1
|
Leaf A, Rothschild JA, Sharpe TM, Sims ST, Macias CJ, Futch GG, Roberts MD, Stout JR, Ormsbee MJ, Aragon AA, Campbell BI, Arent SM, D’Agostino DP, Barrack MT, Kerksick CM, Kreider RB, Kalman DS, Antonio J. International society of sports nutrition position stand: ketogenic diets. J Int Soc Sports Nutr 2024; 21:2368167. [PMID: 38934469 PMCID: PMC11212571 DOI: 10.1080/15502783.2024.2368167] [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: 05/29/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
POSITION STATEMENT The International Society of Sports Nutrition (ISSN) provides an objective and critical review of the use of a ketogenic diet in healthy exercising adults, with a focus on exercise performance and body composition. However, this review does not address the use of exogenous ketone supplements. The following points summarize the position of the ISSN. 1. A ketogenic diet induces a state of nutritional ketosis, which is generally defined as serum ketone levels above 0.5 mM. While many factors can impact what amount of daily carbohydrate intake will result in these levels, a broad guideline is a daily dietary carbohydrate intake of less than 50 grams per day. 2. Nutritional ketosis achieved through carbohydrate restriction and a high dietary fat intake is not intrinsically harmful and should not be confused with ketoacidosis, a life-threatening condition most commonly seen in clinical populations and metabolic dysregulation. 3. A ketogenic diet has largely neutral or detrimental effects on athletic performance compared to a diet higher in carbohydrates and lower in fat, despite achieving significantly elevated levels of fat oxidation during exercise (~1.5 g/min). 4. The endurance effects of a ketogenic diet may be influenced by both training status and duration of the dietary intervention, but further research is necessary to elucidate these possibilities. All studies involving elite athletes showed a performance decrement from a ketogenic diet, all lasting six weeks or less. Of the two studies lasting more than six weeks, only one reported a statistically significant benefit of a ketogenic diet. 5. A ketogenic diet tends to have similar effects on maximal strength or strength gains from a resistance training program compared to a diet higher in carbohydrates. However, a minority of studies show superior effects of non-ketogenic comparators. 6. When compared to a diet higher in carbohydrates and lower in fat, a ketogenic diet may cause greater losses in body weight, fat mass, and fat-free mass, but may also heighten losses of lean tissue. However, this is likely due to differences in calorie and protein intake, as well as shifts in fluid balance. 7. There is insufficient evidence to determine if a ketogenic diet affects males and females differently. However, there is a strong mechanistic basis for sex differences to exist in response to a ketogenic diet.
Collapse
Affiliation(s)
- Alex Leaf
- Alex Leaf LLC, Scientific Affairs, Scottsdale, AZ, USA
| | - Jeffrey A. Rothschild
- Auckland University of Technology, Sports Performance Research Institute New Zealand, Auckland, New Zealand
- High Performance Sport New Zealand, Performance Nutrition, Auckland, New Zealand
| | - Tim M. Sharpe
- University of Western States, Human Nutrition and Functional Medicine, Portland, OR, USA
| | - Stacy T. Sims
- Auckland University of Technology, Sports Performance Research Institute New Zealand, Auckland, New Zealand
- Stanford University, Stanford Lifestyle Medicine, Palo Alto, CA, USA
| | - Chad J. Macias
- University of Western States, Human Nutrition and Functional Medicine, Portland, OR, USA
| | - Geoff G. Futch
- Springfield College, Department of Exercise Science and Athletic Training, Springfield, MA, USA
- FitPro Analytics, Scientific Affairs, Springfield, MA, USA
| | | | - Jeffrey R. Stout
- University of Central Florida, School of Kinesiology and Rehabilitation Sciences, Orlando, FL, USA
| | - Michael J. Ormsbee
- Florida State University, Institute of Sports Sciences & Medicine, Tallahassee, FL, USA
- University of KwaZulu Natal, Discipline of Biokinetics, Exercise and Leisure Sciences, Durban, South Africa
| | | | - Bill I. Campbell
- University of South Florida, Performance and Physique Enhancement Laboratory, Exercise Science Program, Tampa, FL, USA
| | - Shawn M. Arent
- University of South Carolina, Department of Exercise Science, Arnold School of Public Health, Columbia, SC, USA
| | - Dominic P. D’Agostino
- Institute for Human and Machine Cognition, Human Healthspan, Resilience, and Performance, Pensacola, FL, USA
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, FL, USA
| | - Michelle T. Barrack
- California State University, Department of Family and Consumer Sciences, Long Beach, CA, USA
| | - Chad M. Kerksick
- Lindenwood University, Exercise and Performance Nutrition Laboratory, College of Science, Technology, and Health, St. Charles, MO, USA
| | - Richard B. Kreider
- Texas A&M University, Exercise and Sport Nutrition Lab, Department of Kinesiology and Sports Management, College Station, TX, USA
| | - Douglas S. Kalman
- Nova Southeastern University, Department of Nutrition. Dr. Kiran C. Patel College of Osteopathic Medicine. Davie, FL, USA
| | - Jose Antonio
- Department of Health and Human Performance, Nova Southeastern University, Davie, FL, USA
| |
Collapse
|
2
|
Kositanurit W, Siritaweechai N, Varachotisate P, Burana C, Sukswai N, Surintrspanont J, Siriviriyakul P, Kaikaew K, Werawatganon D. Genistein mitigates diet-induced obesity and metabolic dysfunctions in gonadectomized mice with some sex-differential effects. Front Endocrinol (Lausanne) 2024; 15:1392866. [PMID: 39351533 PMCID: PMC11439649 DOI: 10.3389/fendo.2024.1392866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 08/26/2024] [Indexed: 10/04/2024] Open
Abstract
Background Obesity is associated with insulin resistance (IR) and metabolic dysfunction-associated steatotic liver disease (MASLD). Genistein, an isoflavone, is a promising natural compound for preventing and treating obesity and metabolic dysfunctions. We aimed to investigate the sex-specific protective effects of genistein on obesity, IR, and MASLD in a murine model of sex hormone deprivation with diet-induced obesity (DIO), mimicking postmenopausal women or aging men with metabolic syndrome. Methods Gonadectomized and sham-operated C57BL/6NJcl mice were fed a high-fat high-sucrose diet for 4 weeks to induce obesity (7 mice per group). In gonadectomized mice, genistein (16 mg/kg/day) or vehicle (7.5% dimethyl sulfoxide) was orally administered for 45 days. We assessed glucose homeostasis parameters, hepatic histopathology, and hepatic gene expression to investigate the effects of gonadectomy and genistein treatment. Results Gonadectomy exacerbated adiposity in both sexes. Ovariectomy diminished the protective effects of female gonadal hormones on the homeostatic model assessment for insulin resistance (HOMA-IR), serum alanine transaminase levels, hepatic steatosis score, and the expression of hepatic genes associated with MASLD progression and IR, such as Fasn, Srebf1, Saa1, Cd36, Col1a1, Pck1, and Ppargc1a. Genistein treatment in gonadectomized mice significantly reduced body weight gain and the hepatic steatosis score in both sexes. However, genistein treatment significantly attenuated HOMA-IR and the expression of the hepatic genes only in female mice. Conclusion Genistein treatment mitigates DIO-related MASLD in both male and female gonadectomized mice. Regarding hepatic gene expression associated with MASLD and IR, the beneficial effect of genistein was significantly evident only in female mice. This study suggests a potential alternative application of genistein in individuals with obesity and sex hormone deprivation, yet pending clinical trials.
Collapse
Affiliation(s)
- Weerapat Kositanurit
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Natakorn Siritaweechai
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Pachara Varachotisate
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Chuti Burana
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Narittee Sukswai
- Precision Pathology of Neoplasia Research Group, Department of Pathology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Jerasit Surintrspanont
- Precision Pathology of Neoplasia Research Group, Department of Pathology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Pathology, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Prasong Siriviriyakul
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Alternative and Complementary Medicine for Gastrointestinal and Liver Diseases, Chulalongkorn University, Bangkok, Thailand
| | - Kasiphak Kaikaew
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Alternative and Complementary Medicine for Gastrointestinal and Liver Diseases, Chulalongkorn University, Bangkok, Thailand
| | - Duangporn Werawatganon
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Alternative and Complementary Medicine for Gastrointestinal and Liver Diseases, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
3
|
Fernandois D, Rusidzé M, Mueller-Fielitz H, Sauve F, Deligia E, Silva MSB, Evrard F, Franco-García A, Mazur D, Martinez-Corral I, Jouy N, Rasika S, Maurage CA, Giacobini P, Nogueiras R, Dehouck B, Schwaninger M, Lenfant F, Prevot V. Estrogen receptor-α signaling in tanycytes lies at the crossroads of fertility and metabolism. Metabolism 2024; 158:155976. [PMID: 39019342 PMCID: PMC7616427 DOI: 10.1016/j.metabol.2024.155976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND Estrogen secretion by the ovaries regulates the hypothalamic-pituitary-gonadal axis during the reproductive cycle, influencing gonadotropin-releasing hormone (GnRH) and luteinizing hormone (LH) secretion, and also plays a role in regulating metabolism. Here, we establish that hypothalamic tanycytes-specialized glia lining the floor and walls of the third ventricle-integrate estrogenic feedback signals from the gonads and couple reproduction with metabolism by relaying this information to orexigenic neuropeptide Y (NPY) neurons. METHODS Using mouse models, including mice floxed for Esr1 (encoding estrogen receptor alpha, ERα) and those with Cre-dependent expression of designer receptors exclusively activated by designer drugs (DREADDs), along with viral-mediated, pharmacological and indirect calorimetric approaches, we evaluated the role of tanycytes and tanycytic estrogen signaling in pulsatile LH secretion, cFos expression in NPY neurons, estrous cyclicity, body-weight changes and metabolic parameters in adult females. RESULTS In ovariectomized mice, chemogenetic activation of tanycytes significantly reduced LH pulsatile release, mimicking the effects of direct NPY neuron activation. In intact mice, tanycytes were crucial for the estrogen-mediated control of GnRH/LH release, with tanycytic ERα activation suppressing fasting-induced NPY neuron activation. Selective knockout of Esr1 in tanycytes altered estrous cyclicity and fertility in female mice and affected estrogen's ability to inhibit refeeding in fasting mice. The absence of ERα signaling in tanycytes increased Npy transcripts and body weight in intact mice and prevented the estrogen-mediated decrease in food intake as well as increase in energy expenditure and fatty acid oxidation in ovariectomized mice. CONCLUSIONS Our findings underscore the pivotal role of tanycytes in the neuroendocrine coupling of reproduction and metabolism, with potential implications for its age-related deregulation after menopause. SIGNIFICANCE STATEMENT Our investigation reveals that tanycytes, specialized glial cells in the brain, are key interpreters of estrogen signals for orexigenic NPY neurons in the hypothalamus. Disrupting tanycytic estrogen receptors not only alters fertility in female mice but also impairs the ability of estrogens to suppress appetite. This work thus sheds light on the critical role played by tanycytes in bridging the hormonal regulation of cyclic reproductive function and appetite/feeding behavior. This understanding may have potential implications for age-related metabolic deregulation after menopause.
Collapse
Affiliation(s)
- Daniela Fernandois
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, F-59000 Lille, France
| | - Mariam Rusidzé
- Institute of Metabolic and Cardiovascular Diseases (I2MC) Equipe 4, Inserm U1297UPS, CHU, Toulouse, France
| | - Helge Mueller-Fielitz
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
| | - Florent Sauve
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, F-59000 Lille, France
| | - Eleonora Deligia
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, F-59000 Lille, France
| | - Mauro S B Silva
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, F-59000 Lille, France
| | - Florence Evrard
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, F-59000 Lille, France
| | - Aurelio Franco-García
- Group of Cellular and Molecular Pharmacology, Department of Pharmacology, CEIR Campus Mare Nostrum, University of Murcia, Spain, Instituto Murciano de Investigación Biosanitaria (IMIB), Pascual Parrilla, Murcia, Spain
| | - Daniele Mazur
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, F-59000 Lille, France
| | - Ines Martinez-Corral
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, F-59000 Lille, France
| | | | - S Rasika
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, F-59000 Lille, France
| | - Claude-Alain Maurage
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, F-59000 Lille, France
| | - Paolo Giacobini
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, F-59000 Lille, France
| | - Ruben Nogueiras
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Benedicte Dehouck
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, F-59000 Lille, France
| | - Markus Schwaninger
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
| | - Francoise Lenfant
- Institute of Metabolic and Cardiovascular Diseases (I2MC) Equipe 4, Inserm U1297UPS, CHU, Toulouse, France
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, F-59000 Lille, France.
