1
|
Slattery JD, Rambousek JR, Tsui E, Honeycutt MK, Goldberg M, Graham JL, Wietecha TA, Wolden-Hanson T, Williams AL, O’Brien KD, Havel PJ, Blevins JE. Effects of systemic oxytocin and beta-3 receptor agonist (CL 316243) treatment on body weight and adiposity in male diet-induced obese rats. Front Endocrinol (Lausanne) 2025; 16:1503096. [PMID: 40104132 PMCID: PMC11913664 DOI: 10.3389/fendo.2025.1503096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 01/31/2025] [Indexed: 03/20/2025] Open
Abstract
Previous studies have implicated hindbrain oxytocin (OT) receptors in the control of food intake and brown adipose tissue (BAT) thermogenesis. We recently demonstrated that hindbrain [fourth ventricle (4V)] administration of oxytocin (OT) could be used as an adjunct to drugs that directly target beta-3 adrenergic receptors (β3-AR) to elicit weight loss in diet-induced obese (DIO) rodents. What remains unclear is whether systemic OT can be used as an adjunct with the β3-AR agonist, CL 316243, to increase BAT thermogenesis and elicit weight loss in DIO rats. We hypothesized that systemic OT and β3-AR agonist (CL 316243) treatment would produce an additive effect to reduce body weight and adiposity in DIO rats by decreasing food intake and stimulating BAT thermogenesis. To test this hypothesis, we determined the effects of systemic (subcutaneous) infusions of OT (50 nmol/day) or vehicle (VEH) when combined with daily systemic (intraperitoneal) injections of CL 316243 (0.5 mg/kg) or VEH on body weight, adiposity, food intake and brown adipose tissue temperature (TIBAT). OT and CL 316243 monotherapy decreased body weight by 8.0 ± 0.9% (P<0.05) and 8.6 ± 0.6% (P<0.05), respectively, but OT in combination with CL 316243 produced more substantial weight loss (14.9 ± 1.0%; P<0.05) compared to either treatment alone. These effects were associated with decreased adiposity, energy intake and elevated TIBAT during the treatment period. The findings from the current study suggest that the effects of systemic OT and CL 316243 to elicit weight loss are additive and appear to be driven primarily by OT-elicited changes in food intake and CL 316243-elicited increases in BAT thermogenesis.
Collapse
Affiliation(s)
- Jared D. Slattery
- Veterans Affairs (VA) Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, United States
| | - June R. Rambousek
- Veterans Affairs (VA) Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, United States
| | - Edison Tsui
- Veterans Affairs (VA) Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, United States
| | - Mackenzie K. Honeycutt
- Veterans Affairs (VA) Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, United States
| | - Matvey Goldberg
- Veterans Affairs (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
| | - Tomasz A. Wietecha
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington (UW) School of Medicine, Seattle, WA, United States
- University of Washington (UW) Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, WA, United States
| | - Tami Wolden-Hanson
- Veterans Affairs (VA) Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, United States
| | - Amber L. Williams
- Veterans Affairs (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
- University of Washington (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
- Veterans Affairs (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 (UW) School of Medicine, Seattle, WA, United States
| |
Collapse
|
2
|
Slattery JD, Rambousek JR, Tsui E, Honeycutt MK, Goldberg M, Graham JL, Wietecha TA, Wolden-Hanson T, Williams AL, O'Brien KD, Havel PJ, Blevins JE. Effects of systemic oxytocin and beta-3 receptor agonist (CL 316243) treatment on body weight and adiposity in male diet-induced obese rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.27.615550. [PMID: 39502365 PMCID: PMC11537314 DOI: 10.1101/2024.09.27.615550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
Previous studies have implicated hindbrain oxytocin (OT) receptors in the control of food intake and brown adipose tissue (BAT) thermogenesis. We recently demonstrated that hindbrain [fourth ventricle (4V)] administration of oxytocin (OT) could be used as an adjunct to drugs that directly target beta-3 adrenergic receptors (β3-AR) to elicit weight loss in diet-induced obese (DIO) rodents. What remains unclear is whether systemic OT can be used as an adjunct with the β3-AR agonist, CL 316243, to increase BAT thermogenesis and elicit weight loss in DIO rats. We hypothesized that systemic OT and β3-AR agonist (CL 316243) treatment would produce an additive effect to reduce body weight and adiposity in DIO rats by decreasing food intake and stimulating BAT thermogenesis. To test this hypothesis, we determined the effects of systemic (subcutaneous) infusions of OT (50 nmol/day) or vehicle (VEH) when combined with daily systemic (intraperitoneal) injections of CL 316243 (0.5 mg/kg) or VEH on body weight, adiposity, food intake and brown adipose tissue temperature (TIBAT). OT and CL 316243 monotherapy decreased body weight by 8.0±0.9% (P<0.05) and 8.6±0.6% (P<0.05), respectively, but OT in combination with CL 316243 produced more substantial weight loss (14.9±1.0%; P<0.05) compared to either treatment alone. These effects were associated with decreased adiposity, energy intake and elevated TIBAT during the treatment period. The findings from the current study suggest that the effects of systemic OT and CL 316243 to elicit weight loss are additive and appear to be driven primarily by OT-elicited changes in food intake and CL 316243-elicited increases in BAT thermogenesis.
Collapse
Affiliation(s)
- 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
| | - June R Rambousek
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
| | - 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
| | - 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
| | - 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
| | - James L Graham
- Department of Nutrition, University of California, Davis, CA 95616, USA
| | - Tomasz A Wietecha
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
- UW Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, WA 98109
| | - 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
| | - Amber L Williams
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
| | - Kevin D O'Brien
- Division of Cardiology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
- UW Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, WA 98109
| | - Peter J Havel
- Department of Nutrition, University of California, Davis, CA 95616, USA
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - 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
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
3
|
Cao X, Wang Q, Zhang L, Sun H, Xu G, Chen X, Wu Z, Liu H, Yuan G, Wu J, Liu T. Oxytocin alleviates high-fat diet-induced anxiety by decreasing glutamatergic synaptic transmission in the ventral dentate gyrus in adolescent mice. Neuropharmacology 2025; 262:110201. [PMID: 39481751 DOI: 10.1016/j.neuropharm.2024.110201] [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: 06/08/2024] [Revised: 09/21/2024] [Accepted: 10/27/2024] [Indexed: 11/02/2024]
Abstract
A high-fat diet (HFD)-induced obesity is associated with mental disorders in adolescence. However, the mechanisms underlying these associations remain unclear. In this study, we hypothesized that synaptic remodeling occurs in the ventral hippocampus (vHP) of obese mice. To investigate this, we established a postnatal model of HFD-induced obesity in mice and observed increased body weight, elevated plasma luteinizing hormone and testosterone levels, premature puberty, and enhanced anxiety-like behavior in male subjects. We also examined the effect of HFD on the c-Fos protein expression in the ventral dentate gyrus (vDG) and explored the influence of intracerebroventricular (i.c.v) oxytocin injections on HFD-induced anxiety. Our results indicated an increase in c-Fos-positive cells in the vDG following HFD consumption. Additionally, we recorded the spontaneous synaptic activity of miniature excitatory postsynaptic currents (mEPSCs) in the vDG. Notably, HFD resulted in an elevated mEPSC frequency without affecting mEPSC amplitude. Subsequently, investigations demonstrated that i.c.v oxytocin injections reversed anxiety-like behavior induced by HFD. Moreover, the application of oxytocin in a bath solution reduced the mEPSC frequency in the vDG. These findings suggest that postnatal HFD intake induces synaptic dysfunction in the vDG, associated with the hyperactivity of vDG neurons, potentially contributing to the anxiety-like behavior in juvenile obesity.
Collapse
Affiliation(s)
- Xi Cao
- Department of Pediatrics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Qiyuan Wang
- Department of Pediatrics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Lina Zhang
- Department of Pediatrics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Huichao Sun
- Department of Pediatrics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Gang Xu
- Jiangxi Provincial Key Laboratory of Trauma, Burn and Pain Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Xiao Chen
- Department of Pediatrics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Zhihong Wu
- Department of Pediatrics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Huibao Liu
- Department of Pediatrics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Gaole Yuan
- Department of Pediatrics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
| | - Jian Wu
- Department of Pediatrics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
| | - Tao Liu
- Department of Pediatrics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China; Jiangxi Provincial Key Laboratory of Trauma, Burn and Pain Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
4
|
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, Taborsky GJ, Blevins JE. Sympathetic innervation of interscapular brown adipose tissue is not a predominant mediator of Oxytocin (OT)-elicited reductions of body weight gain and adiposity in male diet-induced obese rats. FRONTIERS IN DRUG DELIVERY 2024; 4:1497746. [PMID: 39866535 PMCID: PMC11759500 DOI: 10.3389/fddev.2024.1497746] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
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
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 98108, USA
| | - 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 98108, USA
| | - 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
| | - 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
| | - 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
| | - 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
| | - June R Rambousek
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
| | - 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
| | - 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
| | - Tomasz A Wietecha
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
- UW Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, WA 98109
| | - James L Graham
- Department of Nutrition, University of California, Davis, CA 95616, USA
| | - Geronimo P Tapia
- UTEP Systems Neuroscience Laboratory, Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, 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
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington
| | - Kevin D O'Brien
- Division of Cardiology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
- UW Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, WA 98109
| | - Thomas O Mundinger
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, 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
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington
| | - Vitaly Ryu
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Peter J Havel
- Department of Nutrition, University of California, Davis, CA 95616, USA
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Arshad M Khan
- UTEP Systems Neuroscience Laboratory, Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - 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
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - 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
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
5
|
Usmani SS, Jung HG, Zhang Q, Kim MW, Choi Y, Caglayan AB, Cai D. Targeting the hypothalamus for modeling age-related DNA methylation and developing OXT-GnRH combinational therapy against Alzheimer's disease-like pathologies in male mouse model. Nat Commun 2024; 15:9419. [PMID: 39482312 PMCID: PMC11528003 DOI: 10.1038/s41467-024-53507-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/09/2024] [Indexed: 11/03/2024] Open
Abstract
The hypothalamus plays an important role in aging, but it remains unclear regarding the underlying epigenetics and whether this hypothalamic basis can help address aging-related diseases. Here, by comparing mouse hypothalamus with two other limbic system components, we show that the hypothalamus is characterized by distinctively high-level DNA methylation during young age and by the distinct dynamics of DNA methylation and demethylation when approaching middle age. On the other hand, age-related DNA methylation in these limbic system components commonly and sensitively applies to genes in hypothalamic regulatory pathways, notably oxytocin (OXT) and gonadotropin-releasing hormone (GnRH) pathways. Middle age is associated with transcriptional declines of genes which encode OXT, GnRH and signaling components, which similarly occur in an Alzheimer's disease (AD)-like model. Therapeutically, OXT-GnRH combination is substantially more effective than individual peptides in treating AD-like disorders in male 5×FAD model. In conclusion, the hypothalamus is important for modeling age-related DNA methylation and developing hypothalamic strategies to combat AD.
Collapse
Affiliation(s)
- Salman Sadullah Usmani
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Hyun-Gug Jung
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Qichao Zhang
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Min Woo Kim
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yuna Choi
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ahmet Burak Caglayan
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Dongsheng Cai
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
6
|
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, Taborsky 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] [Key Words] [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
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 98108, USA
| | - 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 98108, USA
| | - 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
| | - 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
| | - 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
| | - 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
| | - June R Rambousek
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
| | - 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
| | - 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
| | - Tomasz A Wietecha
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
- UW Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, WA 98109
| | - James L Graham
- Department of Nutrition, University of California, Davis, CA 95616, USA
| | - Geronimo P Tapia
- UTEP Systems Neuroscience Laboratory, Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, 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
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington
| | - Kevin D O'Brien
- Division of Cardiology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
- UW Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, WA 98109
| | - Thomas O Mundinger
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, 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
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington
| | - Vitaly Ryu
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Peter J Havel
- Department of Nutrition, University of California, Davis, CA 95616, USA
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Arshad M Khan
- UTEP Systems Neuroscience Laboratory, Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - 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
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - 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
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
7
|
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
|
8
|
Worth AA, Feetham CH, Morrissey NA, Luckman SM. Paraventricular oxytocin neurons impact energy intake and expenditure: projections to the bed nucleus of the stria terminalis reduce sucrose consumption. Front Endocrinol (Lausanne) 2024; 15:1449326. [PMID: 39286269 PMCID: PMC11402739 DOI: 10.3389/fendo.2024.1449326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/12/2024] [Indexed: 09/19/2024] Open
Abstract
Background The part played by oxytocin and oxytocin neurons in the regulation of food intake is controversial. There is much pharmacological data to support a role for oxytocin notably in regulating sugar consumption, however, several recent experiments have questioned the importance of oxytocin neurons themselves. Methods Here we use a combination of histological and chemogenetic techniques to investigate the selective activation or inhibition of oxytocin neurons in the hypothalamic paraventricular nucleus (OxtPVH). We then identify a pathway from OxtPVH neurons to the bed nucleus of the stria terminalis using the cell-selective expression of channel rhodopsin. Results OxtPVH neurons increase their expression of cFos after both physiological (fast-induced re-feeding or oral lipid) and pharmacological (systemic administration of cholecystokinin or lithium chloride) anorectic signals. Chemogenetic activation of OxtPVH neurons is sufficient to decrease free-feeding in Oxt Cre:hM3Dq mice, while inhibition in Oxt Cre:hM4Di mice attenuates the response to administration of cholecystokinin. Activation of OxtPVH neurons also increases energy expenditure and core-body temperature, without a significant effect on locomotor activity. Finally, the selective, optogenetic stimulation of a pathway from OxtPVH neurons to the bed nucleus of the stria terminalis reduces the consumption of sucrose. Conclusion Our results support a role for oxytocin neurons in the regulation of whole-body metabolism, including a modulatory action on food intake and energy expenditure. Furthermore, we demonstrate that the pathway from OxtPVH neurons to the bed nucleus of the stria terminalis can regulate sugar consumption.
