Excellence for Nanoscale BioPhotonics and Institute for Photonics and Advanced Sensing, Adelaide, SA, 5005, Australia; Discipline of Pharmacology, Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Abdallah Salem
- Discipline of Pharmacology, Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia
| |
Collapse
|
28
|
Krawczyńska A, Antushevich H, Bochenek J, Wojtulewicz K, Pawlina B, Herman A, Zięba D. Photoperiodic conditions as a factor modulating leptin influence
on pro-inflammatory cytokines
and their receptors gene expression in ewe’s aorta. JOURNAL OF ANIMAL AND FEED SCIENCES 2019. [DOI: 10.22358/jafs/110022/2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
29
|
Zuccarelli L, Galasso L, Turner R, Coffey EJB, Bessone L, Strapazzon G. Human Physiology During Exposure to the Cave Environment: A Systematic Review With Implications for Aerospace Medicine. Front Physiol 2019; 10:442. [PMID: 31068833 PMCID: PMC6491700 DOI: 10.3389/fphys.2019.00442] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 04/01/2019] [Indexed: 11/13/2022] Open
Abstract
Background: Successful long-duration missions outside low-Earth orbit will depend on technical and physiological challenges under abnormal environmental conditions. Caves, characterized by absence of light, confinement, three-dimensional human movement and long-duration isolation, are identifiably one of the earliest examples of scientific enquiry into space analogs. However, little is known about the holistic human physiological response during cave exploration or prolonged habitation. Objectives: The aim of our review was to conduct a systematic bibliographic research review of the effects of short and prolonged exposure to a cave environment on human physiology, with a view to extend the results to implications for human planetary exploration missions. Methods: A systematic search was conducted following the structured PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines for electronic databases. Results: The search retrieved 1,519 studies. There were 50 articles selected for further consideration, of which 31 met our inclusion criteria. Short-term cave exposure studies have investigated visual dysfunction, cardiovascular, endocrine-metabolic, immunologic-hematological and muscular responses in humans. Augmentations of heart rate, muscular damage, initial anticipatory stress reaction and inflammatory responses were reported during caving activity. Prolonged exposure studies mainly investigated whether biological rhythms persist or desist in the absence of standard environmental conditions. Changes were evident in estimated vs. actual rest-activity cycle periods and external desynchronization, body temperature, performance reaction time and heart rate cycles. All studies have shown a marked methodological heterogeneity and lack reproduction under controlled conditions. Conclusions: This review facilitates a further comparison of the proposed physiological impact of a subterranean space analog environment, with existing knowledge in related disciplines pertaining to human operative preparation under challenging environmental conditions. This comprehensive overview should stimulate more reproducible research on this topic and offer the opportunity to advance study design and focus future human research in the cave environment on noteworthy, reproducible projects.
Collapse
Affiliation(s)
- Lucrezia Zuccarelli
- Department of Medicine, University of Udine, Udine, Italy.,Directorate of Human and Robotics, Exploration, European Space Agency, Köln, Germany
| | - Letizia Galasso
- Department of Biomedical Science for Health, University of Milan, Milan, Italy
| | - Rachel Turner
- Institute of Mountain Emergency Medicine, Eurac Research, Bolzano, Italy
| | - Emily J B Coffey
- Department of Psychology, Concordia University, Montreal, QC, Canada
| | - Loredana Bessone
- Directorate of Human and Robotics, Exploration, European Space Agency, Köln, Germany
| | - Giacomo Strapazzon
- Directorate of Human and Robotics, Exploration, European Space Agency, Köln, Germany.,Institute of Mountain Emergency Medicine, Eurac Research, Bolzano, Italy
| |
Collapse
|
30
|
Sergi D, Campbell FM, Grant C, Morris AC, Bachmair EM, Koch C, McLean FH, Muller A, Hoggard N, de Roos B, Porteiro B, Boekschoten MV, McGillicuddy FC, Kahn D, Nicol P, Benzler J, Mayer CD, Drew JE, Roche HM, Muller M, Nogueiras R, Dieguez C, Tups A, Williams LM. SerpinA3N is a novel hypothalamic gene upregulated by a high-fat diet and leptin in mice. GENES AND NUTRITION 2018; 13:28. [PMID: 30519364 PMCID: PMC6263559 DOI: 10.1186/s12263-018-0619-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 11/06/2018] [Indexed: 02/08/2023]
Abstract
Background Energy homeostasis is regulated by the hypothalamus but fails when animals are fed a high-fat diet (HFD), and leptin insensitivity and obesity develops. To elucidate the possible mechanisms underlying these effects, a microarray-based transcriptomics approach was used to identify novel genes regulated by HFD and leptin in the mouse hypothalamus. Results Mouse global array data identified serpinA3N as a novel gene highly upregulated by both a HFD and leptin challenge. In situ hybridisation showed serpinA3N expression upregulation by HFD and leptin in all major hypothalamic nuclei in agreement with transcriptomic gene expression data. Immunohistochemistry and studies in the hypothalamic clonal neuronal cell line, mHypoE-N42 (N42), confirmed that alpha 1-antichymotrypsin (α1AC), the protein encoded by serpinA3, is localised to neurons and revealed that it is secreted into the media. SerpinA3N expression in N42 neurons is upregulated by palmitic acid and by leptin, together with IL-6 and TNFα, and all three genes are downregulated by the anti-inflammatory monounsaturated fat, oleic acid. Additionally, palmitate upregulation of serpinA3 in N42 neurons is blocked by the NFκB inhibitor, BAY11, and the upregulation of serpinA3N expression in the hypothalamus by HFD is blunted in IL-1 receptor 1 knockout (IL-1R1−/−) mice. Conclusions These data demonstrate that serpinA3 expression is implicated in nutritionally mediated hypothalamic inflammation.