| |
Collapse
|
4
|
He L, Li Q, Yang Y, Li J, Luo W, Huang Y, Zhong X. Pharmacovigilance study of GLP-1 receptor agonists for metabolic and nutritional adverse events. Front Pharmacol 2024; 15:1416985. [PMID: 39040467 PMCID: PMC11260617 DOI: 10.3389/fphar.2024.1416985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 06/18/2024] [Indexed: 07/24/2024] Open
Abstract
Aims: Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) are employed extensively in the management of type 2 diabetes and obesity. However, there is a paucity of real-world data on their safety and tolerability for metabolic and nutritional adverse events in large sample populations. This study aimed to analyse the metabolic and nutritional safety signatures of different GLP-1 RAs by exploring the Food and Drug Administration (FDA) Adverse Event Reporting System (FAERS). Methods: AEs data were extracted from the FDA Adverse Event Reporting System database for each GLP-1 RA from the time of its launch until the second quarter of 2023. The reported odds ratio (ROR), proportional reporting ratio (PRR), Empirical Bayesian Geometric Mean and Bayesian Confidence Propagation Neural Network were employed to identify AE signals. Results: A system organ class of metabolism and nutrition disorders was employed to filter AE reports, resulting in the identification of 10,450 reports for exenatide, 2,860 reports for liraglutide, 240 reports for albiglutide, 4,847 reports for dulaglutide, 2,905 reports for semaglutide, 1,089 reports for tirzepatide, and 13 reports for lixisenatide. Semaglutide (ROR, 3.34; 95%CI, 3.22), liraglutide (ROR, 2.78; 95%CI, 2.69), and exenatide (ROR, 2.15; 95%CI, 2.11) were associated with metabolism and nutrition disorders. The number of AE signals detected were as follows: albiglutide (n = 1), lixisenatide (n = 2), tirzepatide (n = 11), exenatide (n = 12), liraglutide (n = 16), semaglutide (n = 20), dulaglutide (n = 22). Dehydration was the most frequent AE contributing to serious outcomes for liraglutide (n = 318, 23.93%), dulaglutide (n = 434, 20.90%), semaglutide (n = 370, 25.10%) and tirzepatide (n = 70, 32.86%). The time to onset (TTO) of AE was statistically different between exenatide and the other GLP-1 RAs (p < 0.001), and the Weibull parameters for dehydration for liraglutide, dulaglutide, and semaglutide analyses all showed an early failure-type profile. Conclusion: Our study suggests that exenatide, liraglutide, and semaglutide are more susceptible to metabolic and nutritional AEs than other GLP-1 RAs. Liraglutide, dulaglutide, semaglutide, and tirzepaptide's potential to induce dehydration, necessitates special attention. Despite certain deficiencies, GLP-1 RAs have considerable potential for the treatment of eating disorders.
Collapse
Affiliation(s)
- Long He
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Qiuyu Li
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yongfeng Yang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jiahao Li
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Wei Luo
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yilan Huang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiaoyan Zhong
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
5
|
Kucukdereli H, Amsalem O, Pottala T, Lim M, Potgieter L, Hasbrouck A, Lutas A, Andermann ML. Repeated stress triggers seeking of a starvation-like state in anxiety-prone female mice. Neuron 2024; 112:2130-2141.e7. [PMID: 38642553 PMCID: PMC11287784 DOI: 10.1016/j.neuron.2024.03.027] [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: 05/18/2023] [Revised: 01/28/2024] [Accepted: 03/27/2024] [Indexed: 04/22/2024]
Abstract
Elevated anxiety often precedes anorexia nervosa and persists after weight restoration. Patients with anorexia nervosa often describe self-starvation as pleasant, potentially because food restriction can be anxiolytic. Here, we tested whether repeated stress can cause animals to prefer a starvation-like state. We developed a virtual reality place preference paradigm in which head-fixed mice can voluntarily seek a starvation-like state induced by optogenetic stimulation of hypothalamic agouti-related peptide (AgRP) neurons. Prior to stress exposure, males but not females showed a mild aversion to AgRP stimulation. Strikingly, following multiple days of stress, a subset of females developed a strong preference for AgRP stimulation that was predicted by high baseline anxiety. Such stress-induced changes in preference were reflected in changes in facial expressions during AgRP stimulation. Our study suggests that stress may cause females predisposed to anxiety to seek a starvation state and provides a powerful experimental framework for investigating the underlying neural mechanisms.
Collapse
Affiliation(s)
- Hakan Kucukdereli
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Oren Amsalem
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Trent Pottala
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Michelle Lim
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Leilani Potgieter
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Amanda Hasbrouck
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Andrew Lutas
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Mark L Andermann
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
6
|
Ribeiro JL, Santos TA, Garcia MT, Carvalho BFDC, Esteves JECS, Moraes RM, Anbinder AL. Heat-killed Limosilactobacillus reuteri ATCC PTA 6475 prevents bone loss in ovariectomized mice: A preliminary study. PLoS One 2024; 19:e0304358. [PMID: 38820403 PMCID: PMC11142514 DOI: 10.1371/journal.pone.0304358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 05/11/2024] [Indexed: 06/02/2024] Open
Abstract
Osteoporosis is an important health problem that occurs due to an imbalance between bone formation and resorption. Hormonal deficiency post-menopause is a significant risk factor. The probiotic Limosilactobacillus reuteri has been reported to prevent ovariectomy (Ovx)-induced bone loss in mice and reduce bone loss in postmenopausal women. Despite the numerous health benefits of probiotics, as they are live bacteria, the administration is not risk-free for certain groups (e.g., neonates and immunosuppressed patients). We evaluated the effects of L. reuteri (ATCC PTA 6475) and its heat-killed (postbiotic) form on Ovx-induced bone loss. Adult female mice (BALB/c) were randomly divided into four groups: group C-control (sham); group OVX-C-Ovx; group OVX-POS-Ovx + heat-killed probiotic; group OVX-PRO-Ovx + probiotic. L. reuteri or the postbiotic was administered to the groups (1.3x109 CFU/day) by gavage. Bacterial morphology after heat treatment was accessed by scanning electron microscopy (SEM). The treatment started one week after Ovx and lasted 28 days (4 weeks). The animals were euthanized at the end of the treatment period. Bone microarchitecture and ileum Occludin and pro-inflammatory cytokines gene expression were evaluated by computed microtomography and qPCR techniques, respectively. The Ovx groups had lower percentage of bone volume (BV/TV) and number of bone trabeculae as well as greater total porosity compared to the control group. Treatment with live and heat-killed L. reuteri resulted in higher BV/TV and trabecular thickness than the Ovx group. The heat treatment caused some cell surface disruptions, but its structure resembled that of the live probiotic in SEM analysis. There were no statistical differences in Occludin, Il-6 and Tnf-α gene expression. Both viable and heat-killed L. reuteri prevented bone loss on ovariectomized mice, independently of gut Occludin and intestinal Il-6 and Tnf-α gene expression.
Collapse
Affiliation(s)
- Jaqueline Lemes Ribeiro
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology of São José dos Campos (São Paulo State University–Unesp), São José dos Campos, São Paulo, Brazil
| | - Thaís Aguiar Santos
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology of São José dos Campos (São Paulo State University–Unesp), São José dos Campos, São Paulo, Brazil
| | - Maíra Terra Garcia
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology of São José dos Campos (São Paulo State University–Unesp), São José dos Campos, São Paulo, Brazil
| | - Bruna Fernandes do Carmo Carvalho
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology of São José dos Campos (São Paulo State University–Unesp), São José dos Campos, São Paulo, Brazil
| | | | - Renata Mendonça Moraes
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology of São José dos Campos (São Paulo State University–Unesp), São José dos Campos, São Paulo, Brazil
| | - Ana Lia Anbinder
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology of São José dos Campos (São Paulo State University–Unesp), São José dos Campos, São Paulo, Brazil
| |
Collapse
|
7
|
Costanzo A. Temporal patterns in taste sensitivity. Nutr Rev 2024; 82:831-847. [PMID: 37558243 PMCID: PMC11082591 DOI: 10.1093/nutrit/nuad097] [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: 08/11/2023] Open
Abstract
Individuals vary in their ability to taste, and some individuals are more sensitive to certain tastes than others. Taste sensitivity is a predictor of various factors, such as diet, eating behavior, appetite regulation, and overall health. Furthermore, taste sensitivity can fluctuate within an individual over short to long periods of time: for example, in daily (diurnal) cycles, monthly (menstrual) cycles (in females), and yearly (seasonal) cycles. Understanding these temporal patterns is important for understanding individual eating habits and food preferences, particularly in the context of personalized and precision nutrition. This review provides a summary of the literature on taste sensitivity patterns across 3 temporal dimensions: daily, monthly, and yearly. Good evidence for diurnal patterns has been observed for sweet taste and fat taste, although the evidence is limited to rodent studies for the latter. Obese populations showed limited variation to sweet and fat taste sensitivities over a day, with limited variation in sweet taste sensitivity being linked to insulin resistance. There were mixed observations of temporal variation in sensitivity to sour and umami tastes, and there were no patterns in sensitivity to bitter taste. Menstrual patterns in sweet taste sensitivity were consistent with patterns in food intake. Other taste modality investigations had mixed findings that had little agreement across studies. Hormonal changes in females influence taste sensitivity to some degree, although the overall patterns are unclear. Seasonal patterns have been less well studied, but there is weak evidence that sweet, salty, and bitter taste sensitivities change across seasons. Differences in seasonal taste patterns have been observed in subgroups susceptible to mental health disorders, requiring further investigation. Patterns of taste sensitivity are evident across multiple temporal dimensions, and more research is needed to determine the influence of these patterns on food intake. Dysregulation of these patterns may also be a marker of certain diseases or health conditions, warranting further investigation. Notably, the alimentary tastes (umami, fat, and carbohydrate) are underrepresented in this research area and require additional investigation.
Collapse
Affiliation(s)
- Andrew Costanzo
- CASS Food Research Centre, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
| |
Collapse
|
8
|
Kuebler IRK, Suárez M, Wakabayashi KT. Sex differences and sex-specific regulation of motivated behavior by Melanin-concentrating hormone: a short review. Biol Sex Differ 2024; 15:33. [PMID: 38570844 PMCID: PMC10993549 DOI: 10.1186/s13293-024-00608-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 03/23/2024] [Indexed: 04/05/2024] Open
Abstract
Recent preclinical research exploring how neuropeptide transmitter systems regulate motivated behavior reveal the increasing importance of sex as a critical biological variable. Neuropeptide systems and their central circuits both contribute to sex differences in a range of motivated behaviors and regulate sex-specific behaviors. In this short review, we explore the current research of how sex as a biological variable influences several distinct motivated behaviors that are modulated by the melanin-concentrating hormone (MCH) neuropeptide system. First, we review how MCH regulates feeding behavior within the context of energy homeostasis differently between male and female rodents. Then, we focus on MCH's role in lactation as a sex-specific process within the context of energy homeostasis. Next, we discuss the sex-specific effects of MCH on maternal behavior. Finally, we summarize the role of MCH in drug-motivated behaviors. While these topics are traditionally investigated from different scientific perspectives, in this short review we discuss how these behaviors share commonalities within the larger context of motivated behaviors, and that sex differences discovered in one area of research may impact our understanding in another. Overall, our review highlights the need for further research into how sex differences in energy regulation associated with reproduction and parental care contribute to regulating motivated behaviors.
Collapse
Affiliation(s)
- Isabel R K Kuebler
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, Lincoln, NE, 68588-0308, USA
| | - Mauricio Suárez
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, Lincoln, NE, 68588-0308, USA
| | - Ken T Wakabayashi
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, Lincoln, NE, 68588-0308, USA.
- Rural Drug Addiction Research Center, University of Nebraska-Lincoln, 660 N 12th St., Lincoln, NE, 68588, USA.
| |
Collapse
|
9
|
Halpage J, DaSilva Pantoja P, Mancarella S. Prolonged tamoxifen-enriched diet is associated with cardiomyopathy and nutritional frailty in mice. Exp Physiol 2024; 109:513-523. [PMID: 38291801 PMCID: PMC10984784 DOI: 10.1113/ep091668] [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: 11/13/2023] [Accepted: 01/01/2024] [Indexed: 02/01/2024]
Abstract
Tamoxifen (TAM) is required for gene recombination in the inducible Cre/lox system. The TAM-enriched diet is considered safe, with negligible impact on animal wellbeing. However, studies reporting the long-term effects of the TAM diet and its potential impact on experimental outcomes are scarce. We conducted a longitudinal study on mice exposed to a 4-week dietary TAM citrate supplementation. Several parameters were recorded, such as body weight, body composition, mortality, and cardiac function. The collagen1a2 (Col1a2) transgenic mouse was used to assess TAM-induced recombination in vivo in cardiac fibroblasts followed by myocardial infarction (MI). The impact of TAM on the MI outcome was also evaluated. The recombination efficiency and cytotoxic effect of the TAM active metabolite, 4-hydroxy-tamoxifen (4-OHT), were assessed in vitro. Mice exposed to a TAM diet showed body weight loss and a 10% increase in mortality (P = 0.045). The TAM diet decreased cardiac function and induced cardiac remodeling, indicated by decreased fractional shortening from 32.23% to 19.23% (P = 0.001) and left ventricular (LV) wall thinning. All measured parameters were reversed to normal when mice were returned to a normal diet. Infarcted Col1a2-CreER mice on the TAM regimen showed gene recombination in fibroblasts, but it was associated with a substantial increase in mortality post-surgery (2.5-fold) compared to the controls. In vitro, 4-OHT induced gene editing in fibroblasts; however, cell growth arrest and cytotoxicity were observed at high concentrations. In conclusion, prolonged exposure to the TAM diet can be detrimental and necessitates careful model selection and interpretation of the results.