Collapse
Affiliation(s)
| | | | | | - Simon M. Luckman
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
9
|
Arata M, Tamura K, Aoki H, Noguchi H, Takeda A, Minato S, Yamamoto S, Kinouchi R, Yoshida K, Yamamoto Y, Kaji T, Iwasa T. The Effects of Testosterone on Hypothalamic and Serum Oxytocin Levels Are Affected by the Estrogen Milieu in Female Rats. Nutrients 2024; 16:2533. [PMID: 39125412 PMCID: PMC11314409 DOI: 10.3390/nu16152533] [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: 05/28/2024] [Revised: 07/30/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
Previous studies have suggested that the effects of androgens on body weight (BW) and appetite are affected by the estrogen milieu in females; however, the mechanism underlying these effects remains unclear. We hypothesized that androgens may affect endogenous oxytocin (OT), which is a hypothalamic anorectic factor, and that these effects of androgens may be altered by the estrogen milieu in females. To investigate this hypothesis, in the present study, we examined the effects of testosterone on peripheral and central OT levels in ovariectomized female rats that did or did not receive estradiol supplementation. Ovariectomized female rats were randomly divided into non-estradiol-supplemented or estradiol-supplemented groups, and half of the rats in each group were concurrently supplemented with testosterone (i.e., rats were divided into four groups, n = 7 per each group). We also measured peripheral and central OT receptor (OTR) gene expression levels. As a result, we found that testosterone increased serum and hypothalamic OT levels and OT receptor mRNA levels in non-estradiol-supplemented rats, whereas it had no effects on these factors in estradiol-supplemented rats. In addition, testosterone reduced food intake, BW gain, and fat weight in non-estradiol-supplemented rats, whereas it did not have any effects on BW, appetite, or fat weight in estradiol-supplemented rats. These findings indicate that the effects of androgens on OT may be affected by the estrogen milieu, and elevated OT levels may be related to the blunting of appetite and prevention of obesity under estrogen-deficient conditions.
Collapse
Affiliation(s)
- Moeka Arata
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan; (M.A.)
| | - Kou Tamura
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan; (M.A.)
| | - Hidenori Aoki
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan; (M.A.)
| | - Hiroki Noguchi
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan; (M.A.)
| | - Asuka Takeda
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan; (M.A.)
| | - Saki Minato
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan; (M.A.)
| | - Shota Yamamoto
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan; (M.A.)
- Department of Renal and Genitourinary Surgery, Graduate School of Medicine, Hokkaido University, Sapporo 060-0808, Japan
| | - Riyo Kinouchi
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan; (M.A.)
| | - Kanako Yoshida
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan; (M.A.)
| | - Yuri Yamamoto
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan; (M.A.)
| | - Takashi Kaji
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan; (M.A.)
| | - Takeshi Iwasa
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan; (M.A.)
| |
Collapse
|
10
|
Lawson EA. Understanding oxytocin in human physiology and pathophysiology: A path towards therapeutics. COMPREHENSIVE PSYCHONEUROENDOCRINOLOGY 2024; 19:100242. [PMID: 38974962 PMCID: PMC11225698 DOI: 10.1016/j.cpnec.2024.100242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 05/06/2024] [Accepted: 05/27/2024] [Indexed: 07/09/2024] Open
Abstract
•Oxytocin is a multifaceted hypothalamic-pituitary hormone involved in energy homeostasis, mental health, and bone metabolism.•Oxytocin deficiency in energy deficit states and in hypopituitarism is associated with worse mental health and bone health.•Oxytocin modulates appetitive neurocircuitry, improves impulse control, and reduces food intake in humans.•Defining the oxytocin system in human physiology and pathophysiology could lead to novel therapeutic strategies.
Collapse
Affiliation(s)
- Elizabeth A. Lawson
- Neuroendocrine Unit, Massachusetts General Hospital, Department of Medicine, Harvard Medical School, 50 Staniford Street, Suite 750B, Boston, MA, 02114, USA
| |
Collapse
|
11
|
Edwards MM, Nguyen HK, Dodson AD, Herbertson AJ, Wolden-Hanson T, Wietecha TA, Honeycutt MK, Slattery JD, O’Brien KD, Graham JL, Havel PJ, Mundinger TO, Sikkema CL, Peskind ER, Ryu V, Taborsky GJ, Blevins JE. Sympathetic innervation of interscapular brown adipose tissue is not a predominant mediator of oxytocin-elicited reductions of body weight and adiposity in male diet-induced obese mice. Front Endocrinol (Lausanne) 2024; 15:1440070. [PMID: 39145314 PMCID: PMC11321955 DOI: 10.3389/fendo.2024.1440070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 06/28/2024] [Indexed: 08/16/2024] Open
Abstract
Previous studies indicate that CNS administration of oxytocin (OT) reduces body weight in high fat diet-induced obese (DIO) rodents by reducing food intake and increasing energy expenditure (EE). We recently demonstrated that hindbrain (fourth ventricular [4V]) administration of OT elicits weight loss and elevates interscapular brown adipose tissue temperature (TIBAT, a surrogate measure of increased EE) in DIO mice. What remains unclear is whether OT-elicited weight loss requires increased sympathetic nervous system (SNS) outflow to IBAT. We hypothesized that OT-induced stimulation of SNS outflow to IBAT contributes to its ability to activate BAT and elicit weight loss in DIO mice. To test this hypothesis, we determined the effect of disrupting SNS activation of IBAT on the ability of 4V OT administration to increase TIBAT and elicit weight loss in DIO mice. We first determined whether bilateral surgical SNS denervation to IBAT was successful as noted by ≥ 60% reduction in IBAT norepinephrine (NE) content in DIO mice. NE content was selectively reduced in IBAT at 1-, 6- and 7-weeks post-denervation by 95.9 ± 2.0, 77.4 ± 12.7 and 93.6 ± 4.6% (P<0.05), respectively and was unchanged in inguinal white adipose tissue, pancreas or liver. We subsequently measured the effects of acute 4V OT (1, 5 µg ≈ 0.99, 4.96 nmol) on TIBAT in DIO mice following sham or bilateral surgical SNS denervation to IBAT. We found that the high dose of 4V OT (5 µg ≈ 4.96 nmol) elevated TIBAT similarly in sham mice as in denervated mice. We subsequently measured the effects of chronic 4V OT (16 nmol/day over 29 days) or vehicle infusions on body weight, adiposity and food intake in DIO mice following sham or bilateral surgical denervation of IBAT. Chronic 4V OT reduced body weight by 5.7 ± 2.23% and 6.6 ± 1.4% in sham and denervated mice (P<0.05), respectively, and this effect was similar between groups (P=NS). OT produced corresponding reductions in whole body fat mass (P<0.05). Together, these findings support the hypothesis that sympathetic innervation of IBAT is not necessary for OT-elicited increases in BAT thermogenesis and reductions of body weight and adiposity in male DIO mice.
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
| | - 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
| | - Tomasz A. Wietecha
- Division of Cardiology, 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
| | - 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
- Division of Cardiology, 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
| | - James L. Graham
- Department of Nutrition, University of California, Davis, Davis, CA, 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
| | - Thomas O. Mundinger
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States
| | - 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, United States
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, United States
| | - 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, United States
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, United States
| | - Vitaly Ryu
- Department of Medicine and Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - 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, United States
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA, 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
|
12
|
Van Drunen R, Dai Y, Wei H, Fekry B, Noori S, Shivshankar S, Bravo R, Zhao Z, Yoo SH, Justice N, Wu JQ, Tong Q, Eckel-Mahan K. Cell-specific regulation of the circadian clock by BMAL1 in the paraventricular nucleus: Implications for regulation of systemic biological rhythms. Cell Rep 2024; 43:114380. [PMID: 38935503 PMCID: PMC11446153 DOI: 10.1016/j.celrep.2024.114380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 03/28/2024] [Accepted: 06/03/2024] [Indexed: 06/29/2024] Open
Abstract
Circadian rhythms are internal biological rhythms driving temporal tissue-specific, metabolic programs. Loss of the circadian transcription factor BMAL1 in the paraventricular nucleus (PVN) of the hypothalamus reveals its importance in metabolic rhythms, but its functions in individual PVN cells are poorly understood. Here, loss of BMAL1 in the PVN results in arrhythmicity of processes controlling energy balance and alters peripheral diurnal gene expression. BMAL1 chromatin immunoprecipitation sequencing (ChIP-seq) and single-nucleus RNA sequencing (snRNA-seq) reveal its temporal regulation of target genes, including oxytocin (OXT), and restoring circulating OXT peaks in BMAL1-PVN knockout (KO) mice rescues absent activity rhythms. While glutamatergic neurons undergo day/night changes in expression of genes involved in cell morphogenesis, astrocytes and oligodendrocytes show gene expression changes in cytoskeletal organization and oxidative phosphorylation. Collectively, our findings show diurnal gene regulation in neuronal and non-neuronal PVN cells and that BMAL1 contributes to diurnal OXT secretion, which is important for systemic diurnal rhythms.
Collapse
Affiliation(s)
- Rachel Van Drunen
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yulin Dai
- Center for Precision Health, McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Haichao Wei
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Baharan Fekry
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Sina Noori
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Samay Shivshankar
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Rafael Bravo
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Zhongming Zhao
- Center for Precision Health, McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Seung-Hee Yoo
- MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Biochemistry and Cell Biology, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Nicholas Justice
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jia Qian Wu
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Qingchun Tong
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Kristin Eckel-Mahan
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
13
|
Önal D, Korkmaz H, Önal G, Pehlivanoğlu B. Body weight modulates the impact of oxytocin on chronic cold-immobilization stress response. Peptides 2024; 177:171202. [PMID: 38555975 DOI: 10.1016/j.peptides.2024.171202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/21/2024] [Accepted: 03/27/2024] [Indexed: 04/02/2024]
Abstract
By activating the stress system, stress modulates various physiological parameters including food intake, energy consumption, and, consequently, body weight. The role of oxytocin in the regulation of stress and obesity cannot be disregarded. Based on these findings, we aimed to investigate the effect of intranasal oxytocin on stress response in high-fat-diet (HFD)--fed and control-diet-fed rats exposed to chronic stress. Cold-immobilization stress was applied for 5 consecutive days to male Sprague-Dawley rats fed either with a control diet (n=20) or HFD (n=20) for 6 weeks. Half of the animals in each group received oxytocin. Stress response was evaluated via plasma and salivary cortisol levels as well as elevated plus maze scores. Prefrontal cortex and hypothalamic oxytocin receptor (OxtR) expression levels were identified using western blot analysis. The results showed higher stress response in HFD-fed animals than in control animals both under basal and post-stress conditions. Oxytocin application had a prominent anxiolytic effect in the control group but an insignificant effect in the HFD group. While OxtR expression levels in the prefrontal cortex did not vary according to the body weight and oxytocin application, OxtR levels in the hypothalamus were higher in the HFD- and/or oxytocin-treated animals. Our results indicated that the peripheral and central effects of oxytocin vary with body weight. Moreover, obesity masks the anxiolytic effects of oxytocin, probably by reinforcing the stress condition via central OxtRs. In conclusion, elucidating the mechanisms underlying the central effect of oxytocin is important to cope with stress and obesity.
Collapse
Affiliation(s)
- Deniz Önal
- Faculty of Medicine, Department of Physiology, Balıkesir University, Balıkesir, Türkiye.
| | - Hilal Korkmaz
- Faculty of Medicine, Department of Physiology, Hacettepe University, Ankara, Türkiye
| | - Gizem Önal
- Oxford Parkinson's Disease Centre and Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Bilge Pehlivanoğlu
- Faculty of Medicine, Department of Physiology, Hacettepe University, Ankara, Türkiye
| |
Collapse
|
14
|
Edwards MM, Nguyen HK, Dodson AD, Herbertson AJ, Wolden-Hanson T, Wietecha T, Honeycutt MK, Slattery JD, O'Brien KD, Graham JL, Havel PJ, Mundinger TO, Sikkema C, Peskind ER, Ryu V, Taborsky GJ, Blevins JE. Sympathetic innervation of interscapular brown adipose tissue is not a predominant mediator of oxytocin-elicited reductions of body weight and adiposity in male diet-induced obese mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596425. [PMID: 38854021 PMCID: PMC11160755 DOI: 10.1101/2024.05.29.596425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Previous studies indicate that CNS administration of oxytocin (OT) reduces body weight in high fat diet-induced obese (DIO) rodents by reducing food intake and increasing energy expenditure (EE). We recently demonstrated that hindbrain (fourth ventricular [4V]) administration of OT elicits weight loss and elevates interscapular brown adipose tissue temperature (T IBAT , a surrogate measure of increased EE) in DIO mice. What remains unclear is whether OT-elicited weight loss requires increased sympathetic nervous system (SNS) outflow to IBAT. We hypothesized that OT-induced stimulation of SNS outflow to IBAT contributes to its ability to activate BAT and elicit weight loss in DIO mice. To test this hypothesis, we determined the effect of disrupting SNS activation of IBAT on the ability of 4V OT administration to increase T IBAT and elicit weight loss in DIO mice. We first determined whether bilateral surgical SNS denervation to IBAT was successful as noted by ≥ 60% reduction in IBAT norepinephrine (NE) content in DIO mice. NE content was selectively reduced in IBAT at 1-, 6- and 7-weeks post-denervation by 95.9±2.0, 77.4±12.7 and 93.6±4.6% ( P <0.05), respectively and was unchanged in inguinal white adipose tissue, pancreas or liver. We subsequently measured the effects of acute 4V OT (1, 5 µg ≈ 0.99, 4.96 nmol) on T IBAT in DIO mice following sham or bilateral surgical SNS denervation to IBAT. We found that the high dose of 4V OT (5 µg ≈ 4.96 nmol) elevated T IBAT similarly in sham mice as in denervated mice. We subsequently measured the effects of chronic 4V OT (16 nmol/day over 29 days) or vehicle infusions on body weight, adiposity and food intake in DIO mice following sham or bilateral surgical denervation of IBAT. Chronic 4V OT reduced body weight by 5.7±2.23% and 6.6±1.4% in sham and denervated mice ( P <0.05), respectively, and this effect was similar between groups ( P =NS). OT produced corresponding reductions in whole body fat mass ( P <0.05). Together, these findings support the hypothesis that sympathetic innervation of IBAT is not necessary for OT-elicited increases in BAT thermogenesis and reductions of body weight and adiposity in male DIO mice.