Collapse
Affiliation(s)
- Domenico Sergi
- 1Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD UK
| | - Fiona M Campbell
- 1Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD UK
| | - Christine Grant
- 1Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD UK
| | - Amanda C Morris
- 1Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD UK
| | | | - Christiane Koch
- 2Department of Animal Physiology, Faculty of Biology, Philipps University Marburg, Karl-von-Frisch Str. 8, 35043 Marburg, Germany.,3Centre for Neuroendocrinology and Brain Health Research Centre, Department of Physiology, School of Medical Sciences, University of Otago, Dunedin, 9054 New Zealand
| | - Fiona H McLean
- 1Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD UK
| | - Aifric Muller
- 1Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD UK
| | - Nigel Hoggard
- 1Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD UK
| | - Baukje de Roos
- 1Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD UK
| | - Begona Porteiro
- 4Department of Physiology, University of Santiago de Compostela, 15705 Santiago de Compostela, Spain.,5CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Mark V Boekschoten
- 6Nutrition, Metabolism and Genomics Group, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| | - Fiona C McGillicuddy
- 7Nutrigenomics Research Group, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Darcy Kahn
- 1Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD UK
| | - Phyllis Nicol
- 1Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD UK
| | - Jonas Benzler
- 2Department of Animal Physiology, Faculty of Biology, Philipps University Marburg, Karl-von-Frisch Str. 8, 35043 Marburg, Germany.,3Centre for Neuroendocrinology and Brain Health Research Centre, Department of Physiology, School of Medical Sciences, University of Otago, Dunedin, 9054 New Zealand
| | - Claus-Dieter Mayer
- 8Biomathematics & Statistics Scotland (BioSS), Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD UK
| | - Janice E Drew
- 1Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD UK
| | - Helen M Roche
- 7Nutrigenomics Research Group, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Michael Muller
- 9Nutrigenomics and Systems Nutrition Group, Norwich Medical School, University of East Anglia, Norwich, NR4 7UQ UK
| | - Ruben Nogueiras
- 4Department of Physiology, University of Santiago de Compostela, 15705 Santiago de Compostela, Spain.,5CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Carlos Dieguez
- 4Department of Physiology, University of Santiago de Compostela, 15705 Santiago de Compostela, Spain.,5CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Alexander Tups
- 2Department of Animal Physiology, Faculty of Biology, Philipps University Marburg, Karl-von-Frisch Str. 8, 35043 Marburg, Germany.,3Centre for Neuroendocrinology and Brain Health Research Centre, Department of Physiology, School of Medical Sciences, University of Otago, Dunedin, 9054 New Zealand
| | - Lynda M Williams
- 1Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD UK
| |
Collapse
|
31
|
Pro-inflammatory cytokines: The link between obesity and osteoarthritis. Cytokine Growth Factor Rev 2018; 44:38-50. [PMID: 30340925 DOI: 10.1016/j.cytogfr.2018.10.002] [Citation(s) in RCA: 546] [Impact Index Per Article: 91.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/06/2018] [Accepted: 10/08/2018] [Indexed: 02/05/2023]
Abstract
Osteoarthritis (OA), characterized by joint malfunction and chronic disability, is the most common form of arthritis. Clinical and animal experiments reveal that age-related OA is associated with many factors such as age, sex, trauma, and obesity. One of the most influential and modifiable risk factors is obesity. Obesity not only increases mechanical stress on the tibiofemoral cartilage, but also leads to a higher prevalence of OA in non-weight-bearing areas. There is a link between obesity and inflammation. Adipose tissues play a crucial role in this context because they are the major source of cytokines, chemokines, and metabolically-active mediators named adipokines. The adipokines, including adiponectin and leptin, have been demonstrated to regulate inflammatory immune responses in cartilage. Obese people and animals show a higher level of serum tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL)-1β and IL-6, all of which are produced by macrophages derived from adipose tissue. These pro-inflammatory cytokines regulate the proliferation and apoptosis of adipocytes, promote lipolysis, inhibit lipid synthesis and decrease blood lipids through autocrine and paracrine mechanisms. Elevated levels of TNF-α, IL-1 and IL-6 have been found in the synovial fluid, synovial membrane, subchondral bone and cartilage of OA patients, confirming their important roles in OA pathogenesis. TNF-α, IL-6 and IL-1 are the factors released by fat to negatively regulate cartilage directly. Moreover, TNF-α, IL-1 and IL-6 can induce the production of other cytokines, matrix metalloproteinases (MMPs) and prostaglandins and inhibit the synthesis of proteoglycans and type II collagen; thus, they play a pivotal role in cartilage matrix degradation and bone resorption in OA. Activated chondrocytes also produce MMP-1, MMP-3, MMP-13, and aggrecanase 1 and 2 (ADAMTS-4, ADAMTS-5). In addition, IL-1, TNF-α and IL-6 may cause OA indirectly by regulating release of adiponectin and leptin from adipocytes. In this review, we first summarize the relationship between obesity and inflammation. Then we summarize the roles of IL-1, TNF-α and IL-6 in OA. We further discuss how IL-1, TNF-α and IL-6 regulate the communication between fat and OA, and their pathological roles in obesity-related OA. Lastly, we discuss the possibility of using the pro-inflammatory signaling pathway as a therapeutic target to develop drugs for obesity-related OA.