Collapse
Affiliation(s)
- Janith Halpage
- Department of PhysiologyUniversity of Tennessee Health Sciences CenterMemphisTennesseeUSA
| | | | - Salvatore Mancarella
- Department of PhysiologyUniversity of Tennessee Health Sciences CenterMemphisTennesseeUSA
| |
Collapse
|
10
|
Reho JJ, Muskus PC, Bennett DM, Grobe CC, Burnett CML, Nakagawa P, Segar JL, Sigmund CD, Grobe JL. Modulatory effects of estrous cycle on ingestive behaviors and energy balance in young adult C57BL/6J mice maintained on a phytoestrogen-free diet. Am J Physiol Regul Integr Comp Physiol 2024; 326:R242-R253. [PMID: 38284128 PMCID: PMC11213288 DOI: 10.1152/ajpregu.00273.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 01/30/2024]
Abstract
The estrous cycle is known to modify food, fluid, and electrolyte intake behaviors and energy homeostasis in various species, in part through fluctuations in estrogen levels. Simultaneously, commonly commercially available rodent dietary formulations greatly vary in soy protein content, and thereby the delivery of biologically active phytoestrogens. To explore the interactions among the estrous cycle, sodium, fluid, and caloric seeking behaviors, and energy homeostasis, young adult C57BL/6J female mice were maintained on a soy protein-free 2920x diet and provided water, or a choice between water and 0.15 mol/L NaCl drink solution. Comprehensive metabolic phenotyping was performed using a multiplexed Promethion (Sable Systems International) system, and estrous stages were determined via daily vaginal cytology. When provided food and water, estrous cycling had no major modulatory effects on intake behaviors or energy balance. When provided a saline solution drink choice, significant modulatory effects of the transition from diestrus to proestrus were observed upon fluid intake patterning, locomotion, and total energy expenditure. Access to saline increased total daily sodium consumption and aspects of energy expenditure, but these effects were not modified by the estrous stage. Collectively, these results indicate that when supplied a phytoestrogen-free diet, the estrous cycle has minor modulatory effects on ingestive behaviors and energy balance in C57BL/6J mice that are sensitive to sodium supply.NEW & NOTEWORTHY When provided a phytoestrogen-free diet, the estrous cycle had very little effect on food and water intake, physical activity, or energy expenditure in C57BL/6J mice. In contrast, when provided an NaCl drink in addition to food and water, the estrous cycle was associated with changes in intake behaviors and energy expenditure. These findings highlight the complex interactions among estrous cycling, dietary formulation, and nutrient presentation upon ingestive behaviors and energy homeostasis in mice.
Collapse
Affiliation(s)
- John J Reho
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Patricia C Muskus
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Darby M Bennett
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Connie C Grobe
- Department of Pediatrics, Division of Neonatology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Colin M L Burnett
- Department of Medicine/Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Pablo Nakagawa
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Jeffrey L Segar
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Department of Pediatrics, Division of Neonatology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Curt D Sigmund
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Justin L Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| |
Collapse
|
11
|
Leidmaa E, Prodan AM, Depner LL, Komorowska-Müller JA, Beins EC, Schuermann B, Kolbe CC, Zimmer A. Astrocytic Dagla Deletion Decreases Hedonic Feeding in Female Mice. Cannabis Cannabinoid Res 2024; 9:74-88. [PMID: 38265773 PMCID: PMC10874831 DOI: 10.1089/can.2023.0194] [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: 01/25/2024] Open
Abstract
Introduction: Endocannabinoids and exogenous cannabinoids are potent regulators of feeding behavior and energy metabolism. Stimulating cannabinoid receptor signaling enhances appetite, particularly for energy-dense palatable foods, and promotes energy storage. To elucidate the underlying cellular mechanisms, we investigate here the potential role of astrocytic endocannabinoid 2-arachidonoylglycerol (2-AG). Astrocytes provide metabolic support for neurons and contribute to feeding regulation but the effect of astrocytic 2-AG on feeding is unknown. Materials and Methods: We generated mice lacking the 2-AG synthesizing enzyme diacylglycerol lipase alpha (Dagla) in astrocytes (GLAST-Dagla KO) and investigated hedonic feeding behavior in male and female mice. Body weight and baseline water and food intake was characterized; additionally, the mice went through milk, saccharine, and sucrose preference tests in fed and fasted states. In female mice, the estrous cycle stages were identified and plasma levels of female sex hormones were measured. Results: We found that the effects of the inducible astrocytic Dagla deletion were sex-specific. Acute milk preference was decreased in female, but not in male mice and the effect was most evident in the estrus stage of the cycle. This prompted us to investigate sex hormone profiles, which were found to be altered in GLAST-Dagla KO females. Specifically, follicle-stimulating hormone was elevated in the estrus stage, luteinizing hormone in the proestrus, and progesterone was increased in both proestrus and estrus stages of the cycle compared with controls. Conclusions: Astrocytic Dagla regulates acute hedonic appetite for palatable food in females and not in males, possibly owing to a deregulated female sex hormone profile. It is plausible that endocannabinoid production by astrocytes at least partly contributes to the greater susceptibility to overeating in females. This finding may also be important for understanding the effects of exogenous cannabinoids on sex hormone profiles.
Collapse
Affiliation(s)
- Este Leidmaa
- Medical Faculty, Institute of Molecular Psychiatry, University of Bonn, Bonn, Germany
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Alexandra Maria Prodan
- Medical Faculty, Institute of Molecular Psychiatry, University of Bonn, Bonn, Germany
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Lena-Louise Depner
- Medical Faculty, Institute of Molecular Psychiatry, University of Bonn, Bonn, Germany
| | | | - Eva Carolina Beins
- Medical Faculty, Institute of Molecular Psychiatry, University of Bonn, Bonn, Germany
- Medical Faculty, Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Britta Schuermann
- Medical Faculty, Institute of Molecular Psychiatry, University of Bonn, Bonn, Germany
| | | | - Andreas Zimmer
- Medical Faculty, Institute of Molecular Psychiatry, University of Bonn, Bonn, Germany
| |
Collapse
|
12
|
Kundakovic M, Tickerhoof M. Epigenetic mechanisms underlying sex differences in the brain and behavior. Trends Neurosci 2024; 47:18-35. [PMID: 37968206 PMCID: PMC10841872 DOI: 10.1016/j.tins.2023.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/21/2023] [Accepted: 09/26/2023] [Indexed: 11/17/2023]
Abstract
Sex differences are found across brain regions, behaviors, and brain diseases. Sexual differentiation of the brain is initiated prenatally but it continues throughout life, as a result of the interaction of three major factors: gonadal hormones, sex chromosomes, and the environment. These factors are thought to act, in part, via epigenetic mechanisms which control chromatin and transcriptional states in brain cells. In this review, we discuss evidence that epigenetic mechanisms underlie sex-specific neurobehavioral changes during critical organizational periods, across the estrous cycle, and in response to diverse environments throughout life. We further identify future directions for the field that will provide novel mechanistic insights into brain sex differences, inform brain disease treatments and women's brain health in particular, and apply to people across genders.
Collapse
Affiliation(s)
- Marija Kundakovic
- Department of Biological Sciences, Fordham University, Bronx, NY 10458, USA.
| | - Maria Tickerhoof
- Department of Biological Sciences, Fordham University, Bronx, NY 10458, USA
| |
Collapse
|
13
|
Kuutti MA, Hyvärinen M, Lankila H, Aukee P, Hietavala EM, Laakkonen EK. Association of eating behavior with symptoms of pelvic floor disorders in middle-aged women: An observational study. WOMEN'S HEALTH (LONDON, ENGLAND) 2024; 20:17455057241305075. [PMID: 39658908 PMCID: PMC11632885 DOI: 10.1177/17455057241305075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 11/08/2024] [Accepted: 11/19/2024] [Indexed: 12/12/2024]
Abstract
BACKGROUND Estrogen deficiency during menopause, aging, reproductive history, and factors increasing intra-abdominal pressure may lead to structural and functional failure in the pelvic floor. Lifestyle choices, such as eating behavior, may contribute to pelvic floor disorders. OBJECTIVES The objective of the study was to investigate associations of eating behavior with symptoms of pelvic floor disorders, that is, stress urinary incontinence, urgency urinary incontinence, fecal incontinence, and constipation or defecation difficulties among middle-aged women. DESIGN A cross-sectional, observational study was performed using a population sample of 1098 Finnish women aged 47-55 years. METHODS Eating behavior, food consumption frequency, demographical, gynecological, and physical activity variables were assessed using self-report questionnaires. Logistic regression models were used to assess the associations of eating behavior, food frequency, and symptoms of pelvic floor disorders. Models were adjusted with demographical, gynecological, and physical activity variables. RESULTS In adjusted models, middle-aged women with disordered eating style were more likely to experience the symptoms of stress urinary incontinence (odds ratio (OR) 1.5, p = 0.002), and constipation or defecation difficulties (OR 1.4, p = 0.041). Adding body mass index into the models abolished associations. Of the studied food items, more frequent consumption of ready-made, highly processed foods (OR 1.5, p = 0.001), and fast foods (OR 1.5, p = 0.005) were independently associated with symptoms of stress urinary incontinence regardless of eating style, whereas consuming ready-made foods (OR 1.4, p = 0.048) was associated with symptoms of urgency urinary incontinence. Daily consumption of fruits (OR 0.8, p = 0.034) was independently associated with symptoms of stress urinary incontinence. Furthermore, we observed that daily consumption of porridge was associated with symptoms of constipation or defecation difficulties (OR 1.7, p = 0.010) independently of eating style. Alcohol consumption (OR 0.9, p = 0.015) was inversely associated with constipation and defecation difficulties. Women with overall higher quality diet had lower odds for stress urinary incontinence (OR 0.9, p = 0.002). CONCLUSION This study provides proof-of-concept evidence to the hypothesis that eating behavior and consuming certain food items are associated with perceived pelvic floor disorders. As a preventive action, eating behavior of women with the risk of these symptoms should be assessed, and guidance toward healthy eating patterns should be provided.
Collapse
Affiliation(s)
- Mari A Kuutti
- Gerontology Research Center and Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Matti Hyvärinen
- Gerontology Research Center and Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Hannamari Lankila
- Gerontology Research Center and Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Pauliina Aukee
- Department of Obstetrics and Gynecology, Wellbeing Services County of Central Finland, Jyväskylä, Finland
| | - Enni-Maria Hietavala
- Gerontology Research Center and Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Eija K Laakkonen
- Gerontology Research Center and Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| |
Collapse
|
14
|
Qu J, Wu D, Ko CW, Zhu Q, Liu M, Tso P. Deficiency of apoA-IV in Female 129X1/SvJ Mice Leads to Diet-Induced Obesity, Insulin Resistance, and Decreased Energy Expenditure. Nutrients 2023; 15:4655. [PMID: 37960308 PMCID: PMC10650794 DOI: 10.3390/nu15214655] [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: 10/02/2023] [Revised: 10/23/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Obesity is one of the main risk factors for cardiovascular diseases, type II diabetes, hypertension, and certain cancers. Obesity in women at the reproductive stage adversely affects contraception, fertility, maternal well-being, and the health of their offspring. Being a major protein component in chylomicrons and high-density lipoproteins, apolipoprotein A-IV (apoA-IV) is involved in lipid metabolism, food intake, glucose homeostasis, prevention against atherosclerosis, and platelet aggregation. The goal of the present study is to determine the impact of apoA-IV deficiency on metabolic functions in 129X1/SvJ female mouse strain. After chronic high-fat diet feeding, apoA-IV-/- mice gained more weight with a higher fat percentage than wild-type (WT) mice, as determined by measuring their body composition. Increased adiposity and adipose cell size were also observed with a microscope, particularly in periovarian fat pads. Based on plasma lipid and adipokine assays, we found that obesity in apoA-IV-/- mice was not associated with hyperlipidemia but with higher leptin levels. Compared to WT mice, apoA-IV deficiency displayed glucose intolerance and elevated insulin levels, according to the data of the glucose tolerance test, and increased HOMA-IR values at fasting, suggesting possible insulin resistance. Lastly, we found obesity in apoA-IV-/- mice resulting from reduced energy expenditure but not food intake. Together, we established a novel and excellent female mouse model for future mechanistic study of obesity and its associated comorbidities.
Collapse
Affiliation(s)
- Jie Qu
- Medpace Reference Laboratories, LLC, 5365 Medpace Way, Cincinnati, OH 45227, USA;
| | - Dong Wu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan 250012, China;
| | - Chih-Wei Ko
- Chroma Medicine, 201 Brookline Ave, Suite 1101, Boston, MA 02215, USA;
| | - Qi Zhu
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati, 2180 E Galbraith Road, Cincinnati, OH 45237, USA; (Q.Z.); (M.L.)
| | - Min Liu
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati, 2180 E Galbraith Road, Cincinnati, OH 45237, USA; (Q.Z.); (M.L.)
| | - Patrick Tso
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati, 2180 E Galbraith Road, Cincinnati, OH 45237, USA; (Q.Z.); (M.L.)
| |
Collapse
|
15
|
Massa MG, Scott RL, Cara AL, Cortes LR, Vander PB, Sandoval NP, Park JW, Ali SL, Velez LM, Wang HB, Ati SS, Tesfaye B, Reue K, van Veen JE, Seldin MM, Correa SM. Feeding neurons integrate metabolic and reproductive states in mice. iScience 2023; 26:107918. [PMID: 37817932 PMCID: PMC10561062 DOI: 10.1016/j.isci.2023.107918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 07/27/2023] [Accepted: 09/12/2023] [Indexed: 10/12/2023] Open
Abstract
Balance between metabolic and reproductive processes is important for survival, particularly in mammals that gestate their young. How the nervous system coordinates this balance is an active area of study. Herein, we demonstrate that somatostatin (SST) neurons of the tuberal hypothalamus alter feeding in a manner sensitive to metabolic and reproductive states in mice. Whereas chemogenetic activation of SST neurons increased food intake across sexes, ablation decreased food intake only in female mice during proestrus. This ablation effect was only apparent in animals with low body mass. Fat transplantation and bioinformatics analysis of SST neuronal transcriptomes revealed white adipose as a key modulator of these effects. These studies indicate that SST hypothalamic neurons integrate metabolic and reproductive cues by responding to varying levels of circulating estrogens to modulate feeding differentially based on energy stores. Thus, gonadal steroid modulation of neuronal circuits can be context dependent and gated by metabolic status.