Collapse
|
15
|
Maejima Y, Yokota S, Yamachi M, Misaka S, Ono T, Oizumi H, Mizuno K, Hidema S, Nishimori K, Aoyama M, de Wet H, Shimomura K. Traditional Japanese medicine Kamikihito ameliorates sucrose preference, chronic inflammation and obesity induced by a high fat diet in middle-aged mice. Front Endocrinol (Lausanne) 2024; 15:1387964. [PMID: 38742193 PMCID: PMC11089234 DOI: 10.3389/fendo.2024.1387964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 04/09/2024] [Indexed: 05/16/2024] Open
Abstract
The high prevalence of obesity has become a pressing global public health problem and there exists a strong association between increased BMI and mortality at a BMI of 25 kg/m2 or higher. The prevalence of obesity is higher among middle-aged adults than among younger groups and the combination of aging and obesity exacerbate systemic inflammation. Increased inflammatory cytokines such as interleukin 6 and tumor necrosis factor alpha (TNFα) are hallmarks of obesity, and promote the secretion of hepatic C-reactive protein (CRP) which further induces systematic inflammation. The neuropeptide oxytocin has been shown to have anti-obesity and anti-inflammation effects, and also suppress sweet-tasting carbohydrate consumption in mammals. Previously, we have shown that the Japanese herbal medicine Kamikihito (KKT), which is used to treat neuropsychological stress disorders in Japan, functions as an oxytocin receptors agonist. In the present study, we further investigated the effect of KKT on body weight (BW), food intake, inflammation, and sweet preferences in middle-aged obese mice. KKT oral administration for 12 days decreased the expression of pro-inflammatory cytokines in the liver, and the plasma CRP and TNFα levels in obese mice. The effect of KKT administration was found to be different between male and female mice. In the absence of sucrose, KKT administration decreased food intake only in male mice. However, while having access to a 30% sucrose solution, both BW and food intake was decreased by KKT administration in male and female mice; but sucrose intake was decreased in female mice alone. In addition, KKT administration decreased sucrose intake in oxytocin deficient lean mice, but not in the WT lean mice. The present study demonstrates that KKT ameliorates chronic inflammation, which is strongly associated with aging and obesity, and decreases food intake in male mice as well as sucrose intake in female mice; in an oxytocin receptor dependent manner.
Collapse
Affiliation(s)
- Yuko Maejima
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
- Department of Obesity and Inflammation research, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Shoko Yokota
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Megumi Yamachi
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Shingen Misaka
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Tomoyuki Ono
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Hiroaki Oizumi
- Tsumura Kampo Research Laboratories, Kampo Research and Development Division, Tsumura & Co., Ibaraki, Japan
| | - Keita Mizuno
- Tsumura Kampo Research Laboratories, Kampo Research and Development Division, Tsumura & Co., Ibaraki, Japan
| | - Shizu Hidema
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Katsuhiko Nishimori
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Masato Aoyama
- Department of Animal Science, Faculty of Agriculture, Utsunomiya University, Utsunomiya, Japan
| | - Heidi de Wet
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Kenju Shimomura
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
- Department of Obesity and Inflammation research, Fukushima Medical University School of Medicine, Fukushima, Japan
| |
Collapse
|
16
|
Maejima Y, Yokota S, Hidema S, Nishimori K, de Wet H, Shimomura K. Systemic Co-Administration of Low-Dose Oxytocin and Glucagon-Like Peptide 1 Additively Decreases Food Intake and Body Weight. Neuroendocrinology 2024; 114:639-657. [PMID: 38599201 DOI: 10.1159/000538792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 04/08/2024] [Indexed: 04/12/2024]
Abstract
INTRODUCTION GLP-1 receptor agonists are the number one drug prescribed for the treatment of obesity and type 2 diabetes. These drugs are not, however, without side effects, and in an effort to maximize therapeutic effect while minimizing adverse effects, gut hormone co-agonists received considerable attention as new drug targets in the fight against obesity. Numerous previous reports identified the neuropeptide oxytocin (OXT) as a promising anti-obesity drug. The aims of this study were to evaluate OXT as a possible co-agonist for GLP-1 and examine the effects of its co-administration on food intake (FI) and body weight (BW) in mice. METHODS FI and c-Fos levels were measured in the feeding centers of the brain in response to an intraperitoneal injection of saline, OXT, GLP-1, or OXT/GLP-1. The action potential frequency and cytosolic Ca2+ ([Ca2+]i) in response to OXT, GLP-1, or OXT/GLP-1 were measured in ex vivo paraventricular nucleus (PVN) neuronal cultures. Finally, FI and BW changes were compared in diet-induced obese mice treated with saline, OXT, GLP-1, or OXT/GLP-1 for 13 days. RESULTS Single injection of OXT/GLP-1 additively decreased FI and increased c-Fos expression specifically in the PVN and supraoptic nucleus. Seventy percent of GLP-1 receptor-positive neurons in the PVN also expressed OXT receptors, and OXT/GLP-1 co-administration dramatically increased firing and [Ca2+]i in the PVN OXT neurons. The chronic OXT/GLP-1 co-administration decreased BW without changing FI. CONCLUSION Chronic OXT/GLP-1 co-administration decreases BW, possibly via the activation of PVN OXT neurons. OXT might be a promising candidate as an incretin co-agonist in obesity treatment.
Collapse
Affiliation(s)
- Yuko Maejima
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
- Departments of Obesity and Inflammation Research, Fukushima Medical University School of Medicine, Fukushima, Japan
- Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Oxford, UK
| | - Shoko Yokota
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Shizu Hidema
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Katsuhiko Nishimori
- Departments of Obesity and Inflammation Research, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Heidi de Wet
- Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Oxford, UK
| | - Kenju Shimomura
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
- Departments of Obesity and Inflammation Research, Fukushima Medical University School of Medicine, Fukushima, Japan
| |
Collapse
|
17
|
Masaki R, Yamamoto Y, Tamura K, Aoki H, Noguchi H, Takeda A, Minato S, Tanano R, Yamanaka E, Maeda T, Sugimoto T, Sasada H, Inui H, Kagawa T, Yoshida A, Mineda A, Kinouchi R, Yoshida K, Kaji T, Iwasa T. Comparison of endogenous hypothalamic and serum OT levels between young and middle-aged perimenopausal female rats. THE JOURNAL OF MEDICAL INVESTIGATION 2024; 71:246-250. [PMID: 39462559 DOI: 10.2152/jmi.71.246] [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] [Indexed: 10/29/2024]
Abstract
Oxytocin (OT) regulates food intake and body weight, particularly in obese individuals. Decreases in the effects of OT have recently been implicated in metabolic disturbances, and the administration of estradiol (E2) increased serum OT levels. Although weight gain is frequently observed in perimenopausal women, endogenous OT levels remain unclear. Therefore, we herein compared endogenous levels of hypothalamic and serum OT between young and middle-aged perimenopausal female rats and examined the relationship between serum estrogen and leptin levels. Body weight and visceral and subcutaneous fat weights were higher in middle-aged rats. Although no significant differences were observed in serum OT and E2 levels, serum leptin levels and hypothalamic mRNA levels of OT and the OT receptor (OTR) were significantly higher in middle-aged rats than in young rats. Serum OT levels did not correlate with hypothalamic OT mRNA levels or serum E2 levels. E2 maintains serum OT levels in perimenopausal rats, and other factors may elevate hypothalamic OT/OTR mRNA levels. Increases in body and fat weights in perimenopausal rats may be attributed to factors other than OT. Therefore, the administration of OT alone may not be sufficient to prevent metabolic disorders induced by the perimenopausal status. J. Med. Invest. 71 : 246-250, August, 2024.
Collapse
Affiliation(s)
- Rie Masaki
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Yuri Yamamoto
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Kou Tamura
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Hidenori Aoki
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Hiroki Noguchi
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Asuka Takeda
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Saki Minato
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Risa Tanano
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Erika Yamanaka
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Takaaki Maeda
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Tatsuo Sugimoto
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Hikari Sasada
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Hiroaki Inui
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Tomohiro Kagawa
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Atsuko Yoshida
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Ayuka Mineda
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Riyo Kinouchi
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Kanako Yoshida
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Takashi Kaji
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Takeshi Iwasa
- Department of Obstetrics and Gynecology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| |
Collapse
|
18
|
Civelek E, Ozturk Civelek D, Akyel YK, Kaleli Durman D, Okyar A. Circadian Dysfunction in Adipose Tissue: Chronotherapy in Metabolic Diseases. BIOLOGY 2023; 12:1077. [PMID: 37626963 PMCID: PMC10452180 DOI: 10.3390/biology12081077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023]
Abstract
Essential for survival and reproduction, the circadian timing system (CTS) regulates adaptation to cyclical changes such as the light/dark cycle, temperature change, and food availability. The regulation of energy homeostasis possesses rhythmic properties that correspond to constantly fluctuating needs for energy production and consumption. Adipose tissue is mainly responsible for energy storage and, thus, operates as one of the principal components of energy homeostasis regulation. In accordance with its roles in energy homeostasis, alterations in adipose tissue's physiological processes are associated with numerous pathologies, such as obesity and type 2 diabetes. These alterations also include changes in circadian rhythm. In the current review, we aim to summarize the current knowledge regarding the circadian rhythmicity of adipogenesis, lipolysis, adipokine secretion, browning, and non-shivering thermogenesis in adipose tissue and to evaluate possible links between those alterations and metabolic diseases. Based on this evaluation, potential therapeutic approaches, as well as clock genes as potential therapeutic targets, are also discussed in the context of chronotherapy.
Collapse
Affiliation(s)
- Erkan Civelek
- Department of Pharmacology, Faculty of Pharmacy, Istanbul University, 34116 Istanbul, Turkey; (E.C.); (D.K.D.)
| | - Dilek Ozturk Civelek
- Department of Pharmacology, Faculty of Pharmacy, Bezmialem Vakıf University, 34093 Istanbul, Turkey;
| | - Yasemin Kubra Akyel
- Department of Medical Pharmacology, School of Medicine, Istanbul Medipol University, 34815 Istanbul, Turkey;
| | - Deniz Kaleli Durman
- Department of Pharmacology, Faculty of Pharmacy, Istanbul University, 34116 Istanbul, Turkey; (E.C.); (D.K.D.)
| | - Alper Okyar
- Department of Pharmacology, Faculty of Pharmacy, Istanbul University, 34116 Istanbul, Turkey; (E.C.); (D.K.D.)
| |
Collapse
|
19
|
Zhang Y, Zhu Y, Wang J, Jin L, Guo M, Chen L, Zhang L, Li Y, Wan B, Zhang R, Jia W, Hu C. Neuregulin4 Acts on Hypothalamic ErBb4 to Excite Oxytocin Neurons and Preserve Metabolic Homeostasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204824. [PMID: 37060105 PMCID: PMC10238187 DOI: 10.1002/advs.202204824] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 03/10/2023] [Indexed: 06/04/2023]
Abstract
Neuregulin 4 (Nrg4) is an adipose tissue-enriched secreted factor that modulates glucose and lipid metabolism. Nrg4 is closely associated with obesity and preserves diet-induced metabolic disorders. However, the specific mechanisms via which Nrg4 regulates metabolic homeostasis remain incompletely understood. Here, this work finds that the Nrg4 receptor, ErbB4, is highly expressed in the hypothalamus, and the phosphorylation of hypothalamic ErbB4 is reduced in diet-induced obesity (DIO) mice. Peripheral Nrg4 can act on ErbB4 via blood circulation and excite neurons in the paraventricular nucleus of hypothalamus (PVN). Central administration of recombinant Nrg4 protein (rNrg4) reduces obesity and related metabolic disorders by influencing energy expenditure and intake. Overexpression of ErbB4 in the PVN protects against obesity, whereas its knock down in oxytocin (Oxt) neuron accelerates obesity. Furthermore, Nrg4-ErbB4 signaling excites Oxt release, and ablation of Oxt neuron considerably attenuates the effect of Nrg4 on energy balance. These data suggest that the hypothalamus is a key target of Nrg4, which partially explains the multifaceted roles of Nrg4 in metabolism.