Collapse
|
32
|
Besedovsky HO. The immune system as a sensorial system that can modulate brain functions and reset homeostasis. Ann N Y Acad Sci 2018; 1437:5-14. [PMID: 30126011 DOI: 10.1111/nyas.13935] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 06/22/2018] [Accepted: 06/27/2018] [Indexed: 12/17/2022]
Abstract
Evidence indicates that activated immune cells release products, typically cytokines, that can convey information to the brain about the type of ongoing peripheral immune responses. This evidence led colleagues and me to categorize the immune system as another sensorial system that, upon receiving this information, can emit neuroendocrine signals with immunoregulatory functions that can also reset homeostatic mechanisms. Here, I discuss evidence and clues indicating (1) possible mechanisms by which cytokines, such as those of the interleukin 1 (IL-1) family, can reset energy homeostasis to balance the high fuel requirement of the immune system and the brain; and (2) the possibility that the tripartite synapse, which includes astrocytes as a third component, processes and integrates immune signals at brain levels with other sensorial signals that the central nervous system permanently receives.
Collapse
Affiliation(s)
- Hugo O Besedovsky
- Research Group Immunophysiology, Division of Neurophysiology, Institute of Physiology and Pathophysiology, Marburg, Germany
| |
Collapse
|
33
|
Little HC, Tan SY, Cali FM, Rodriguez S, Lei X, Wolfe A, Hug C, Wong GW. Multiplex Quantification Identifies Novel Exercise-regulated Myokines/Cytokines in Plasma and in Glycolytic and Oxidative Skeletal Muscle. Mol Cell Proteomics 2018; 17:1546-1563. [PMID: 29735541 DOI: 10.1074/mcp.ra118.000794] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 05/02/2018] [Indexed: 12/15/2022] Open
Abstract
Exercise is known to confer major health benefits, but the underlying mechanisms are not well understood. The systemic effects of exercise on multi-organ systems are thought to be partly because of myokines/cytokines secreted by skeletal muscle. The extent to which exercise alters cytokine expression and secretion in different muscle fiber types has not been systematically examined. Here, we assessed changes in 66 mouse cytokines in serum, and in glycolytic (plantaris) and oxidative (soleus) muscles, in response to sprint, endurance, or chronic wheel running. Both acute and short-term exercise significantly altered a large fraction of cytokines in both serum and muscle, twenty-three of which are considered novel exercise-regulated myokines. Most of the secreted cytokine receptors profiled were also altered by physical activity, suggesting an exercise-regulated mechanism that modulates the generation of soluble receptors found in circulation. A greater overlap in cytokine profile was seen between endurance and chronic wheel running. Between fiber types, both acute and chronic exercise induced significantly more cytokine changes in oxidative compared with glycolytic muscle. Further, changes in a subset of circulating cytokines were not matched by their changes in muscle, but instead reflected altered expression in liver and adipose tissues. Last, exercise-induced changes in cytokine mRNA and protein were only minimally correlated in soleus and plantaris. In sum, our results indicate that exercise regulates many cytokines whose pleiotropic actions may be linked to positive health outcomes. These data provide a framework to further understand potential crosstalk between skeletal muscle and other organ compartments.
Collapse
Affiliation(s)
- Hannah C Little
- From the ‡Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205.,§Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Stefanie Y Tan
- From the ‡Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205.,§Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Francesca M Cali
- From the ‡Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205.,§Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Susana Rodriguez
- From the ‡Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205.,§Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Xia Lei
- From the ‡Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205.,§Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Andrew Wolfe
- ¶Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
| | - Christopher Hug
- ‖Division of Pulmonary Medicine, Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts
| | - G William Wong
- From the ‡Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; .,§Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| |
Collapse
|
34
|
Abstract
The experiment was conducted to examine the effect of a high fat diet (HFD) on airway hyperresponsiveness (AHR) in mice. Twenty-three adult male C57BL/6 J mice were fed with HFD or regular chow diet for two weeks. The total respiratory resistance was measured by forced oscillation technique at baseline and after methacholine aerosol challenge at 1, 3, 10 and 30 mg/mL. Bronchoalveolar lavage (BAL) was performed. Lipid levels and lipid peroxidation in lung tissue were measured along with gene expression of multiple cytokines. Lungs were digested, and IL-1β secretion by pulmonary macrophages was determined. HFD feeding resulted in 11% higher body weight compared to chow. HFD did not affect respiratory resistance at baseline, but significantly augmented airway responses to methacholine compared to chow diet (40.5 ± 17.7% increase at 30 mg/ml methacholine, p < 0.05). HFD induced a 3.2 ± 0.6 fold increase in IL-1β gene expression (p < 0.001) and a 38 fold increase in IL-1β secretion in the lungs. There was no change in BAL and no change in any other cytokines, lipid levels or lipid peroxidation. Hence, HFD induced AHR in mice prior to the development of significant obesity which was associated with up-regulation of pulmonary IL-1β.
Collapse
|
35
|
Pourghaderi M, Hosseinzadeh-Attar MJ, Jalali M, Siassi F, Nabavizadeh F, Eshraghian MR. The Effect of Peripheral Injection of Leptin on Biomarkers of Aging in Calorie Restricted Rats. NUTRITION AND FOOD SCIENCES RESEARCH 2018. [DOI: 10.29252/nfsr.5.2.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
36
|
Abstract
Interactions between the brain and distinct adipose depots have a key role in maintaining energy balance, thereby promoting survival in response to metabolic challenges such as cold exposure and starvation. Recently, there has been renewed interest in the specific central neuronal circuits that regulate adipose depots. Here, we review anatomical, genetic and pharmacological studies on the neural regulation of adipose function, including lipolysis, non-shivering thermogenesis, browning and leptin secretion. In particular, we emphasize the role of leptin-sensitive neurons and the sympathetic nervous system in modulating the activity of brown, white and beige adipose tissues. We provide an overview of advances in the understanding of the heterogeneity of the brain regulation of adipose tissues and offer a perspective on the challenges and paradoxes that the community is facing regarding the actions of leptin on this system.