Collapse
Affiliation(s)
- Megan G. Massa
- Department of Integrative Biology and Physiology, University of California – Los Angeles, Los Angeles, CA 90095, USA
- Neuroscience Interdepartmental Doctoral Program, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Rachel L. Scott
- Department of Integrative Biology and Physiology, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Alexandra L. Cara
- Department of Integrative Biology and Physiology, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Laura R. Cortes
- Department of Integrative Biology and Physiology, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Paul B. Vander
- Department of Integrative Biology and Physiology, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Norma P. Sandoval
- Department of Integrative Biology and Physiology, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Jae W. Park
- Department of Integrative Biology and Physiology, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Sahara L. Ali
- Department of Integrative Biology and Physiology, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Leandro M. Velez
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Huei-Bin Wang
- Department of Integrative Biology and Physiology, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Shomik S. Ati
- Department of Integrative Biology and Physiology, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Bethlehem Tesfaye
- Department of Integrative Biology and Physiology, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Karen Reue
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - J. Edward van Veen
- Department of Integrative Biology and Physiology, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Marcus M. Seldin
- Department of Biological Chemistry, School of Medicine, University of California – Irvine, Irvine, CA 92697, USA
| | - Stephanie M. Correa
- Department of Integrative Biology and Physiology, University of California – Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
16
|
Weidlinger S, Winterberger K, Pape J, Weidlinger M, Janka H, von Wolff M, Stute P. Impact of estrogens on resting energy expenditure: A systematic review. Obes Rev 2023; 24:e13605. [PMID: 37544655 DOI: 10.1111/obr.13605] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 06/13/2023] [Accepted: 06/23/2023] [Indexed: 08/08/2023]
Abstract
The fear of weight gain is one of the main reasons for women not to initiate or to early discontinue hormonal contraception or menopausal hormone therapy. Resting energy expenditure is by far the largest component and the most important determinant of total energy expenditure. Given that low resting energy expenditure is a confirmed predictive factor for weight gain and consecutively for the development of obesity, research into the influence of sex steroids on resting energy expenditure is a particularly exciting area. The objective of this systematic review was to evaluate the effects of medication with natural and synthetic estrogens on resting energy expenditure in healthy normal weight and overweight women. Through complex systematic literature searches, a total of 10 studies were identified that investigated the effects of medication with estrogens on resting energy expenditure. Our results demonstrate that estrogen administration increases resting energy expenditure by up to +208 kcal per day in the context of contraception and by up to +222 kcal per day in the context of menopausal hormone therapy, suggesting a preventive effect of circulating estrogen levels and estrogen administration on weight gain and obesity development.
Collapse
Affiliation(s)
- Susanna Weidlinger
- Department of Obstetrics and Gynecology, University Hospital of Bern, Bern, Switzerland
| | - Katja Winterberger
- Department of Obstetrics and Gynecology, University Hospital of Bern, Bern, Switzerland
| | - Janna Pape
- Department of Obstetrics and Gynecology, University Hospital of Bern, Bern, Switzerland
| | | | - Heidrun Janka
- Medical Library, University Library Bern, University of Bern, Bern, Switzerland
| | - Michael von Wolff
- Department of Obstetrics and Gynecology, University Hospital of Bern, Bern, Switzerland
| | - Petra Stute
- Department of Obstetrics and Gynecology, University Hospital of Bern, Bern, Switzerland
| |
Collapse
|
17
|
Shan W, Ding J, Xu J, Du Q, Chen C, Liao Q, Yang X, Lou J, Jin Z, Chen M, Xie R. Estrogen regulates duodenal glucose absorption by affecting estrogen receptor-α on glucose transporters. Mol Cell Endocrinol 2023:112028. [PMID: 37769868 DOI: 10.1016/j.mce.2023.112028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 10/03/2023]
Abstract
The mechanisms of estrogen in glucose metabolism are well established; however, its role in glucose absorption remains unclear. In this study, we investigated the effects of estrogen on glucose absorption in humans, mice, and SCBN intestinal epithelial cells. We first observed a correlation between estrogen and blood glucose in young women and found that glucose tolerance was significantly less in the premenstrual phase than in the preovulatory phase. Similarly, with decreased serum estradiol levels in ovariectomized mice, estrogen receptors alpha (ERα) and beta (ERβ) in the duodenum were reduced, and weight and abdominal fat increased significantly. The expression of sodium/glucose cotransporter 1 (SGLT1) and glucose transporter 2 (GLUT2) and glucose absorption in the duodenum decreased significantly. Estrogen significantly upregulated SGLT1 and GLUT2 expression in SCBN cells. Silencing of ERα, but not ERβ, reversed this trend, suggesting that ERα may be key to estrogen-regulating glucose transporters. A mechanistic study revealed that downstream, estrogen regulates the protein kinase C (PKC) pathway. Overall, our findings indicate that estrogen promotes glucose absorption, and estrogen and ERα deficiency can inhibit SGLT1 and GLUT2 expression through the PKC signaling pathway, thereby reducing glucose absorption.
Collapse
Affiliation(s)
- Weixi Shan
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, China; Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi, Guizhou, 563003, China
| | - Jianhong Ding
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, China; Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi, Guizhou, 563003, China
| | - Jingyu Xu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, China; Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi, Guizhou, 563003, China
| | - Qian Du
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, China; Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi, Guizhou, 563003, China
| | - Changmei Chen
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, China; Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi, Guizhou, 563003, China
| | - Qiushi Liao
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, China; Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi, Guizhou, 563003, China
| | - Xiaoxu Yang
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, China; Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi, Guizhou, 563003, China
| | - Jun Lou
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, China; Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi, Guizhou, 563003, China
| | - Zhe Jin
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, China; Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi, Guizhou, 563003, China
| | - Mingkai Chen
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuhan, 430060, Hubei, China.
| | - Rui Xie
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, China; Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi, Guizhou, 563003, China.
| |
Collapse
|
18
|
Abstract
Females are often underrepresented in the scientific literature, but awareness of the need for female-specific research is increasing. Review articles have been published on the effects of the menstrual cycle on aspects of exercise performance and physiology, yet to date no research has reviewed the effect of menstrual cycle phase on dietary energy intake. Fluctuations in endogenous sex hormones across the menstrual cycle influence a range of physiological processes, including those involved in nutritional status. Observational research typically quantifies female athletes' nutritional intakes at a single time point; however, this may provide inaccurate information if dietary intake fluctuates across the menstrual cycle. Similarly, this may have implications for interventional research, where dietary intake is often poorly controlled or monitored. This review aimed to synthesize the published literature on dietary energy intakes of naturally menstruating females in various phases of the menstrual cycle. The review critiques the relevant literature in light of recent publications on good practice for female research, explores the impact of the menstrual cycle on energy intake, identifies gaps within the evidence base, and informs future research. Overall, energy intake appears to be lower in the follicular phase compared with the luteal phase, with a particular decrease in the days leading up to and including ovulation. The magnitude of these fluctuations is not yet clearly quantifiable and most likely varies, both between individuals, and from cycle to cycle. This review notes the lack of high-quality research investigating the energy intakes of females across the menstrual cycle, and the very limited data available for female athletes and others who undertake large amounts of physical activity. It also highlights the need for researchers to take into consideration anovulatory cycles and the potential effects of premenstrual disorders on dietary intake.
Collapse
Affiliation(s)
- Michaela M Rogan
- are with the Department of Human Nutrition, University of Otago, Dunedin, New Zealand
| | - Katherine E Black
- K.E. Black, Department of Human Nutrition, University of Otago, PO Box 54, Dunedin, Otago, New Zealand. E-mail:
| |
Collapse
|
19
|
Zhao Z, Gobrogge K. Neurodevelopmental Model Explaining Associations between Sex Hormones, Personality, and Eating Pathology. Brain Sci 2023; 13:859. [PMID: 37371339 PMCID: PMC10296733 DOI: 10.3390/brainsci13060859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/22/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
Clinical scientists have been investigating the relationships between sex hormones, personality, and eating disorders for decades. However, there is a lack of direct research that addresses whether personality mediates or moderates the relationships between sex hormones and eating pathology. Moreover, the neural mechanisms that underlie the interactive associations between these variables remain unclear. This review aims to summarize the associations between these constructs, describe a neural mechanism mediating these relationships, and offer clinical strategies for the early identification and intervention of eating disorders. The gathered evidence shows that aggressiveness, impulsivity, and obsessive-compulsiveness may mediate or moderate the relationships between sex hormones and eating pathology, but only among females. Furthermore, sex hormone receptor density in the mesocorticolimbic dopamine pathway may explain the neural mechanism of these associations. Future research should use more comprehensive personality measurements and assess the mediation and moderation effects of temperament while taking the hormone levels of women across menstrual cycles into account. Additionally, electroencephalography and functional magnetic resonance imaging should be implemented to directly assess brain activity and corroborate these findings.
Collapse
Affiliation(s)
- Ziyu Zhao
- Department of Psychological & Brain Sciences, Boston University, Boston, MA 02215, USA;
| | - Kyle Gobrogge
- Department of Psychological & Brain Sciences, Boston University, Boston, MA 02215, USA;
- Undergraduate Program in Neuroscience, College of Art & Sciences, Boston University, Boston, MA 02215, USA
| |
Collapse
|
20
|
Kucukdereli H, Amsalem O, Pottala T, Lim M, Potgieter L, Hasbrouck A, Lutas A, Andermann ML. Chronic stress triggers seeking of a starvation-like state in anxiety-prone female mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.16.541013. [PMID: 37292650 PMCID: PMC10245771 DOI: 10.1101/2023.05.16.541013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Elevated anxiety often precedes anorexia nervosa and persists after weight restoration. Patients with anorexia nervosa often describe hunger as pleasant, potentially because food restriction can be anxiolytic. Here, we tested whether chronic stress can cause animals to prefer a starvation-like state. We developed a virtual reality place preference paradigm in which head-fixed mice can voluntarily seek a starvation-like state induced by optogenetic stimulation of hypothalamic agouti-related peptide (AgRP) neurons. Prior to stress induction, male but not female mice showed mild aversion to AgRP stimulation. Strikingly, following chronic stress, a subset of females developed a strong preference for AgRP stimulation that was predicted by high baseline anxiety. Such stress-induced changes in preference were reflected in changes in facial expressions during AgRP stimulation. Our study suggests that stress may cause females predisposed to anxiety to seek a starvation state, and provides a powerful experimental framework for investigating the underlying neural mechanisms.
Collapse
|
21
|
Opoku AA, Abushama M, Konje JC. Obesity and menopause. Best Pract Res Clin Obstet Gynaecol 2023:102348. [PMID: 37244787 DOI: 10.1016/j.bpobgyn.2023.102348] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 04/30/2023] [Indexed: 05/29/2023]
Abstract
The global obesity pandemic continues to rise, with figures from the World Health Organization showing that 13% of the world's adult population was obese in 2016. Obesity has significant implications, with an increased risk of cardiovascular diseases, diabetes mellitus, metabolic syndrome, and several malignancies. The menopausal transition is associated with increased obesity, a transition from a gynecoid to an android body shape, and increased abdominal and visceral fat, which further worsens the associated cardiometabolic risks. Whether this increased obesity is a consequence of menopause, age, genetics, or environmental factors has long been debated. Increasing life expectancy means women spend a significant part of their lives in the menopause. As such, understanding this complex interplay of obesity and menopause is important to providing the right advice/management. We review the current evidence on obesity and menopause, focusing on the implications of increased obesity during menopause, the impact of menopause on obesity, and the effect of available treatments on associated morbidities.
