1
|
Edwards MM, Nguyen HK, Dodson AD, Herbertson AJ, Honeycutt MK, Slattery JD, Rambousek JR, Tsui E, Wolden-Hanson T, Wietecha TA, Graham JL, Tapia GP, Sikkema CL, O'Brien KD, Mundinger TO, Peskind ER, Ryu V, Havel PJ, Khan AM, Tabosky GJ, Blevins JE. Sympathetic innervation of interscapular brown adipose tissue is not a predominant mediator of OT-elicited reductions of body weight gain and adiposity in male diet-induced obese rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.12.612710. [PMID: 39345420 PMCID: PMC11430106 DOI: 10.1101/2024.09.12.612710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Recent studies indicate that central administration of oxytocin (OT) reduces body weight (BW) in high fat diet-induced obese (DIO) rodents by reducing energy intake and increasing energy expenditure (EE). Previous studies in our lab have shown that administration of OT into the fourth ventricle (4V; hindbrain) elicits weight loss and stimulates interscapular brown adipose tissue temperature (TIBAT) in DIO rats. We hypothesized that OT-elicited stimulation of sympathetic nervous system (SNS) activation of IBAT contributes to its ability to activate BAT and reduce BW in DIO rats. To test this, we determined the effect of disrupting SNS activation of IBAT on OT-elicited stimulation of TIBAT and reduction of BW in DIO rats. We first confirmed that bilateral surgical SNS denervation to IBAT was successful based on having achieved ≥ 60% reduction in IBAT norepinephrine (NE) content from DIO rats. NE content was selectively reduced in IBAT by 94.7 ± 2.7, 96.8 ± 1.8 and 85.9 ± 6.1% (P<0.05) at 1, 6 and 7-weeks post-denervation, respectively, and was unchanged in liver or inguinal white adipose tissue. We then measured the impact of bilateral surgical SNS denervation to IBAT on the ability of acute 4V OT (1, 5 μg) to stimulate TIBAT in DIO rats. We found that the high dose of 4V OT (5 μg) stimulated TIBAT similarly between sham and denervated rats (P=NS) and that the effects of 4V OT to stimulate TIBAT did not require beta-3 adrenergic receptor signaling. We subsequently measured the effect of bilateral surgical denervation of IBAT on the effect of chronic 4V OT (16 nmol/day) or vehicle infusion to reduce BW, adiposity and energy intake in DIO rats. Chronic 4V OT reduced BW gain by -7.2 ± 9.6 g and -14.1 ± 8.8 g in sham and denervated rats (P<0.05 vs vehicle treatment), respectively, and this effect was similar between groups (P=NS). These effects were associated with reductions in adiposity and energy intake (P<0.05). Collectively, these findings support the hypothesis that sympathetic innervation of IBAT is not required for central OT to increase BAT thermogenesis and reduce BW gain and adiposity in male DIO rats.
Collapse
|
2
|
Dodson AD, Herbertson AJ, Honeycutt MK, Vered R, Slattery JD, Goldberg M, Tsui E, Wolden-Hanson T, Graham JL, Wietecha TA, O’Brien KD, Havel PJ, Sikkema CL, Peskind ER, Mundinger TO, Taborsky GJ, Blevins JE. Sympathetic Innervation of Interscapular Brown Adipose Tissue Is Not a Predominant Mediator of Oxytocin-Induced Brown Adipose Tissue Thermogenesis in Female High Fat Diet-Fed Rats. Curr Issues Mol Biol 2024; 46:11394-11424. [PMID: 39451559 PMCID: PMC11506511 DOI: 10.3390/cimb46100679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024] Open
Abstract
Recent studies have indicated that hindbrain [fourth ventricle (4V)] administration of the neurohypophyseal hormone, oxytocin (OT), reduces body weight, energy intake and stimulates interscapular brown adipose tissue temperature (TIBAT) in male diet-induced obese (DIO) rats. What remains unclear is whether chronic hindbrain (4V) OT can impact body weight in female high fat diet-fed (HFD) rodents and whether this involves activation of brown adipose tissue (BAT). We hypothesized that OT-elicited stimulation of sympathetic nervous system (SNS) activation of interscapular brown adipose tissue (IBAT) contributes to its ability to activate BAT and reduce body weight in female high HFD-fed rats. To test this hypothesis, we determined the effect of disrupting SNS activation of IBAT on OT-elicited stimulation of TIBAT and reduction of body weight in DIO rats. We first measured the impact of bilateral surgical SNS denervation to IBAT on the ability of acute 4V OT (0.5, 1, and 5 µg ≈ 0.5, 0.99, and 4.96 nmol) to stimulate TIBAT in female HFD-fed rats. We found that the high dose of 4V OT (5 µg ≈ 4.96 nmol) stimulated TIBAT similarly between sham rats and denervated rats (p = NS). We subsequently measured the effect of bilateral surgical denervation of IBAT on the effect of chronic 4V OT (16 nmol/day ≈ 16.1 μg/day) or vehicle infusion to reduce body weight, adiposity and energy intake in female HFD-fed rats (N = 7-8/group). Chronic 4V OT reduced body weight gain (sham: -18.0 ± 4.9 g; denervation: -15.9 ± 3.7 g) and adiposity (sham: -13.9 ± 3.7 g; denervation: -13.6 ± 2.4 g) relative to vehicle treatment (p < 0.05) and these effects were similar between groups (p = NS). These effects were attributed, in part, to reduced energy intake evident during weeks 2 (p < 0.05) and 3 (p < 0.05). To test whether these results translate to other female rodent species, we also examined the effect of chronic 4V infusion of OT on body weight and adiposity in two strains of female HFD-fed mice. Similar to what we found in the HFD-fed rat model, we also found that chronic 4V OT (16 nmol/day) infusion resulted in reduced body weight gain, adiposity and energy intake in female DIO C57BL/6J and DBA/2J mice (p < 0.05 vs. vehicle). Together, these findings suggest that (1) sympathetic innervation of IBAT is not necessary for OT-elicited increases in BAT thermogenesis and weight loss in female HFD-fed rats and (2) the effects of OT to reduce weight gain and adiposity translate to other female mouse models of diet-induced obesity (DIO).
Collapse
Affiliation(s)
- Andrew D. Dodson
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (A.D.D.); (A.J.H.); (M.K.H.); (R.V.); (J.D.S.); (M.G.); (E.T.); (T.W.-H.); (C.L.S.); (E.R.P.)
| | - Adam J. Herbertson
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (A.D.D.); (A.J.H.); (M.K.H.); (R.V.); (J.D.S.); (M.G.); (E.T.); (T.W.-H.); (C.L.S.); (E.R.P.)
| | - Mackenzie K. Honeycutt
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (A.D.D.); (A.J.H.); (M.K.H.); (R.V.); (J.D.S.); (M.G.); (E.T.); (T.W.-H.); (C.L.S.); (E.R.P.)
| | - Ron Vered
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (A.D.D.); (A.J.H.); (M.K.H.); (R.V.); (J.D.S.); (M.G.); (E.T.); (T.W.-H.); (C.L.S.); (E.R.P.)
| | - Jared D. Slattery
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (A.D.D.); (A.J.H.); (M.K.H.); (R.V.); (J.D.S.); (M.G.); (E.T.); (T.W.-H.); (C.L.S.); (E.R.P.)
| | - Matvey Goldberg
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (A.D.D.); (A.J.H.); (M.K.H.); (R.V.); (J.D.S.); (M.G.); (E.T.); (T.W.-H.); (C.L.S.); (E.R.P.)
| | - Edison Tsui
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (A.D.D.); (A.J.H.); (M.K.H.); (R.V.); (J.D.S.); (M.G.); (E.T.); (T.W.-H.); (C.L.S.); (E.R.P.)
| | - Tami Wolden-Hanson
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (A.D.D.); (A.J.H.); (M.K.H.); (R.V.); (J.D.S.); (M.G.); (E.T.); (T.W.-H.); (C.L.S.); (E.R.P.)
| | - James L. Graham
- Department of Nutrition, University of California, Davis, CA 95616, USA; (J.L.G.); (P.J.H.)
| | - Tomasz A. Wietecha
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA; (T.A.W.); (T.O.M.)
- UW Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, WA 98109, USA;
| | - Kevin D. O’Brien
- UW Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, WA 98109, USA;
- Division of Cardiology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Peter J. Havel
- Department of Nutrition, University of California, Davis, CA 95616, USA; (J.L.G.); (P.J.H.)
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Carl L. Sikkema
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (A.D.D.); (A.J.H.); (M.K.H.); (R.V.); (J.D.S.); (M.G.); (E.T.); (T.W.-H.); (C.L.S.); (E.R.P.)
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Elaine R. Peskind
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (A.D.D.); (A.J.H.); (M.K.H.); (R.V.); (J.D.S.); (M.G.); (E.T.); (T.W.-H.); (C.L.S.); (E.R.P.)
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Thomas O. Mundinger
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA; (T.A.W.); (T.O.M.)
| | - Gerald J. Taborsky
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (A.D.D.); (A.J.H.); (M.K.H.); (R.V.); (J.D.S.); (M.G.); (E.T.); (T.W.-H.); (C.L.S.); (E.R.P.)
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA; (T.A.W.); (T.O.M.)
| | - James E. Blevins
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (A.D.D.); (A.J.H.); (M.K.H.); (R.V.); (J.D.S.); (M.G.); (E.T.); (T.W.-H.); (C.L.S.); (E.R.P.)
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA; (T.A.W.); (T.O.M.)
| |
Collapse
|
3
|
Sakamoto K, Butera MA, Zhou C, Maurizi G, Chen B, Ling L, Shawkat A, Patlolla L, Thakker K, Calle V, Morgan DA, Rahmouni K, Schwartz GJ, Tahiri A, Buettner C. Overnutrition causes insulin resistance and metabolic disorder through increased sympathetic nervous system activity. Cell Metab 2024:S1550-4131(24)00376-0. [PMID: 39437790 DOI: 10.1016/j.cmet.2024.09.012] [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: 01/09/2024] [Revised: 06/19/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024]
Abstract
The mechanisms underlying obesity-induced insulin resistance remain incompletely understood, as impaired cellular insulin signaling, traditionally considered the primary driver of insulin resistance, does not always accompany impaired insulin action. Overnutrition rapidly increases plasma norepinephrine (NE), suggesting overactivation of the sympathetic nervous system (SNS). However, the role of the SNS in obesity is controversial, as both increased and decreased SNS activity (SNA) have been reported. Here, we show that reducing catecholamine (CA) release from the SNS protects against overnutrition-induced insulin resistance as well as hyperglucagonemia, adipose tissue dysfunction, and fatty liver disease, as we demonstrate utilizing a mouse model of inducible and peripherally restricted deletion of tyrosine hydroxylase (th; THΔper). A key mechanism through which heightened SNA induces insulin resistance is by triggering adipose tissue lipolysis. Increased SNA emerges as a critical driver in the pathogenesis of overnutrition-induced insulin resistance and metabolic disease independent of cellular insulin signaling.
Collapse
Affiliation(s)
- Kenichi Sakamoto
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mary A Butera
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chunxue Zhou
- Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Giulia Maurizi
- Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bandy Chen
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Li Ling
- Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adham Shawkat
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Likhitha Patlolla
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Kavira Thakker
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Victor Calle
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Donald A Morgan
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Kamal Rahmouni
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Gary J Schwartz
- Department of Medicine & Neuroscience, Albert Einstein College of Medicine, New York, NY, USA
| | - Azeddine Tahiri
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Christoph Buettner
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
4
|
Wang Y, Duan C, Du X, Zhu Y, Wang L, Hu J, Sun Y. Vagus Nerve and Gut-Brain Communication. Neuroscientist 2024:10738584241259702. [PMID: 39041416 DOI: 10.1177/10738584241259702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
The vagus nerve, as an important component of the gut-brain axis, plays a crucial role in the communication between the gut and brain. It influences food intake, fat metabolism, and emotion by regulating the gut-brain axis, which is closely associated with the development of gastrointestinal, psychiatric, and metabolism-related disorders. In recent years, significant progress has been made in understanding the vagus-mediated regulatory pathway, highlighting its profound implications in the development of many diseases. Here, we summarize the latest advancements in vagus-mediated gut-brain pathways and the novel interventions targeting the vagus nerve. This will provide valuable insights for future research on treatment of obesity and gastrointestinal and depressive disorders based on vagus nerve stimulation.
Collapse
Affiliation(s)
- Yiyang Wang
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chenxi Duan
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xinyi Du
- Institute of Materiobiology, College of Sciences, Shanghai University, Shanghai, China
| | - Ying Zhu
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China
- Institute of Materiobiology, College of Sciences, Shanghai University, Shanghai, China
| | - Lihua Wang
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China
- Institute of Materiobiology, College of Sciences, Shanghai University, Shanghai, China
| | - Jun Hu
- Institute of Materiobiology, College of Sciences, Shanghai University, Shanghai, China
- The Interdisciplinary Research Center, Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China
| | - Yanhong Sun
- Institute of Materiobiology, College of Sciences, Shanghai University, Shanghai, China
| |
Collapse
|
5
|
Ehrhardt RA, Giesy SL, Hileman SM, Houseknecht KL, Boisclair YR. Effects of the central melanocortin system on feed intake, metabolic hormones and insulin action in the sheep. J Anim Sci 2023; 101:skad398. [PMID: 38035762 PMCID: PMC10734672 DOI: 10.1093/jas/skad398] [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: 09/05/2023] [Accepted: 11/29/2023] [Indexed: 12/02/2023] Open
Abstract
Voluntary feed intake is insufficient to meet the nutrient demands associated with late pregnancy in prolific ewes and early lactation in high-yielding dairy cows. Under these conditions, peripheral signals such as growth hormone and ceramides trigger adaptations aimed at preserving metabolic well-being. Recent work in rodents has shown that the central nervous system-melanocortin (CNS-MC) system, consisting of alpha-melanocyte-stimulating hormone (α-MSH) and agouti-related peptide (AGRP) acting respectively as agonist and antagonist on central MC receptors, contributes to the regulation of some of the same adaptations. To assess the effects of the CNC-MC on peripheral adaptations in ruminants, ewes were implanted with an intracerebroventricular cannula in the third ventricle and infused over days with artificial cerebrospinal fluid (aCSF), the α-MSH analog melanotan-I (MTI), or AGRP. Infusion of MTI at 0.03 nmol/h reduced intake, expressed as a fold of maintenance energy requirement (M), from 1.8 to 1.1 M (P < 0.0001), whereas AGRP at 0.3 nmol/h increased intake from 1.8 to 2.0 M (P < 0.01); these doses were used in all subsequent experiments. To assess the effect of MTI on plasma variables, sheep were fed ad libitum and infused with aCSF or MTI or pair-fed to MTI-treated sheep and infused with aCSF (aCSFPF). Feed intake of the MTI and aCSFPF groups was 40% lower than the aCSF group (P < 0.0001). MTI increased plasma triiodothyronine and thyroxine in an intake-independent manner (P < 0.05 or less) but was devoid of effects on plasma glucose, insulin, and cortisol. None of these variables were altered by AGRP infusion in sheep fed at a fixed intake of 1.6 M. To assess the effect of CNS-MC activation on insulin action, ewes were infused with aCSF or MTI over the last 3 d of a 14-d period when energy intake was limited to 0.3 M and studied under basal conditions and during hyperinsulinemic-euglycemic clamps. MTI had no effect on plasma glucose, plasma insulin, or glucose entry rate under basal conditions but blunted the ability of insulin to inhibit endogenous glucose production during hyperinsulinemic-euglycemic clamps (P < 0.0001). Finally, MTI tended to reduce plasma leptin in sheep fed at 0.3 M (P < 0.08), and this effect became significant at 0.6 M (P < 0.05); MTI had no effect on plasma adiponectin irrespective of feeding level. These data suggest a role for the CNC-MC in regulating metabolic efficiency and peripheral insulin action.