Collapse
Affiliation(s)
- Alexandre Caron
- Division of Hypothalamic Research and Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Syann Lee
- Division of Hypothalamic Research and Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joel K. Elmquist
- Division of Hypothalamic Research and Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Laurent Gautron
- Division of Hypothalamic Research and Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
37
|
The effects of overnight nutrient intake on hypothalamic inflammation in a free-choice diet-induced obesity rat model. Appetite 2018; 120:527-535. [DOI: 10.1016/j.appet.2017.10.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/03/2017] [Accepted: 10/03/2017] [Indexed: 12/31/2022]
|
38
|
Ami N, Sato H, Hayakawa Y. Paclitaxel-induced hypothermia and hypoperfusion increase breast cancer metastasis and angiogenesis in mice. Oncol Lett 2017; 15:2330-2334. [PMID: 29434941 PMCID: PMC5776906 DOI: 10.3892/ol.2017.7621] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 11/06/2017] [Indexed: 01/07/2023] Open
Abstract
Housing temperature has been shown to influence thermoregulation and behavior of preclinical cancer models; and anti-cancer drugs typically reduce peripheral blood flow and body temperature. In the present study, the effects of paclitaxel (PTX)-induced reduction of body temperature and peripheral blood flow on metastatic 4T1 breast cancer was investigated in a mouse model and the modification of these effects by thermoneutral temperature was also assessed. A single dose of PTX decreased the body temperature and peripheral blood flow in mice housed at a standard temperature (23°C). Furthermore, although lung metastasis and angiogenesis of inoculated 4T1 cells increased in mice pretreated with PTX, mice housed at a thermoneutral temperature (30°C) could compensate their body temperature and peripheral blood flow compared with control mice, and also suppressed 4T1 angiogenesis and metastasis to lung. The present results imply that maintenance of body temperature or efficient energy supply for thermogenesis may prevent tumor relapse or metastasis after chemotherapy.
Collapse
Affiliation(s)
- Nozomi Ami
- R&D Center, Terumo Corp., Nakai-machi, Kanagawa 259-0151, Japan.,Division of Pathogenic Biochemistry, Institute of Natural Medicine, University of Toyama, Toyama-shi, Toyama 930-0194, Japan
| | - Hideki Sato
- R&D Center, Terumo Corp., Nakai-machi, Kanagawa 259-0151, Japan
| | - Yoshihiro Hayakawa
- Division of Pathogenic Biochemistry, Institute of Natural Medicine, University of Toyama, Toyama-shi, Toyama 930-0194, Japan
| |
Collapse
|
39
|
Stahn AC, Werner A, Opatz O, Maggioni MA, Steinach M, von Ahlefeld VW, Moore A, Crucian BE, Smith SM, Zwart SR, Schlabs T, Mendt S, Trippel T, Koralewski E, Koch J, Choukèr A, Reitz G, Shang P, Röcker L, Kirsch KA, Gunga HC. Increased core body temperature in astronauts during long-duration space missions. Sci Rep 2017; 7:16180. [PMID: 29170507 PMCID: PMC5701078 DOI: 10.1038/s41598-017-15560-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/27/2017] [Indexed: 12/12/2022] Open
Abstract
Humans’ core body temperature (CBT) is strictly controlled within a narrow range. Various studies dealt with the impact of physical activity, clothing, and environmental factors on CBT regulation under terrestrial conditions. However, the effects of weightlessness on human thermoregulation are not well understood. Specifically, studies, investigating the effects of long-duration spaceflight on CBT at rest and during exercise are clearly lacking. We here show that during exercise CBT rises higher and faster in space than on Earth. Moreover, we observed for the first time a sustained increased astronauts’ CBT also under resting conditions. This increase of about 1 °C developed gradually over 2.5 months and was associated with augmented concentrations of interleukin-1 receptor antagonist, a key anti-inflammatory protein. Since even minor increases in CBT can impair physical and cognitive performance, both findings have a considerable impact on astronauts’ health and well-being during future long-term spaceflights. Moreover, our findings also pinpoint crucial physiological challenges for spacefaring civilizations, and raise questions about the assumption of a thermoregulatory set point in humans, and our evolutionary ability to adapt to climate changes on Earth.