Collapse
Affiliation(s)
- Yi Zhang
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Yangyang Zhu
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
- Institute for Metabolic DiseaseFengxian Central Hospital Affiliated to Southern Medical UniversityShanghai201449P. R. China
| | - Jinghui Wang
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
- Department of EndocrinologyAffiliated Hospital of Nantong UniversityNantong226001P. R. China
| | - Li Jin
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Mingwei Guo
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241P. R. China
| | - Liwei Chen
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Lina Zhang
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Yangyang Li
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Baocheng Wan
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Rong Zhang
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Weiping Jia
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Cheng Hu
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Centre for DiabetesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
- Institute for Metabolic DiseaseFengxian Central Hospital Affiliated to Southern Medical UniversityShanghai201449P. R. China
| |
Collapse
|
20
|
Ragozzino FJ, Peterson B, Karatsoreos IN, Peters JH. Circadian regulation of glutamate release pathways shapes synaptic throughput in the brainstem nucleus of the solitary tract (NTS). J Physiol 2023; 601:1881-1896. [PMID: 36975145 PMCID: PMC10192157 DOI: 10.1113/jp284370] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/06/2023] [Indexed: 03/29/2023] Open
Abstract
Circadian regulation of autonomic reflex pathways pairs physiological function with the daily light cycle. The brainstem nucleus of the solitary tract (NTS) is a key candidate for rhythmic control of the autonomic nervous system. Here we investigated circadian regulation of NTS neurotransmission and synaptic throughput using patch-clamp electrophysiology in brainstem slices from mice. We found that spontaneous quantal glutamate release onto NTS neurons showed strong circadian rhythmicity, with the highest rate of release during the light phase and the lowest in the dark, that were sufficient to drive day/night differences in constitutive postsynaptic action potential firing. In contrast, afferent evoked action potential throughput was enhanced during the dark and diminished in the light. Afferent-driven synchronous release pathways showed a similar decrease in release probability that did not explain the enhanced synaptic throughput during the night. However, analysis of postsynaptic membrane properties revealed diurnal changes in conductance, which, when coupled with the circadian changes in glutamate release pathways, tuned synaptic throughput between the light and dark phases. These coordinated pre-/postsynaptic changes encode nuanced control over synaptic performance and pair NTS action potential firing and vagal throughput with time of day. KEY POINTS: Vagal afferent neurons relay information from peripheral organs to the brainstem nucleus of the solitary tract (NTS) to initiate autonomic reflex pathways as well as providing important controls of food intake, digestive function and energy balance. Vagally mediated reflexes and behaviours are under strong circadian regulation. Diurnal fluctuations in presynaptic vesicle release pathways and postsynaptic membrane conductances provide nuanced control over NTS action potential firing and vagal synaptic throughput. Coordinated pre-/postsynaptic changes represent a fundamental mechanism mediating daily changes in vagal afferent signalling and autonomic function.
Collapse
Affiliation(s)
- Forrest J. Ragozzino
- Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - BreeAnne Peterson
- Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Ilia N. Karatsoreos
- Department of Psychological and Brain Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - James H. Peters
- Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| |
Collapse
|
21
|
Zayan U, Caccialupi Da Prato L, Muscatelli F, Matarazzo V. Modulation of the thermosensory system by oxytocin. Front Mol Neurosci 2023; 15:1075305. [PMID: 36698777 PMCID: PMC9868264 DOI: 10.3389/fnmol.2022.1075305] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/06/2022] [Indexed: 01/11/2023] Open
Abstract
Oxytocin (OT) is a neurohormone involved early in neurodevelopment and is implicated in multiple functions, including sensory modulation. Evidence of such modulation has been observed for different sensory modalities in both healthy and pathological conditions. This review summarizes the pleiotropic modulation that OT can exercise on an often overlooked sensory system: thermosensation. This system allows us to sense temperature variations and compensate for the variation to maintain a stable core body temperature. Oxytocin modulates autonomic and behavioral mechanisms underlying thermoregulation at both central and peripheral levels. Hyposensitivity or hypersensitivity for different sensory modalities, including thermosensitivity, is a common feature in autism spectrum disorder (ASD), recapitulated in several ASD mouse models. These sensory dysregulations occur early in post-natal development and are correlated with dysregulation of the oxytocinergic system. In this study, we discussed the potential link between thermosensory atypia and the dysregulation of the oxytocinergic system in ASD.
Collapse
|
22
|
Changes in Serum Oxytocin Levels under Physiological and Supraphysiological Gonadal Steroid Hormone Conditions in Women of Reproductive Age: A Preliminary Study. Nutrients 2022; 14:nu14245350. [PMID: 36558508 PMCID: PMC9787714 DOI: 10.3390/nu14245350] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Oxytocin (OT) affects many behavioral, psychological, and physiological functions, including appetite and body weight regulation. Central and peripheral OT levels are markedly affected by gonadal steroids, especially estrogen, and the anorectic effects of estrogen are partially mediated by OT in rodents. In this study, the relationship between the estrogen milieu and serum OT levels was evaluated in women of reproductive age under physiological (n = 9) and supraphysiological estrogenic conditions (n = 7). Consequently, it was found that serum OT levels were increased in physiological (the ovulatory phase) and supraphysiological (on the day of the human chorionic gonadotropin trigger in an ovarian stimulation cycle) estrogenic conditions, and that serum OT levels were positively correlated with serum estradiol levels. On the other hand, serum OT levels were negatively correlated with serum progesterone levels, and there was no correlation between serum and follicular OT levels. These results suggest that OT levels may be positively and negatively regulated by estrogen and progesterone, respectively, in humans. However, the physiological roles of these actions of gonadal steroids on OT remain unclear.
Collapse
|
23
|
Fukushima A, Kataoka N, Nakamura K. An oxytocinergic neural pathway that stimulates thermogenic and cardiac sympathetic outflow. Cell Rep 2022; 40:111380. [PMID: 36130511 DOI: 10.1016/j.celrep.2022.111380] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 03/29/2022] [Accepted: 08/28/2022] [Indexed: 11/17/2022] Open
Abstract
Oxytocin alters autonomic functions besides social behaviors. However, the central neuronal links between hypothalamic oxytocinergic neurons and the autonomic nervous system remain unclear. Here we show that oxytocinergic neurons in the rat paraventricular hypothalamic nucleus (PVH), a pivotal site for energy homeostasis, innervate sympathetic premotor neurons in the rostral medullary raphe region (rMR) to stimulate brown adipose tissue (BAT) thermogenesis and cardiovascular functions. Oxytocin receptor stimulation in the rMR evokes BAT thermogenesis and tachycardia. In vivo optogenetic stimulation of the PVH→rMR long-range oxytocinergic pathway, using a virus-mediated system for amplified gene expression in oxytocinergic neurons, not only elicits BAT thermogenic and cardiac responses but also potentiates sympathetic responses evoked by glutamatergic transmission in the rMR. The PVH→rMR oxytocinergic pathway connects the hypothalamic circuit for energy homeostasis to thermogenic and cardiac sympathetic outflow, and, therefore, its defects may cause obesity and impaired thermoregulation, as seen in Prader-Willi syndrome.
Collapse
Affiliation(s)
- Akihiro Fukushima
- Department of Integrative Physiology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Naoya Kataoka
- Department of Integrative Physiology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Nagoya University Institute for Advanced Research, Nagoya 464-8601, Japan
| | - Kazuhiro Nakamura
- Department of Integrative Physiology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| |
Collapse
|
24
|
Olszewski PK, Noble EE, Paiva L, Ueta Y, Blevins JE. Oxytocin as a potential pharmacological tool to combat obesity. J Neuroendocrinol 2022; 34:e13106. [PMID: 35192207 PMCID: PMC9372234 DOI: 10.1111/jne.13106] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/06/2022] [Accepted: 02/08/2022] [Indexed: 11/30/2022]
Abstract
The neuropeptide oxytocin (OT) has emerged as an important anorexigen in the regulation of food intake and energy balance. It has been shown that the release of OT and activation of hypothalamic OT neurons coincide with food ingestion. Its effects on feeding have largely been attributed to limiting meal size through interactions in key regulatory brain regions governing the homeostatic control of food intake such as the hypothalamus and hindbrain in addition to key feeding reward areas such as the nucleus accumbens and ventral tegmental area. Furthermore, the magnitude of an anorexigenic response to OT and feeding-related activation of the brain OT circuit are modified by the composition and flavor of a diet, as well as by a social context in which a meal is consumed. OT is particularly effective in reducing consumption of carbohydrates and sweet tastants. Pharmacologic, genetic, and pair-feeding studies indicate that OT-elicited weight loss cannot be fully explained by reductions of food intake and that the overall impact of OT on energy balance is also partly a result of OT-elicited changes in lipolysis, energy expenditure, and glucose regulation. Peripheral administration of OT mimics many of its effects when it is given into the central nervous system, raising the questions of whether and to what extent circulating OT acts through peripheral OT receptors to regulate energy balance. Although OT has been found to elicit weight loss in female mice, recent studies have indicated that sex and estrous cycle may impact oxytocinergic modulation of food intake. Despite the overall promising basic research data, attempts to use OT in the clinical setting to combat obesity and overeating have generated somewhat mixed results. The focus of this mini-review is to briefly summarize the role of OT in feeding and metabolism, address gaps and inconsistencies in our knowledge, and discuss some of the limitations to the potential use of chronic OT that should help guide future research on OT as a tailor-made anti-obesity therapeutic.
Collapse
Affiliation(s)
- Pawel K Olszewski
- Faculty of Science and Engineering, University of Waikato, Waikato, New Zealand
- Department of Food Science and Nutrition, College of Food, Agricultural and Natural Resource Sciences, University of Minnesota, St Paul, Minnesota, USA
- Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - Emily E Noble
- Department of Nutritional Sciences, University of Georgia, Athens, Georgia, USA
| | - Luis Paiva
- Instituto de Ciencia Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Yoichi Ueta
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| | - James E Blevins
- Department of Veterans Affairs Medical Center, VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Seattle, Washington, USA
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
25
|
Changes in Endogenous Oxytocin Levels and the Effects of Exogenous Oxytocin Administration on Body Weight Changes and Food Intake in Polycystic Ovary Syndrome Model Rats. Int J Mol Sci 2022; 23:ijms23158207. [PMID: 35897783 PMCID: PMC9330807 DOI: 10.3390/ijms23158207] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/24/2022] [Accepted: 07/24/2022] [Indexed: 12/10/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is frequently seen in females of reproductive age and is associated with metabolic disorders that are exacerbated by obesity. Although body weight reduction programs via diet and lifestyle changes are recommended for modifying reproductive and metabolic phenotypes, the drop-out rate is high. Thus, an efficacious, safe, and continuable treatment method is needed. Recent studies have shown that oxytocin (OT) reduces body weight gain and food intake, and promotes lipolysis in some mammals, including humans (especially obese individuals), without any adverse effects. In the present study, we evaluated the changes in endogenous OT levels, and the effects of acute and chronic OT administration on body weight changes, food intake, and fat mass using novel dihydrotestosterone-induced PCOS model rats. We found that the serum OT level was lower in PCOS model rats than in control rats, whereas the hypothalamic OT mRNA expression level did not differ between them. Acute intraperitoneal administration of OT during the dark phase reduced the body weight gain and food intake in PCOS model rats, but these effects were not observed in control rats. In contrast, chronic administration of OT decreased the food intake in both the PCOS model rats and control rats. These findings indicate that OT may be a candidate medicine that is efficacious, safe, and continuable for treating obese PCOS patients.
Collapse
|
26
|
Florea T, Palimariciuc M, Cristofor AC, Dobrin I, Chiriță R, Bîrsan M, Dobrin RP, Pădurariu M. Oxytocin: Narrative Expert Review of Current Perspectives on the Relationship with Other Neurotransmitters and the Impact on the Main Psychiatric Disorders. Medicina (B Aires) 2022; 58:medicina58070923. [PMID: 35888641 PMCID: PMC9318841 DOI: 10.3390/medicina58070923] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/04/2022] [Accepted: 07/08/2022] [Indexed: 12/23/2022] Open
Abstract
Is a cyclic neuropeptide produced primarily in the hypothalamus and plays an important neuromodulatory role for other neurotransmitter systems, with an impact on behavior, response to danger, stress, and complex social interactions, such as pair bonding and child care. This narrative expert review examines the literature on oxytocin as a brain hormone. We focused on oxytocin structure, distribution, genetics, and the oxytocin receptor system, as well as the relationship of oxytocin with other neurotransmitters and the resulting impacts on the main psychiatric disorders. Oxytocin levels have been correlated over time with mental illness, with numerous studies focusing on oxytocin and the pathophysiology of the main psychiatric disorders, such as autism, schizophrenia, personality disorders, mood, and eating disorders. We highlight the role oxytocin plays in improving symptoms such as anxiety, depression, and social behavior, as the literature suggests. Risk factors and causes for psychiatric disorders range from genetic to environmental and social factors. Oxytocin could impact the latter, being linked with other neurotransmitter systems that are responsible for integrating different situations during the development phases of individuals. Also, these systems have an important role in how the body responds to stressors or bonding with others, helping with the creation of social support groups that could speed up recovery in many situations. Oxytocin has the potential to become a key therapeutic agent for future treatment and prevention strategies concerning the main psychiatric disorders.
Collapse
Affiliation(s)
- Tudor Florea
- Department of Medicine III, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 16 Universității Street, 700115 Iasi, Romania; (T.F.); (M.P.); (A.C.C.); (I.D.); (R.C.)
| | - Matei Palimariciuc
- Department of Medicine III, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 16 Universității Street, 700115 Iasi, Romania; (T.F.); (M.P.); (A.C.C.); (I.D.); (R.C.)