Collapse
Affiliation(s)
- Richard A Ehrhardt
- Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI 48824, USA
| | - Sarah L Giesy
- Department of Animal Science, Cornell University, Ithaca, NY 14853, USA
| | - Stanley M Hileman
- Department of Physiology, Pharmacology, and Toxicology, West Virginia University, Morgantown, WV 26506, USA
| | - Karen L Houseknecht
- Department of Biomedical Sciences, University of New England, Portland, ME 04103, USA
| | - Yves R Boisclair
- Department of Animal Science, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
6
|
Qian X, Meng X, Zhang S, Zeng W. Neuroimmune regulation of white adipose tissues. FEBS J 2022; 289:7830-7853. [PMID: 34564950 DOI: 10.1111/febs.16213] [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: 05/05/2021] [Revised: 08/21/2021] [Accepted: 09/24/2021] [Indexed: 01/14/2023]
Abstract
The white adipose tissues (WAT) are located in distinct depots throughout the body. They serve as an energy reserve, providing fatty acids for other tissues via lipolysis when needed, and function as an endocrine organ to regulate systemic metabolism. Their activities are coordinated through intercellular communications among adipocytes and other cell types such as residential and infiltrating immune cells, which are collectively under neuronal control. The adipocytes and immune subtypes including macrophages/monocytes, eosinophils, neutrophils, group 2 innate lymphoid cells (ILC2s), T and B cells, dendritic cells (DCs), and natural killer (NK) cells display cellular and functional diversity in response to the energy states and contribute to metabolic homeostasis and pathological conditions. Accumulating evidence reveals that neuronal innervations control lipid deposition and mobilization via regulating lipolysis, adipocyte size, and cellularity. Vice versa, the neuronal innervations and activity are influenced by cellular factors in the WAT. Though the literature describing adipose tissue cells is too extensive to cover in detail, we strive to highlight a selected list of neuronal and immune components in this review. The cell-to-cell communications and the perspective of neuroimmune regulation are emphasized to enlighten the potential therapeutic opportunities for treating metabolic disorders.
Collapse
Affiliation(s)
- Xinmin Qian
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Xia Meng
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Shan Zhang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Wenwen Zeng
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China.,Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China
| |
Collapse
|
7
|
Puente-Ruiz SC, Jais A. Reciprocal signaling between adipose tissue depots and the central nervous system. Front Cell Dev Biol 2022; 10:979251. [PMID: 36200038 PMCID: PMC9529070 DOI: 10.3389/fcell.2022.979251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
In humans, various dietary and social factors led to the development of increased brain sizes alongside large adipose tissue stores. Complex reciprocal signaling mechanisms allow for a fine-tuned interaction between the two organs to regulate energy homeostasis of the organism. As an endocrine organ, adipose tissue secretes various hormones, cytokines, and metabolites that signal energy availability to the central nervous system (CNS). Vice versa, the CNS is a critical regulator of adipose tissue function through neural networks that integrate information from the periphery and regulate sympathetic nerve outflow. This review discusses the various reciprocal signaling mechanisms in the CNS and adipose tissue to maintain organismal energy homeostasis. We are focusing on the integration of afferent signals from the periphery in neuronal populations of the mediobasal hypothalamus as well as the efferent signals from the CNS to adipose tissue and its implications for adipose tissue function. Furthermore, we are discussing central mechanisms that fine-tune the immune system in adipose tissue depots and contribute to organ homeostasis. Elucidating this complex signaling network that integrates peripheral signals to generate physiological outputs to maintain the optimal energy balance of the organism is crucial for understanding the pathophysiology of obesity and metabolic diseases such as type 2 diabetes.
Collapse
|
8
|
Almeida DL, Moreira VM, Cardoso LE, Junior MDF, Pavanelo A, Ribeiro TA, da Silva Franco CC, Tófolo LP, Peres MNC, Ribeiro MVG, Ferreira ARO, Gomes RM, Miranda RA, Trevenzoli IH, Armitage JA, Palma-Rigo K, de Freitas Mathias PC. Lean in one way, in obesity another: effects of moderate exercise in brown adipose tissue of early overfed male Wistar rats. Int J Obes (Lond) 2022; 46:137-143. [PMID: 34552207 DOI: 10.1038/s41366-021-00969-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 09/01/2021] [Accepted: 09/13/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND Early postnatal overfeeding (PO) induces long-term overweight and reduces brown adipose tissue (BAT) thermogenesis. Exercise has been suggested as a possible intervention to increase BAT function. In this study, we investigated chronical effects of moderate-intensity exercise in BAT function in postnatal overfed male Wistar rats METHODS: Litters' delivery was on postnatal-day 0 - PN0. At PN2, litters were adjusted to nine (normal litter - NL) or three pups (small litter - SL) per dam. Animals were weaned on PN21 and in PN30 randomly divided into sedentary (NL-Sed and SL-Sed) or exercised (NL-Exe and SL-Exe), N of 14 litters per group. Exercise protocol started (PN30) with an effort test; training sessions were performed three times weekly at 60% of the VO2max achieved in effort test, until PN80. On PN81, a temperature transponder was implanted beneath the interscapular BAT, whose temperature was assessed in periods of lights-on and -off from PN87 to PN90. Sympathetic nerve activation of BAT was registered at PN90. Animals were euthanized at PN91 and tissues collected RESULTS: PO impaired BAT thermogenesis in lights-on (pPO < 0.0001) and -off (pPO < 0.01). Exercise increased BAT temperature in lights-on (pExe < 0.0001). In NL-Exe, increased BAT activity was associated with higher sympathetic activity (pExe < 0.05), β3-AR (pExe < 0.001), and UCP1 (pExe < 0.001) content. In SL-Exe, increasing BAT thermogenesis is driven by a combination of tissue morphology remodeling (pExe < 0.0001) with greater effect in increasing UCP1 (pExe < 0.001) and increased β3-AR (pExe < 0.001) content. CONCLUSION Moderate exercise chronically increased BAT thermogenesis in both, NL and SL groups. In NL-Exe by increasing Sympathetic activity, and in SL-Exe by a combination of increased β3-AR and UCP1 content with morphologic remodeling of BAT. Chronically increasing BAT thermogenesis in obese subjects may lead to higher overall energy expenditure, favoring the reduction of obesity and related comorbidities.
Collapse
Affiliation(s)
- Douglas Lopes Almeida
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil.
- Department of Physiology, State University of Londrina, Londrina, Paraná, Brazil.
| | - Veridiana Mota Moreira
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
- Department of Physiology, State University of Sergipe, São Cristóvão, Sergipe, Brazil
| | - Lucas Eduardo Cardoso
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | | | - Audrei Pavanelo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Tatiane Aparecida Ribeiro
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Claudinéia Conationi da Silva Franco
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Laize Perón Tófolo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Maria Natália Chimirri Peres
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Maiara Vanusa Guedes Ribeiro
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Anna Rebeka Oliveira Ferreira
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Rodrigo Mello Gomes
- Physiological Sciences Department, Federal University of Goiás, Av Esperança, Goiânia/GO, Brazil
| | - Rosiane Aparecida Miranda
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, 550 Av, Pedro Calmon, Rio de Janeiro, Brazil
| | - Isis Hara Trevenzoli
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, 550 Av, Pedro Calmon, Rio de Janeiro, Brazil
| | - James Andrew Armitage
- Deakin University, School of Medicine, Optometry, 75 Pigdons Rd, Waurn Ponds, VIC, Australia
| | - Kesia Palma-Rigo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| | - Paulo Cesar de Freitas Mathias
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Cell Biology and Genetics, State University of Maringá, 5790 Av, Colombo, Maringá/PR, Brazil
| |
Collapse
|
9
|
Li F, Cui X, Jing J, Wang S, Shi H, Xue B, Shi H. Brown Fat Dnmt3b Deficiency Ameliorates Obesity in Female Mice. Life (Basel) 2021; 11:life11121325. [PMID: 34947856 PMCID: PMC8703316 DOI: 10.3390/life11121325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/26/2021] [Accepted: 11/26/2021] [Indexed: 11/16/2022] Open
Abstract
Obesity results from a chronic energy imbalance due to energy intake exceeding energy expenditure. Activation of brown fat thermogenesis has been shown to combat obesity. Epigenetic regulation, including DNA methylation, has emerged as a key regulator of brown fat thermogenic function. Here we aimed to study the role of Dnmt3b, a DNA methyltransferase involved in de novo DNA methylation, in the regulation of brown fat thermogenesis and obesity. We found that the specific deletion of Dnmt3b in brown fat promotes the thermogenic and mitochondrial program in brown fat, enhances energy expenditure, and decreases adiposity in female mice fed a regular chow diet. With a lean phenotype, the female knockout mice also exhibit increased insulin sensitivity. In addition, Dnmt3b deficiency in brown fat also prevents diet-induced obesity and insulin resistance in female mice. Interestingly, our RNA-seq analysis revealed an upregulation of the PI3K-Akt pathway in the brown fat of female Dnmt3b knockout mice. However, male Dnmt3b knockout mice have no change in their body weight, suggesting the existence of sexual dimorphism in the brown fat Dnmt3b knockout model. Our data demonstrate that Dnmt3b plays an important role in the regulation of brown fat function, energy metabolism and obesity in female mice.
Collapse
Affiliation(s)
- Fenfen Li
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (F.L.); (X.C.); (J.J.); (S.W.)
| | - Xin Cui
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (F.L.); (X.C.); (J.J.); (S.W.)
| | - Jia Jing
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (F.L.); (X.C.); (J.J.); (S.W.)
| | - Shirong Wang
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (F.L.); (X.C.); (J.J.); (S.W.)
| | - Huidong Shi
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| | - Bingzhong Xue
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (F.L.); (X.C.); (J.J.); (S.W.)
- Correspondence: (B.X.); (H.S.)
| | - Hang Shi
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (F.L.); (X.C.); (J.J.); (S.W.)
- Correspondence: (B.X.); (H.S.)
| |
Collapse
|
10
|
Edwards MM, Nguyen HK, Dodson AD, Herbertson AJ, Wietecha TA, Wolden-Hanson T, Graham JL, Honeycutt MK, Slattery JD, O’Brien KD, Havel PJ, Blevins JE. Effects of Combined Oxytocin and Beta-3 Receptor Agonist (CL 316243) Treatment on Body Weight and Adiposity in Male Diet-Induced Obese Rats. Front Physiol 2021; 12:725912. [PMID: 34566687 PMCID: PMC8457402 DOI: 10.3389/fphys.2021.725912] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/05/2021] [Indexed: 11/13/2022] Open
Abstract
Previous studies have indicated that oxytocin (OT) reduces body weight in diet-induced obese (DIO) rodents through reductions in energy intake and increases in energy expenditure. We recently demonstrated that hindbrain [fourth ventricular (4V)] administration of OT evokes weight loss and elevates interscapular brown adipose tissue temperature (T IBAT ) in DIO rats. What remains unclear is whether OT can be used as an adjunct with other drugs that directly target beta-3 receptors in IBAT to promote BAT thermogenesis and reduce body weight in DIO rats. We hypothesized that the combined treatment of OT and the beta-3 agonist, CL 316243, would produce an additive effect to decrease body weight and adiposity in DIO rats by reducing energy intake and increasing BAT thermogenesis. We assessed the effects of 4V infusions of OT (16 nmol/day) or vehicle (VEH) in combination with daily intraperitoneal injections of CL 316243 (0.5 mg/kg) or VEH on food intake, T IBAT , body weight and body composition. OT and CL 316243 alone reduced body weight by 7.8 ± 1.3% (P < 0.05) and 9.1 ± 2.1% (P < 0.05), respectively, but the combined treatment produced more pronounced weight loss (15.5 ± 1.2%; P < 0.05) than either treatment alone. These effects were associated with decreased adiposity, adipocyte size, energy intake and increased uncoupling protein 1 (UCP-1) content in epididymal white adipose tissue (EWAT) (P < 0.05). In addition, CL 316243 alone (P < 0.05) and in combination with OT (P < 0.05) elevated T IBAT and IBAT UCP-1 content and IBAT thermogenic gene expression. These findings are consistent with the hypothesis that the combined treatment of OT and the beta-3 agonist, CL 316243, produces an additive effect to decrease body weight. The findings from the current study suggest that the effects of the combined treatment on energy intake, fat mass, adipocyte size and browning of EWAT were not additive and appear to be driven, in part, by transient changes in energy intake in response to OT or CL 316243 alone as well as CL 316243-elicited reduction of fat mass and adipocyte size and induction of browning of EWAT.