Collapse
Affiliation(s)
- Alexander C Stahn
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Physiology, Center for Space Medicine and Extreme Environments, CharitéCrossOver (CCO), Charitéplatz 1, Berlin, 10117, Germany.,Division of Sleep and Chronobiology, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, 1019 Blockley Hall, 423 Guardian Drive, Philadelphia, PA, 19104-6021, USA
| | - Andreas Werner
- German Air Force, Centre of Aerospace Medicine, Aviation Physiology Training Centre, Aviation Physiology Diagnostics and Science, Steinborner Str. 43, 01936, Königsbrück, Germany
| | - Oliver Opatz
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Physiology, Center for Space Medicine and Extreme Environments, CharitéCrossOver (CCO), Charitéplatz 1, Berlin, 10117, Germany
| | - Martina A Maggioni
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Physiology, Center for Space Medicine and Extreme Environments, CharitéCrossOver (CCO), Charitéplatz 1, Berlin, 10117, Germany.,Department of Biomedical Sciences for Health, Università degli Studi di Milano, via Luigi Mangiagalli 31, 20133, Milan, Italy
| | - Mathias Steinach
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Physiology, Center for Space Medicine and Extreme Environments, CharitéCrossOver (CCO), Charitéplatz 1, Berlin, 10117, Germany
| | - Victoria Weller von Ahlefeld
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Physiology, Center for Space Medicine and Extreme Environments, CharitéCrossOver (CCO), Charitéplatz 1, Berlin, 10117, Germany
| | - Alan Moore
- Department of Health and Kinesiology, Lamar University, Beaumont, TX, 77710, USA
| | - Brian E Crucian
- Biomedical Research and Environmental Sciences Division, NASA Johnson Space Center, Houston, TX, 77058, USA
| | - Scott M Smith
- Biomedical Research and Environmental Sciences Division, NASA Johnson Space Center, Houston, TX, 77058, USA
| | - Sara R Zwart
- Preventive Medicine and Community Health, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555, USA
| | - Thomas Schlabs
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Physiology, Center for Space Medicine and Extreme Environments, CharitéCrossOver (CCO), Charitéplatz 1, Berlin, 10117, Germany
| | - Stefan Mendt
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Physiology, Center for Space Medicine and Extreme Environments, CharitéCrossOver (CCO), Charitéplatz 1, Berlin, 10117, Germany
| | - Tobias Trippel
- Charité Medizinische Klinik, Charité Universitätsmedizin Berlin, Kardiologie, Augustenburger Platz 1, Berlin, 13353, Germany
| | - Eberhard Koralewski
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Physiology, Center for Space Medicine and Extreme Environments, CharitéCrossOver (CCO), Charitéplatz 1, Berlin, 10117, Germany
| | - Jochim Koch
- Drägerwerk AG & Co. KGaA, Moislinger Allee 53-55, Lubeck, 23558, Germany
| | - Alexander Choukèr
- Department of Anaesthesiology, Hospital of the University of Munich, Marchioninistrasse 15, München, 81377, Germany
| | - Günther Reitz
- DLR, Institut für Luft- und Raumfahrtmedizin, Abteilung Strahlenbiologie, Linder Höhe, Köln, 51147, Germany.,Nuclear Physics Institute of the Czech Academy of Sciences, Department of Radiation Dosimetry, Na Truhlářce 39/64, Praha 8, 180 00, Czech Republic
| | - Peng Shang
- Key Laboratory for Space Bioscience & Biotechnology, Institute of Special Environnments Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Lothar Röcker
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Physiology, Center for Space Medicine and Extreme Environments, CharitéCrossOver (CCO), Charitéplatz 1, Berlin, 10117, Germany
| | - Karl A Kirsch
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Physiology, Center for Space Medicine and Extreme Environments, CharitéCrossOver (CCO), Charitéplatz 1, Berlin, 10117, Germany
| | - Hanns-Christian Gunga
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Physiology, Center for Space Medicine and Extreme Environments, CharitéCrossOver (CCO), Charitéplatz 1, Berlin, 10117, Germany.
| |
Collapse
|
40
|
Flatow EA, Komegae EN, Fonseca MT, Brito CF, Musteata FM, Antunes-Rodrigues J, Steiner AA. Elucidating the role of leptin in systemic inflammation: a study targeting physiological leptin levels in rats and their macrophages. Am J Physiol Regul Integr Comp Physiol 2017; 313:R572-R582. [DOI: 10.1152/ajpregu.00171.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/18/2017] [Accepted: 07/26/2017] [Indexed: 01/24/2023]
Abstract
To elucidate the role of leptin in acute systemic inflammation, we investigated how its infusion at low, physiologically relevant doses affects the responses to bacterial lipopolysaccharide (LPS) in rats subjected to 24 h of food deprivation. Leptin was infused subcutaneously (0–20 μg·kg−1·h−1) or intracerebroventricularly (0–1 μg·kg−1·h−1). Using hypothermia and hypotension as biomarkers of systemic inflammation, we identified the phase extending from 90 to 240 min post-LPS as the most susceptible to modulation by leptin. In this phase, leptin suppressed the rise in plasma TNF-α and accelerated the recoveries from hypothermia and hypotension. Suppression of TNF-α was not accompanied by changes in other cytokines or prostaglandins. Leptin suppressed TNF-α when infused peripherally but not when infused into the brain. Importantly, the leptin dose that suppressed TNF-α corresponded to the lowest dose that limited food consumption; this dose elevated plasma leptin within the physiological range (to 5.9 ng/ml). We then conducted in vitro experiments to investigate whether an action of leptin on macrophages could parallel our in vivo observations. The results revealed that, when sensitized by food deprivation, LPS-stimulated peritoneal macrophages can be inhibited by leptin at concentrations that are lower than those reported to promote cytokine release. It is concluded that physiological levels of leptin do not exert a proinflammatory effect but rather an anti-inflammatory effect involving selective suppression of TNF-α via an action outside the brain. The mechanism of this effect might involve a previously unrecognized, suppressive action of leptin on macrophage subpopulations sensitized by food deprivation, but future studies are warranted.