Collapse
Affiliation(s)
- Albert A Opoku
- Consultant Obstetrician and Gynecologist, Hamad Medical Corporation, Al Wakra Hospital, P O Box 82228, Al Wakra, Qatar; Assistant Professor in Clinical Obstetrics and Gynecology, Weill Cornell, Medicine, P O Box 24144, Doha, Qatar.
| | - Mandy Abushama
- Assistant Professor in Clinical Obstetrics and Gynecology, Weill Cornell Medicine, Qatar; Senior Consultant Obstetrician and Gynecologist, Feto Maternal Center, Al Markhiya, Doha, Qatar.
| | - Justin C Konje
- Senior Consultant Obstetrician and Gynecologist, Feto Maternal Center, Al Markhiya, Doha, Qatar; Professor of Obstetrics and Gynecology, Weill Cornell Medicine, Qatar; Emeritus Professor, Obstetrics and Gynecology, Department of Health Sciences, University of Leicester, United Kingdom.
| |
Collapse
|
22
|
Massa MG, Scott RL, Cara AL, Cortes LR, Sandoval NP, Park JW, Ali S, Velez LM, Tesfaye B, Reue K, van Veen JE, Seldin M, Correa SM. Feeding Neurons Integrate Metabolic and Reproductive States in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.25.525595. [PMID: 36747631 PMCID: PMC9900829 DOI: 10.1101/2023.01.25.525595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Trade-offs between metabolic and reproductive processes are important for survival, particularly in mammals that gestate their young. Puberty and reproduction, as energetically taxing life stages, are often gated by metabolic availability in animals with ovaries. How the nervous system coordinates these trade-offs is an active area of study. We identify somatostatin neurons of the tuberal nucleus (TNSST) as a node of the feeding circuit that alters feeding in a manner sensitive to metabolic and reproductive states in mice. Whereas chemogenetic activation of TNSST neurons increased food intake across sexes, selective ablation decreased food intake only in female mice during proestrus. Interestingly, this ablation effect was only apparent in animals with a low body mass. Fat transplantation and bioinformatics analysis of TNSST neuronal transcriptomes revealed white adipose as a key modulator of the effects of TNSST neurons on food intake. Together, these studies point to a mechanism whereby TNSST hypothalamic neurons modulate feeding by responding to varying levels of circulating estrogens differentially based on energy stores. This research provides insight into how neural circuits integrate reproductive and metabolic signals, and illustrates how gonadal steroid modulation of neuronal circuits can be context-dependent and gated by metabolic status.
Collapse
Affiliation(s)
- Megan G Massa
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
| | - Rachel L Scott
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
| | - Alexandra L Cara
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
| | - Laura R Cortes
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
| | - Norma P Sandoval
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
| | - Jae W Park
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
| | - Sahara Ali
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
| | - Leandro M Velez
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA
| | - Bethlehem Tesfaye
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
| | - Karen Reue
- Department of Human Genetics, David Geffen School of Medicine at University of California, Los Angeles, CA
| | - J Edward van Veen
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
| | - Marcus Seldin
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA
| | - Stephanie M Correa
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
| |
Collapse
|
23
|
Marsh ML, Oliveira MN, Vieira-Potter VJ. Adipocyte Metabolism and Health after the Menopause: The Role of Exercise. Nutrients 2023; 15:444. [PMID: 36678314 PMCID: PMC9862030 DOI: 10.3390/nu15020444] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/19/2023] Open
Abstract
Postmenopausal women represent an important target population in need of preventative cardiometabolic approaches. The loss of estrogen following the menopause eliminates protections against metabolic dysfunction, largely due to its role in the health and function of adipose tissue. In addition, some studies associate the menopause with reduced physical activity, which could potentially exacerbate the deleterious cardiometabolic risk profile accompanying the menopause. Meanwhile, exercise has adipocyte-specific effects that may alleviate the adverse impact of estrogen loss through the menopausal transition period and beyond. Exercise thus remains the best therapeutic agent available to mitigate menopause-associated metabolic dysfunction and represents a vital behavioral strategy to prevent and alleviate health decline in this population.
Collapse
|
24
|
Johnson CS, Mermelstein PG. The interaction of membrane estradiol receptors and metabotropic glutamate receptors in adaptive and maladaptive estradiol-mediated motivated behaviors in females. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 168:33-91. [PMID: 36868633 DOI: 10.1016/bs.irn.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Estrogen receptors were initially identified as intracellular, ligand-regulated transcription factors that result in genomic change upon ligand binding. However, rapid estrogen receptor signaling initiated outside of the nucleus was also known to occur via mechanisms that were less clear. Recent studies indicate that these traditional receptors, estrogen receptor α and estrogen receptor β, can also be trafficked to act at the surface membrane. Signaling cascades from these membrane-bound estrogen receptors (mERs) can rapidly alter cellular excitability and gene expression, particularly through the phosphorylation of CREB. A principal mechanism of neuronal mER action has been shown to occur through glutamate-independent transactivation of metabotropic glutamate receptors (mGlu), which elicits multiple signaling outcomes. The interaction of mERs with mGlu has been shown to be important in many diverse functions in females, including driving motivated behaviors. Experimental evidence suggests that a large part of estradiol-induced neuroplasticity and motivated behaviors, both adaptive and maladaptive, occurs through estradiol-dependent mER activation of mGlu. Herein we will review signaling through estrogen receptors, both "classical" nuclear receptors and membrane-bound receptors, as well as estradiol signaling through mGlu. We will focus on how the interactions of these receptors and their downstream signaling cascades are involved in driving motivated behaviors in females, discussing a representative adaptive motivated behavior (reproduction) and maladaptive motivated behavior (addiction).
Collapse
Affiliation(s)
- Caroline S Johnson
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Paul G Mermelstein
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States.
| |
Collapse
|
25
|
Strehle LD, Russart KLG, Burch VA, Grant CV, Pyter LM. Ovarian status modulates endocrine and neuroinflammatory responses to a murine mammary tumor. Am J Physiol Regul Integr Comp Physiol 2022; 323:R432-R444. [PMID: 35993563 PMCID: PMC9512114 DOI: 10.1152/ajpregu.00124.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 08/01/2022] [Accepted: 08/05/2022] [Indexed: 11/22/2022]
Abstract
Patients with breast cancer have increased circulating inflammatory markers and mammary tumors increase neuroinflammation in rodent models. Menopausal status is not only important in the context of breast cancer as circulating estrogen influences tumor progression, but also because estrogen is anti-inflammatory and an essential modulator of endocrine function in the brain and body. Here, we manipulated "menopause" status (ovary-intact and ovariectomized) in an estrogen receptor (ER)+ mouse mammary tumor model to determine the extent to which ovarian status modulates: 1) tumor effects on estrogen concentrations and signaling in the brain, 2) tumor effects on estrogen-associated neurobiology and inflammation, and 3) the ability for tumor resection to resolve the effects of a tumor. We hypothesized that reduced circulating estradiol (E2) after an ovariectomy exacerbates tumor-induced peripheral and central inflammation. Notably, we observed ovarian-dependent modulation on tumor-induced peripheral outcomes, including E2-dependent processes and, to a lesser degree, circulating inflammatory markers. In the brain, ovariectomy exacerbated neuroinflammatory markers in select brain regions and modulated E2-related neurobiology due to a tumor and/or resection. Overall, our data suggest that ovarian status has moderate implications for tumor-induced alterations in neuroendocrinology and neuroinflammation and mild effects on peripheral inflammatory outcomes in this murine mammary tumor model.
Collapse
Affiliation(s)
- Lindsay D Strehle
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio
| | - Kathryn L G Russart
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Valerie A Burch
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio
| | - Corena V Grant
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio
| | - Leah M Pyter
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, Ohio
- Department of Neuroscience, The Ohio State University, Columbus, Ohio
| |
Collapse
|
26
|
Ghosh-Swaby OR, Reichelt AC, Sheppard PAS, Davies J, Bussey TJ, Saksida LM. Metabolic hormones mediate cognition. Front Neuroendocrinol 2022; 66:101009. [PMID: 35679900 DOI: 10.1016/j.yfrne.2022.101009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 05/18/2022] [Accepted: 06/02/2022] [Indexed: 11/16/2022]
Abstract
Recent biochemical and behavioural evidence indicates that metabolic hormones not only regulate energy intake and nutrient content, but also modulate plasticity and cognition in the central nervous system. Disruptions in metabolic hormone signalling may provide a link between metabolic syndromes like obesity and diabetes, and cognitive impairment. For example, altered metabolic homeostasis in obesity is a strong determinant of the severity of age-related cognitive decline and neurodegenerative disease. Here we review the evidence that eating behaviours and metabolic hormones-particularly ghrelin, leptin, and insulin-are key players in the delicate regulation of neural plasticity and cognition. Caloric restriction and antidiabetic therapies, both of which affect metabolic hormone levels can restore metabolic homeostasis and enhance cognitive function. Thus, metabolic hormone pathways provide a promising target for the treatment of cognitive decline.
Collapse
Affiliation(s)
- Olivia R Ghosh-Swaby
- Schulich School of Medicine and Dentistry, Neuroscience Program, Western University, London, ON, Canada
| | - Amy C Reichelt
- Faculty of Health and Medical Sciences, Adelaide Medical School, Adelaide, Australia
| | - Paul A S Sheppard
- Schulich School of Medicine and Dentistry, Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Jeffrey Davies
- Swansea University Medical School, Swansea University, Swansea, UK
| | - Timothy J Bussey
- Schulich School of Medicine and Dentistry, Neuroscience Program, Western University, London, ON, Canada; Schulich School of Medicine and Dentistry, Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Lisa M Saksida
- Schulich School of Medicine and Dentistry, Neuroscience Program, Western University, London, ON, Canada; Schulich School of Medicine and Dentistry, Department of Physiology and Pharmacology, Western University, London, ON, Canada.
| |
Collapse
|
27
|
Oliveira LA, Della Lucia CM, Rezende FAC, Ferreira LG, Anastácio LR, Souza TCDM, Daniel MM, Liboredo JC. Food Craving and Its Associated Factors during COVID-19 Outbreak in Brazil. AMERICAN JOURNAL OF HEALTH EDUCATION 2022. [DOI: 10.1080/19325037.2022.2071360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
28
|
Maury-Sintjago E, Rodríguez-Fernández A, Parra-Flores J, Ruíz-De la Fuente M. Obese Women Have a High Carbohydrate Intake without Changes in the Resting Metabolic Rate in the Luteal Phase. Nutrients 2022; 14:1997. [PMID: 35631136 PMCID: PMC9147294 DOI: 10.3390/nu14101997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/02/2022] [Accepted: 05/03/2022] [Indexed: 02/07/2023] Open
Abstract
Hormonal changes are caused by the menstrual cycle phases, which influence resting metabolic rate and eating behavior. The aim of the study was to determine resting metabolic rate (RMR) and its association with dietary intake according to the menstrual cycle phase in lean and obese Chilean women. This cross-sectional analytical study included 30 adult women (15 lean and 15 with obesity). Body composition was measured with a tetrapolar bioelectrical impedance meter. Nutritional status was determined by adiposity. A 24-h recall of three nonconsecutive days verifies dietary intake. The RMR was measured by indirect calorimetry. All measurements were performed in both the follicular and luteal phases of the menstrual cycle. Statistical analyses were performed with STATA software at a significance level, which was α = 0.05. The RMR (β = 121.6 kcal/d), temperature (β = 0.36 °C), calorie intake (β = 317.1 kcal/d), and intake of lipids (β = 13.8 g/d) were associated with the luteal phase in lean women. Only extracellular water (β = 1.11%) and carbohydrate consumption (β = 45.2 g/d) were associated in women with obesity. Lean women showed increased RMR, caloric intake, and lipid intake during the luteal phase. For women with obesity, carbohydrate intake increased but not RMR.
Collapse
Affiliation(s)
- Eduard Maury-Sintjago
- Department of Nutrition and Public Health, Universidad del Bío-Bío, Chillan 3780000, Chile; (E.M.-S.); (A.R.-F.); (J.P.-F.)
- GABO Grupo de Investigación en Auxología, Bioantropología y Ontogenia, FACSA, Universidad del Bío-Bío, Chillan 3780000, Chile
| | - Alejandra Rodríguez-Fernández
- Department of Nutrition and Public Health, Universidad del Bío-Bío, Chillan 3780000, Chile; (E.M.-S.); (A.R.-F.); (J.P.-F.)
- GABO Grupo de Investigación en Auxología, Bioantropología y Ontogenia, FACSA, Universidad del Bío-Bío, Chillan 3780000, Chile
| | - Julio Parra-Flores
- Department of Nutrition and Public Health, Universidad del Bío-Bío, Chillan 3780000, Chile; (E.M.-S.); (A.R.-F.); (J.P.-F.)
- GABO Grupo de Investigación en Auxología, Bioantropología y Ontogenia, FACSA, Universidad del Bío-Bío, Chillan 3780000, Chile
| | - Marcela Ruíz-De la Fuente
- Department of Nutrition and Public Health, Universidad del Bío-Bío, Chillan 3780000, Chile; (E.M.-S.); (A.R.-F.); (J.P.-F.)