Collapse
Affiliation(s)
- Melise M. Edwards
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, United States
| | - Ha K. Nguyen
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, United States
| | - Andrew D. Dodson
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, United States
| | - Adam J. Herbertson
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, United States
| | - Tomasz A. Wietecha
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States
- UW Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, WA, United States
| | - Tami Wolden-Hanson
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, United States
| | - James L. Graham
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Mackenzie K. Honeycutt
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, United States
| | - Jared D. Slattery
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, United States
| | - Kevin D. O’Brien
- UW Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, WA, United States
- Division of Cardiology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States
| | - Peter J. Havel
- Department of Nutrition, University of California, Davis, Davis, CA, United States
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - James E. Blevins
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, United States
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
11
|
Gómez-Valadés AG, Pozo M, Varela L, Boudjadja MB, Ramírez S, Chivite I, Eyre E, Haddad-Tóvolli R, Obri A, Milà-Guasch M, Altirriba J, Schneeberger M, Imbernón M, Garcia-Rendueles AR, Gama-Perez P, Rojo-Ruiz J, Rácz B, Alonso MT, Gomis R, Zorzano A, D'Agostino G, Alvarez CV, Nogueiras R, Garcia-Roves PM, Horvath TL, Claret M. Mitochondrial cristae-remodeling protein OPA1 in POMC neurons couples Ca 2+ homeostasis with adipose tissue lipolysis. Cell Metab 2021; 33:1820-1835.e9. [PMID: 34343501 PMCID: PMC8432968 DOI: 10.1016/j.cmet.2021.07.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 05/14/2021] [Accepted: 07/09/2021] [Indexed: 01/21/2023]
Abstract
Appropriate cristae remodeling is a determinant of mitochondrial function and bioenergetics and thus represents a crucial process for cellular metabolic adaptations. Here, we show that mitochondrial cristae architecture and expression of the master cristae-remodeling protein OPA1 in proopiomelanocortin (POMC) neurons, which are key metabolic sensors implicated in energy balance control, is affected by fluctuations in nutrient availability. Genetic inactivation of OPA1 in POMC neurons causes dramatic alterations in cristae topology, mitochondrial Ca2+ handling, reduction in alpha-melanocyte stimulating hormone (α-MSH) in target areas, hyperphagia, and attenuated white adipose tissue (WAT) lipolysis resulting in obesity. Pharmacological blockade of mitochondrial Ca2+ influx restores α-MSH and the lipolytic program, while improving the metabolic defects of mutant mice. Chemogenetic manipulation of POMC neurons confirms a role in lipolysis control. Our results unveil a novel axis that connects OPA1 in POMC neurons with mitochondrial cristae, Ca2+ homeostasis, and WAT lipolysis in the regulation of energy balance.
Collapse
Affiliation(s)
- Alicia G Gómez-Valadés
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.
| | - Macarena Pozo
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Luis Varela
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Mehdi Boutagouga Boudjadja
- Faculty of Biology, Medicine and Health, School of Medical Sciences, University of Manchester, M13 9PT Manchester, UK
| | - Sara Ramírez
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Iñigo Chivite
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Elena Eyre
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Roberta Haddad-Tóvolli
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Arnaud Obri
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Maria Milà-Guasch
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Jordi Altirriba
- Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Marc Schneeberger
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Mónica Imbernón
- Department of Physiology, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela, Instituto de Investigación Sanitaria (IDIS), 15782 Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Angela R Garcia-Rendueles
- Neoplasia & Endocrine Differentiation, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela, Instituto de Investigación Sanitaria (IDIS), 15782 Santiago de Compostela, Spain
| | - Pau Gama-Perez
- Departament de Ciències Fisiològiques, Universitat de Barcelona, 08907 Barcelona, Spain; Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), 08908 L'Hospitalet de Llobregat, Spain
| | - Jonathan Rojo-Ruiz
- Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), 47003 Valladolid, Spain
| | - Bence Rácz
- Department of Anatomy and Histology, University of Veterinary Medicine, 1078 Budapest, Hungary
| | - Maria Teresa Alonso
- Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), 47003 Valladolid, Spain
| | - Ramon Gomis
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; Department of Endocrinology and Nutrition, Hospital Clínic, School of Medicine, University of Barcelona, 08036 Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Antonio Zorzano
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain; Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; Institute for Research in Biomedicine (IRB Barcelona), 08028 Barcelona, Spain
| | - Giuseppe D'Agostino
- Faculty of Biology, Medicine and Health, School of Medical Sciences, University of Manchester, M13 9PT Manchester, UK
| | - Clara V Alvarez
- Neoplasia & Endocrine Differentiation, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela, Instituto de Investigación Sanitaria (IDIS), 15782 Santiago de Compostela, Spain
| | - Rubén Nogueiras
- Department of Physiology, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela, Instituto de Investigación Sanitaria (IDIS), 15782 Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Pablo M Garcia-Roves
- Departament de Ciències Fisiològiques, Universitat de Barcelona, 08907 Barcelona, Spain; Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), 08908 L'Hospitalet de Llobregat, Spain
| | - Tamas L Horvath
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Anatomy and Histology, University of Veterinary Medicine, 1078 Budapest, Hungary
| | - Marc Claret
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain; School of Medicine, Universitat de Barcelona, 08036 Barcelona, Spain.
| |
Collapse
|
12
|
Scherer T, Sakamoto K, Buettner C. Brain insulin signalling in metabolic homeostasis and disease. Nat Rev Endocrinol 2021; 17:468-483. [PMID: 34108679 DOI: 10.1038/s41574-021-00498-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/22/2021] [Indexed: 02/06/2023]
Abstract
Insulin signalling in the central nervous system regulates energy homeostasis by controlling metabolism in several organs and by coordinating organ crosstalk. Studies performed in rodents, non-human primates and humans over more than five decades using intracerebroventricular, direct hypothalamic or intranasal application of insulin provide evidence that brain insulin action might reduce food intake and, more importantly, regulates energy homeostasis by orchestrating nutrient partitioning. This Review discusses the metabolic pathways that are under the control of brain insulin action and explains how brain insulin resistance contributes to metabolic disease in obesity, the metabolic syndrome and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Thomas Scherer
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria.
| | - Kenichi Sakamoto
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Christoph Buettner
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA.
| |
Collapse
|
13
|
Wang S, Cao Q, Cui X, Jing J, Li F, Shi H, Xue B, Shi H. Dnmt3b Deficiency in Myf5 +-Brown Fat Precursor Cells Promotes Obesity in Female Mice. Biomolecules 2021; 11:1087. [PMID: 34439754 PMCID: PMC8393658 DOI: 10.3390/biom11081087] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 11/17/2022] Open
Abstract
Increasing energy expenditure through activation of brown fat thermogenesis is a promising therapeutic strategy for the treatment of obesity. Epigenetic regulation has emerged as a key player in regulating brown fat development and thermogenic program. Here, we aimed to study the role of DNA methyltransferase 3b (Dnmt3b), a DNA methyltransferase involved in de novo DNA methylation, in the regulation of brown fat function and energy homeostasis. We generated a genetic model with Dnmt3b deletion in brown fat-skeletal lineage precursor cells (3bKO mice) by crossing Dnmt3b-floxed (fl/fl) mice with Myf5-Cre mice. Female 3bKO mice are prone to diet-induced obesity, which is associated with decreased energy expenditure. Dnmt3b deficiency also impairs cold-induced thermogenic program in brown fat. Surprisingly, further RNA-seq analysis reveals a profound up-regulation of myogenic markers in brown fat of 3bKO mice, suggesting a myocyte-like remodeling in brown fat. Further motif enrichment and pyrosequencing analysis suggests myocyte enhancer factor 2C (Mef2c) as a mediator for the myogenic alteration in Dnmt3b-deficient brown fat, as indicated by decreased methylation at its promoter. Our data demonstrate that brown fat Dnmt3b is a key regulator of brown fat development, energy metabolism and obesity in female mice.
Collapse
Affiliation(s)
- Shirong Wang
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (S.W.); (Q.C.); (X.C.); (J.J.); (F.L.)
| | - Qiang Cao
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (S.W.); (Q.C.); (X.C.); (J.J.); (F.L.)
| | - Xin Cui
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (S.W.); (Q.C.); (X.C.); (J.J.); (F.L.)
| | - Jia Jing
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (S.W.); (Q.C.); (X.C.); (J.J.); (F.L.)
| | - Fenfen Li
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (S.W.); (Q.C.); (X.C.); (J.J.); (F.L.)
| | - Huidong Shi
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| | - Bingzhong Xue
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (S.W.); (Q.C.); (X.C.); (J.J.); (F.L.)
| | - Hang Shi
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (S.W.); (Q.C.); (X.C.); (J.J.); (F.L.)
| |
Collapse
|
14
|
Münzberg H, Floyd E, Chang JS. Sympathetic Innervation of White Adipose Tissue: to Beige or Not to Beige? Physiology (Bethesda) 2021; 36:246-255. [PMID: 34159808 DOI: 10.1152/physiol.00038.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Obesity research progresses in understanding neuronal circuits and adipocyte biology to regulate metabolism. However, the interface of neuro-adipocyte interaction is less studied. We summarize the current knowledge of adipose tissue innervation and interaction with adipocytes and emphasize adipocyte transitions from white to brown adipocytes and vice versa. We further highlight emerging concepts for the differential neuronal regulation of brown/beige versus white adipocyte and the interdependence of both for metabolic regulation.
Collapse
Affiliation(s)
- Heike Münzberg
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Elizabeth Floyd
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Ji Suk Chang
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| |
Collapse
|
15
|
Campos RDMM, Toscano AE, Gouveia HJCB, Lacerda DC, Pereira SDC, Paz IAADSG, Dantas Alves JS, Manhães-de-Castro R. Neonatal fluoxetine exposure delays reflex ontogeny, somatic development, and food intake similarly in male and female rats. Can J Physiol Pharmacol 2021; 99:490-498. [PMID: 32941740 DOI: 10.1139/cjpp-2020-0261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Serotonin (5-HT) acts as a neuromodulator and plays a critical role in brain development. Changes in 5-HT signaling during the perinatal period can affect neural development and may result in behavioral changes in adulthood; however, further investigations are necessary including both sexes to study possible differences. Thus, the aim of this study was to investigate the impact of neonatal treatment with fluoxetine on the development of male and female offspring. The animals were divided into four groups according to sex and treatment. The experimental groups received fluoxetine at 10 mg·kg-1 (1 μL/g of body weight (bw)) and the animals of control group received saline solution 0.9% (1 μL/g of bw) from postnatal days 1-21. In the neonatal period, reflex ontogeny, somatic development, physical features, and food intake were recorded. In the postnatal period (until day 31) bw and post-weaning food intake were recorded. Chronic administration of fluoxetine in the neonatal period caused a delay in the reflex ontogeny and somatic development, as well as reduction of lactation, post-weaning bw, and post-weaning food intake in rats. No difference was found between the sexes. These changes reaffirm that serotonin plays an important role in regulating the plasticity of the brain during the early development period, but without sex differences.
Collapse
Affiliation(s)
| | - Ana Elisa Toscano
- Department of Nursing, CAV, Federal University of Pernambuco, Vitória de Santo Antão, Pernambuco, Brazil
| | | | - Diego Cabral Lacerda
- Post-Graduation Program in Nutrition, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Sabrina da Conceição Pereira
- Post-Graduation in Neuropsychiatry and Behavioral Sciences, Health Sciences Center, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | | | | | - Raul Manhães-de-Castro
- Studies in Nutrition and Phenotypic Plasticity, Department of Nutrition, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| |
Collapse
|
16
|
Edwards MM, Nguyen HK, Herbertson AJ, Dodson AD, Wietecha T, Wolden-Hanson T, Graham JL, O'Brien KD, Havel PJ, Blevins JE. Chronic hindbrain administration of oxytocin elicits weight loss in male diet-induced obese mice. Am J Physiol Regul Integr Comp Physiol 2021; 320:R471-R487. [PMID: 33470901 PMCID: PMC8238148 DOI: 10.1152/ajpregu.00294.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/15/2021] [Accepted: 01/16/2021] [Indexed: 02/08/2023]
Abstract
Previous studies indicate that oxytocin (OT) administration reduces body weight in high-fat diet (HFD)-induced obese (DIO) rodents through both reductions in food intake and increases in energy expenditure. We recently demonstrated that chronic hindbrain [fourth ventricular (4V)] infusions of OT evoke weight loss in DIO rats. Based on these findings, we hypothesized that chronic 4V OT would elicit weight loss in DIO mice. We assessed the effects of 4V infusions of OT (16 nmol/day) or vehicle over 28 days on body weight, food intake, and body composition. OT reduced body weight by approximately 4.5% ± 1.4% in DIO mice relative to OT pretreatment body weight (P < 0.05). These effects were associated with reduced adiposity and adipocyte size [inguinal white adipose tissue (IWAT)] (P < 0.05) and attributed, in part, to reduced energy intake (P < 0.05) at a dose that did not increase kaolin intake (P = NS). OT tended to increase uncoupling protein-1 expression in IWAT (0.05 < P < 0.1) suggesting that OT stimulates browning of WAT. To assess OT-elicited changes in brown adipose tissue (BAT) thermogenesis, we examined the effects of 4V OT on interscapular BAT temperature (TIBAT). 4V OT (1 µg) elevated TIBAT at 0.75 (P = 0.08), 1, and 1.25 h (P < 0.05) postinjection; a higher dose (5 µg) elevated TIBAT at 0.75-, 1-, 1.25-, 1.5-, 1.75- (P < 0.05), and 2-h (0.05 < P < 0.1) postinjection. Together, these findings support the hypothesis that chronic hindbrain OT treatment evokes sustained weight loss in DIO mice by reducing energy intake and increasing BAT thermogenesis at a dose that is not associated with evidence of visceral illness.