- Institute of Psychiatry “Socola”, 36 Bucium Street, 700282 Iasi, Romania;
| | - Ana Caterina Cristofor
- Department of Medicine III, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 16 Universității Street, 700115 Iasi, Romania; (T.F.); (M.P.); (A.C.C.); (I.D.); (R.C.)
- Institute of Psychiatry “Socola”, 36 Bucium Street, 700282 Iasi, Romania;
| | - Irina Dobrin
- Department of Medicine III, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 16 Universității Street, 700115 Iasi, Romania; (T.F.); (M.P.); (A.C.C.); (I.D.); (R.C.)
- Institute of Psychiatry “Socola”, 36 Bucium Street, 700282 Iasi, Romania;
| | - Roxana Chiriță
- Department of Medicine III, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 16 Universității Street, 700115 Iasi, Romania; (T.F.); (M.P.); (A.C.C.); (I.D.); (R.C.)
- Institute of Psychiatry “Socola”, 36 Bucium Street, 700282 Iasi, Romania;
| | - Magdalena Bîrsan
- Department of Drug Industry and Pharmaceutical Biotechnology, Faculty of Pharmacy, Grigore T. Popa University of Medicine and Pharmacy of Iaşi, 16 Universităţii Street, 700115 Iaşi, Romania;
| | - Romeo Petru Dobrin
- Department of Medicine III, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 16 Universității Street, 700115 Iasi, Romania; (T.F.); (M.P.); (A.C.C.); (I.D.); (R.C.)
- Institute of Psychiatry “Socola”, 36 Bucium Street, 700282 Iasi, Romania;
- Correspondence:
| | - Manuela Pădurariu
- Institute of Psychiatry “Socola”, 36 Bucium Street, 700282 Iasi, Romania;
| |
Collapse
|
27
|
Keeling E, Lynn SA, Koh YM, Scott JA, Kendall A, Gatherer M, Page A, Cagampang FR, Lotery AJ, Ratnayaka JA. A High Fat "Western-style" Diet Induces AMD-Like Features in Wildtype Mice. Mol Nutr Food Res 2022; 66:e2100823. [PMID: 35306732 PMCID: PMC9287010 DOI: 10.1002/mnfr.202100823] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 03/01/2022] [Indexed: 12/20/2022]
Abstract
Scope The intake of a “Western‐style” diet rich in fats is linked with developing retinopathies including age‐related macular degeneration (AMD). Wildtype mice are given a high fat diet (HFD) to determine how unhealthy foods can bring about retinal degeneration. Methods and results Following weaning, female C57BL/6 mice are maintained on standard chow (7% kcal fat, n = 29) or a HFD (45% kcal fat, n = 27) for 12 months. Animals were sacrificed following electroretinography (ERG) and their eyes analyzed by histology, confocal immunofluorescence, and transmission electron microscopy. HFD mice become obese, but showed normal retinal function compared to chow‐fed controls. However, diminished β3tubulin labeling of retinal cross‐sections indicated fewer/damaged neuronal processes in the inner plexiform layer. AMD‐linked proteins clusterin and TIMP3 accumulated in the retinal pigment epithelium (RPE) and Bruch's membrane (BrM). Neutral lipids also deposited in the outer retinae of HFD mice. Ultrastructural analysis revealed disorganized photoreceptor outer segments, collapsed/misaligned RPE microvilli, vacuoles, convoluted basolateral RPE infolds and BrM changes. Basal laminar‐like deposits were also present alongside abnormal choroidal endothelial cells. Conclusions We show that prolonged exposure to an unhealthy “Western‐style” diet alone can recapitulate early‐intermediate AMD‐like features in wildtype mice, highlighting the importance of diet and nutrition in the etiology of sight‐loss.
Collapse
Affiliation(s)
- Eloise Keeling
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton, SO16 6YD, UK
| | - Savannah A Lynn
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton, SO16 6YD, UK
| | - Yen Min Koh
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton, SO16 6YD, UK
| | - Jenny A Scott
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton, SO16 6YD, UK
| | - Aaron Kendall
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton, SO16 6YD, UK
| | - Maureen Gatherer
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton, SO16 6YD, UK
| | - Anton Page
- Biomedical Imaging Unit, University of Southampton, MP12, Tremona Road, Southampton, SO16 6YD, UK
| | - Felino R Cagampang
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, UK
| | - Andrew J Lotery
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton, SO16 6YD, UK.,Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton, SO16 6YD, UK
| | - J Arjuna Ratnayaka
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton, SO16 6YD, UK
| |
Collapse
|
28
|
Dunigan AI, Roseberry AG. Actions of feeding-related peptides on the mesolimbic dopamine system in regulation of natural and drug rewards. ADDICTION NEUROSCIENCE 2022; 2:100011. [PMID: 37220637 PMCID: PMC10201992 DOI: 10.1016/j.addicn.2022.100011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The mesolimbic dopamine system is the primary neural circuit mediating motivation, reinforcement, and reward-related behavior. The activity of this system and multiple behaviors controlled by it are affected by changes in feeding and body weight, such as fasting, food restriction, or the development of obesity. Multiple different peptides and hormones that have been implicated in the control of feeding and body weight interact with the mesolimbic dopamine system to regulate many different dopamine-dependent, reward-related behaviors. In this review, we summarize the effects of a selected set of feeding-related peptides and hormones acting within the ventral tegmental area and nucleus accumbens to alter feeding, as well as food, drug, and social reward.
Collapse
Affiliation(s)
- Anna I. Dunigan
- Department of Biology and Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| | - Aaron G. Roseberry
- Department of Biology and Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
29
|
Maikoo S, Wilkins A, Qulu L. The effect of oxytocin and an enriched environment on anxiety-like behaviour and corticosterone levels in a prenatally stressed febrile seizure rat model. IBRO Neurosci Rep 2022; 13:47-56. [PMID: 36590100 PMCID: PMC9795298 DOI: 10.1016/j.ibneur.2022.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 04/30/2022] [Accepted: 05/03/2022] [Indexed: 02/01/2023] Open
Abstract
Background Febrile seizures (FS) are a neurological abnormality which occur following a fever that has resulted from a systemic infection and are characterised by convulsions. These convulsions occur due to abnormally increased signalling of interleukin-1 beta, resulting in increased neuronal hyper-excitability. Furthermore, exposure to prenatal stress has been shown to exacerbate seizure duration, elicit anxiety-like behaviour and corticosterone levels. Oxytocin is a neuropeptide with anxiolytic, social bonding, and stress regulation effects. Therefore, the aim of the study was to assess whether oxytocin can attenuate the anxiety-like behaviour and increased corticosterone in rat offspring exposed to prenatal stress and FS. Method Sprague Dawley rats were mated. On GND14, prenatal stress was induced on pregnant dams for 1 hr/7 days. On PND 14, rat pups were injected with lipopolysaccharide (LPS, 200 μg/kg, i.p.) followed 2.5 h later by an i.p. injection of kainic acid (KA, 1.75 mg/kg). Oxytocin (1 mg/kg) was induced via different routes (intraperitoneal or intranasal) as well an enriched environment between PND 22-26. The enriched environment included larger cages (1560 cm2) with only 4 pups per cage, compared to those groups not receiving enrichment (646 cm2), as well as cardboard rolls and plastic toys. On PND 27-33 the light/dark box and elevated plus maze were used to assess anxiety-like behaviour. On PND 34 all rats were euthanized using a sharp guillotine, trunk blood and hypothalamic tissue were collected for neurochemical analysis (ELISA kit). Results Our findings confirmed that exposure to both prenatal stress and febrile seizures resulted anxiety-like behaviour and significantly higher plasma corticosterone concentrations compared to their counterparts. Environmental enrichment was significantly effective in attenuating the increased basal corticosterone levels and anxiety-like behaviour seen in the prenatally stressed FS rat. Although direct administration of oxytocin showed higher significance in reducing corticosterone plasma levels when compared to the enriched environment. Furthermore, hypothalamic oxytocin levels were not significant in rat exposed to environmental enrichment while oxytocin treatment showed a significant effect when compared to their counterparts. Conclusion Therefore, oxytocin administration during early postnatal development shows great potential in reversing the effects of prenatal stress and its subsequent exacerbation of FS.
Collapse
Affiliation(s)
- Shreyal Maikoo
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Andria Wilkins
- University of KwaZulu-Natal, College of Health Sciences, Department of Human Physiology, Durban, South Africa
| | - Lihle Qulu
- Stellenbosch University - Tygerberg Campus: Stellenbosch University Faculty of Medicine and Health Sciences, Capetown, South Africa,Corresponding author.
| |
Collapse
|
30
|
Angelidi AM, Belanger MJ, Kokkinos A, Koliaki CC, Mantzoros CS. Novel Noninvasive Approaches to the Treatment of Obesity: From Pharmacotherapy to Gene Therapy. Endocr Rev 2022; 43:507-557. [PMID: 35552683 PMCID: PMC9113190 DOI: 10.1210/endrev/bnab034] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Indexed: 02/08/2023]
Abstract
Recent insights into the pathophysiologic underlying mechanisms of obesity have led to the discovery of several promising drug targets and novel therapeutic strategies to address the global obesity epidemic and its comorbidities. Current pharmacologic options for obesity management are largely limited in number and of modest efficacy/safety profile. Therefore, the need for safe and more efficacious new agents is urgent. Drugs that are currently under investigation modulate targets across a broad range of systems and tissues, including the central nervous system, gastrointestinal hormones, adipose tissue, kidney, liver, and skeletal muscle. Beyond pharmacotherapeutics, other potential antiobesity strategies are being explored, including novel drug delivery systems, vaccines, modulation of the gut microbiome, and gene therapy. The present review summarizes the pathophysiology of energy homeostasis and highlights pathways being explored in the effort to develop novel antiobesity medications and interventions but does not cover devices and bariatric methods. Emerging pharmacologic agents and alternative approaches targeting these pathways and relevant research in both animals and humans are presented in detail. Special emphasis is given to treatment options at the end of the development pipeline and closer to the clinic (ie, compounds that have a higher chance to be added to our therapeutic armamentarium in the near future). Ultimately, advancements in our understanding of the pathophysiology and interindividual variation of obesity may lead to multimodal and personalized approaches to obesity treatment that will result in safe, effective, and sustainable weight loss until the root causes of the problem are identified and addressed.
Collapse
Affiliation(s)
- Angeliki M Angelidi
- Section of Endocrinology, VA Boston Healthcare System, Harvard Medical School, Boston, MA, USA
- Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Matthew J Belanger
- Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Alexander Kokkinos
- First Department of Propaedeutic Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Chrysi C Koliaki
- First Department of Propaedeutic Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Christos S Mantzoros
- Section of Endocrinology, VA Boston Healthcare System, Harvard Medical School, Boston, MA, USA
- Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
31
|
Sucrose intake by rats affected by both intraperitoneal oxytocin administration and time of day. Psychopharmacology (Berl) 2022; 239:429-442. [PMID: 34731267 DOI: 10.1007/s00213-021-06014-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/20/2021] [Indexed: 10/19/2022]
Abstract
RATIONALE Daily limited access to palatable food or drink at a fixed time is commonly used in rodent models of bingeing. Under these conditions, entrainment may modulate intake patterns. Oxytocin is involved in circadian patterns of intake and, when administered peripherally, reduces sucrose intake. However, oxytocin's effects on intake under limited-access conditions and its potential interaction with entrainment have not been explored. OBJECTIVES This study examined the role of entrainment on intake patterns, oxytocin's effects on sucrose intakes and locomotor activity and whether oxytocin's effects were mediated by its actions at the oxytocin receptor. METHODS Sated rats received daily 1-h access to 10% sucrose solution either at a fixed or varied time of day. Rats received intraperitoneal oxytocin (0 mg/kg, 0.3 mg/kg, 1 mg/kg, 3 mg/kg) prior to sucrose access, and spontaneous locomotor activity was assessed in an open-field test. Rats were then pre-treated with an oxytocin receptor antagonist, L368,899, prior to oxytocin before sucrose access. RESULTS Intake patterns did not differ between fixed- or varied-time presentations; rats consumed more sucrose solution in the middle as opposed to the early-dark phase. Oxytocin dose-dependently reduced sucrose intakes, but also reduced locomotor activity. There was some evidence of partial blockade of oxytocin-induced sucrose intake reductions by L368,899, but the results were unclear. CONCLUSIONS Time of day and oxytocin impact sucrose solution intake under daily limited access in rats and the sedative-like effects of oxytocin should be considered in future studies on oxytocin and food intake.
Collapse
|
32
|
Takayanagi Y, Onaka T. Roles of Oxytocin in Stress Responses, Allostasis and Resilience. Int J Mol Sci 2021; 23:ijms23010150. [PMID: 35008574 PMCID: PMC8745417 DOI: 10.3390/ijms23010150] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/17/2021] [Accepted: 12/19/2021] [Indexed: 02/06/2023] Open
Abstract
Oxytocin has been revealed to work for anxiety suppression and anti-stress as well as for psychosocial behavior and reproductive functions. Oxytocin neurons are activated by various stressful stimuli. The oxytocin receptor is widely distributed within the brain, and oxytocin that is released or diffused affects behavioral and neuroendocrine stress responses. On the other hand, there has been an increasing number of reports on the role of oxytocin in allostasis and resilience. It has been shown that oxytocin maintains homeostasis, shifts the set point for adaptation to a changing environment (allostasis) and contributes to recovery from the shifted set point by inducing active coping responses to stressful stimuli (resilience). Recent studies have suggested that oxytocin is also involved in stress-related disorders, and it has been shown in clinical trials that oxytocin provides therapeutic benefits for patients diagnosed with stress-related disorders. This review includes the latest information on the role of oxytocin in stress responses and adaptation.