- GABO Grupo de Investigación en Auxología, Bioantropología y Ontogenia, FACSA, Universidad del Bío-Bío, Chillan 3780000, Chile
| |
Collapse
|
29
|
The Impact of Binge-Like Palatable Food Intake on the Endogenous Glucagon-Like Peptide-1 System in Female Rats. Behav Brain Res 2022; 428:113869. [PMID: 35378108 DOI: 10.1016/j.bbr.2022.113869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 03/08/2022] [Accepted: 03/29/2022] [Indexed: 11/21/2022]
Abstract
Binge eating involves consumption of large amounts of food and a loss of control over the amount consumed. The incidence of binge eating disorder is higher in females than males, hinting at important sex differences in binge eating behavior, but the neural underpinnings of binge eating still remain unresolved. Recent work in male rats has shown that a history of binge-like palatable food intake suppresses hindbrain expression of preproglucagon (PPG), the precursor for glucagon-like peptide-1 (GLP-1). Given the roles of GLP-1 in reducing feeding and food reward, this could be a mechanism underlying binge-like eating in rodents. However, whether similar effects occur in female rats is unknown. Here, we tested the hypothesis that a history of binge-like palatable food intake in female rats would reduce PPG expression in the nucleus tractus solitarius (NTS), a key central site of GLP-1 production. Female rats given access to vegetable shortening every fourth day (4D) engaged in binge-like feeding, demonstrated by consuming significantly more shortening during the first hour of fat access compared to counterparts with ad libitum (AL) fat access. After several weeks of fat access under these schedules, PPG and GLP-1 receptor (GLP-1R) expression were measured in the NTS and ileum. Surprisingly, and in contrast to previous findings in male rats, there were no significant differences in expression of PPG or GLP-1R in either site in 4D versus AL rats, nor were there effects on plasma GLP-1 levels. These findings highlight key differences in the effects of binge-like intake on the central GLP-1 system in female compared to male rats.
Collapse
|
30
|
Maher EE, Overby PF, Bull AH, Beckmann JS, Leyrer-Jackson JM, Koebele SV, Bimonte-Nelson HA, Gipson CD. Natural and synthetic estrogens specifically alter nicotine demand and cue-induced nicotine seeking in female rats. Neuropharmacology 2021; 198:108756. [PMID: 34416269 DOI: 10.1016/j.neuropharm.2021.108756] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/05/2021] [Accepted: 08/14/2021] [Indexed: 11/17/2022]
Abstract
Women have more difficulty maintaining smoking cessation than men, and experience greater withdrawal symptomatology as well as higher prevalence of relapse. Further, currently available treatments for smoking cessation, such as the nicotine patch and varenicline, have been shown to be less effective in women. Fluctuations in ovarian hormones across the menstrual cycle can affect craving and smoking relapse propensity. In addition, many women who smoke use some form of oral contraceptives, which most often contain ethinyl estradiol (EE), a synthetic, orally bio-available estrogen that is currently prescribed to women chronically and has been shown to alter smoking reward in women. The current study examined the impact of 17β-estradiol (E2), the prominent endogenous form of the steroid hormone estrogen, as well as EE, on nicotine self-administration, demand, and reinstatement following ovariectomy (OVX) or sham surgery. OVX vehicle-treated female rats consumed less nicotine, had lower intensity of demand, and reinstated less compared to sham vehicle-treated female rats. OVX-E2 and OVX-EE treatment groups showed a rebound of nicotine intake later in training, and Q0 levels of consumption were partially rescued in both groups. Further, E2 but not EE reversed the abolishment of reinstated nicotine seeking induced by OVX. Taken together, these results demonstrate that natural and synthetic estrogens play a critical role in mediating the neurobehavioral effects of nicotine, and future studies are essential for our understanding of how synthetic hormones contained within oral contraceptives interact with smoking.
Collapse
Affiliation(s)
- Erin E Maher
- Department of Family and Community Medicine, University of Kentucky, Lexington, KY, USA
| | - Paula F Overby
- Department of Psychology, Arizona State University, Tempe, AZ, USA
| | - Amanda H Bull
- Department of Psychology, Arizona State University, Tempe, AZ, USA
| | - Joshua S Beckmann
- Department of Psychology, University of Kentucky, Lexington, KY, USA
| | | | - Stephanie V Koebele
- Department of Psychology, Arizona State University, Tempe, AZ, USA; Arizonia Alzheimer's Consortium, Phoenix, AZ, USA
| | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ, USA; Arizonia Alzheimer's Consortium, Phoenix, AZ, USA
| | - Cassandra D Gipson
- Department of Family and Community Medicine, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
31
|
Yeh JH, Tung YT, Yeh YS, Chien YW. Effects of Dietary Fatty Acid Composition on Lipid Metabolism and Body Fat Accumulation in Ovariectomized Rats. Nutrients 2021; 13:nu13062022. [PMID: 34208400 PMCID: PMC8231186 DOI: 10.3390/nu13062022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/05/2021] [Accepted: 06/10/2021] [Indexed: 01/21/2023] Open
Abstract
Background: Obesity is a state of excess energy storage resulting in body fat accumulation, and postmenopausal obesity is a rising issue. In this study using ovariectomized (OVX) rats, we mimicked low estrogen levels in a postmenopausal state in order to investigate the effects of different amounts and types of dietary fatty acids on body fat accumulation and body lipid metabolism. Methods: At 9 weeks of age, rats (n = 40) were given an ovariectomy, eight of which were sham-operated to serve as a control group (S). We then divided OVX rats into four different intervention groups: diet with 5% soybean oil (C), and diet with 5% (L), 15% (M), and 20% (H) (w/w) experimental oil, containing 60% monounsaturated fatty acids (MUFAs) and with a polyunsaturated/saturated fatty acid (P/S) ratio of 5. Results: After OVX, compared to the S group, the C group showed significantly higher body weight, and insulin and leptin levels. Compared to the C group, the H group had lower hepatic triglyceride level and FAS enzyme activity, and higher hepatic ACO and CPT-1 gene expressions and enzyme activities. Conclusions: An OVX leads to severe weight gain and lipid metabolism abnormalities, while according to previous studies, high fat diet may worsen the situation. However, during our experiment, we discovered that the experimental oil mixture with 60% MUFAs and P/S = 5 may ameliorate these imbalances.
Collapse
Affiliation(s)
- Jhih-Han Yeh
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 11031, Taiwan;
| | - Yu-Tang Tung
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung 40227, Taiwan;
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Sheng Yeh
- Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO 63112, USA;
| | - Yi-Wen Chien
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 11031, Taiwan;
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei 11031, Taiwan
- Nutrition Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Research Center of Geriatric Nutrition, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan
- Correspondence: ; Tel.: +886-2-2736-1661 (ext. 6556); Fax: +886-2-2737-3112
| |
Collapse
|
32
|
Chen Q, Wang B, Wang S, Qian X, Li X, Zhao J, Zhang H, Chen W, Wang G. Modulation of the Gut Microbiota Structure with Probiotics and Isoflavone Alleviates Metabolic Disorder in Ovariectomized Mice. Nutrients 2021; 13:1793. [PMID: 34070274 PMCID: PMC8225012 DOI: 10.3390/nu13061793] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/12/2021] [Accepted: 05/21/2021] [Indexed: 02/06/2023] Open
Abstract
The decrease in ovarian hormone secretion that occurs during menopause results in an increase in body weight and adipose tissue mass. Probiotics and soy isoflavones (SIFs) could affect the gut microbiota and exert anti-obesity effects. The objective of this study was to investigate the effects of probiotics and a diet containing SIF (SIF diet) on ovariectomized mice with menopausal obesity, including the gut microbiome. The results demonstrate that Bifidobacterium longum 15M1 can reverse menopausal obesity, whilst the combination of Lactobacillus plantarum 30M5 and a SIF diet was more effective in alleviating menopausal lipid metabolism disorder than either components alone. Probiotics and SIFs play different anti-obesity roles in menopausal mice. Furthermore, 30M5 alters the metabolites of the gut microbiota that increase the circulating estrogen level, upregulates the expression of estrogen receptor α in abdominal adipose tissue and improves the production of short-chain fatty acids (SCFAs). A SIF diet can significantly alter the structure of the fecal bacterial community and enrich the pathways related to SCFAs production. Moreover, 30M5 and a SIF diet acted synergistically to effectively resolve abnormal serum lipid levels in ovariectomized mice, and these effects appear to be associated with regulation of the diversity and structure of the intestinal microbiota to enhance SCFAs production and promote estrogen circulation.
Collapse
Affiliation(s)
- Qian Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (Q.C.); (B.W.); (S.W.); (X.Q.); (X.L.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Botao Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (Q.C.); (B.W.); (S.W.); (X.Q.); (X.L.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Shunhe Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (Q.C.); (B.W.); (S.W.); (X.Q.); (X.L.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xin Qian
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (Q.C.); (B.W.); (S.W.); (X.Q.); (X.L.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xiu Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (Q.C.); (B.W.); (S.W.); (X.Q.); (X.L.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (Q.C.); (B.W.); (S.W.); (X.Q.); (X.L.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (Q.C.); (B.W.); (S.W.); (X.Q.); (X.L.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
- Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (Q.C.); (B.W.); (S.W.); (X.Q.); (X.L.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Gang Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (Q.C.); (B.W.); (S.W.); (X.Q.); (X.L.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| |
Collapse
|
33
|
Yamada C. Relationship between Orexigenic Peptide Ghrelin Signal, Gender Difference and Disease. Int J Mol Sci 2021; 22:ijms22073763. [PMID: 33916403 PMCID: PMC8038632 DOI: 10.3390/ijms22073763] [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: 03/08/2021] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 12/11/2022] Open
Abstract
Growth hormone secretagogue receptor 1a (GHS-R1a), which is one of the G protein-coupled receptors (GPCRs), is involved in various physiological actions such as energy consumption, growth hormone secretion promoting action, and cardiovascular protective action. The ligand was searched for as an orphan receptor for a while, but the ligand was found to be acylated ghrelin (ghrelin) discovered by Kangawa and Kojima et al. in 1999. Recently, it has also been reported that dysregulation of GHS-R1a mediates reduced feeding in various diseases. On the other hand, since the physiological effects of ghrelin have been studied exclusively in male mice, few studies have been conducted on gender differences in ghrelin reactivity. In this review, we describe (1) the characteristics of GHS-R1a, (2) the role of ghrelin in hypophagia due to stress or anticancer drugs, and (3) the gender differences in the physiological effects of GHS-R1a and the influence of stress on it.
Collapse
Affiliation(s)
- Chihiro Yamada
- Tsumura Kampo Research Laboratories, Tsumura & Co., Ibaraki 300-1192, Japan
| |
Collapse
|
34
|
Dahir NS, Calder AN, McKinley BJ, Liu Y, Gilbertson TA. Sex differences in fat taste responsiveness are modulated by estradiol. Am J Physiol Endocrinol Metab 2021; 320:E566-E580. [PMID: 33427045 PMCID: PMC7988783 DOI: 10.1152/ajpendo.00331.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Sex as a biological variable has been the focus of increasing interest. Relatively few studies have focused, however, on differences in peripheral taste function between males and females. Nonetheless, there are reports of sex-dependent differences in chemosensitivity in the gustatory system. The involvement of endogenous changes in ovarian hormones has been suggested to account for taste discrepancies. Additionally, whether sex differences exist in taste receptor expression, activation, and subsequent signaling pathways that may contribute to different taste responsiveness is not well understood. In this study, we show the presence of both the nuclear and plasma membrane forms of estrogen receptor (ER) mRNA and protein in mouse taste cells. Furthermore, we provide evidence that estrogen increases taste cell activation during the application of fatty acids, the chemical cue for fat taste, in taste receptor cells. We found that genes important for the transduction pathway of fatty acids vary between males and females and that these differences also exist across the various taste papillae. In vivo support for the effect of estrogens in taste cells was provided by comparing the fatty acid responsiveness in male, intact female, and ovariectomized (OVX) female mice with and without hormone replacement. In general, females detected fatty acids at lower concentrations, and the presence of circulating estrogens increased this apparent fat taste sensitivity. Taken together, these data indicate that increased circulating estrogens in the taste system may play a significant role in physiology and chemosensory cellular activation and, in turn, may alter taste-driven behavior.NEW & NOTEWORTHY Using molecular, cellular, and behavioral analyses, this study shows that sex differences occur in fat taste in a mouse model. Female mice are more responsive to fatty acids, leading to an overall decrease in intake and fatty acid preference. These differences are linked to sex hormones, as estradiol enhances taste cell responsiveness to fatty acids during periods of low circulating estrogen following ovariectomy and in males. Estradiol is ineffective in altering fatty acid signaling during a high-estrogen period and in ovariectomized mice on hormone replacement. Thus, taste receptor cells are a direct target for actions of estrogen, and there are multiple receptors with differing patterns of expression in taste cells.
Collapse
Affiliation(s)
- Naima S Dahir
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida
- Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, Florida
| | - Ashley N Calder
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida
- Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, Florida
| | | | - Yan Liu
- Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, Florida
| | - Timothy A Gilbertson
- Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, Florida
| |
Collapse
|
35
|
Mikhail ME, Keel PK, Burt SA, Sisk CL, Neale M, Boker S, Klump KL. Trait negative affect interacts with ovarian hormones to predict risk for emotional eating. Clin Psychol Sci 2021; 9:114-128. [PMID: 33758690 PMCID: PMC7983867 DOI: 10.1177/2167702620951535] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Ovarian hormones significantly influence dysregulated eating in females. However, most women do not develop appreciable disordered eating, suggesting that ovarian hormones may not affect all women equally. We examined whether individual differences in trait negative affect (NA) moderate ovarian hormone-dysregulated eating associations in 446 women who provided saliva samples for hormone measurements and ratings of NA and emotional eating daily for 45 consecutive days. Women were at greatest risk for emotional eating when they had high trait NA and experienced a hormonal milieu characterized by low estradiol or high progesterone. While effects were evident in all women, the combination of high trait NA and high progesterone was particularly risky for women with a history of clinically significant binge eating episodes. These findings provide initial evidence that affective and hormonal risk interact to promote emotional eating, and that effects may be amplified in women with clinically significant binge eating.