Collapse
MESH Headings
- Adipocytes, Brown/drug effects
- Adipocytes, Brown/metabolism
- Adipocytes, Brown/pathology
- Adipocytes, White/drug effects
- Adipocytes, White/metabolism
- Adipocytes, White/pathology
- Adiposity/drug effects
- Animals
- Anti-Obesity Agents/administration & dosage
- Diet, High-Fat
- Disease Models, Animal
- Eating/drug effects
- Energy Intake/drug effects
- Infusions, Intraventricular
- Leptin/blood
- Male
- Mice, Inbred C57BL
- Obesity/drug therapy
- Obesity/metabolism
- Obesity/pathology
- Obesity/physiopathology
- Oxytocin/administration & dosage
- Rhombencephalon/drug effects
- Rhombencephalon/physiopathology
- Thermogenesis/drug effects
- Uncoupling Protein 1/metabolism
- Weight Loss/drug effects
- Mice
Collapse
Affiliation(s)
- Melise M Edwards
- Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Veteran Affairs Puget Sound Health Care System, Seattle, Washington
| | - Ha K Nguyen
- Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Veteran Affairs Puget Sound Health Care System, Seattle, Washington
| | - Adam J Herbertson
- Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Veteran Affairs Puget Sound Health Care System, Seattle, Washington
| | - Andrew D Dodson
- Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Veteran Affairs Puget Sound Health Care System, Seattle, Washington
| | - Tomasz Wietecha
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, Washington
- UW Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, Washington
| | - Tami Wolden-Hanson
- Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Veteran Affairs Puget Sound Health Care System, Seattle, Washington
| | - James L Graham
- Department of Nutrition, University of California, Davis, California
| | - Kevin D O'Brien
- UW Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, Washington
- Division of Cardiology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Peter J Havel
- Department of Nutrition, University of California, Davis, California
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, California
| | - James E Blevins
- Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Veteran Affairs Puget Sound Health Care System, Seattle, Washington
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| |
Collapse
|
17
|
McMillan TR, Forster MAM, Short LI, Rudecki AP, Cline DL, Gray SL. Melanotan II, a melanocortin agonist, partially rescues the impaired thermogenic capacity of pituitary adenylate cyclase-activating polypeptide deficient mice. Exp Physiol 2020; 106:427-437. [PMID: 33332767 DOI: 10.1113/ep088838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 11/27/2020] [Indexed: 12/16/2022]
Abstract
NEW FINDINGS What is the central question of this study? Can chronic treatment of pituitary adenylate cyclase-activating polypeptide (PACAP) deficient mice with the melanocortin agonist melanotan II during cold acclimation rescue the impaired thermogenic capacity previously observed in PACAP deficient mice? What is the main finding and its importance? Using a genetic model of PACAP deficiency, this study provides evidence that PACAP acts upstream of the melanocortin system in regulating sympathetic nerve activity to brown adipose tissue in mice. ABSTRACT Impaired adipose tissue function in obesity, including reduced thermogenic potential, has detrimental consequences for metabolic health. Hormonal regulation of adaptive thermogenesis is being explored as a potential therapeutic target for human obesity. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide expressed in nuclei of the hypothalamus known to regulate energy expenditure, and functional studies reveal a role for PACAP in the central regulation of thermogenesis, although mechanisms are not well understood. We hypothesized that PACAP acts upstream of the melanocortin system to regulate sympathetic nerve activity to stimulate thermogenesis. To assess this, female PACAP-/- and PACAP+/+ mice were given daily peripheral injections of a melanocortin receptor agonist, melanotan II (MTII), for 3 weeks during cold acclimation, and the effect of MTII on thermogenic capacity and adipose tissue remodelling was examined by physiological and histological analyses. MTII partially rescued the impaired thermogenic capacity in PACAP-/- mice as compared to PACAP+/+ mice as determined by measuring noradrenaline-induced metabolic rate. In addition, MTII treatment during cold acclimation corrected the previously identified deficit in lipid utilization in response to adrenergic stimulation in PACAP-/- null mice, suggesting impaired lipid mobilization may contribute to the impaired thermogenic capacity of PACAP-/- mice. Results presented here provide physiological evidence to suggest that PACAP acts upstream of melanocortin receptors to facilitate sympathetically induced mechanisms of adaptive thermogenesis in response to cold acclimation.
Collapse
Affiliation(s)
- Thecla Rae McMillan
- Northern Medical Program, University of Northern British Columbia, Prince George, British Columbia, Canada
| | - Maeghan A M Forster
- Northern Medical Program, University of Northern British Columbia, Prince George, British Columbia, Canada
| | - Landon I Short
- Northern Medical Program, University of Northern British Columbia, Prince George, British Columbia, Canada
| | - Alexander P Rudecki
- Northern Medical Program, University of Northern British Columbia, Prince George, British Columbia, Canada
| | - Daemon L Cline
- Northern Medical Program, University of Northern British Columbia, Prince George, British Columbia, Canada
| | - Sarah L Gray
- Northern Medical Program, University of Northern British Columbia, Prince George, British Columbia, Canada
| |
Collapse
|
18
|
Holland J, Sorrell J, Yates E, Smith K, Arbabi S, Arnold M, Rivir M, Morano R, Chen J, Zhang X, Dimarchi R, Woods SC, Sanchez-Gurmaches J, Wohleb E, Perez-Tilve D. A Brain-Melanocortin-Vagus Axis Mediates Adipose Tissue Expansion Independently of Energy Intake. Cell Rep 2020; 27:2399-2410.e6. [PMID: 31116984 PMCID: PMC6550338 DOI: 10.1016/j.celrep.2019.04.089] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 03/18/2019] [Accepted: 04/18/2019] [Indexed: 02/07/2023] Open
Abstract
The melanocortin system is a brain circuit that influences energy balance by regulating energy intake and expenditure. In addition, the brain-melanocortin system controls adipose tissue metabolism to optimize fuel mobilization and storage. Specifically, increased brain-melanocortin signaling or negative energy balance promotes lipid mobilization by increasing sympathetic nervous system input to adipose tissue. In contrast, calorie-independent mechanisms favoring energy storage are less understood. Here, we demonstrate that reduction of brain-melanocortin signaling actively promotes fat mass gain by activating the lipogenic program and adipocyte and endothelial cell proliferation in white fat depots independently of caloric intake via efferent nerve fibers conveyed by the common hepatic branch of the vagus nerve. Those vagally regulated obesogenic signals also contribute to the fat mass gain following chronic high-fat diet feeding. These data reveal a physiological mechanism whereby the brain controls energy stores that may contribute to increased susceptibility to obesity. Brain-melanocortin signaling controls fat mass indirectly by regulating energy balance and by direct control of lipid mobilization from adipose tissue via sympathetic nervous system activity. Holland et al. show that reduced brain-melanocortin signaling promotes white adipose tissue expansion via signals conveyed by efferent innervation of the vagus nerve.
Collapse
Affiliation(s)
- Jenna Holland
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Joyce Sorrell
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Emily Yates
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kathleen Smith
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Shahriar Arbabi
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Myrtha Arnold
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | - Marita Rivir
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Rachel Morano
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jenny Chen
- Genomics, Epigenomics and Sequencing Core, Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Xiang Zhang
- Genomics, Epigenomics and Sequencing Core, Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Richard Dimarchi
- Novo Nordisk Research Center Indianapolis, IN, USA; Department of Chemistry, Indiana University, Bloomington, IN, USA
| | - Stephen C Woods
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Joan Sanchez-Gurmaches
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Division of Endocrinology and Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH, USA
| | - Eric Wohleb
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Diego Perez-Tilve
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
19
|
Manceau R, Majeur D, Alquier T. Neuronal control of peripheral nutrient partitioning. Diabetologia 2020; 63:673-682. [PMID: 32030470 DOI: 10.1007/s00125-020-05104-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 12/20/2019] [Indexed: 12/16/2022]
Abstract
The appropriate utilisation, storage and conversion of nutrients in peripheral tissues, referred to as nutrient partitioning, is a fundamental process to adapt to nutritional and metabolic challenges and is thus critical for the maintenance of a healthy energy balance. Alterations in this process during nutrient excess can have deleterious effects on glucose and lipid homeostasis and contribute to the development of obesity and type 2 diabetes. Nutrient partitioning is a complex integrated process under the control of hormonal and neural signals. Neural control relies on the capacity of the brain to sense circulating metabolic signals and mount adaptive neuroendocrine and autonomic responses. This review aims to discuss the hypothalamic neurocircuits and molecular mechanisms controlling nutrient partitioning and their potential contribution to metabolic maladaptation and disease.
Collapse
Affiliation(s)
- Romane Manceau
- Montréal Diabetes Research Center and Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 rue Saint-Denis, Montréal, QC, H2X 0A9, Canada
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Danie Majeur
- Montréal Diabetes Research Center and Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 rue Saint-Denis, Montréal, QC, H2X 0A9, Canada
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Thierry Alquier
- Montréal Diabetes Research Center and Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 rue Saint-Denis, Montréal, QC, H2X 0A9, Canada.
- Department of Medicine, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
20
|
Zhu Q, Weng J, Shen M, Fish J, Shen Z, Coschigano KT, Davidson WS, Tso P, Shi H, Lo CC. Apolipoprotein A-IV Enhances Fatty Acid Uptake by Adipose Tissues of Male Mice via Sympathetic Activation. Endocrinology 2020; 161:5802681. [PMID: 32157301 PMCID: PMC7100924 DOI: 10.1210/endocr/bqaa042] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 03/04/2020] [Indexed: 12/31/2022]
Abstract
Apolipoprotein A-IV (ApoA-IV) synthesized by the gut regulates lipid metabolism. Sympathetic innervation of adipose tissues also controls lipid metabolism. We hypothesized that ApoA-IV required sympathetic innervation to increase fatty acid (FA) uptake by adipose tissues and brown adipose tissue (BAT) thermogenesis. After 3 weeks feeding of either a standard chow diet or a high-fat diet (HFD), mice with unilateral denervation of adipose tissues received intraperitoneal administration of recombinant ApoA-IV protein and intravenous infusion of lipid mixture with radioactive triolein. In chow-fed mice, ApoA-IV administration increased FA uptake by intact BAT but not the contralateral denervated BAT or intact white adipose tissue (WAT). Immunoblots showed that, in chow-fed mice, ApoA-IV increased expression of lipoprotein lipase and tyrosine hydroxylase in both intact BAT and inguinal WAT (IWAT), while ApoA-IV enhanced protein levels of β3 adrenergic receptor, adipose triglyceride lipase, and uncoupling protein 1 in the intact BAT only. In HFD-fed mice, ApoA-IV elevated FA uptake by intact epididymal WAT (EWAT) but not intact BAT or IWAT. ApoA-IV increased sympathetic activity assessed by norepinephrine turnover (NETO) rate in BAT and EWAT of chow-fed mice, whereas it elevated NETO only in EWAT of HFD-fed mice. These observations suggest that, in chow-fed mice, ApoA-IV activates sympathetic activity of BAT and increases FA uptake by BAT via innervation, while in HFD-fed mice, ApoA-IV stimulates sympathetic activity of EWAT to shunt FAs into the EWAT.
Collapse
Affiliation(s)
- Qi Zhu
- Department of Biology, Miami University, Oxford, OH
| | - Jonathan Weng
- Department of Biomedical Sciences and Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine, Athens, OH
| | - Minqian Shen
- Department of Biology, Miami University, Oxford, OH
| | - Jace Fish
- Department of Biomedical Sciences and Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine, Athens, OH
| | - Zhujun Shen
- Department of Biomedical Sciences and Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine, Athens, OH
| | - Karen T Coschigano
- Department of Biomedical Sciences and Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine, Athens, OH
| | - W Sean Davidson
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH
| | - Patrick Tso
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH
| | - Haifei Shi
- Department of Biology, Miami University, Oxford, OH
| | - Chunmin C Lo
- Department of Biomedical Sciences and Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine, Athens, OH
- Correspondence: Chunmin C Lo, Department of Biomedical Sciences, Irvine Hall 228, 1 Ohio University, Athens, OH 45701-2979. E-mail:
| |
Collapse
|
21
|
Lee NJ, Qi Y, Enriquez RF, Clarke I, Ip CK, Wee N, Baldock PA, Herzog H. Energy partitioning between fat and bone mass is controlled via a hypothalamic leptin/NPY relay. Int J Obes (Lond) 2020; 44:2149-2164. [PMID: 32152498 DOI: 10.1038/s41366-020-0550-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 01/28/2020] [Accepted: 02/06/2020] [Indexed: 01/21/2023]
Abstract
BACKGROUND/OBJECTIVES Maintaining energy balance is important to ensure a healthy organism. However, energy partitioning, coordinating the distribution of sufficient energy to different organs and tissues is equally important, but the control of this process is largely unknown. In obesity, an increase in fat mass necessitates the production of additional bone mass to cope with the increase in bodyweight and processes need to be in place to communicate this new weight bearing demand. Here, we investigate the interaction between leptin and NPY, two factors critically involved in the regulation of both energy metabolism and bone mass, in this process. METHODS We assessed the co-localization of leptin receptors on NPY neurons using RNAScope followed by a systematic examination of body composition and energy metabolism profiling in male and female mice lacking leptin receptors specifically in NPY neurons (Leprlox/lox;NPYCre/+). The effect of short-term switching between chow and high-fat diet was also examined in these mice. RESULTS We uncovered that leptin receptor expression is greater on a subpopulation of NPY neurons in the arcuate that do not express AgRP. We further show that Leprlox/lox;NPYCre/+ mice exhibit significantly increased adiposity while bone mass is diminished. These body composition changes occur in the absence of alterations in food intake or energy expenditure, demonstrating a prominent role for leptin signaling in NPY neurons in the control of energy partitioning. Importantly however, when fed a high-fat diet, these mice display a switch in energy partitioning whereby they exhibit a significantly enhanced ability to increase their bone mass to match the increased bodyweight caused by higher caloric intake concurrent with attenuated adiposity. CONCLUSIONS Taken together, these results demonstrate that leptin signaling in NPY neurons is critical for coordinating energy partitioning between fat and bone mass especially during situations of changes in energy balance.