Collapse
Affiliation(s)
- Elizabeth A. Flatow
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Evilin N. Komegae
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Monique T. Fonseca
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Camila F. Brito
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Florin M. Musteata
- Department of Pharmaceutical Sciences, Albany College of Pharmacy and Health Sciences, Albany, New York; and
| | - José Antunes-Rodrigues
- Department of Physiology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Alexandre A. Steiner
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
41
|
Abstract
The hypothalamus is an evolutionarily conserved brain structure that regulates an organism's basic functions, such as homeostasis and reproduction. Several hypothalamic nuclei and neuronal circuits have been the focus of many studies seeking to understand their role in regulating these basic functions. Within the hypothalamic neuronal populations, the arcuate melanocortin system plays a major role in controlling homeostatic functions. The arcuate pro-opiomelanocortin (POMC) neurons in particular have been shown to be critical regulators of metabolism and reproduction because of their projections to several brain areas both in and outside of the hypothalamus, such as autonomic regions of the brain stem and spinal cord. Here, we review and discuss the current understanding of POMC neurons from their development and intracellular regulators to their physiological functions and pathological dysregulation.
Collapse
Affiliation(s)
- Chitoku Toda
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, Connecticut 06520; .,Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Anna Santoro
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, Connecticut 06520; .,Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Jung Dae Kim
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, Connecticut 06520; .,Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Sabrina Diano
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, Connecticut 06520; .,Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut 06520.,Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06520.,Section of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut 06520
| |
Collapse
|
42
|
Bando M, Iwakura H, Ueda Y, Ariyasu H, Inaba H, Furukawa Y, Furuta H, Nishi M, Akamizu T. IL-1β directly suppress ghrelin mRNA expression in ghrelin-producing cells. Mol Cell Endocrinol 2017; 447:45-51. [PMID: 28237719 DOI: 10.1016/j.mce.2017.02.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/20/2017] [Accepted: 02/20/2017] [Indexed: 02/05/2023]
Abstract
In animal models, ghrelin production is suppressed by LPS administration. To elucidate the detailed molecular mechanisms involved in the phenomenon, we investigated the effects of LPS and LPS-inducible cytokines, including TNF-α, IL-1β, and IL-6, on the expression of ghrelin in the ghrelin-producing cell line MGN3-1. These cells expressed IL-1R, and IL-1β significantly suppressed ghrelin mRNA levels. The suppressive effects of IL-1β were attenuated by knockdown of IKKβ, suggesting the involvement of the NF-κB pathway. These results suggested that IL-1β is a major regulator of ghrelin expression during inflammatory processes.
Collapse
Affiliation(s)
- Mika Bando
- The First Department of Medicine, Wakayama Medical University, Japan
| | - Hiroshi Iwakura
- The First Department of Medicine, Wakayama Medical University, Japan.
| | - Yoko Ueda
- The First Department of Medicine, Wakayama Medical University, Japan
| | - Hiroyuki Ariyasu
- The First Department of Medicine, Wakayama Medical University, Japan
| | - Hidefumi Inaba
- The First Department of Medicine, Wakayama Medical University, Japan
| | - Yasushi Furukawa
- The First Department of Medicine, Wakayama Medical University, Japan
| | - Hiroto Furuta
- The First Department of Medicine, Wakayama Medical University, Japan
| | - Masahiro Nishi
- The First Department of Medicine, Wakayama Medical University, Japan
| | - Takashi Akamizu
- The First Department of Medicine, Wakayama Medical University, Japan
| |
Collapse
|
43
|
Laursen TL, Zak RB, Shute RJ, Heesch MWS, Dinan NE, Bubak MP, La Salle DT, Slivka DR. Leptin, adiponectin, and ghrelin responses to endurance exercise in different ambient conditions. Temperature (Austin) 2017; 4:166-175. [PMID: 28680932 DOI: 10.1080/23328940.2017.1294235] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Excessive positive energy balance is a major factor leading to obesity. The ability to alter the appetite-regulating hormones leptin, adiponectin, and ghrelin may help decrease excessive energy intake. Exercise and exposure to extreme temperatures can independently affect these appetite-regulating hormones. PURPOSE To determine the effect of exercising in different environmental conditions on the circulating concentrations of leptin, adiponectin, and ghrelin. METHODS Eleven recreationally-trained male participants completed 3 separate 1 h cycling bouts at 60% Wmax in hot, cold, and room temperature conditions (33°C, 7°C, 20°C), followed by a 3 h recovery at room temperature. Blood was drawn pre-exercise, post-exercise, and 3 h post-exercise. Hematocrit and hemoglobin were measured to account for change in plasma volume. RESULTS Leptin concentrations were lower at post and 3 h post-exercise compared with pre-exercise, with and without correction for plasma volume shifts, regardless of temperature (p < 0.05). Adiponectin was higher post-exercise compared with pre-exercise (p = 0.021) but not 3 h post-exercise (p = 0.084) without correction for plasma volume shifts. However, adiponectin concentrations were not different at any time point when plasma volume shifts were accounted for (p > 0.05). Total ghrelin and acylated ghrelin concentrations were not affected at post and 3 h post-exercise compared with pre-exercise, with and without correcting for plasma volume shifts, regardless of ambient temperature (p > 0.05). No differences in leptin, adiponectin, or ghrelin were found between trials (p > 0.05). CONCLUSION Temperature does not affect the circulating concentrations of appetite-regulating hormones during an acute bout of endurance exercise.