Collapse
Affiliation(s)
- Megan E. Mikhail
- Department of Psychology, Michigan State University, East Lansing, MI 48824-1116
| | - Pamela K. Keel
- Department of Psychology, Florida State University, Tallahassee, FL 32306-4301
| | - S. Alexandra Burt
- Department of Psychology, Michigan State University, East Lansing, MI 48824-1116
| | - Cheryl L. Sisk
- Neuroscience Program, Michigan State University, East Lansing, MI 48824-1116
| | - Michael Neale
- Departments of Psychiatry, Human Genetics, and Psychology, Virginia Commonwealth University, Richmond, VA 23298
| | - Steven Boker
- Department of Psychology, University of Virginia, Charlottesville, VA 22904-4400
| | - Kelly L. Klump
- Department of Psychology, Michigan State University, East Lansing, MI 48824-1116
| |
Collapse
|
36
|
Mustac T, Yuabov A, Macanian J, Aminov S, Fazylov D, Lulu EB, Nashed M, Albakry A, Jean-Philippe-Morisset B, Bodnar RJ. Acute d-fenfluramine, but not fluoxetine decreases sweet intake in BALB/c, C57BL/6 and SWR inbred mouse strains. Physiol Behav 2020; 224:113029. [PMID: 32590091 DOI: 10.1016/j.physbeh.2020.113029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/21/2020] [Accepted: 06/22/2020] [Indexed: 11/29/2022]
Abstract
Dopamine, opioid and muscarinic receptor antagonists differentially reduce sucrose and saccharin intakes across inbred mouse strains. Whereas these systems stimulate sweet intake, serotonin signaling inhibits food intake. The present study examined whether fluoxetine (0.1-10 mg/kg) or d-fenfluramine (0.1-6 mg/kg) differentially inhibited sucrose or saccharin intake in BALB/c, C57BL/6 and SWR mice. Fluoxetine marginally altered sucrose intake in all strains. d-fenfluramine significantly, but quite similarly reduced (ID40) sucrose and saccharin intake in BALB/c (5.7 vs. 5.8 mg/kg), C57BL/6 (4.4 vs. 4.3 mg/kg) and SWR (4.6 vs. 5.6 mg/kg) mice, suggesting serotonin-induced inhibition of orosensory mechanisms in all three inbred mouse strains.
Collapse
Affiliation(s)
- Tatjana Mustac
- Department of Psychology, Queens College, City University of New York (CUNY)
| | - Asnat Yuabov
- Department of Psychology, Queens College, City University of New York (CUNY)
| | - Jason Macanian
- Department of Psychology, Queens College, City University of New York (CUNY)
| | - Sonya Aminov
- Department of Psychology, Queens College, City University of New York (CUNY)
| | - David Fazylov
- Department of Psychology, Queens College, City University of New York (CUNY)
| | - Eden Ben Lulu
- Department of Psychology, Queens College, City University of New York (CUNY)
| | - Mirna Nashed
- Department of Psychology, Queens College, City University of New York (CUNY)
| | - Ahmed Albakry
- Department of Psychology, Queens College, City University of New York (CUNY)
| | | | - Richard J Bodnar
- Department of Psychology, Queens College, City University of New York (CUNY); CUNY Neuroscience Collaborative and Psychology Doctoral Program, CUNY Graduate Center, New York, NY, USA.
| |
Collapse
|
37
|
Choo E, Koh A, Goodman J, Bushnell J, Mielke-Maday H, Merte B, Dando R. Decrease in sweet taste response and T1R3 sweet taste receptor expression in pregnant mice highlights a potential mechanism for increased caloric consumption in pregnancy. Physiol Behav 2020; 228:113191. [PMID: 33007356 DOI: 10.1016/j.physbeh.2020.113191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 08/11/2020] [Accepted: 09/28/2020] [Indexed: 01/22/2023]
Abstract
While much is known on how the maternal diet affects offspring fitness, less is known on the role of taste in guiding and promoting food intake during this crucial period. Women have intense food cravings and exhibit altered taste preferences during pregnancy, however the mechanistic details underlying these changes are presently unclear. We performed longitudinal brief-access taste testing in female mice before, during, and after pregnancy, along with quantitative PCR on taste buds and morphological analysis of taste tissues from pregnant and non-pregnant mice. Sucrose licking response decreased progressively during pregnancy compared to that prior to mating, with partial recovery in the post-partum period. No change in taste morphology was evident between pregnant and non-pregnant mice, however a notable decrease in T1R3 sweet taste receptor mRNA expression was recorded in pregnant dams. We conclude that altered taste preferences during pregnancy likely result from changes in the expression profile of taste buds in the mother, which may promote a less healthy diet while expecting.
Collapse
Affiliation(s)
- Ezen Choo
- Biomedical & Biological Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| | - Anna Koh
- Department of Food Science, Cornell University, Ithaca, NY 14853
| | - Jason Goodman
- Department of Food Science, Cornell University, Ithaca, NY 14853
| | | | | | - Bryan Merte
- College of Arts and Sciences, Cornell University, Ithaca, NY 14853
| | - Robin Dando
- Department of Food Science, Cornell University, Ithaca, NY 14853.
| |
Collapse
|
38
|
Navarro VM. Metabolic regulation of kisspeptin - the link between energy balance and reproduction. Nat Rev Endocrinol 2020; 16:407-420. [PMID: 32427949 PMCID: PMC8852368 DOI: 10.1038/s41574-020-0363-7] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/16/2020] [Indexed: 12/17/2022]
Abstract
Hypothalamic kisspeptin neurons serve as the nodal regulatory centre of reproductive function. These neurons are subjected to a plethora of regulatory factors that ultimately affect the release of kisspeptin, which modulates gonadotropin-releasing hormone (GnRH) release from GnRH neurons to control the reproductive axis. The presence of sufficient energy reserves is critical to achieve successful reproduction. Consequently, metabolic factors impose a very tight control over kisspeptin synthesis and release. This Review offers a synoptic overview of the different steps in which kisspeptin neurons are subjected to metabolic regulation, from early developmental stages to adulthood. We cover an ample array of known mechanisms that underlie the metabolic regulation of KISS1 expression and kisspeptin release. Furthermore, the novel role of kisspeptin neurons as active players within the neuronal circuits that govern energy balance is discussed, offering evidence of a bidirectional role of these neurons as a nexus between metabolism and reproduction.
Collapse
Affiliation(s)
- Víctor M Navarro
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Harvard Graduate Program in Neuroscience, Boston, MA, USA.
| |
Collapse
|
39
|
Caldwell AE, Zaman A, Ostendorf DM, Pan Z, Swanson BB, Phelan S, Wyatt HR, Bessesen DH, Melanson EL, Catenacci VA. Impact of Combined Hormonal Contraceptive Use on Weight Loss: A Secondary Analysis of a Behavioral Weight-Loss Trial. Obesity (Silver Spring) 2020; 28:1040-1049. [PMID: 32441474 PMCID: PMC7556729 DOI: 10.1002/oby.22787] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 02/24/2020] [Accepted: 02/27/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE This study aimed to perform a preliminary investigation of the impact of combined hormonal contraceptive (CHC) use on weight loss during an 18-month behavioral weight-loss trial. METHODS Adults (n = 170; 18-55 years; BMI 27-42 kg/m2 ) received a weight-loss intervention that included a reduced-calorie diet, a progressive exercise prescription, and group-based behavioral support. Premenopausal women (n = 110) were classified as CHC users (CHC, n = 17) or non-CHC users (non-CHC, n = 93). Changes in weight were examined within groups using a linear mixed model, adjusted for age and randomized group assignment. RESULTS At 6 M, weight was reduced from baseline in both CHC (mean, -6.7 kg; 95% CI: -9.8 to -3.7 kg) and non-CHC (-9.1 kg; -9.1 to -6.4 kg). Between 6 and 18 M, CHC regained weight (4.9 kg; 0.9 to 8.9 kg), while weight remained relatively unchanged in non-CHC (-0.1 kg; -1.8 to 1.6 kg). At 18 M, weight was relatively unchanged from baseline in CHC (-1.8 kg; -7.3 to 3.6 kg) and was reduced from baseline in non-CHC (-7.9 kg; -10.2 to -5.5 kg). CONCLUSIONS In this secondary data analysis, CHC use was associated with weight regain after initial weight loss. Prospective studies are needed to further understand the extent to which CHC use influences weight loss and maintenance.
Collapse
Affiliation(s)
- Ann E Caldwell
- Anschutz Health and Wellness Center, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Adnin Zaman
- Anschutz Health and Wellness Center, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Danielle M Ostendorf
- Anschutz Health and Wellness Center, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Zhaoxing Pan
- Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Bryan B Swanson
- Department of Chemistry and Biochemistry, Colorado College, Colorado Springs, Colorado, USA
| | - Suzanne Phelan
- Kinesiology and Public Health Department, California Polytechnic State University, San Luis Obispo, California, USA
| | - Holly R Wyatt
- Anschutz Health and Wellness Center, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Daniel H Bessesen
- Anschutz Health and Wellness Center, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Edward L Melanson
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Division of Geriatric Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Geriatric Research, Education, and Clinical Center, Eastern Colorado Veterans Affairs Medical Center, Denver, Colorado, USA
| | - Victoria A Catenacci
- Anschutz Health and Wellness Center, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
40
|
Massa MG, Correa SM. Sexes on the brain: Sex as multiple biological variables in the neuronal control of feeding. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165840. [PMID: 32428559 DOI: 10.1016/j.bbadis.2020.165840] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 05/05/2020] [Accepted: 05/13/2020] [Indexed: 12/21/2022]
Abstract
Neuronal interactions at the level of vagal, homeostatic, and hedonic circuitry work to regulate the neuronal control of feeding. This integrative system appears to vary across sex and gender in the animal and human worlds. Most feeding research investigating these variations across sex and gender focus on how the organizational and activational mechanisms of hormones contribute to these differences. However, in limited studies spanning both the central and peripheral nervous systems, sex differences in feeding have been shown to manifest not just at the level of the hormonal, but also at the chromosomal, epigenetic, cellular, and even circuitry levels to alter food intake. In this review, we provide a brief orientation to the current understanding of how these neuronal systems interact before dissecting selected studies from the recent literature to exemplify how feeding physiology at all levels can be affected by the various components of sex.
Collapse
Affiliation(s)
- Megan G Massa
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, United States of America; Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, Los Angeles, CA, United States of America; Neuroscience Interdepartmental Doctoral Program, University of California, Los Angeles, CA, United States of America.
| | - Stephanie M Correa
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, United States of America; Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, Los Angeles, CA, United States of America.
| |
Collapse
|
41
|
Bhardwaj P, Ikeda T, Zhou XK, Wang H, Zheng XE, Giri DD, Elemento O, Verma A, Miyazawa M, Mukherjee S, Falcone DJ, Wendel NK, Scherr DS, Dannenberg AJ. Supplemental estrogen and caloric restriction reduce obesity-induced periprostatic white adipose inflammation in mice. Carcinogenesis 2020; 40:914-923. [PMID: 31067318 DOI: 10.1093/carcin/bgz088] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 04/13/2019] [Accepted: 05/07/2019] [Indexed: 01/22/2023] Open
Abstract
Obesity is associated with an increased incidence of high-grade prostate cancer (PC) and worse prognosis for PC patients. Recently, we showed in men that obesity-related periprostatic white adipose tissue (WAT) inflammation, characterized by macrophages surrounding dead or dying adipocytes forming crown-like structures, was associated with high-grade PC. Possibly, interventions that suppress periprostatic WAT inflammation will improve outcomes for men with PC. Here, we tested the hypothesis that supplemental 17β-estradiol (E2) could decrease periprostatic WAT inflammation in obese male mice. Mice were fed a high-fat diet to induce periprostatic WAT inflammation before being treated with supplemental E2. E2 supplementation suppressed caloric intake, induced weight loss, decreased periprostatic WAT inflammation and downregulated the expression of genes linked to inflammation including Cd68, Mcp1 and Tnf. Similar to the effects of E2 supplementation, treatment with diethylstilbestrol, a synthetic estrogen, also suppressed caloric intake and reduced periprostatic WAT inflammation. To determine whether the observed effects of supplemental estrogen could be reproduced by caloric restriction (CR) alone, obese mice were put on a 30% CR diet. Like estrogen treatment, CR was effective in reducing body weight, periprostatic WAT inflammation and the expression of pro-inflammatory genes. Transcriptomic analyses of periprostatic fat showed that obesity was associated with enrichment in inflammatory response pathways, which were normalized by both supplemental E2 and CR. Taken together, these findings strengthen the rationale for future efforts to determine whether either CR or supplemental estrogen will decrease periprostatic WAT inflammation and thereby improve outcomes for men with PC.