Collapse
Affiliation(s)
- Nicola J Lee
- Neuroscience Division, Garvan Institute of Medical Research, St Vincent's Hospital Sydney, Sydney, NSW, 2010, Australia.,St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia
| | - Yue Qi
- Neuroscience Division, Garvan Institute of Medical Research, St Vincent's Hospital Sydney, Sydney, NSW, 2010, Australia
| | - Ronaldo F Enriquez
- Neuroscience Division, Garvan Institute of Medical Research, St Vincent's Hospital Sydney, Sydney, NSW, 2010, Australia
| | - Ireni Clarke
- Neuroscience Division, Garvan Institute of Medical Research, St Vincent's Hospital Sydney, Sydney, NSW, 2010, Australia
| | - Chi Kin Ip
- Neuroscience Division, Garvan Institute of Medical Research, St Vincent's Hospital Sydney, Sydney, NSW, 2010, Australia.,St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia
| | - Natalie Wee
- Bone Division, Garvan Institute of Medical Research, St Vincent's Hospital Sydney, Sydney, NSW, 2010, Australia
| | - Paul A Baldock
- St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia.,Bone Division, Garvan Institute of Medical Research, St Vincent's Hospital Sydney, Sydney, NSW, 2010, Australia
| | - Herbert Herzog
- Neuroscience Division, Garvan Institute of Medical Research, St Vincent's Hospital Sydney, Sydney, NSW, 2010, Australia. .,St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia.
| |
Collapse
|
22
|
Yu H, Chhabra KH, Thompson Z, Jones GL, Kiran S, Shangguan G, Low MJ. Hypothalamic POMC deficiency increases circulating adiponectin despite obesity. Mol Metab 2020; 35:100957. [PMID: 32244188 PMCID: PMC7082555 DOI: 10.1016/j.molmet.2020.01.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/22/2020] [Accepted: 01/31/2020] [Indexed: 02/06/2023] Open
Abstract
Objective The steep rise in the prevalence of obesity and its related metabolic syndrome have become a major worldwide health concerns. Melanocortin peptides from hypothalamic arcuate nucleus (Arc) POMC neurons induce satiety to limit food intake. Consequently, Arc Pomc-deficient mice (ArcPomc−/−) exhibit hyperphagia and obesity. Previous studies demonstrated that the circulating levels of adiponectin, a protein abundantly produced and secreted by fat cells, negatively correlate with obesity in both rodents and humans. However, we found that ArcPomc−/− mice have increased circulating adiponectin levels despite obesity. Therefore, we investigated the physiological function and underlying mechanisms of hypothalamic POMC in regulating systemic adiponectin levels. Methods Circulating adiponectin was measured in obese ArcPomc−/− mice at ages 4–52 weeks. To determine whether increased adiponectin was a direct result of ArcPomc deficiency or a secondary effect of obesity, we examined plasma adiponectin levels in calorie-restricted mice with or without a history of obesity and in ArcPomc−/− mice before and after genetic restoration of Pomc expression in the hypothalamus. To delineate the mechanisms causing increased adiponectin in ArcPomc−/− mice, we determined sympathetic outflow to adipose tissue by assessing epinephrine, norepinephrine, and tyrosine hydroxylase protein levels and measured the circulating adiponectin in the mice after acute norepinephrine or propranolol treatments. In addition, adiponectin mRNA and protein levels were measured in discrete adipose tissue depots to ascertain which fat depots contributed the most to the high level of adiponectin in the ArcPomc−/− mice. Finally, we generated compound Adiopoq−/−:ArcPomc−/− mice and compared their growth, body composition, and glucose homeostasis to the individual knockout mouse strains and their wild-type controls. Results Obese ArcPomc−/− female mice had unexpectedly increased plasma adiponectin compared to wild-type siblings at all ages greater than 8 weeks. Despite chronic calorie restriction to achieve normal body weights, higher adiponectin levels persisted in the ArcPomc−/− female mice. Genetic restoration of Pomc expression in the Arc or acute treatment of the ArcPomc−/− female mice with melanotan II reduced adiponectin levels to control littermate values. The ArcPomc−/− mice had defective thermogenesis and decreased epinephrine, norepinephrine, and tyrosine hydroxylase protein levels in their fat pads, indicating reduced sympathetic outflow to adipose tissue. Injections of norepinephrine into the ArcPomc−/− female mice reduced circulating adiponectin levels, whereas injections of propranolol significantly increased adiponectin levels. Despite the beneficial effects of adiponectin on metabolism, the deletion of adiponectin alleles in the ArcPomc−/− mice did not exacerbate their metabolic abnormalities. Conclusion In summary, to the best of our knowledge, this study provides the first evidence that despite obesity, the ArcPomc−/− mouse model has high circulating adiponectin levels, which demonstrated that increased fat mass is not necessarily correlated with hypoadiponectinemia. Our investigation also found a previously unknown physiological pathway connecting POMC neurons via the sympathetic nervous system to circulating adiponectin, thereby shedding light on the biological regulation of adiponectin. Obese female hypothalamic-specific Pomc-deficient mice have unexpectedly elevated circulating adiponectin. Restoration of Pomc expression in the hypothalamus reduces plasma adiponectin. Low sympathetic output to subcutaneous fat depots in the Pomc-deficient mice contributes to high adiponectin levels. Deletion of adiponectin in hypothalamic-specific Pomc-deficient mice does not alter their metabolic phenotype.
Collapse
Affiliation(s)
- Hui Yu
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA.
| | - Kavaljit H Chhabra
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA; Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Zoe Thompson
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Graham L Jones
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Sylee Kiran
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA; School of Literature, Science, and Arts, University of Michigan, Ann Arbor, MI, USA
| | - Gary Shangguan
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Malcolm J Low
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA; Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
23
|
Zhu Q, Liu X, Glazier BJ, Krolick KN, Yang S, He J, Lo CC, Shi H. Differential Sympathetic Activation of Adipose Tissues by Brain-Derived Neurotrophic Factor. Biomolecules 2019; 9:biom9090452. [PMID: 31492038 PMCID: PMC6769916 DOI: 10.3390/biom9090452] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/17/2019] [Accepted: 09/02/2019] [Indexed: 12/12/2022] Open
Abstract
Centrally administered brain-derived neurotrophic factor (BDNF) decreases body adiposity beyond what can be accounted for by decreased food intake, implying enhanced lipid metabolism by BDNF. Consistent with this notion, intracerebroventricular (icv) injection of BDNF in rats increased the expression of lipolytic enzymes in white adipose tissues (WAT) and increased circulating concentrations of lipolytic products without changing the levels of adrenal gland hormones. This suggests that central BDNF-induced lipid mobilization is likely due to sympathetic neural activation, rather than activation of the adrenocortical or adrenomedullary system. We hypothesized that BDNF activated sympathetic innervation of adipose tissues to regulate lipolysis. Rats with unilateral denervation of interscapular brown adipose tissue (BAT) and different WAT depots received icv injections of saline or BDNF. Both intact and denervated adipose tissues were exposed to the same circulating factors, but denervated adipose tissues did not receive neural signals. Norepinephrine (NE) turnover (NETO) of BAT and WAT was assessed as a measure of sympathetic activity. Findings revealed that central BDNF treatment induced a change in NETO in some but not all the adipose tissues tested. Specifically, greater NETO rates were found in BAT and gonadal epididymal WAT (EWAT), but not in inguinal WAT (IWAT) or retroperitoneal WAT (RWAT), of BDNF-treated rats compared to saline-treated rats. Furthermore, intact innervation was necessary for BDNF-induced NETO in BAT and EWAT. In addition, BDNF increased the expression of lipolytic enzymes in both intact and denervated EWAT and IWAT, suggesting that BDNF-induced WAT lipolysis was independent of intact innervation. To summarize, centrally administered BDNF selectively provoked sympathetic drives to BAT and EWAT that was dependent on intact innervation, while BDNF also increased lipolysis in a manner independent of intact innervation.
Collapse
Affiliation(s)
- Qi Zhu
- Department of Biology, Miami University, Oxford, OH 45056, USA
| | - Xian Liu
- Department of Biology, Miami University, Oxford, OH 45056, USA
| | | | | | - Shangyuwen Yang
- Department of Biology, Miami University, Oxford, OH 45056, USA
| | - Jingyan He
- Department of Biology, Miami University, Oxford, OH 45056, USA
| | - Chunmin C Lo
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Diabetes Institute, Ohio University, Athens, OH 45701, USA.
| | - Haifei Shi
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| |
Collapse
|
24
|
Bernasochi GB, Bell JR, Simpson ER, Delbridge LM, Boon WC. Impact of Estrogens on the Regulation of White, Beige, and Brown Adipose Tissue Depots. Compr Physiol 2019; 9:457-475. [DOI: 10.1002/cphy.c180009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
25
|
Wen S, Wang C, Gong M, Zhou L. An overview of energy and metabolic regulation. SCIENCE CHINA-LIFE SCIENCES 2018; 62:771-790. [PMID: 30367342 DOI: 10.1007/s11427-018-9371-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 08/23/2018] [Indexed: 12/21/2022]
Abstract
The physiology and behaviors related to energy balance are monitored by the nervous and humoral systems. Because of the difficulty in treating diabetes and obesity, elucidating the energy balance mechanism and identifying critical targets for treatment are important research goals. Therefore, the purpose of this article is to describe energy regulation by the central nervous system (CNS) and peripheral humoral pathway. Homeostasis and rewarding are the basis of CNS regulation. Anorexigenic or orexigenic effects reflect the activities of the POMC/CART or NPY/AgRP neurons within the hypothalamus. Neurotransmitters have roles in food intake, and responsive brain nuclei have different functions related to food intake, glucose monitoring, reward processing. Peripheral gut- or adipose-derived hormones are the major source of peripheral humoral regulation systems. Nutrients or metabolites and gut microbiota affect metabolism via a discrete pathway. We also review the role of peripheral organs, the liver, adipose tissue, and skeletal muscle in peripheral regulation. We discuss these topics and how the body regulates metabolism.
Collapse
Affiliation(s)
- Song Wen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China
| | - Chaoxun Wang
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China
| | - Min Gong
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China
| | - Ligang Zhou
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China.
| |
Collapse
|
26
|
Carnagarin R, Matthews VB, Herat LY, Ho JK, Schlaich MP. Autonomic Regulation of Glucose Homeostasis: a Specific Role for Sympathetic Nervous System Activation. Curr Diab Rep 2018; 18:107. [PMID: 30232652 DOI: 10.1007/s11892-018-1069-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW Cardiometabolic disorders such as obesity, metabolic syndrome and diabetes are increasingly common and associated with adverse cardiovascular outcomes. The mechanisms driving these developments are incompletely understood but likely to include autonomic dysregulation. The latest evidence for such a role is briefly reviewed here. RECENT FINDINGS Recent findings highlight the relevance of autonomic regulation in glucose metabolism and identify sympathetic activation, in concert with parasympathetic withdrawal, as a major contributor to the development of metabolic disorders and an important mediator of the associated adverse cardiovascular consequences. Methods targeting sympathetic overactivity using pharmacological and device-based approaches are available and appear as logical additional approaches to curb the burden of metabolic disorders and alleviate the associated morbidity from cardiovascular causes. While the available data are encouraging, the role of therapeutic inhibition of sympathetic overdrive in the prevention of the metabolic disorders and the associated adverse outcomes requires adequate testing in properly sized randomised controlled trials.
Collapse
Affiliation(s)
- Revathy Carnagarin
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit / Medical Research Foundation, University of Western Australia, Level 3, MRF Building, Rear 50 Murray St, Perth, WA, 6000, Australia
| | - Vance B Matthews
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit / Medical Research Foundation, University of Western Australia, Level 3, MRF Building, Rear 50 Murray St, Perth, WA, 6000, Australia
| | - Lakshini Y Herat
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit / Medical Research Foundation, University of Western Australia, Level 3, MRF Building, Rear 50 Murray St, Perth, WA, 6000, Australia
| | - Jan K Ho
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit / Medical Research Foundation, University of Western Australia, Level 3, MRF Building, Rear 50 Murray St, Perth, WA, 6000, Australia
| | - Markus P Schlaich
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit / Medical Research Foundation, University of Western Australia, Level 3, MRF Building, Rear 50 Murray St, Perth, WA, 6000, Australia.
- Departments of Cardiology and Nephrology, Royal Perth Hospital, Perth, Australia.
- Neurovascular Hypertension & Kidney Disease Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia.
| |
Collapse
|
27
|
da L.D. Barros M, Manhães-de-Castro R, Alves DT, Quevedo OG, Toscano AE, Bonnin A, Galindo L. Long term effects of neonatal exposure to fluoxetine on energy balance: A systematic review of experimental studies. Eur J Pharmacol 2018; 833:298-306. [DOI: 10.1016/j.ejphar.2018.06.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 06/01/2018] [Accepted: 06/07/2018] [Indexed: 02/01/2023]
|
28
|
Pozo M, Claret M. Hypothalamic Control of Systemic Glucose Homeostasis: The Pancreas Connection. Trends Endocrinol Metab 2018; 29:581-594. [PMID: 29866501 DOI: 10.1016/j.tem.2018.05.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 05/10/2018] [Accepted: 05/12/2018] [Indexed: 12/22/2022]
Abstract
Maintenance of glucose homeostasis is mandatory for organismal survival. It is accomplished by complex and coordinated interplay between glucose detection mechanisms and multiple effector systems. The brain, in particular homeostatic regions such as the hypothalamus, plays a crucial role in orchestrating such a highly integral response. We review here current understanding of how the hypothalamus senses glucose availability and participates in systemic glucose homeostasis. We provide an update of the relevant signaling pathways and neuronal subsets involved, as well as of the mechanisms modulating metabolic processes in peripheral tissues such as liver, skeletal muscle, fat, and especially the pancreas. We also discuss the relevance of these networks in human biology and prevalent metabolic conditions such as diabetes and obesity.
Collapse
Affiliation(s)
- Macarena Pozo
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Marc Claret
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08036 Barcelona, Spain.
| |
Collapse
|
29
|
Güemes A, Georgiou P. Review of the role of the nervous system in glucose homoeostasis and future perspectives towards the management of diabetes. Bioelectron Med 2018; 4:9. [PMID: 32232085 PMCID: PMC7098234 DOI: 10.1186/s42234-018-0009-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/10/2018] [Indexed: 12/16/2022] Open
Abstract
Diabetes is a disease caused by a breakdown in the glucose metabolic process resulting in abnormal blood glucose fluctuations. Traditionally, control has involved external insulin injection in response to elevated blood glucose to substitute the role of the beta cells in the pancreas which would otherwise perform this function in a healthy individual. The central nervous system (CNS), however, also plays a vital role in glucose homoeostasis through the control of pancreatic secretion and insulin sensitivity which could potentially be used as a pathway for enhancing glucose control. In this review, we present an overview of the brain regions, peripheral nerves and molecular mechanisms by which the CNS regulates glucose metabolism and the potential benefits of modulating them for diabetes management. Development of technologies to interface to the nervous system will soon become a reality through bioelectronic medicine and we present the emerging opportunities for the treatment of type 1 and type 2 diabetes.