Collapse
Affiliation(s)
- Terence L Laursen
- School of Health, Physical Education, and Recreation, Exercise Physiology Laboratory, University of Nebraska-Omaha, Omaha, NE, USA
| | - Roksana B Zak
- School of Health, Physical Education, and Recreation, Exercise Physiology Laboratory, University of Nebraska-Omaha, Omaha, NE, USA
| | - Robert J Shute
- School of Health, Physical Education, and Recreation, Exercise Physiology Laboratory, University of Nebraska-Omaha, Omaha, NE, USA
| | | | - Nicholas E Dinan
- School of Health, Physical Education, and Recreation, Exercise Physiology Laboratory, University of Nebraska-Omaha, Omaha, NE, USA
| | - Matthew P Bubak
- School of Health, Physical Education, and Recreation, Exercise Physiology Laboratory, University of Nebraska-Omaha, Omaha, NE, USA
| | - D Taylor La Salle
- School of Health, Physical Education, and Recreation, Exercise Physiology Laboratory, University of Nebraska-Omaha, Omaha, NE, USA
| | - Dustin R Slivka
- School of Health, Physical Education, and Recreation, Exercise Physiology Laboratory, University of Nebraska-Omaha, Omaha, NE, USA
| |
Collapse
|
44
|
Tuomikoski P, Savolainen-Peltonen H. Vasomotor symptoms and metabolic syndrome. Maturitas 2017; 97:61-65. [PMID: 28159064 DOI: 10.1016/j.maturitas.2016.12.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 12/16/2016] [Indexed: 12/28/2022]
Abstract
A vast majority of menopausal women suffer from vasomotor symptoms, such as hot flushes and night sweats, the mean duration of which may be up to 7-10 years. In addition to a decreased quality of life, vasomotor symptoms may have an impact on overall health. Vasomotor symptoms are associated with overactivity of the sympathetic nervous system, and sympathetic overdrive in turn is associated with metabolic syndrome, which is a known risk factor for cardiovascular disease. Menopausal hot flushes have a complex relationship to different features of the metabolic syndrome and not all data point towards an association between vasomotor symptoms and metabolic syndrome. Thus, it is still unclear whether vasomotor symptoms are an independent risk factor for metabolic syndrome. Research in this area is constantly evolving and we present here the most recent data on the possible association between menopausal vasomotor symptoms and the metabolic syndrome.
Collapse
Affiliation(s)
| | - Hanna Savolainen-Peltonen
- University of Helsinki, Helsinki, Finland; Helsinki University Hospital, Department of Obstetrics and Gynecology, 00029 Helsinki, Finland
| |
Collapse
|
45
|
Wauman J, Zabeau L, Tavernier J. The Leptin Receptor Complex: Heavier Than Expected? Front Endocrinol (Lausanne) 2017; 8:30. [PMID: 28270795 PMCID: PMC5318964 DOI: 10.3389/fendo.2017.00030] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/01/2017] [Indexed: 12/31/2022] Open
Abstract
Under normal physiological conditions, leptin and the leptin receptor (ObR) regulate the body weight by balancing food intake and energy expenditure. However, this adipocyte-derived hormone also directs peripheral processes, including immunity, reproduction, and bone metabolism. Leptin, therefore, can act as a metabolic switch connecting the body's nutritional status to high energy consuming processes. We provide an extensive overview of current structural insights on the leptin-ObR interface and ObR activation, coupling to signaling pathways and their negative regulation, and leptin functioning under normal and pathophysiological conditions (obesity, autoimmunity, cancer, … ). We also discuss possible cross-talk with other receptor systems on the receptor (extracellular) and signaling cascade (intracellular) levels.
Collapse
Affiliation(s)
- Joris Wauman
- Cytokine Receptor Laboratory, Faculty of Medicine and Health Sciences, Department of Biochemistry, Ghent University, Ghent, Belgium
- VIB Medical Biotechnology Center, VIB, Ghent, Belgium
| | - Lennart Zabeau
- Cytokine Receptor Laboratory, Faculty of Medicine and Health Sciences, Department of Biochemistry, Ghent University, Ghent, Belgium
- VIB Medical Biotechnology Center, VIB, Ghent, Belgium
| | - Jan Tavernier
- Cytokine Receptor Laboratory, Faculty of Medicine and Health Sciences, Department of Biochemistry, Ghent University, Ghent, Belgium
- VIB Medical Biotechnology Center, VIB, Ghent, Belgium
- *Correspondence: Jan Tavernier,
| |
Collapse
|
46
|
Abstract
In the current study, we examined the effects of LPS and inflammatory cytokines including IL-1β, TNF-α, and IL-6 on the expression of ghrelin in MGN3-1 cells. We found that IL-1β, and TNF-α with lesser extent, significantly suppressed ghrelin mRNA expression in the cells. MGN3-1 cells expressed IL-1β receptor and IL-1β significantly stimulated NF-κB, p38, JNK, and ERK pathways. Knockdown of IKK2 by siRNA significantly attenuated the suppression of ghrelin mRNA by IL-1β. These results indicate that IL-1β directly suppressed ghrelin mRNA via NF-κB pathway at least partially, which may have a role in the regulation of appetite during inflammation.