Collapse
|
33
|
Anekonda VT, Thompson BW, Ho JM, Roberts ZS, Edwards MM, Nguyen HK, Dodson AD, Wolden-Hanson T, Chukri DW, Herbertson AJ, Graham JL, Havel PJ, Wietecha TA, O’Brien KD, Blevins JE. Hindbrain Administration of Oxytocin Reduces Food Intake, Weight Gain and Activates Catecholamine Neurons in the Hindbrain Nucleus of the Solitary Tract in Rats. J Clin Med 2021; 10:5078. [PMID: 34768597 PMCID: PMC8584350 DOI: 10.3390/jcm10215078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 11/16/2022] Open
Abstract
Existing studies show that CNS oxytocin (OT) signaling is important in the control of energy balance, but it is unclear which neurons may contribute to these effects. Our goals were to examine (1) the dose-response effects of acute OT administration into the third (3V; forebrain) and fourth (4V; hindbrain) ventricles to assess sensitivity to OT in forebrain and hindbrain sites, (2) the extent to which chronic 4V administration of OT reduces weight gain associated with the progression of diet-induced obesity, and (3) whether nucleus tractus solitarius (NTS) catecholamine neurons are downstream targets of 4V OT. Initially, we examined the dose-response effects of 3V and 4V OT (0.04, 0.2, 1, or 5 μg). 3V and 4V OT (5 μg) suppressed 0.5-h food intake by 71.7 ± 6.0% and 60 ± 12.9%, respectively. 4V OT (0.04, 0.2, 1 μg) reduced food intake by 30.9 ± 12.9, 42.1 ± 9.4, and 56.4 ± 9.0%, respectively, whereas 3V administration of OT (1 μg) was only effective at reducing 0.5-h food intake by 38.3 ± 10.9%. We subsequently found that chronic 4V OT infusion, as with chronic 3V infusion, reduced body weight gain (specific to fat mass) and tended to reduce plasma leptin in high-fat diet (HFD)-fed rats, in part, through a reduction in energy intake. Lastly, we determined that 4V OT increased the number of hindbrain caudal NTS Fos (+) neurons (156 ± 25) relative to vehicle (12 ± 3). The 4V OT also induced Fos in tyrosine hydroxylase (TH; marker of catecholamine neurons) (+) neurons (25 ± 7%) relative to vehicle (0.8 ± 0.3%). Collectively, these findings support the hypothesis that OT within the hindbrain is effective at reducing food intake, weight gain, and adiposity and that NTS catecholamine neurons in addition to non-catecholaminergic neurons are downstream targets of CNS OT.
Collapse
Affiliation(s)
- Vishwanath T. Anekonda
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (V.T.A.); (B.W.T.); (J.M.H.); (Z.S.R.); (M.M.E.); (H.K.N.); (A.D.D.); (T.W.-H.); (D.W.C.); (A.J.H.)
| | - Benjamin W. Thompson
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (V.T.A.); (B.W.T.); (J.M.H.); (Z.S.R.); (M.M.E.); (H.K.N.); (A.D.D.); (T.W.-H.); (D.W.C.); (A.J.H.)
| | - Jacqueline M. Ho
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (V.T.A.); (B.W.T.); (J.M.H.); (Z.S.R.); (M.M.E.); (H.K.N.); (A.D.D.); (T.W.-H.); (D.W.C.); (A.J.H.)
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA;
| | - Zachary S. Roberts
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (V.T.A.); (B.W.T.); (J.M.H.); (Z.S.R.); (M.M.E.); (H.K.N.); (A.D.D.); (T.W.-H.); (D.W.C.); (A.J.H.)
| | - 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 98108, USA; (V.T.A.); (B.W.T.); (J.M.H.); (Z.S.R.); (M.M.E.); (H.K.N.); (A.D.D.); (T.W.-H.); (D.W.C.); (A.J.H.)
| | - 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 98108, USA; (V.T.A.); (B.W.T.); (J.M.H.); (Z.S.R.); (M.M.E.); (H.K.N.); (A.D.D.); (T.W.-H.); (D.W.C.); (A.J.H.)
| | - 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; (V.T.A.); (B.W.T.); (J.M.H.); (Z.S.R.); (M.M.E.); (H.K.N.); (A.D.D.); (T.W.-H.); (D.W.C.); (A.J.H.)
| | - 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; (V.T.A.); (B.W.T.); (J.M.H.); (Z.S.R.); (M.M.E.); (H.K.N.); (A.D.D.); (T.W.-H.); (D.W.C.); (A.J.H.)
| | - Daniel W. Chukri
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (V.T.A.); (B.W.T.); (J.M.H.); (Z.S.R.); (M.M.E.); (H.K.N.); (A.D.D.); (T.W.-H.); (D.W.C.); (A.J.H.)
| | - 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; (V.T.A.); (B.W.T.); (J.M.H.); (Z.S.R.); (M.M.E.); (H.K.N.); (A.D.D.); (T.W.-H.); (D.W.C.); (A.J.H.)
| | - James L. Graham
- Department of Nutrition and Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (J.L.G.); (P.J.H.)
| | - Peter J. Havel
- Department of Nutrition and Department of Molecular Biosciences, School of Veterinary Medicine, 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;
- 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
| | - 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; (V.T.A.); (B.W.T.); (J.M.H.); (Z.S.R.); (M.M.E.); (H.K.N.); (A.D.D.); (T.W.-H.); (D.W.C.); (A.J.H.)
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA;
| |
Collapse
|
34
|
Niu J, Tong J, Blevins JE. Oxytocin as an Anti-obesity Treatment. Front Neurosci 2021; 15:743546. [PMID: 34720864 PMCID: PMC8549820 DOI: 10.3389/fnins.2021.743546] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 09/16/2021] [Indexed: 12/19/2022] Open
Abstract
Obesity is a growing health concern, as it increases risk for heart disease, hypertension, type 2 diabetes, cancer, COVID-19 related hospitalizations and mortality. However, current weight loss therapies are often associated with psychiatric or cardiovascular side effects or poor tolerability that limit their long-term use. The hypothalamic neuropeptide, oxytocin (OT), mediates a wide range of physiologic actions, which include reproductive behavior, formation of prosocial behaviors and control of body weight. We and others have shown that OT circumvents leptin resistance and elicits weight loss in diet-induced obese rodents and non-human primates by reducing both food intake and increasing energy expenditure (EE). Chronic intranasal OT also elicits promising effects on weight loss in obese humans. This review evaluates the potential use of OT as a therapeutic strategy to treat obesity in rodents, non-human primates, and humans, and identifies potential mechanisms that mediate this effect.
Collapse
Affiliation(s)
- JingJing Niu
- VA Puget Sound Health Care System, Office of Research and Development, Medical Research Service, Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States
| | - Jenny Tong
- VA Puget Sound Health Care System, Office of Research and Development, Medical Research Service, Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States
| | - James E. Blevins
- VA Puget Sound Health Care System, Office of Research and Development, Medical Research Service, Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
35
|
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: 1.3] [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
|
36
|
Çatli G, Acar S, Cingöz G, Rasulova K, Yarim AK, Uzun H, Küme T, Kızıldağ S, Dündar BN, Abacı A. Oxytocin receptor gene polymorphism and low serum oxytocin level are associated with hyperphagia and obesity in adolescents. Int J Obes (Lond) 2021; 45:2064-2073. [PMID: 34091593 DOI: 10.1038/s41366-021-00876-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 05/12/2021] [Accepted: 05/27/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND/OBJECTIVES In recent years, oxytocin (OXT) and polymorphisms in the oxytocin receptor (OXTR) gene have been reported to play roles in obesity pathogenesis. However, there was no study evaluating OXTR gene variants in childhood obesity. The aim of the study was to investigate the relation of OXTR gene polymorphisms and serum OXT levels with metabolic and anthropometric parameters in obese and healthy adolescents. SUBJECTS/METHODS The study was a multi-centered case-control study, which was conducted on obese and healthy adolescents aged between 12 and 17 years. Serum OXT and leptin levels were measured, and OXTR gene variants were studied by qPCR (rs53576) and RFLP (rs2254298) methods. RESULTS A total of 250 obese and 250 healthy adolescents were included in this study. In the obese group, serum OXT level was lower and leptin level was higher than the control group. In the obese group, frequencies of homozygous mutant (G/G) and heterozygous (A/G) genotypes for rs53576 polymorphism were higher than the control group. Homozygous mutant(G/G) and heterozygous (A/G) genotypes for rs53576 polymorphism were found to increase the risk of obesity compared to the wild type (A/A) genotype [OR = 6.05 and OR = 3.06; p < 0.001, respectively]. In patients with homozygous mutant (G/G) and heterozygous (A/G) genotype for rs53576 polymorphism, serum OXT levels were lower than the wild type (A/A) genotype. In the obese group, hyperphagia score was higher than the control group and correlated negatively with serum OXT level. CONCLUSIONS This study revealed that low serum OXT level, which is associated with hyperphagia may be an underlying cause for obesity in adolescents. For rs53576 polymorphism of the OXTR gene, obesity risk is higher in patients with homozygous mutant(G/G) and heterozygous(A/G)genotypes.
Collapse
Affiliation(s)
- Gönül Çatli
- Department of Pediatric Endocrinology, Izmir KatipÇelebi University, Faculty of Medicine, İzmir, Turkey.
| | - Sezer Acar
- Department of Pediatric Endocrinology, Dokuz Eylül University, Faculty of Medicine, İzmir, Turkey
| | - Gülten Cingöz
- Department of Pediatrics, Sağlik Bilimleri University, Tepecik Training and Research Hospital, İzmir, Turkey
| | - Khayala Rasulova
- Department of Medical Biology and Genetics, Dokuz Eylül University, Faculty of Medicine, İzmir, Turkey
| | - Ayça Kanat Yarim
- Department of Medical Biology and Genetics, Dokuz Eylül University, Faculty of Medicine, İzmir, Turkey
| | - Hamide Uzun
- Department of Nutrition and Dietetics, Sağlik Bilimleri University, Tepecik Training and Research Hospital, İzmir, Turkey
| | - Tuncay Küme
- Department of Biochemistry, Dokuz Eylül University, Faculty of Medicine, İzmir, Turkey
| | - Sefa Kızıldağ
- Department of Medical Biology and Genetics, Dokuz Eylül University, Faculty of Medicine, İzmir, Turkey
| | - Bumin Nuri Dündar
- Department of Pediatric Endocrinology, Izmir KatipÇelebi University, Faculty of Medicine, İzmir, Turkey
| | - Ayhan Abacı
- Department of Pediatric Endocrinology, Dokuz Eylül University, Faculty of Medicine, İzmir, Turkey
| |
Collapse
|
37
|
Assinder SJ, Boumelhem BB. Oxytocin stimulates lipolysis, prostaglandin E 2 synthesis, and leptin secretion in 3T3-L1 adipocytes. Mol Cell Endocrinol 2021; 534:111381. [PMID: 34216640 DOI: 10.1016/j.mce.2021.111381] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 06/01/2021] [Accepted: 06/29/2021] [Indexed: 11/26/2022]
Abstract
A model of oxytocin in the regulation of metabolic status has described one of oxytocin synthesis and release from the neurohypophysis in response to leptin, to suppress further leptin release. In addition, a lipogenic role for oxytocin has been suggested, consistent with an insulinergic action. This model, however, may be incorrect. Oxytocin reduces fat mass in the absence of either leptin or leptin receptor signalling, thereby challenging the interdependence between leptin and oxytocin. An oxytocin induced production of the anti-lipolytic prostaglandin E2 (PGE2) might account for this. Media from 3T3-L1 differentiated adipocytes treated with oxytocin (0-50 nmol.L-1) for 24 hrs were assayed for PGE2, leptin, adiponectin, and glycerol. Harvested cells were analysed for lipid droplet triglyceride and cytosolic free fatty acid (FFA) by flow cytometry, and for altered expression of lipolytic and lipogenic associated gene ontology transcripts by cDNA array. Both PGE2 and leptin secretion were significantly increased by oxytocin treatment whilst adiponectin secretion was not. A significant increase in cytosolic FFA was detected following oxytocin treatment, similar to that determined following treatment with isoproterenol (positive control). A significant increase in glycerol release to the culture media confirmed a lipolytic effect. No enrichment of lipolytic and lipogenic associated gene ontology transcripts was determined, but significant overrepresentation of chemosensory olfactory transcripts was. In conclusion, oxytocin stimulates lipolysis in 3T3-L1 adipocytes, mediated by autocrine/paracrine actions of PGE2 and leptin. To confirm that this response is mediated solely by the oxytocin receptor, further experiments would require those effects being blocked by a specific oxytocin antagonist.