Collapse
Affiliation(s)
- Amparo Güemes
- Centre for Bio-Inspired Technology, Department of Electrical and Electronic Engineering, Imperial College London, South Kensington Campus, London, UK
| | - Pantelis Georgiou
- Centre for Bio-Inspired Technology, Department of Electrical and Electronic Engineering, Imperial College London, South Kensington Campus, London, UK
| |
Collapse
|
30
|
Reichenbach A, Stark R, Mequinion M, Lockie SH, Lemus MB, Mynatt RL, Luquet S, Andrews ZB. Carnitine acetyltransferase (Crat) in hunger-sensing AgRP neurons permits adaptation to calorie restriction. FASEB J 2018; 32:fj201800634R. [PMID: 29932868 PMCID: PMC6219829 DOI: 10.1096/fj.201800634r] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 06/12/2018] [Indexed: 12/13/2022]
Abstract
Hunger-sensing agouti-related peptide (AgRP) neurons ensure survival by adapting metabolism and behavior to low caloric environments. This adaption is accomplished by consolidating food intake, suppressing energy expenditure, and maximizing fat storage (nutrient partitioning) for energy preservation. The intracellular mechanisms responsible are unknown. Here we report that AgRP carnitine acetyltransferase (Crat) knockout (KO) mice exhibited increased fatty acid utilization and greater fat loss after 9 d of calorie restriction (CR). No differences were seen in mice with ad libitum food intake. Eleven days ad libitum feeding after CR resulted in greater food intake, rebound weight gain, and adiposity in AgRP Crat KO mice compared with wild-type controls, as KO mice act to restore pre-CR fat mass. Collectively, this study highlights the importance of Crat in AgRP neurons to regulate nutrient partitioning and fat mass during chronically reduced caloric intake. The increased food intake, body weight gain, and adiposity in KO mice after CR also highlights the detrimental and persistent metabolic consequence of impaired substrate utilization associated with CR. This finding may have significant implications for postdieting weight management in patients with metabolic diseases.-Reichenbach, A., Stark, R., Mequinion, M., Lockie, S. H., Lemus, M. B., Mynatt, R. L., Luquet, S., Andrews, Z. B. Carnitine acetyltransferase (Crat) in hunger-sensing AgRP neurons permits adaptation to calorie restriction.
Collapse
Affiliation(s)
- Alex Reichenbach
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Romana Stark
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Mathieu Mequinion
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Sarah H. Lockie
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Moyra B. Lemus
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Randall L. Mynatt
- Gene Nutrient Interactions Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
- Transgenic Core Facility, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA; and
| | - Serge Luquet
- Université of Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionelle et Adaptative, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche 8251, Paris, France
| | - Zane B. Andrews
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Physiology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
31
|
Harris RBS. Denervation as a tool for testing sympathetic control of white adipose tissue. Physiol Behav 2018; 190:3-10. [PMID: 28694155 PMCID: PMC5758439 DOI: 10.1016/j.physbeh.2017.07.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 07/05/2017] [Accepted: 07/06/2017] [Indexed: 10/19/2022]
Abstract
This review summarizes the evidence derived from studies utilizing denervation procedures to demonstrate sympathetic control of white adipose tissue metabolism and body fat mass. A majority of the work demonstrating neural control of white fat was performed in the Bartness laboratory with Siberian hamsters as the predominant experimental model. These animals experience dramatic changes in body fat mass in response to changes in photoperiod, however, the mechanisms identified in hamsters have been reproduced or further elucidated by experiments with other animal models. Evidence for the role of sympathetic innervation contributing to the control of white adipocyte lipolysis and preadipocyte proliferation is summarized. In addition, evidence from denervation experiments for neural communication between different white fat depots as well as for a feedback control loop between sensory afferents from individual fat depots and sympathetic efferents to the same or distant white fat depots is discussed.
Collapse
Affiliation(s)
- Ruth B S Harris
- Medical College of Georgia, Augusta University, Augusta, GA 30912, United States.
| |
Collapse
|
32
|
Piskunova YV, Kazantceva AY, Baklanov AV, Bazhan NM. Mutation yellow in agouti loci prevents age-related increase of skeletal muscle genes regulating free fatty acids oxidation. Vavilovskii Zhurnal Genet Selektsii 2018. [DOI: 10.18699/vj18.358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The lethal yellow mutation in agouti loci (Ay mutation) reduces the activity of melanocortin (MC) receptors and causes hyperphagia, obesity and type two diabetes mellitus in aging mice (Ay mice). It is unknown if changes in distinct elements of the metabolic system such as white adipose tissue (WAT) and brown adipose tissue (BAT), and skeletal muscle will manifest before the development of obesity. The aim of this work was to measure the relative gene expression of key proteins that regulate carbohydrate-lipid metabolism in WAT, BAT and skeletal muscle in Ay mice before the development of obesity. C57Bl/6J mice bearing a dominant autosomal mutation Ay (Ay /a mice) and mice of the standard genotype (a/a mice, control) have been studied in three age groups: 10, 15 and 30 weeks. The relative mRNA level of genes was measured by real-time PCR in skeletal muscles (uncoupling protein 3 (Ucp3) and carnitine palmitoyl transferase 1b (Cpt1b) (free fatty acids oxidation), solute carrier family 2 (facilitated glucose transporter), member 4 (Slc2a4) (glucose uptake)), in WAT lipoprotein lipase (Lpl) (triglyceride deposition), hormone-sensitive lipase (Lipe) (lipid mobilization), and Slc2a4 (glucose uptake)), and in BAT: uncoupling protein 1 (Ucp1) (energy expenditure). The expression of Cpt1b was reduced in young Ay mice (10 weeks), there was no transient peak of transcription of Cpt1b, Ucp3 in skeletal muscle tissue and Lipe, Slc2a4 in WAT in early adult Ay mice (15 weeks), which was noted in а/а mice. Reduction of the transcriptional activity of the studied genes in skeletal muscle and white adipose tissue can initiate the development of melanocortin obesity in Ay mice.
Collapse
Affiliation(s)
| | | | | | - N. M. Bazhan
- Novosibirsk State University; Institute of Cytology and Genetics SB RAS
| |
Collapse
|
33
|
Caron A, Dungan Lemko HM, Castorena CM, Fujikawa T, Lee S, Lord CC, Ahmed N, Lee CE, Holland WL, Liu C, Elmquist JK. POMC neurons expressing leptin receptors coordinate metabolic responses to fasting via suppression of leptin levels. eLife 2018. [PMID: 29528284 PMCID: PMC5866097 DOI: 10.7554/elife.33710] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Leptin is critical for energy balance, glucose homeostasis, and for metabolic and neuroendocrine adaptations to starvation. A prevalent model predicts that leptin’s actions are mediated through pro-opiomelanocortin (POMC) neurons that express leptin receptors (LEPRs). However, previous studies have used prenatal genetic manipulations, which may be subject to developmental compensation. Here, we tested the direct contribution of POMC neurons expressing LEPRs in regulating energy balance, glucose homeostasis and leptin secretion during fasting using a spatiotemporally controlled Lepr expression mouse model. We report a dissociation between leptin’s effects on glucose homeostasis versus energy balance in POMC neurons. We show that these neurons are dispensable for regulating food intake, but are required for coordinating hepatic glucose production and for the fasting-induced fall in leptin levels, independent of changes in fat mass. We also identify a role for sympathetic nervous system regulation of the inhibitory adrenergic receptor (ADRA2A) in regulating leptin production. Collectively, our findings highlight a previously unrecognized role of POMC neurons in regulating leptin levels.
Collapse
Affiliation(s)
- Alexandre Caron
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | | | - Carlos M Castorena
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Teppei Fujikawa
- Department of Cellular and Integrative Physiology, UT Health San Antonio, San Antonio, United States
| | - Syann Lee
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Caleb C Lord
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Newaz Ahmed
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Charlotte E Lee
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - William L Holland
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Chen Liu
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, United States
| | - Joel K Elmquist
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
34
|
Braun K, Oeckl J, Westermeier J, Li Y, Klingenspor M. Non-adrenergic control of lipolysis and thermogenesis in adipose tissues. ACTA ACUST UNITED AC 2018. [PMID: 29514884 DOI: 10.1242/jeb.165381] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The enormous plasticity of adipose tissues, to rapidly adapt to altered physiological states of energy demand, is under neuronal and endocrine control. In energy balance, lipolysis of triacylglycerols and re-esterification of free fatty acids are opposing processes operating in parallel at identical rates, thus allowing a more dynamic transition from anabolism to catabolism, and vice versa. In response to alterations in the state of energy balance, one of the two processes predominates, enabling the efficient mobilization or storage of energy in a negative or positive energy balance, respectively. The release of noradrenaline from the sympathetic nervous system activates lipolysis in a depot-specific manner by initiating the canonical adrenergic receptor-Gs-protein-adenylyl cyclase-cyclic adenosine monophosphate-protein kinase A pathway, targeting proteins of the lipolytic machinery associated with the interface of the lipid droplets. In brown and brite adipocytes, lipolysis stimulated by this signaling pathway is a prerequisite for the activation of non-shivering thermogenesis. Free fatty acids released by lipolysis are direct activators of uncoupling protein 1-mediated leak respiration. Thus, pro- and anti-lipolytic mediators are bona fide modulators of thermogenesis in brown and brite adipocytes. In this Review, we discuss adrenergic and non-adrenergic mechanisms controlling lipolysis and thermogenesis and provide a comprehensive overview of pro- and anti-lipolytic mediators.
Collapse
Affiliation(s)
- Katharina Braun
- Chair of Molecular Nutritional Medicine, Technical University of Munich, TUM School of Life Sciences Weihenstephan, Gregor-Mendel-Str. 2, D-85354 Freising, Germany.,EKFZ - Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Gregor-Mendel-Str. 2, D-85354 Freising, Germany.,ZIEL - Institute for Food & Health, Technical University of Munich, Gregor-Mendel-Str. 2, D-85354 Freising, Germany
| | - Josef Oeckl
- Chair of Molecular Nutritional Medicine, Technical University of Munich, TUM School of Life Sciences Weihenstephan, Gregor-Mendel-Str. 2, D-85354 Freising, Germany.,EKFZ - Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Gregor-Mendel-Str. 2, D-85354 Freising, Germany.,ZIEL - Institute for Food & Health, Technical University of Munich, Gregor-Mendel-Str. 2, D-85354 Freising, Germany
| | - Julia Westermeier
- Chair of Molecular Nutritional Medicine, Technical University of Munich, TUM School of Life Sciences Weihenstephan, Gregor-Mendel-Str. 2, D-85354 Freising, Germany.,EKFZ - Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Gregor-Mendel-Str. 2, D-85354 Freising, Germany
| | - Yongguo Li
- Chair of Molecular Nutritional Medicine, Technical University of Munich, TUM School of Life Sciences Weihenstephan, Gregor-Mendel-Str. 2, D-85354 Freising, Germany.,EKFZ - Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Gregor-Mendel-Str. 2, D-85354 Freising, Germany
| | - Martin Klingenspor
- Chair of Molecular Nutritional Medicine, Technical University of Munich, TUM School of Life Sciences Weihenstephan, Gregor-Mendel-Str. 2, D-85354 Freising, Germany .,EKFZ - Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Gregor-Mendel-Str. 2, D-85354 Freising, Germany.,ZIEL - Institute for Food & Health, Technical University of Munich, Gregor-Mendel-Str. 2, D-85354 Freising, Germany
| |
Collapse
|
35
|
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
|
36
|
Nguyen NLT, Xue B, Bartness TJ. Sensory denervation of inguinal white fat modifies sympathetic outflow to white and brown fat in Siberian hamsters. Physiol Behav 2018; 190:28-33. [PMID: 29447836 DOI: 10.1016/j.physbeh.2018.02.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 02/10/2018] [Accepted: 02/10/2018] [Indexed: 10/18/2022]
Abstract
White adipose tissue (WAT) and brown adipose tissue (BAT) have sympathetic nervous system (SNS) and sensory innervations. Previous studies from our laboratory revealed central neuroanatomical evidence of WAT sensory and BAT SNS crosstalk with double labeling of inguinal WAT (IWAT) sensory and interscapular BAT (IBAT) SNS neurons. We previously demonstrated that WAT lipolysis increases IBAT temperature, but this effect is absent when IWAT afferents are surgically denervated, which severs both sensory and SNS nerves. It is possible that WAT sensory feedback can regulate SNS drive to itself and other WAT and BAT depots, and thus contribute to the existence of differential SNS outflow to fat during different energy challenges. Here we selectively denervated IWAT sensory nerves in Siberian hamsters using capsaicin and measured norepinephrine turnover (NETO) i.e., SNS drive to WAT and BAT depots, IBAT uncoupling protein 1 (UCP1) expression, body mass, fat mass, blood glucose, and food consumed after a 24-h cold exposure. IWAT sensory denervation decreased both IWAT and IBAT NETO and IBAT UCP1 expression. IWAT sensory denervation, however, increased mesenteric WAT (MWAT) NETO after the 24-h cold exposure and did not modify epididymal WAT (EWAT) and retroperitoneal WAT (RWAT) NETO compared with respective controls. Body mass, fat mass, blood glucose, and food consumed were unchanged across groups. RWAT and EWAT mass decreased in capsaicin-injected hamsters, but did not in the vehicle hamsters. These results functionally demonstrate the existence of IWAT sensory and IBAT SNS crosstalk and that a disruption in this sensory-SNS feedback mechanism modifies SNS drive to IWAT, IBAT, and MWAT, but not EWAT and RWAT.
Collapse
Affiliation(s)
- Ngoc Ly T Nguyen
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; Center for Obesity Reversal, Georgia State University, Atlanta, GA 30303, USA
| | - Bingzhong Xue
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; Center for Obesity Reversal, Georgia State University, Atlanta, GA 30303, USA.
| | - Timothy J Bartness
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; Center for Obesity Reversal, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
37
|
Chechi K, van Marken Lichtenbelt W, Richard D. Brown and beige adipose tissues: phenotype and metabolic potential in mice and men. J Appl Physiol (1985) 2018; 124:482-496. [PMID: 28302705 PMCID: PMC5867364 DOI: 10.1152/japplphysiol.00021.2017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/09/2017] [Accepted: 03/13/2017] [Indexed: 01/06/2023] Open
Abstract
With the recent rediscovery of brown fat in adult humans, our outlook on adipose tissue biology has undergone a paradigm shift. While we attempt to identify, recruit, and activate classic brown fat stores in humans, identification of beige fat has also raised the possibility of browning our white fat stores. Whether such transformation of human white fat depots can be achieved to enhance the whole body oxidative potential remains to be seen. Evidence to date, however, largely points toward a major oxidative role only for classic brown fat depots, at least in rodents. White fat stores seem to provide the main fuel for sustaining thermogenesis via lipolysis. Interestingly, molecular markers consistent with both classic brown and beige fat identity can be observed in human supraclavicular depot, thereby complicating the discussion on beige fat in humans. Here, we review the recent advances made in our understanding of brown and beige fat in humans and mice. We further provide an overview of their plausible physiological relevance to whole body energy metabolism.