Collapse
Affiliation(s)
- Hiroshi Iwakura
- The First Department of Medicine, Wakayama Medical University, Wakayama 841-8509, Japan
| | - Mika Bando
- The First Department of Medicine, Wakayama Medical University, Wakayama 841-8509, Japan
| | - Yoko Ueda
- The First Department of Medicine, Wakayama Medical University, Wakayama 841-8509, Japan
| | - Takashi Akamizu
- The First Department of Medicine, Wakayama Medical University, Wakayama 841-8509, Japan
| |
Collapse
|
47
|
Ping X, Han D, Jiang Z, Li C. Circadian patterns of plasma leptin, insulin and glucose concentration in the toad-headed lizard Phrynocephalus versicolor. BIOL RHYTHM RES 2016. [DOI: 10.1080/09291016.2016.1210283] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Xiaoge Ping
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Dawei Han
- College of Biological Sciences, China Agricultural University, Beijing, China
- Department of Biology, Truman State University, Kirksville, MO, USA
| | - Zhigang Jiang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Chunwang Li
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
48
|
Liao WH, Henneberg M, Langhans W. Immunity-Based Evolutionary Interpretation of Diet-Induced Thermogenesis. Cell Metab 2016; 23:971-979. [PMID: 27304499 DOI: 10.1016/j.cmet.2016.05.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Diet-induced thermogenesis (DIT) has often been argued to be a physiological defense against obesity, but no empirical proof of its effectiveness in limiting human body weight gain is available. We here propose an immune explanation of DIT-i.e., that it results from the coevolution of host and gut microbiota (especially Firmicutes) that ferment ingested food and proliferate, causing periodic, vagally mediated increases in thermogenesis aimed at curtailing their expansion. Because of this evolutionary adaptive significance related to the immune system, DIT is not effective as an "adaptation" to maintain a certain body mass. Were DIT an effective adaptation to prevent obesity, the current obesity epidemic might not have occurred.
Collapse
Affiliation(s)
- Wan-Hui Liao
- Physiology and Behavior Laboratory, Department of Health Sciences and Technology, ETH Zurich, 8603 Schwerzenbach, Switzerland; Institute of Anatomy, University of Zurich, 8057 Zurich, Switzerland.
| | - Maciej Henneberg
- Institute of Evolutionary Medicine, Medical Faculty, University of Zurich, 8057 Zurich, Switzerland; Biological Anthropology and Comparative Anatomy Unit, University of Adelaide, Adelaide 5005, Australia
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Department of Health Sciences and Technology, ETH Zurich, 8603 Schwerzenbach, Switzerland.
| |
Collapse
|
49
|
Inflammatory transcription factors as activation markers and functional readouts in immune-to-brain communication. Brain Behav Immun 2016; 54:1-14. [PMID: 26348582 DOI: 10.1016/j.bbi.2015.09.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 08/31/2015] [Accepted: 09/04/2015] [Indexed: 02/06/2023] Open
Abstract
Immune-to-brain communication pathways involve humoral mediators, including cytokines, central modulation by neuronal afferents and immune cell trafficking to the brain. During systemic inflammation these pathways contribute to mediating brain-controlled sickness symptoms including fever. Experimentally, activation of these signaling pathways can be mimicked and studied when injecting animals with pathogen associated molecular patterns (PAMPS). One central component of the brain inflammatory response, which leads, for example, to fever induction, is transcriptional activation of brain cells via cytokines and PAMPS. We and others have studied the spatiotemporal activation and the physiological significance of transcription factors for the induction of inflammation within the brain and the manifestation of fever. Evidence has revealed a role of nuclear factor (NF)κB in the initiation, signal transducer and activator of transcription (STAT)3 in the maintenance and NF-interleukin (IL)6 in the maintenance or even termination of brain-inflammation and fever. Moreover, psychological stressors, such as exposure to a novel environment, leads to increased body core temperature and genomic NF-IL6-activation, suggesting a potential use of NF-IL6-immunohistochemistry as a multimodal brain cell activation marker and a role for NF-IL6 for differential brain activity. In addition, the nutritional status, as reflected by circulating levels of the cytokine-like hormone leptin, influence immune-to-brain communication and age-dependent changes in LPS-induced fever. Overall, transcription factors remain therapeutically important targets for the treatment of brain-inflammation and fever induction during infectious/non-infectious inflammatory and psychological stress. However, the exact physiological role and significance of these transcription factors requires to be further investigated.
Collapse
|
50
|
Kwon O, Kim KW, Kim MS. Leptin signalling pathways in hypothalamic neurons. Cell Mol Life Sci 2016; 73:1457-77. [PMID: 26786898 PMCID: PMC11108307 DOI: 10.1007/s00018-016-2133-1] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 12/20/2015] [Accepted: 01/07/2016] [Indexed: 12/25/2022]
Abstract
Leptin is the most critical hormone in the homeostatic regulation of energy balance among those so far discovered. Leptin primarily acts on the neurons of the mediobasal part of hypothalamus to regulate food intake, thermogenesis, and the blood glucose level. In the hypothalamic neurons, leptin binding to the long form leptin receptors on the plasma membrane initiates multiple signaling cascades. The signaling pathways known to mediate the actions of leptin include JAK-STAT signaling, PI3K-Akt-FoxO1 signaling, SHP2-ERK signaling, AMPK signaling, and mTOR-S6K signaling. Recent evidence suggests that leptin signaling in hypothalamic neurons is also linked to primary cilia function. On the other hand, signaling molecules/pathways mitigating leptin actions in hypothalamic neurons have been extensively investigated in an effort to treat leptin resistance observed in obesity. These include SOCS3, tyrosine phosphatase PTP1B, and inflammatory signaling pathways such as IKK-NFκB and JNK signaling, and ER stress-mitochondrial signaling. In this review, we discuss leptin signaling pathways in the hypothalamus, with a particular focus on the most recently discovered pathways.
Collapse
Affiliation(s)
- Obin Kwon
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
- Appetite Regulation Laboratory, Asan Institute for Life Science, Seoul, 05505, Korea
| | - Ki Woo Kim
- Department of Pharmacology, Yonsei University Wonju College of Medicine, Wonju, 26426, Korea
- Department of Global Medical Science, Yonsei University Wonju College of Medicine, Wonju, 26426, Korea
| | - Min-Seon Kim
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea.
- Appetite Regulation Laboratory, Asan Institute for Life Science, Seoul, 05505, Korea.
| |
Collapse
|