Collapse
Affiliation(s)
- Stephen J Assinder
- Discipline of Physiology, School of Medical Science and Bosch Institute, Faculty of Medicine and Health, University of Sydney, Australia.
| | - Badwi B Boumelhem
- Discipline of Physiology, School of Medical Science and Bosch Institute, Faculty of Medicine and Health, University of Sydney, Australia
| |
Collapse
|
38
|
Kerem L, Lawson EA. The Effects of Oxytocin on Appetite Regulation, Food Intake and Metabolism in Humans. Int J Mol Sci 2021; 22:7737. [PMID: 34299356 PMCID: PMC8306733 DOI: 10.3390/ijms22147737] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 12/18/2022] Open
Abstract
The hypothalamic peptide oxytocin and its receptor are involved in a range of physiological processes, including parturition, lactation, cell growth, wound healing, and social behavior. More recently, increasing evidence has established the effects of oxytocin on food intake, energy expenditure, and peripheral metabolism. In this review, we provide a comprehensive description of the central oxytocinergic system in which oxytocin acts to shape eating behavior and metabolism. Next, we discuss the peripheral beneficial effects oxytocin exerts on key metabolic organs, including suppression of visceral adipose tissue inflammation, skeletal muscle regeneration, and bone tissue mineralization. A brief summary of oxytocin actions learned from animal models is presented, showing that weight loss induced by chronic oxytocin treatment is related not only to its anorexigenic effects, but also to the resulting increase in energy expenditure and lipolysis. Following an in-depth discussion on the technical challenges related to endogenous oxytocin measurements in humans, we synthesize data related to the association between endogenous oxytocin levels, weight status, metabolic syndrome, and bone health. We then review clinical trials showing that in humans, acute oxytocin administration reduces food intake, attenuates fMRI activation of food motivation brain areas, and increases activation of self-control brain regions. Further strengthening the role of oxytocin in appetite regulation, we review conditions of hypothalamic insult and certain genetic pathologies associated with oxytocin depletion that present with hyperphagia, extreme weight gain, and poor metabolic profile. Intranasal oxytocin is currently being evaluated in human clinical trials to learn whether oxytocin-based therapeutics can be used to treat obesity and its associated sequela. At the end of this review, we address the fundamental challenges that remain in translating this line of research to clinical care.
Collapse
Affiliation(s)
- Liya Kerem
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA;
- Division of Pediatric Endocrinology, Massachusetts General Hospital for Children, Boston, MA 02114, USA
| | - Elizabeth A. Lawson
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA;
| |
Collapse
|
39
|
Burmester V, Nicholls D, Buckle A, Stanojevic B, Crous-Bou M. Review of eating disorders and oxytocin receptor polymorphisms. J Eat Disord 2021; 9:85. [PMID: 34256847 PMCID: PMC8278600 DOI: 10.1186/s40337-021-00438-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/25/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND AND AIMS Oxytocin, a nine amino acid peptide synthesised in the hypothalamus, has been widely recognised for its role in anxiolysis, bonding, sociality, and appetite. It binds to the oxytocin receptor (OXTR)-a G-protein coupled receptor-that is stimulated by the actions of oestrogen both peripherally and centrally. Studies have implicated OXTR genotypes in conferring either a risk or protective effect in autism, schizophrenia, and eating disorders (ED). There are numerous DNA variations of this receptor, with the most common DNA variation being in the form of the single nucleotide polymorphisms (SNPs). Two OXTR SNPs have been most studied in relation to ED: rs53576 and rs2254298. Each SNP has the same allelic variant that produces genotypes AA, AG, and GG. In this critical review we will evaluate the putative role of rs53576 and rs2254298 SNPs in ED. Additionally, this narrative review will consider the role of gene-environment interactions in the development of ED pathology. FINDINGS The OXTR SNPs rs53576 and rs2254298 show independent associations between the A allele and restrictive eating behaviours. Conversely, the G allele of the OXTR rs53576 SNP is associated with binging behaviours, findings that were also evident in neuroanatomy. One study found the A allele of both OXTR SNPs to confer risk for more severe ED symptomatology while the G allele conferred some protective effect. An interaction between poor maternal care and rs2254298 AG/AA genotype conferred increased risk for binge eating and purging in women. CONCLUSIONS Individual OXTR SNP are unlikely in themselves to explain complex eating disorders but may affect the expression of and/or effectiveness of the OXTR. A growing body of G x E work is indicating that rs53576G homozygosity becomes disadvantageous for later mental health under early adverse conditions but further research to extend these findings to eating pathology is needed. The GWAS approach would benefit this area of knowledge.
Collapse
Affiliation(s)
- Victoria Burmester
- Department of Brain Sciences, Division of Psychiatry, Imperial College London, Burlington Danes, The Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK.
| | - Dasha Nicholls
- Department of Brain Sciences, Division of Psychiatry, Imperial College London, Burlington Danes, The Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Alexis Buckle
- Department of Brain Sciences, Division of Psychiatry, Imperial College London, Burlington Danes, The Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Boban Stanojevic
- Comprehensive Cancer Centre, Faculty of Life Sciences & Medicine, King's College London, Rayne Institute, 111 Coldharbour Ln, London, SE5 9RR, UK.,Laboratory for Radiobiology and Molecular Genetics, "Vinca" Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| | - Marta Crous-Bou
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology (ICO) - Bellvitge Biomedical Research Institute (IDIBELL). L'Hospitalet de Llobregat, Barcelona, Spain.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
40
|
Camerino C. Oxytocin Involvement in Body Composition Unveils the True Identity of Oxytocin. Int J Mol Sci 2021; 22:ijms22126383. [PMID: 34203705 PMCID: PMC8232088 DOI: 10.3390/ijms22126383] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/28/2021] [Accepted: 05/29/2021] [Indexed: 01/11/2023] Open
Abstract
The origin of the Oxytocin/Vasopressin system dates back about 600 million years. Oxytocin (Oxt) together with Vasopressin (VP) regulate a diversity of physiological functions that are important for osmoregulation, reproduction, metabolism, and social behavior. Oxt/VP-like peptides have been identified in several invertebrate species and they are functionally related across the entire animal kingdom. Functional conservation enables future exploitation of invertebrate models to study Oxt’s functions not related to pregnancy and the basic mechanisms of central Oxt/VP signaling. Specifically, Oxt is well known for its effects on uteri contractility and milk ejection as well as on metabolism and energy homeostasis. Moreover, the striking evidence that Oxt is linked to energy regulation is that Oxt- and Oxytocin receptor (Oxtr)-deficient mice show late onset obesity. Interestingly Oxt−/− or Oxtr−/− mice develop weight gain without increasing food intake, suggesting that a lack of Oxt reduce metabolic rate. Oxt is expressed in a diversity of skeletal muscle phenotypes and regulates thermogenesis and bone mass. Oxt may increases skeletal muscle tonicity and/or increases body temperature. In this review, the author compared the three most recent theories on the effects of Oxt on body composition.
Collapse
Affiliation(s)
- Claudia Camerino
- Department of Biomedical Sciences and Human Oncology (Section of Pharmacology), School of Medicine, University of Bari Aldo Moro, P.za G. Cesare 11, 70100 Bari, Italy;
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| |
Collapse
|
41
|
Damen L, Grootjen LN, Juriaans AF, Donze SH, Huisman TM, Visser JA, Delhanty PJ, Hokken‐Koelega AC. Oxytocin in young children with Prader-Willi syndrome: Results of a randomized, double-blind, placebo-controlled, crossover trial investigating 3 months of oxytocin. Clin Endocrinol (Oxf) 2021; 94:774-785. [PMID: 33296519 PMCID: PMC8246775 DOI: 10.1111/cen.14387] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/20/2020] [Accepted: 11/29/2020] [Indexed: 12/24/2022]
Abstract
CONTEXT Prader-Willi syndrome (PWS) is characterized by hypothalamic dysfunction, hyperphagia and a typical behavioural phenotype, with characteristics of autism spectrum disorder (ASD) like stubbornness, temper tantrums and compulsivity. It has been suggested that the oxytocin system in patients with PWS is dysfunctional. In ASD, intranasal oxytocin treatment has favourable effects on behaviour. OBJECTIVE To evaluate the effects of 3 months of twice daily intranasal oxytocin (dose range 16-40 IU/day), compared to placebo, on behaviour and hyperphagia in children with PWS. DESIGN Randomized, double-blind, placebo-controlled, crossover study in the Dutch PWS Reference Center. PATIENTS Twenty-six children with PWS aged 3-11 years. MAIN OUTCOME MEASURES (Change in) behaviour and hyperphagia measured by Oxytocin Questionnaire and Dykens hyperphagia questionnaire. RESULTS In the total group, no significant effects of oxytocin on social behaviour or hyperphagia were found. However, in boys, the Oxytocin Questionnaire scores improved significantly during oxytocin treatment, compared to a deterioration during placebo (4.5 (-0.8 to 15.3) vs. -4.0 (-11.3 to 0.8), P = .025). The Dykens hyperphagia questionnaire scores remained similar during oxytocin treatment, while there was a deterioration during placebo (0.0 (-0.8 to 4.3) vs. -3.5 (-6.0 to 0.0), P = .046). Patients with a deletion had significant improvements in both questionnaire scores during oxytocin treatment, but deteriorations during placebo. Oxytocin treatment was well tolerated, and there were no serious adverse events. CONCLUSIONS Intranasal oxytocin treatment has positive effects on social and eating behaviour in 3-11 years aged boys with PWS and in children with a deletion without safety concerns. Intranasal oxytocin in children with PWS might be considered, but individual effects should be carefully evaluated and treatment discontinued if no effects are found.
Collapse
Affiliation(s)
- Layla Damen
- Dutch Growth Research FoundationRotterdamthe Netherlands
- Department of PediatricsSubdivision of EndocrinologyErasmus University Medical CenterSophia Children’s HospitalRotterdamthe Netherlands
- Dutch Reference Center for Prader‐Willi Syndromethe Netherlands
| | - Lionne N. Grootjen
- Dutch Growth Research FoundationRotterdamthe Netherlands
- Department of PediatricsSubdivision of EndocrinologyErasmus University Medical CenterSophia Children’s HospitalRotterdamthe Netherlands
- Dutch Reference Center for Prader‐Willi Syndromethe Netherlands
| | - Alicia F. Juriaans
- Dutch Growth Research FoundationRotterdamthe Netherlands
- Department of PediatricsSubdivision of EndocrinologyErasmus University Medical CenterSophia Children’s HospitalRotterdamthe Netherlands
- Dutch Reference Center for Prader‐Willi Syndromethe Netherlands
| | - Stephany H. Donze
- Dutch Growth Research FoundationRotterdamthe Netherlands
- Department of PediatricsSubdivision of EndocrinologyErasmus University Medical CenterSophia Children’s HospitalRotterdamthe Netherlands
- Dutch Reference Center for Prader‐Willi Syndromethe Netherlands
| | - T. Martin Huisman
- Department of Internal MedicineErasmus University Medical CenterRotterdamthe Netherlands
| | - Jenny A. Visser
- Department of Internal MedicineErasmus University Medical CenterRotterdamthe Netherlands
| | - Patric J.D. Delhanty
- Department of Internal MedicineErasmus University Medical CenterRotterdamthe Netherlands
| | - Anita C.S. Hokken‐Koelega
- Dutch Growth Research FoundationRotterdamthe Netherlands
- Department of PediatricsSubdivision of EndocrinologyErasmus University Medical CenterSophia Children’s HospitalRotterdamthe Netherlands
- Dutch Reference Center for Prader‐Willi Syndromethe Netherlands
| |
Collapse
|
42
|
Šterk M, Križančić Bombek L, Skelin Klemen M, Slak Rupnik M, Marhl M, Stožer A, Gosak M. NMDA receptor inhibition increases, synchronizes, and stabilizes the collective pancreatic beta cell activity: Insights through multilayer network analysis. PLoS Comput Biol 2021; 17:e1009002. [PMID: 33974632 PMCID: PMC8139480 DOI: 10.1371/journal.pcbi.1009002] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 05/21/2021] [Accepted: 04/26/2021] [Indexed: 12/15/2022] Open
Abstract
NMDA receptors promote repolarization in pancreatic beta cells and thereby reduce glucose-stimulated insulin secretion. Therefore, NMDA receptors are a potential therapeutic target for diabetes. While the mechanism of NMDA receptor inhibition in beta cells is rather well understood at the molecular level, its possible effects on the collective cellular activity have not been addressed to date, even though proper insulin secretion patterns result from well-synchronized beta cell behavior. The latter is enabled by strong intercellular connectivity, which governs propagating calcium waves across the islets and makes the heterogeneous beta cell population work in synchrony. Since a disrupted collective activity is an important and possibly early contributor to impaired insulin secretion and glucose intolerance, it is of utmost importance to understand possible effects of NMDA receptor inhibition on beta cell functional connectivity. To address this issue, we combined confocal functional multicellular calcium imaging in mouse tissue slices with network science approaches. Our results revealed that NMDA receptor inhibition increases, synchronizes, and stabilizes beta cell activity without affecting the velocity or size of calcium waves. To explore intercellular interactions more precisely, we made use of the multilayer network formalism by regarding each calcium wave as an individual network layer, with weighted directed connections portraying the intercellular propagation. NMDA receptor inhibition stabilized both the role of wave initiators and the course of waves. The findings obtained with the experimental antagonist of NMDA receptors, MK-801, were additionally validated with dextrorphan, the active metabolite of the approved drug dextromethorphan, as well as with experiments on NMDA receptor KO mice. In sum, our results provide additional and new evidence for a possible role of NMDA receptor inhibition in treatment of type 2 diabetes and introduce the multilayer network paradigm as a general strategy to examine effects of drugs on connectivity in multicellular systems.
Collapse
Affiliation(s)
- Marko Šterk
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| | | | | | - Marjan Slak Rupnik
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- Alma Mater Europaea–ECM, Maribor, Slovenia
| | - Marko Marhl
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
- Faculty of Education, University of Maribor, Maribor, Slovenia
| | - Andraž Stožer
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Marko Gosak
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| |
Collapse
|
43
|
Hong SM, Ko JK, Moon JJ, Kim YR. Oxytocin: A Potential Therapeutic for Obesity. J Obes Metab Syndr 2021; 30:115-123. [PMID: 33820878 PMCID: PMC8277591 DOI: 10.7570/jomes20098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/20/2020] [Accepted: 01/21/2021] [Indexed: 12/28/2022] Open
Abstract
Oxytocin is a neuropeptide involved in the homeostasis of food consumption and energy; it affects hedonic eating. Studies in obese or binge-eating patients reported the hypophagic effect of oxytocin, which reduced caloric intake after administration. Several studies have demonstrated the effect of oxytocin’s increasing energy intake, decreasing food consumption, and contributing to weight loss. Oxytocin’s effects on food intake and metabolism suggest its therapeutic potential for treating obesity and binge eating.