Collapse
Affiliation(s)
- Kanta Chechi
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Ville de Québec, Quebec , Canada
| | - Wouter van Marken Lichtenbelt
- Department of Human Biology, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center , Maastricht , The Netherlands
| | - Denis Richard
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Ville de Québec, Quebec , Canada
| |
Collapse
|
38
|
Fernandez MO, Hsueh K, Park HT, Sauceda C, Hwang V, Kumar D, Kim S, Rickert E, Mahata S, Webster NJG. Astrocyte-Specific Deletion of Peroxisome-Proliferator Activated Receptor- γ Impairs Glucose Metabolism and Estrous Cycling in Female Mice. J Endocr Soc 2017; 1:1332-1350. [PMID: 29264458 PMCID: PMC5686676 DOI: 10.1210/js.2017-00242] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 09/15/2017] [Indexed: 01/21/2023] Open
Abstract
Mice lacking peroxisome-proliferator activated receptor-γ (PPARγ) in neurons do not become leptin resistant when placed on a high-fat diet (HFD). In male mice, this results in decreased food intake and increased energy expenditure, causing reduced body weight, but this difference in body weight is not observed in female mice. In addition, estrous cycles are disturbed and the ovaries present with hemorrhagic follicles. We observed that PPARγ was more highly expressed in astrocytes than neurons, so we created an inducible, conditional knockout of PPARγ in astrocytes (AKO). The AKO mice had impaired glucose tolerance and hepatic steatosis that did not worsen with HFD. Expression of gluconeogenic genes was elevated in the mouse livers, as was expression of several genes involved in lipogenesis, lipid transport, and storage. The AKO mice also had a reproductive phenotype with fewer estrous cycles, elevated plasma testosterone levels, reduced corpora lutea formation, and alterations in hypothalamic and ovarian gene expression. Thus, the phenotypes of the AKO mice were very different from those seen in the neuronal knockout mice, suggesting distinct roles for PPARγ in these two cell types.
Collapse
Affiliation(s)
- Marina O Fernandez
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, California 92093.,Laboratory of Neuroendocrinology, Instituto de Biología y Medicina Experimental, CONICET. Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Katherine Hsueh
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, California 92093
| | - Hyun Tae Park
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, California 92093.,Department of Obstetrics and Gynecology, Korea University Anam Hospital, Seoul 136-705, Korea
| | - Consuelo Sauceda
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, California 92093
| | - Vicky Hwang
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, California 92093
| | - Deepak Kumar
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, California 92093
| | - Sun Kim
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, California 92093
| | - Emily Rickert
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, California 92093
| | - Sumana Mahata
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, California 92093
| | - Nicholas J G Webster
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, California 92093.,Medical Research Service, VA San Diego Healthcare System, San Diego, California 92161.,Moores Cancer Center, University of California San Diego, La Jolla, California 92093
| |
Collapse
|
39
|
Gupta R, Ma Y, Wang M, Whim MD. AgRP-Expressing Adrenal Chromaffin Cells Are Involved in the Sympathetic Response to Fasting. Endocrinology 2017; 158:2572-2584. [PMID: 28531318 PMCID: PMC5551550 DOI: 10.1210/en.2016-1268] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 05/16/2017] [Indexed: 01/23/2023]
Abstract
Fasting evokes a homeostatic response that maintains circulating levels of energy-rich metabolites and increases the drive to eat. Centrally, this reflex activates a small population of hypothalamic neurons that are characterized by the expression of AgRP, a neuropeptide with an extremely restricted distribution. Apart from the hypothalamus, the only other site with substantial expression is the adrenal gland, but there is disagreement about which cells synthesize AgRP. Using immunohistochemistry, flow cytometry, and reverse transcription-polymerase chain reaction, we show AgRP is present in the mouse adrenal medulla and is expressed by neuroendocrine chromaffin cells that also synthesize the catecholamines and neuropeptide Y. Short-term fasting led to an increase in adrenal AgRP expression. Because AgRP can act as an antagonist at MC3/4 receptors, we tested whether melanotan II, an MC3/4 receptor agonist, could regulate pre- and postsynaptic signaling within the adrenal medulla. Melanotan II decreased the paired-pulse ratio of evoked synaptic currents recorded in chromaffin cells; this effect was blocked by exogenous AgRP. In contrast, neither melanotan II nor AgRP altered the optogenetically evoked release of catecholamines from isolated chromaffin cells. These results are consistent with the idea that AgRP regulates the strength of the sympathetic input by modulation of presynaptic MC3/4 receptors located on preganglionic neurons. We conclude that a small population of neuroendocrine cells in the adrenal medulla, and the arcuate nucleus of the hypothalamus, express AgRP and neuropeptide Y and are functionally involved in the systemic response to fasting.
Collapse
Affiliation(s)
- Rajesh Gupta
- Department of Cell Biology & Anatomy, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | - Yunbing Ma
- Department of Cell Biology & Anatomy, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | - Manqi Wang
- Department of Cell Biology & Anatomy, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | - Matthew D. Whim
- Department of Cell Biology & Anatomy, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| |
Collapse
|
40
|
Roberts ZS, Wolden-Hanson T, Matsen ME, Ryu V, Vaughan CH, Graham JL, Havel PJ, Chukri DW, Schwartz MW, Morton GJ, Blevins JE. Chronic hindbrain administration of oxytocin is sufficient to elicit weight loss in diet-induced obese rats. Am J Physiol Regul Integr Comp Physiol 2017; 313:R357-R371. [PMID: 28747407 DOI: 10.1152/ajpregu.00169.2017] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 06/30/2017] [Accepted: 07/02/2017] [Indexed: 02/06/2023]
Abstract
Oxytocin (OT) administration elicits weight loss in diet-induced obese (DIO) rodents, nonhuman primates, and humans by reducing energy intake and increasing energy expenditure. Although the neurocircuitry underlying these effects remains uncertain, OT neurons in the paraventricular nucleus are positioned to control both energy intake and sympathetic nervous system outflow to interscapular brown adipose tissue (BAT) through projections to the hindbrain nucleus of the solitary tract and spinal cord. The current work was undertaken to examine whether central OT increases BAT thermogenesis, whether this effect involves hindbrain OT receptors (OTRs), and whether such effects are associated with sustained weight loss following chronic administration. To assess OT-elicited changes in BAT thermogenesis, we measured the effects of intracerebroventricular administration of OT on interscapular BAT temperature in rats and mice. Because fourth ventricular (4V) infusion targets hindbrain OTRs, whereas third ventricular (3V) administration targets both forebrain and hindbrain OTRs, we compared responses to OT following chronic 3V infusion in DIO rats and mice and chronic 4V infusion in DIO rats. We report that chronic 4V infusion of OT into two distinct rat models recapitulates the effects of 3V OT to ameliorate DIO by reducing fat mass. While reduced food intake contributes to this effect, our finding that 4V OT also increases BAT thermogenesis suggests that increased energy expenditure may contribute as well. Collectively, these findings support the hypothesis that, in DIO rats, OT action in the hindbrain evokes sustained weight loss by reducing energy intake and increasing BAT thermogenesis.
Collapse
Affiliation(s)
- Zachary S Roberts
- Veterans Affairs Puget Sound Health Care System, Office of Research and Development, Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, Washington
| | - Tami Wolden-Hanson
- Veterans Affairs Puget Sound Health Care System, Office of Research and Development, Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, Washington
| | - Miles E Matsen
- University of Washington Diabetes Institute, University of Washington School of Medicine, Seattle, Washington
| | - Vitaly Ryu
- Department of Biology, Georgia State University, Atlanta, Georgia; and.,Center for Obesity Reversal, Georgia State University, Atlanta, Georgia
| | - Cheryl H Vaughan
- Department of Biology, Georgia State University, Atlanta, Georgia; and.,Center for Obesity Reversal, Georgia State University, Atlanta, Georgia
| | - James L Graham
- Departments of Nutrition and Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, California
| | - Peter J Havel
- Departments of Nutrition and Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, California
| | - Daniel W Chukri
- Veterans Affairs Puget Sound Health Care System, Office of Research and Development, Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, Washington
| | - Michael W Schwartz
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, Washington.,University of Washington Diabetes Institute, University of Washington School of Medicine, Seattle, Washington
| | - Gregory J Morton
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, Washington.,University of Washington Diabetes Institute, University of Washington School of Medicine, Seattle, Washington
| | - James E Blevins
- Veterans Affairs Puget Sound Health Care System, Office of Research and Development, Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, Washington; .,Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| |
Collapse
|
41
|
Molecular Mechanisms of Sodium-Sensitive Hypertension in the Metabolic Syndrome. Curr Hypertens Rep 2017; 19:60. [DOI: 10.1007/s11906-017-0759-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
42
|
Shin AC, Filatova N, Lindtner C, Chi T, Degann S, Oberlin D, Buettner C. Insulin Receptor Signaling in POMC, but Not AgRP, Neurons Controls Adipose Tissue Insulin Action. Diabetes 2017; 66:1560-1571. [PMID: 28385803 PMCID: PMC5440019 DOI: 10.2337/db16-1238] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 03/22/2017] [Indexed: 12/31/2022]
Abstract
Insulin is a key regulator of adipose tissue lipolysis, and impaired adipose tissue insulin action results in unrestrained lipolysis and lipotoxicity, which are hallmarks of the metabolic syndrome and diabetes. Insulin regulates adipose tissue metabolism through direct effects on adipocytes and through signaling in the central nervous system by dampening sympathetic outflow to the adipose tissue. Here we examined the role of insulin signaling in agouti-related protein (AgRP) and pro-opiomelanocortin (POMC) neurons in regulating hepatic and adipose tissue insulin action. Mice lacking the insulin receptor in AgRP neurons (AgRP IR KO) exhibited impaired hepatic insulin action because the ability of insulin to suppress hepatic glucose production (hGP) was reduced, but the ability of insulin to suppress lipolysis was unaltered. To the contrary, in POMC IR KO mice, insulin lowered hGP but failed to suppress adipose tissue lipolysis. High-fat diet equally worsened glucose tolerance in AgRP and POMC IR KO mice and their respective controls but increased hepatic triglyceride levels only in POMC IR KO mice, consistent with impaired lipolytic regulation resulting in fatty liver. These data suggest that although insulin signaling in AgRP neurons is important in regulating glucose metabolism, insulin signaling in POMC neurons controls adipose tissue lipolysis and prevents high-fat diet-induced hepatic steatosis.
Collapse
Affiliation(s)
- Andrew C Shin
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Nika Filatova
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Claudia Lindtner
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Tiffany Chi
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Seta Degann
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Douglas Oberlin
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Christoph Buettner
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
43
|
Burke LK, Darwish T, Cavanaugh AR, Virtue S, Roth E, Morro J, Liu SM, Xia J, Dalley JW, Burling K, Chua S, Vidal-Puig T, Schwartz GJ, Blouet C. mTORC1 in AGRP neurons integrates exteroceptive and interoceptive food-related cues in the modulation of adaptive energy expenditure in mice. eLife 2017; 6. [PMID: 28532548 PMCID: PMC5441868 DOI: 10.7554/elife.22848] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 04/18/2017] [Indexed: 01/19/2023] Open
Abstract
Energy dissipation through interscapular brown adipose tissue (iBAT) thermogenesis is an important contributor to adaptive energy expenditure. However, it remains unresolved how acute and chronic changes in energy availability are detected by the brain to adjust iBAT activity and maintain energy homeostasis. Here, we provide evidence that AGRP inhibitory tone to iBAT represents an energy-sparing circuit that integrates environmental food cues and internal signals of energy availability. We establish a role for the nutrient-sensing mTORC1 signaling pathway within AGRP neurons in the detection of environmental food cues and internal signals of energy availability, and in the bi-directional control of iBAT thermogenesis during nutrient deficiency and excess. Collectively, our findings provide insights into how mTORC1 signaling within AGRP neurons surveys energy availability to engage iBAT thermogenesis, and identify AGRP neurons as a neuronal substrate for the coordination of energy intake and adaptive expenditure under varying physiological and environmental contexts. DOI:http://dx.doi.org/10.7554/eLife.22848.001 Losing weight through dieting can be difficult. Weight loss strategies often prove ineffective because the body works like a thermostat and couples what we eat to the number of calories we burn. When we eat less, our bodies compensate and burn fewer calories, which makes losing weight harder. The brain is the master regulator of this caloric thermostat, but it is not clear how it adjusts our energy expenditure to account for how much we have eaten. A structure deep within the brain called the hypothalamus, which helps regulate appetite, is thought to be involved in the caloric thermostat. Activating a group of neurons within the hypothalamus called the agouti-related neuropeptide (AGRP) neurons causes animals to consume large quantities of food. By contrast, inhibiting AGRP neurons causes animals to stop eating almost entirely. Burke et al. studied AGRP neurons in mice. The experiments show that artificially activating the neurons in mice that don’t have access to food increases the animals’ activity levels but reduces the rate at which they burn calories, which helps the mice to maintain their existing weight. Allowing the mice to eat, or even just to see and smell food, switches off this effect and returns energy expenditure to normal. Finally, exposing mice to a high-fat diet for several days inhibits their AGRP neurons, and causes the animals to burn calories at a faster rate. By using up excess calories, this change also helps the animals maintain their existing body weight. The findings of Burke et al. show that AGRP neurons are a key component of the caloric thermostat. By adjusting the rate at which the body burns calories, AGRP neurons can compensate for any changes in food intake and so limit changes in body weight. This work opens up the possibility of developing therapies that disconnect energy expenditure from energy intake to help maintain long-term weight loss. DOI:http://dx.doi.org/10.7554/eLife.22848.002
Collapse
Affiliation(s)
- Luke K Burke
- MRC Metabolic Diseases Unit, Metabolic Research Laboratories, University of Cambridge, Cambridge, United Kingdom.,WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Tamana Darwish
- MRC Metabolic Diseases Unit, Metabolic Research Laboratories, University of Cambridge, Cambridge, United Kingdom.,WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Althea R Cavanaugh
- Departments of Medicine and Neuroscience, The Albert Einstein College of Medicine, New York, United States
| | - Sam Virtue
- MRC Metabolic Diseases Unit, Metabolic Research Laboratories, University of Cambridge, Cambridge, United Kingdom.,WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Emma Roth
- MRC Metabolic Diseases Unit, Metabolic Research Laboratories, University of Cambridge, Cambridge, United Kingdom.,WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Joanna Morro
- MRC Metabolic Diseases Unit, Metabolic Research Laboratories, University of Cambridge, Cambridge, United Kingdom.,WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Shun-Mei Liu
- Departments of Medicine and Neuroscience, The Albert Einstein College of Medicine, New York, United States
| | - Jing Xia
- Department of Psychology, Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, United Kingdom
| | - Jeffrey W Dalley
- Department of Psychology, Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, United Kingdom.,Department of Psychiatry, Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, United Kingdom
| | - Keith Burling
- MRC Metabolic Diseases Unit, Metabolic Research Laboratories, University of Cambridge, Cambridge, United Kingdom.,WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Streamson Chua
- Departments of Medicine and Neuroscience, The Albert Einstein College of Medicine, New York, United States
| | - Toni Vidal-Puig
- MRC Metabolic Diseases Unit, Metabolic Research Laboratories, University of Cambridge, Cambridge, United Kingdom.,WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Gary J Schwartz
- Departments of Medicine and Neuroscience, The Albert Einstein College of Medicine, New York, United States
| | - Clémence Blouet
- MRC Metabolic Diseases Unit, Metabolic Research Laboratories, University of Cambridge, Cambridge, United Kingdom.,WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
44
|
Ramirez-Plascencia OD, Saderi N, Escobar C, Salgado-Delgado RC. Feeding during the rest phase promotes circadian conflict in nuclei that control energy homeostasis and sleep-wake cycle in rats. Eur J Neurosci 2017; 45:1325-1332. [DOI: 10.1111/ejn.13563] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 03/10/2017] [Accepted: 03/13/2017] [Indexed: 01/11/2023]
Affiliation(s)
- Oscar D. Ramirez-Plascencia
- Facultad de Ciencias; Universidad Autónoma de San Luis Potosí; Av. Salvador Nava Martínez S/N Zona Universitaria Poniente cp. 78290 San Luis Potosí, S.L.P Mexico
| | - Nadia Saderi
- Facultad de Ciencias; Universidad Autónoma de San Luis Potosí; Av. Salvador Nava Martínez S/N Zona Universitaria Poniente cp. 78290 San Luis Potosí, S.L.P Mexico
| | - Carolina Escobar
- Departamento de Anatomía; Facultad de Medicina; Universidad Nacional Autónoma de México; Mexico City Mexico
| | - Roberto C. Salgado-Delgado
- Facultad de Ciencias; Universidad Autónoma de San Luis Potosí; Av. Salvador Nava Martínez S/N Zona Universitaria Poniente cp. 78290 San Luis Potosí, S.L.P Mexico
| |
Collapse
|
45
|
Almundarij TI, Gavini CK, Novak CM. Suppressed sympathetic outflow to skeletal muscle, muscle thermogenesis, and activity energy expenditure with calorie restriction. Physiol Rep 2017; 5:5/4/e13171. [PMID: 28242830 PMCID: PMC5328781 DOI: 10.14814/phy2.13171] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 01/29/2017] [Indexed: 12/21/2022] Open
Abstract
During weight loss, adaptive thermogenesis occurs where energy expenditure (EE) is suppressed beyond that predicted for the smaller body size. Here, we investigated the contributions of resting and nonresting EE to the reduced total EE seen after 3 weeks of 50% calorie restriction (CR) in rats, focusing on activity‐associated EE, muscle thermogenesis, and sympathetic outflow. Prolonged food restriction resulted in a 42% reduction in daily EE, through a 40% decrease in resting EE, and a 48% decline in nonresting EE. These decreases in EE were significant even when the reductions in body weight and lean mass were taken into account. Along with a decreased caloric need for low‐to‐moderate‐intensity treadmill activity with 50% CR, baseline and activity‐related muscle thermogenesis were also suppressed, though the ability to increase muscle thermogenesis above baseline levels was not compromised. When sympathetic drive was measured by assessing norepinephrine turnover (NETO), 50% CR was found to decrease NETO in three of the four muscle groups examined, whereas elevated NETO was found in white adipose tissue of food‐restricted rats. Central activation of melanocortin 4 receptors in the ventromedial hypothalamus stimulated this pathway, enhancing activity EE; this was not compromised by 50% CR. These data suggest that suppressed activity EE contributes to adaptive thermogenesis during energy restriction. This may stem from decreased sympathetic drive to skeletal muscle, increasing locomotor efficiency and reducing skeletal muscle thermogenesis. The capacity to increase activity EE in response to central stimuli is retained, however, presenting a potential target for preventing weight regain.
Collapse
Affiliation(s)
- Tariq I Almundarij
- College of Agriculture and Veterinary Medicine, Al Qassim University, Buraydah, Al-Qassim Province, Saudi Arabia.,Department of Biological Sciences, Kent State University, Kent, Ohio
| | - Chaitanya K Gavini
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois.,School of Biomedical Sciences, Kent State University, Kent, Ohio
| | - Colleen M Novak
- Department of Biological Sciences, Kent State University, Kent, Ohio .,School of Biomedical Sciences, Kent State University, Kent, Ohio
| |
Collapse
|
46
|
Ryu V, Watts AG, Xue B, Bartness TJ. Bidirectional crosstalk between the sensory and sympathetic motor systems innervating brown and white adipose tissue in male Siberian hamsters. Am J Physiol Regul Integr Comp Physiol 2017; 312:R324-R337. [PMID: 28077392 DOI: 10.1152/ajpregu.00456.2015] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 11/28/2016] [Accepted: 12/31/2016] [Indexed: 01/31/2023]
Abstract
The brain networks connected to the sympathetic motor and sensory innervations of brown (BAT) and white (WAT) adipose tissues were originally described using two transneuronally transported viruses: the retrogradely transported pseudorabies virus (PRV), and the anterogradely transported H129 strain of herpes simplex virus-1 (HSV-1 H129). Further complexity was added to this network organization when combined injections of PRV and HSV-1 H129 into either BAT or WAT of the same animal generated sets of coinfected neurons in the brain, spinal cord, and sympathetic and dorsal root ganglia. These neurons are well positioned to act as sensorimotor links in the feedback circuits that control each fat pad. We have now determined the extent of sensorimotor crosstalk between interscapular BAT (IBAT) and inguinal WAT (IWAT). PRV152 and HSV-1 H129 were each injected into IBAT or IWAT of the same animal: H129 into IBAT and PRV152 into IWAT. The reverse configuration was applied in a different set of animals. We found single-labeled neurons together with H129+PRV152 coinfected neurons in multiple brain sites, with lesser numbers in the sympathetic and dorsal root ganglia that innervate IBAT and IWAT. We propose that these coinfected neurons mediate sensory-sympathetic motor crosstalk between IBAT and IWAT. Comparing the relative numbers of coinfected neurons between the two injection configurations showed a bias toward IBAT-sensory and IWAT-sympathetic motor feedback loops. These coinfected neurons provide a neuroanatomical framework for functional interactions between IBAT thermogenesis and IWAT lipolysis that occurs with cold exposure, food restriction/deprivation, exercise, and more generally with alterations in adiposity.
Collapse
Affiliation(s)
- Vitaly Ryu
- Department of Biology, Obesity Reversal Center, Georgia State University, Atlanta, Georgia; and
| | - Alan G Watts
- Department of Biological Sciences, University of Southern California, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, California
| | - Bingzhong Xue
- Department of Biology, Obesity Reversal Center, Georgia State University, Atlanta, Georgia; and
| | - Timothy J Bartness
- Department of Biology, Obesity Reversal Center, Georgia State University, Atlanta, Georgia; and
| |
Collapse
|
47
|
Conceição EPS, Moura EG, Oliveira E, Guarda DS, Figueiredo MS, Quitete FT, Calvino C, Miranda RA, Mathias PCF, Manhães AC, Lisboa PC. Dietary calcium supplementation in adult rats reverts brown adipose tissue dysfunction programmed by postnatal early overfeeding. J Nutr Biochem 2017; 39:117-125. [DOI: 10.1016/j.jnutbio.2016.09.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Revised: 07/05/2016] [Accepted: 09/02/2016] [Indexed: 11/28/2022]
|
48
|
Roh E, Kim MS. Brain Regulation of Energy Metabolism. Endocrinol Metab (Seoul) 2016; 31:519-524. [PMID: 28029023 PMCID: PMC5195827 DOI: 10.3803/enm.2016.31.4.519] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 09/20/2016] [Accepted: 09/30/2016] [Indexed: 11/11/2022] Open
Abstract
In healthy individuals, energy intake is in balance with energy expenditure, which helps to maintain a normal body weight. The brain's inability to control energy homeostasis underlies the pathology of hyperphagia and obesity. The brain detects body energy excess and deficit by sensing the levels of circulating metabolic hormones and nutrients and by receiving metabolic information from the periphery via the autonomic nervous system. A specialized neuronal network coordinates energy intake behavior and the metabolic processes affecting energy expenditure. Here, we briefly review neuronal mechanisms by which our body maintains energy balance.
Collapse
Affiliation(s)
- Eun Roh
- Department of Biomedical Science, University of Ulsan College of Medicine, Seoul, Korea
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Min Seon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
49
|
Almundarij TI, Smyers ME, Spriggs A, Heemstra LA, Beltz L, Dyne E, Ridenour C, Novak CM. Physical Activity, Energy Expenditure, and Defense of Body Weight in Melanocortin 4 Receptor-Deficient Male Rats. Sci Rep 2016; 6:37435. [PMID: 27886210 PMCID: PMC5122857 DOI: 10.1038/srep37435] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 10/28/2016] [Indexed: 01/28/2023] Open
Abstract
Melanocortin 4 receptor (MC4R) variants contribute to human obesity, and rats lacking functional MC4R (Mc4rK314X/K314X) are obese. We investigated the hypothesis that low energy expenditure (EE) and physical activity contribute to this obese phenotype in male rats, and determined whether lack of functional MC4R conferred protection from weight loss during 50% calorie restriction. Though Mc4rK314X/K314X rats showed low brown adipose Ucp1 expression and were less physically active than rats heterozygous for the mutation (Mc4r+/K314X) or wild-type (Mc4r+/+) rats, we found no evidence of lowered EE in Mc4rK314X/K314X rats once body weight was taken into account using covariance. Mc4rK314X/K314X rats had a significantly higher respiratory exchange ratio. Compared to Mc4r+/+ rats, Mc4rK314X/K314X and Mc4r+/K314X rats lost less lean mass during calorie restriction, and less body mass when baseline weight was accounted for. Limited regional overexpression of Mc3r was found in the hypothalamus. Although lower physical activity levels in rats with nonfunctional MC4R did not result in lower total EE during free-fed conditions, rats lacking one or two functional copies of Mc4r showed conservation of mass, particularly lean mass, during energy restriction. This suggests that variants affecting MC4R function may contribute to individual differences in the metabolic response to food restriction.
Collapse
Affiliation(s)
- Tariq I Almundarij
- College of Agriculture and Veterinary Medicine, Al-Qassim University, Buraydah, Al-Qassim Province, Saudi Arabia.,Department of Biological Sciences, Kent State University, Kent, OH, 44242, US
| | - Mark E Smyers
- School of Biomedical Sciences, Kent State University, Kent, OH, 44242, US
| | - Addison Spriggs
- Department of Biological Sciences, Kent State University, Kent, OH, 44242, US
| | - Lydia A Heemstra
- Department of Biological Sciences, Kent State University, Kent, OH, 44242, US
| | - Lisa Beltz
- Department of Natural Sciences, Malone University, Canton, OH, 44709, US
| | - Eric Dyne
- School of Biomedical Sciences, Kent State University, Kent, OH, 44242, US.,Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, US
| | - Caitlyn Ridenour
- Department of Natural Sciences, Malone University, Canton, OH, 44709, US
| | - Colleen M Novak
- Department of Biological Sciences, Kent State University, Kent, OH, 44242, US.,School of Biomedical Sciences, Kent State University, Kent, OH, 44242, US
| |
Collapse
|
50
|
Nguyen NLT, Barr CL, Ryu V, Cao Q, Xue B, Bartness TJ. Separate and shared sympathetic outflow to white and brown fat coordinately regulates thermoregulation and beige adipocyte recruitment. Am J Physiol Regul Integr Comp Physiol 2016; 312:R132-R145. [PMID: 27881398 DOI: 10.1152/ajpregu.00344.2016] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 10/14/2016] [Accepted: 11/05/2016] [Indexed: 11/22/2022]
Abstract
White adipose tissue (WAT) and brown adipose tissue (BAT) are innervated and regulated by the sympathetic nervous system (SNS). It is not clear, however, whether there are shared or separate central SNS outflows to WAT and BAT that regulate their function. We injected two isogenic strains of pseudorabies virus, a retrograde transneuronal viral tract tracer, with unique fluorescent reporters into interscapular BAT (IBAT) and inguinal WAT (IWAT) of the same Siberian hamsters to define SNS pathways to both. To test the functional importance of SNS coordinated control of BAT and WAT, we exposed hamsters with denervated SNS nerves to IBAT to 4°C for 16-24 h and measured core and fat temperatures and norepinephrine turnover (NETO) and uncoupling protein 1 (UCP1) expression in fat tissues. Overall, there were more SNS neurons innervating IBAT than IWAT across the neuroaxis. However, there was a greater percentage of singly labeled IWAT neurons in midbrain reticular nuclei than singly labeled IBAT neurons. The hindbrain had ~30-40% of doubly labeled neurons while the forebrain had ~25% suggesting shared SNS circuitry to BAT and WAT across the brain. The raphe nucleus, a key region in thermoregulation, had ~40% doubly labeled neurons. Hamsters with IBAT SNS denervation maintained core body temperature during acute cold challenge and had increased beige adipocyte formation in IWAT. They also had increased IWAT NETO, temperature, and UCP1 expression compared with intact hamsters. These data provide strong neuroanatomical and functional evidence of WAT and BAT SNS cross talk for thermoregulation and beige adipocyte formation.
Collapse
Affiliation(s)
- Ngoc Ly T Nguyen
- Department of Biology, Georgia State University, Atlanta, Georgia.,Center for Obesity Reversal, Georgia State University, Atlanta, Georgia; and
| | - Candace L Barr
- Neuroscience Institute, Georgia State University, Atlanta, Georgia
| | - Vitaly Ryu
- Department of Biology, Georgia State University, Atlanta, Georgia.,Center for Obesity Reversal, Georgia State University, Atlanta, Georgia; and
| | - Qiang Cao
- Department of Biology, Georgia State University, Atlanta, Georgia.,Center for Obesity Reversal, Georgia State University, Atlanta, Georgia; and
| | - Bingzhong Xue
- Department of Biology, Georgia State University, Atlanta, Georgia; .,Center for Obesity Reversal, Georgia State University, Atlanta, Georgia; and.,Neuroscience Institute, Georgia State University, Atlanta, Georgia
| | - Timothy J Bartness
- Department of Biology, Georgia State University, Atlanta, Georgia.,Center for Obesity Reversal, Georgia State University, Atlanta, Georgia; and.,Neuroscience Institute, Georgia State University, Atlanta, Georgia
| |
Collapse
|