Collapse
Affiliation(s)
- Soo Min Hong
- Department of Endocrinology and Metabolism, Seoul Paik Hospital, Inje University, Seoul, Korea
| | - Jeong-Kyung Ko
- Institute of Eating Disorders and Mental Health, Inje University, Seoul, Korea
| | - Jung-Joon Moon
- Department of Psychiatry, Busan Paik Hospital, Inje University, Busan, Korea
| | - Youl-Ri Kim
- Institute of Eating Disorders and Mental Health, Inje University, Seoul, Korea.,Department of Psychiatry, Seoul Paik Hospital, Inje University, Seoul, Korea
| |
Collapse
|
44
|
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: 2.5] [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
|
45
|
Erdenebayar O, Kato T, Kawakita T, Kasai K, Kadota Y, Yoshida K, Iwasa T, Irahara M. Effects of peripheral oxytocin administration on body weight, food intake, adipocytes, and biochemical parameters in peri- and postmenopausal female rats. Endocr J 2021; 68:7-16. [PMID: 32879161 DOI: 10.1507/endocrj.ej19-0586] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Recent studies have revealed that the administration of oxytocin has beneficial effects on the regulation of body weight, food intake, and metabolic functions, especially in obese individuals. Obesity is common in women after the menopause and drives many components of metabolic syndrome. Weight gain in menopausal women has been frequently reported. Although obesity and associated metabolic disorders are frequently observed in peri- and postmenopausal women, there are few medical interventions for these conditions. In this study, we evaluated the effects of chronic oxytocin administration on appetite, body weight, and fat mass in peri- and postmenopausal female rats. Sixteen naturally premenopausal or menopausal rats were intraperitoneally injected with oxytocin (1,000 μg/day) for 12 days. The daily changes in their body weight and food intake were measured at the same time as the oxytocin and vehicle injections. Intraperitoneally administering oxytocin for 12 days significantly reduced food intake, body weight, and visceral adipocyte size. In addition, oxytocin administration caused reductions in serum triglyceride and low-density lipoprotein-cholesterol levels, while it did not disturb hepatic or renal functions or locomotor activity. This is the first study to show the effects of oxytocin on the metabolic and feeding functions of peri- and postmenopausal female rats. Oxytocin might be a useful treatment for metabolic disorders caused by the menopause or aging.
Collapse
Affiliation(s)
- Otgontsetseg Erdenebayar
- Department of Obstetrics and Gynecology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Takeshi Kato
- Department of Obstetrics and Gynecology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Takako Kawakita
- Department of Obstetrics and Gynecology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Kana Kasai
- Department of Obstetrics and Gynecology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yuri Kadota
- Department of Obstetrics and Gynecology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Kanako Yoshida
- Department of Obstetrics and Gynecology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Takeshi Iwasa
- Department of Obstetrics and Gynecology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Minoru Irahara
- Department of Obstetrics and Gynecology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| |
Collapse
|
46
|
Ohbayashi K, Oyama Y, Yamaguchi C, Asano T, Yada T, Iwasaki Y. Gastrointestinal Distension by Pectin-Containing Carbonated Solution Suppresses Food Intake and Enhances Glucose Tolerance via GLP-1 Secretion and Vagal Afferent Activation. Front Endocrinol (Lausanne) 2021; 12:676869. [PMID: 34168616 PMCID: PMC8217665 DOI: 10.3389/fendo.2021.676869] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 05/10/2021] [Indexed: 12/13/2022] Open
Abstract
Diet-induced gastrointestinal distension is known to evoke satiation and suppress postprandial hyperglycemia; however, the underlying mechanisms remain poorly understood. This study explored how gastrointestinal distension regulates energy homeostasis by using inflating stomach formulation (ISF), the carbonated solution containing pectin that forms stable gel bubbles under acidic condition in the stomach. Here we show that, in mice, oral administration of ISF induced distension of stomach and proximal intestine temporarily, stimulated intestinal glucagon-like peptide-1 (GLP-1) secretion, and activated vagal afferents and brainstem. ISF suppressed food intake and improved glucose tolerance via enhancing insulin sensitivity. The anorexigenic effect was partially inhibited, and the beneficial glycemic effect was blunted by pharmacological GLP-1 receptor blockade and chemical denervation of capsaicin-sensitive sensory nerves. In HFD-fed obese mice showing arrhythmic feeding and obesity, subchronic ISF treatment at the light period (LP) onset for 10 days attenuated LP hyperphagia and visceral fat accumulation. These results demonstrate that gastrointestinal distension by ISF stimulates GLP-1 secretion and the vagal afferent signaling to the brain, thereby regulating feeding behavior and glucose tolerance. Furthermore, subchronic ISF treatment ameliorates HFD-induced visceral obesity. We propose the diet that induces gastrointestinal distension as a novel treatment of hyperphagic obesity and diabetes.
Collapse
Affiliation(s)
- Kento Ohbayashi
- Laboratory of Animal Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Yukiko Oyama
- Self-Medication R&D Laboratories, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Chiharu Yamaguchi
- Self-Medication R&D Laboratories, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Toshiki Asano
- Self-Medication R&D Laboratories, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Toshihiko Yada
- Division of Integrative Physiology, Kansai Electric Power Medical Research Institute, Kobe, Japan
- Division of Diabetes and Endocrinology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yusaku Iwasaki
- Laboratory of Animal Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
- *Correspondence: Yusaku Iwasaki,
| |
Collapse
|
47
|
Abstract
The hypothalamic peptide oxytocin has been increasingly recognized as a hormone and neurotransmitter with important effects on energy intake, metabolism, and body weight and is under investigation as a potential novel therapeutic agent for obesity. The main neurons producing oxytocin and expressing the oxytocin receptor are strategically located in brain areas known to be critically involved in homeostatic energy balance as well as hedonic and motivational aspects of eating behavior. In this chapter, we will review the central and peripheral physiology of oxytocin and the interaction of oxytocin with key hormones and neural circuitries that affect food intake and metabolism. Next, we will synthesize the available data on endogenous oxytocin levels related to caloric intake, body weight, and metabolic status. We will then review the effects of exogenous oxytocin administration on eating behavior, body weight, and metabolism in humans, including in healthy individuals as well as specific populations with suspected perturbations involving oxytocin pathways. Finally, we will address the promise and fundamental challenges of translating this line of research to clinical care.
Collapse
Affiliation(s)
- Liya Kerem
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States; Division of Pediatric Endocrinology, Massachusetts General Hospital for Children, Boston, MA, United States
| | - Elizabeth A Lawson
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
48
|
Wald HS, Chandra A, Kalluri A, Ong ZY, Hayes MR, Grill HJ. NTS and VTA oxytocin reduces food motivation and food seeking. Am J Physiol Regul Integr Comp Physiol 2020; 319:R673-R683. [PMID: 33026822 DOI: 10.1152/ajpregu.00201.2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Oxytocin (OT) is a neuropeptide whose central receptor-mediated actions include reducing food intake. One mechanism of its behavioral action is the amplification of the feeding inhibitory effects of gastrointestinal (GI) satiation signals processed by hindbrain neurons. OT treatment also reduces carbohydrate intake in humans and rodents, and correspondingly, deficits in central OT receptor (OT-R) signaling increase sucrose self-administration. This suggests that additional processes contribute to central OT effects on feeding. This study investigated the hypothesis that central OT reduces food intake by decreasing food seeking and food motivation. As central OT-Rs are expressed widely, a related focus was to assess the role of one or more OT-R-expressing nuclei in food motivation and food-seeking behavior. OT was delivered to the lateral ventricle (LV), nucleus tractus solitarius (NTS), or ventral tegmental area (VTA), and a progressive ratio (PR) schedule of operant reinforcement and an operant reinstatement paradigm were used to measure motivated feeding behavior and food-seeking behavior, respectively. OT delivered to the LV, NTS, or VTA reduced 1) motivation to work for food and 2) reinstatement of food-seeking behavior. Results provide a novel and additional interpretation for central OT-driven food intake inhibition to include the reduction of food motivation and food seeking.
Collapse
Affiliation(s)
- Hallie S Wald
- Department of Psychology, Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ananya Chandra
- Department of Psychology, Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anita Kalluri
- Department of Psychology, Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Zhi Yi Ong
- School of Psychology, University of New South Wales, Sydney, Australia
| | - Matthew R Hayes
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Harvey J Grill
- Department of Psychology, Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
49
|
Yuan J, Zhang R, Wu R, Gu Y, Lu Y. The effects of oxytocin to rectify metabolic dysfunction in obese mice are associated with increased thermogenesis. Mol Cell Endocrinol 2020; 514:110903. [PMID: 32531419 DOI: 10.1016/j.mce.2020.110903] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 06/03/2020] [Accepted: 06/03/2020] [Indexed: 01/12/2023]
Abstract
Oxytocin, a protein hormone mainly produced by hypothalamus, has been shown to repress body weight gain in obese animals, in part, by reducing food intake and increasing energy expenditure. Till now, activation of brown fat tissue (BAT) thermogenesis and white adipose tissue (WAT) browning are considered as two main factors for oxytocin-induced energy expenditure. However, the underlying molecular mechanisms are still not understood well. Here, we observed that oxytocin expression in the hypothalamus and its receptor in adipose tissues were induced by cold exposure in mice. In differentiated adipocytes, oxytocin stimulated brown adipocyte specific gene expression by inducing PRDM16. In high fat diet induced obese mice, oxytocin delivery by osmotic minipumps increased body core temperature and decreased body weight gain. Glucose and insulin tolerance were improved by oxytocin. Hyperinsulinemia and fatty liver were ameliorated in oxytocin-treated animals. Moreover, oxytocin treatment induced thermogenic gene expressions in BAT, inguinal WAT (iWAT), and skeletal muscle. Taken together, our findings revealed a new aspect of oxytocin, i.e. oxytocin induces iWAT browning and stimulates thermogenesis in BAT, iWAT and skeletal muscle, through which oxytocin promotes thermogenesis and thus combats obesity and metabolic dysfunctions.
Collapse
Affiliation(s)
- Jin Yuan
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, Jiangsu, 210011, China; Department of Endocrinology and Metabolism, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China, 20 Xisi Road, Nantong, Jiangsu, 226001, China
| | - Rongping Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China, 20 Xisi Road, Nantong, Jiangsu, 226001, China
| | - Runze Wu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China, 20 Xisi Road, Nantong, Jiangsu, 226001, China
| | - Yunjuan Gu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China, 20 Xisi Road, Nantong, Jiangsu, 226001, China.
| | - Yibing Lu
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, Jiangsu, 210011, China.
| |
Collapse
|
50
|
Maejima Y, Horita S, Otsuka A, Hidema S, Nishimori K, Shimomura K. Oral oxytocin delivery with proton pump inhibitor pretreatment decreases food intake. Peptides 2020; 128:170312. [PMID: 32298773 DOI: 10.1016/j.peptides.2020.170312] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 04/06/2020] [Accepted: 04/06/2020] [Indexed: 12/25/2022]
Abstract
Oxytocin (Oxt) is considered as a potential agent to treat multiple neuropsychiatric disorders, obesity and metabolic syndrome. Although the mechanisms underlying these effects remain unclear, nasal administration is considered to be a potential way to deliver Oxt into blood vessels. The development of an easier, more stable and efficient way is expected. A recent study demonstrated that orally administered Oxt can be transmitted into blood if it is prevented from degradation in stomach and reaches the intestinal tract. In this study, we pretreated mice with a proton pump inhibitor (PPI), omeprazole (20 mg/kg), and administered capsulized Oxt (0.25 mg), so that the Oxt can be prevented from degradation by pepsin due to the low pH in stomach and reach the intestinal tract. Functionally, these mice showed a similar decrease in food intake to those who underwent intraperitoneal administration. We also confirmed that this method dramatically increased plasma Oxt levels and the expression of neural activation marker c-Fos protein in the paraventricular and suprachiasmatic nucleus. Our study showed that by pretreating mice with PPI, Oxt in a gelatin-coated capsule can prevent Oxt from degradation by pepsin in stomach, and reach the bloodstream in an effective concentration. These results indicate that our method is a promising oral delivery of Oxt and should be investigated further for other peptide agents based on peripheral injection or nasal administration.
Collapse
Affiliation(s)
- Yuko Maejima
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, 960-1295, Fukushima, Japan.
| | - Shoichiro Horita
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, 960-1295, Fukushima, Japan
| | - Ayano Otsuka
- Department of Molecular and Cell Biology, Graduate School of Agricultural Science, Tohoku University Sendai-shi, 981-8555, Miyagi, Japan
| | - Shizu Hidema
- Department of Molecular and Cell Biology, Graduate School of Agricultural Science, Tohoku University Sendai-shi, 981-8555, Miyagi, Japan
| | - Katsuhiko Nishimori
- Department of Molecular and Cell Biology, Graduate School of Agricultural Science, Tohoku University Sendai-shi, 981-8555, Miyagi, Japan
| | - Kenju Shimomura
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, 960-1295, Fukushima, Japan
| |
Collapse
|