Collapse
Affiliation(s)
- Priya Bhardwaj
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Takahiro Ikeda
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Xi Kathy Zhou
- Department of Healthcare Policy and Research, Weill Cornell Medicine, New York, NY, USA
| | - Hanhan Wang
- Department of Healthcare Policy and Research, Weill Cornell Medicine, New York, NY, USA
| | - Xi Emily Zheng
- Department of Healthcare Policy and Research, Weill Cornell Medicine, New York, NY, USA
| | - Dilip D Giri
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Olivier Elemento
- Departments of Physiology and Biophysics, Computational Biomedicine and Caryl and Israel Englander Institute for Precision Medicine
| | - Akanksha Verma
- Departments of Physiology and Biophysics, Computational Biomedicine and Caryl and Israel Englander Institute for Precision Medicine
| | - Miki Miyazawa
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | - Domenick J Falcone
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Nils K Wendel
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Douglas S Scherr
- Department of Urology, Weill Cornell Medicine-New York Presbyterian, New York, NY, USA
| | | |
Collapse
|
42
|
Maske CB, Coiduras II, Ondriezek ZE, Terrill SJ, Williams DL. Intermittent High-Fat Diet Intake Reduces Sensitivity to Intragastric Nutrient Infusion and Exogenous Amylin in Female Rats. Obesity (Silver Spring) 2020; 28:942-952. [PMID: 32237211 PMCID: PMC7180114 DOI: 10.1002/oby.22779] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 02/11/2020] [Accepted: 02/13/2020] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Intermittent (INT) access to a high-fat diet (HFD) can induce excessive-intake phenotypes in rodents. This study hypothesized that impaired satiation responses contribute to elevated intake in an INT-HFD access model. METHODS First, this study characterized the intake and meal patterns of female rats that were subjected to an INT HFD in which a 45% HFD was presented for 20 hours every fourth day. To examine nutrient-induced satiation, rats received intragastric infusions of saline or Ensure Plus prior to darkness-onset food access. A similar design was used to examine sensitivity to the satiating effect of amylin. This study then examined whether an INT HFD influences amylin-induced c-Fos in feeding-relevant brain areas. RESULTS Upon INT HFD access, rats consumed meals of larger size. The anorexic response to intragastric Ensure infusion and exogenous amylin treatment was blunted in INT rats on both chow-only and INT-HFD days of the diet regimen, compared with chow-maintained and continuous-HFD rats. An INT HFD did not influence amylin-induced c-Fos in the area postrema, nucleus of the solitary tract, and lateral parabrachial nucleus. CONCLUSIONS Impaired satiation responses, mediated in part by reduced sensitivity to amylin, may explain the elevated intake observed upon INT HFD access and may play a role in disorders of INT overconsumption, including binge eating.
Collapse
Affiliation(s)
- Calyn B Maske
- Program in Neuroscience, Department of Psychology, Florida State University, Tallahassee, Florida, USA
| | - Isabel I Coiduras
- Program in Neuroscience, Department of Psychology, Florida State University, Tallahassee, Florida, USA
| | - Zeleen E Ondriezek
- Program in Neuroscience, Department of Psychology, Florida State University, Tallahassee, Florida, USA
| | - Sarah J Terrill
- Program in Neuroscience, Department of Psychology, Florida State University, Tallahassee, Florida, USA
| | - Diana L Williams
- Program in Neuroscience, Department of Psychology, Florida State University, Tallahassee, Florida, USA
| |
Collapse
|
43
|
Kwak SY, Chung I, Kang J, Perakakis N, Yoo EH, Lee J, Jung HT, Mun BR, Choi WS, Kim OY, Kim S, Kim EK, Oh H, Mantzoros CS, Chung JH, Kim HS, Shin MJ. Sex specific effect of ATPase inhibitory factor 1 on body weight: studies in high fat diet induced obese mice and genetic association studies in humans. Metabolism 2020; 105:154171. [PMID: 32006557 DOI: 10.1016/j.metabol.2020.154171] [Citation(s) in RCA: 4] [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: 10/23/2019] [Revised: 01/21/2020] [Accepted: 01/24/2020] [Indexed: 10/25/2022]
Abstract
BACKGROUND Based on the metabolic effect of exogenous ATPase inhibitory factor 1 (IF1) on glucose metabolism, we tested whether IF1 treatment is effective in ameliorating weight gain and whether its effects are sex specific. METHODS HFD-fed C57BL/6 mice were treated with IF1 (5 mg/kg body weight, injected intraperitoneally). The underlying mechanisms of effect of IF1 on body weight were investigated in vitro and in vivo. Associations between genotypes of IF1 and obesity and relevant phenotype were further tested at the population level. RESULTS Chronic treatment with IF1 significantly decreased body weight gain by regulating food intake of HFD-fed male mice. IF1 activated the AKT/mTORC pathway and modulated the expression of appetite genes in the hypothalamus of HFD-fed male mice and its effect was confirmed in hypothalamic cell lines as well as hypothalamic primary cells. This required the interaction of IF1 with β-F1-ATPase on the plasma membrane of hypothalamic cells, which led to an increase in extracellular ATP production. In addition, IF1 treatment showed sympathetic nerve activation as measured by serum norepinephrine levels and UCP-1 expression in the subcutaneous fat of HFD-fed male mice. Notably, administration of recombinant IF1 to HFD-fed ovariectomized female mice showed remarkable reductions in food intake as well as body weight, which was not observed in wild-type 5-week female mice. Lastly, sex-specific genotype associations of IF1 with obesity prevalence and metabolic traits were demonstrated at the population level in humans. IF1 genetic variant (rs3767303) was significantly associated with lower prevalence of obesity and lower levels of body mass index, waist circumference, hemoglobin A1c, and glucose response area only in male participants. CONCLUSION IF1 is involved in weight regulation by controlling food intake and potentially sympathetic nerve activation in a sex-specific manner.
Collapse
Affiliation(s)
- So-Young Kwak
- Department of Public Health Sciences, BK21PLUS Program in Embodiment: Health-Society Interaction, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - InHyeok Chung
- Department of Public Health Sciences, BK21PLUS Program in Embodiment: Health-Society Interaction, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Joon Kang
- Department of Biotechnology, CHA University, Gyeonggi-do 11160, Republic of Korea
| | - Nikolaos Perakakis
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA 02215, USA
| | - Eun Hye Yoo
- Department of Public Health Sciences, BK21PLUS Program in Embodiment: Health-Society Interaction, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Juhee Lee
- Department of Public Health Sciences, BK21PLUS Program in Embodiment: Health-Society Interaction, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Hun Taek Jung
- Department of Biotechnology, CHA University, Gyeonggi-do 11160, Republic of Korea
| | - Bo-Ram Mun
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Won-Seok Choi
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Oh Yoen Kim
- Department of Food and Nutrition, Dong-A University, Busan 49315, Republic of Korea
| | - Seolsong Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea
| | - Eun-Kyoung Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea; Neurometabolomics Research Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea
| | - Hannah Oh
- Department of Public Health Sciences, BK21PLUS Program in Embodiment: Health-Society Interaction, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA 02215, USA
| | - Ji Hyung Chung
- Department of Biotechnology, CHA University, Gyeonggi-do 11160, Republic of Korea.
| | - Hyeon Soo Kim
- Department of Anatomy, Korea University College of Medicine, Seoul 02841, Republic of Korea.
| | - Min-Jeong Shin
- Department of Public Health Sciences, BK21PLUS Program in Embodiment: Health-Society Interaction, Graduate School, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
44
|
Butler MJ, Perrini AA, Eckel LA. Estradiol treatment attenuates high fat diet-induced microgliosis in ovariectomized rats. Horm Behav 2020; 120:104675. [PMID: 31923417 PMCID: PMC7117977 DOI: 10.1016/j.yhbeh.2020.104675] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 11/20/2019] [Accepted: 12/28/2019] [Indexed: 01/03/2023]
Abstract
Consumption of a high fat diet (HFD) increases circulating free fatty acids, which can enter the brain and promote a state of microgliosis, as defined by a change in microglia number and/or morphology. Most studies investigating diet-induced microgliosis have been conducted in male rodents despite well-documented sex differences in the neural control of food intake and neuroimmune signaling. This highlights the need to investigate how sex hormones may modulate the behavioral and cellular response to HFD consumption. Estradiol is of particular interest since it exerts a potent anorexigenic effect and has both anti-inflammatory and neuroprotective effects in the brain. As such, the aim of the current study was to investigate whether estradiol attenuates the development of HFD-induced microgliosis in female rats. Estradiol- and vehicle-treated ovariectomized rats were fed either a low-fat chow diet or a 60% HFD for 4 days, after which they were perfused and brain sections were processed via immunohistochemistry for microglia-specific Iba1 protein. Four days of HFD consumption promoted microgliosis, as measured via an increase in the number of microglia in the arcuate nucleus (ARC) of the hypothalamus and nucleus of the solitary tract (NTS), and a decrease in microglial branching in the ARC, NTS, lateral hypothalamus (LH), and ventromedial hypothalamus. Estradiol replacement attenuated the HFD-induced changes in microglia accumulation and morphology in the ARC, LH, and NTS. We conclude that estradiol has protective effects against HFD-induced microgliosis in a region-specific manner in hypothalamic and hindbrain areas implicated in the neural control of food intake.
Collapse
Affiliation(s)
- Michael J Butler
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, United States; Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, 43210, United States
| | - Alexis A Perrini
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, United States
| | - Lisa A Eckel
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, United States.
| |
Collapse
|
45
|
Abstract
Neuroinflammation confers changes in brain function (i.e., behavior) that are hypothesized to be adaptive in the short-term, but detrimental (e.g., depression, anxiety) if they persist. Both peripheral tumor growth (outside of the brain) and natural aging independently cause neuroinflammation in rodents, which is corroborated by clinical studies. Mammary tumor effects on neuroinflammation and behavior, however, are typically studied in young rodents, whereas most breast cancer patients are middle-aged. Therefore, the existing literature likely underestimates the resulting neuroinflammation that may occur in clinical cancer populations. The present study tested the hypothesis that aging exacerbates mammary tumor-induced neuroinflammation in female mice. Aging (16 months and ovariectomized) increased body and spleen masses, whereas tumors grew faster and increased spleen mass in young mice (12 weeks) only. Tumors (IL-6, IL-10, TNFα, MCP-1, CXCL1, IP-10) and aging (IL-10, IFNγ) independently increased circulating inflammatory markers, although these variables were only significantly additive in one case (TNFα). In contrast to our prediction, the interaction between tumors and aging resulted in reduced mRNA and protein expression of select inflammatory markers in the hippocampus of tumor-bearing aged mice relative to aged controls. These results indicate that tumors reduce inflammatory activation in the brains of aged mice, a deficit that is likely disadvantageous. Further understanding of how aging and cancer interact to affect brain function is necessary to provide clinically-relevant results and identify mechanisms underlying persistent behavioral issues hampering adult cancer patients. Tumors grew more slowly in aged mice. Tumors and aging independently increased circulating inflammatory markers. Tumors reduced mRNA and protein expression of inflammatory markers in the hippocampus in aged mice. Reduced inflammatory activation in the brains of aged mice is likely not adaptive.
Collapse
|
46
|
Pan X, Taylor MJ, Cohen E, Hanna N, Mota S. Circadian Clock, Time-Restricted Feeding and Reproduction. Int J Mol Sci 2020; 21:ijms21030831. [PMID: 32012883 PMCID: PMC7038040 DOI: 10.3390/ijms21030831] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 01/23/2020] [Accepted: 01/24/2020] [Indexed: 12/29/2022] Open
Abstract
The goal of this review was to seek a better understanding of the function and differential expression of circadian clock genes during the reproductive process. Through a discussion of how the circadian clock is involved in these steps, the identification of new clinical targets for sleep disorder-related diseases, such as reproductive failure, will be elucidated. Here, we focus on recent research findings regarding circadian clock regulation within the reproductive system, shedding new light on circadian rhythm-related problems in women. Discussions on the roles that circadian clock plays in these reproductive processes will help identify new clinical targets for such sleep disorder-related diseases.
Collapse
Affiliation(s)
- Xiaoyue Pan
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York, NY 11501, USA
- Diabetes and Obesity Research Center, NYU Winthrop Hospital, Mineola, New York, NY 11501, USA
- Correspondence:
| | - Meredith J. Taylor
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York, NY 11501, USA
- Diabetes and Obesity Research Center, NYU Winthrop Hospital, Mineola, New York, NY 11501, USA
| | - Emma Cohen
- Diabetes and Obesity Research Center, NYU Winthrop Hospital, Mineola, New York, NY 11501, USA
| | - Nazeeh Hanna
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York, NY 11501, USA
- Department of Pediatrics, NYU Winthrop Hospital, Mineola, New York, NY 11501, USA
| | - Samantha Mota
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York, NY 11501, USA
- Diabetes and Obesity Research Center, NYU Winthrop Hospital, Mineola, New York, NY 11501, USA
| |
Collapse
|
47
|
Zhang X, He Y, Lin Q, Huang L, Zhang Q, Xu Y. Adverse effects of subchronic exposure to cooking oil fumes on the gonads and the GPR30-mediated signaling pathway in female rats. Mol Cell Toxicol 2019. [DOI: 10.1007/s13273-019-00053-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Abstract
Background
Cooking oil fumes (COFs) are composed of particulate matter, polycyclic aromatic hydrocarbons, volatile organic compounds, aldehydes, and ketones, and are currently a global health concern. Some agents in COFs are mutagenic and carcinogenic. However, only a few reports have addressed the hazardous effects of COF exposure on the female reproductive system. In this study, we explored the effects of subchronic exposure to COFs on female gonads in vivo and the possible involvement of the G-protein-coupled receptor 30 signaling pathway.
Methods
COFs were generated by heating commercially available canola oil in an iron pot. Adult female Wistar rats at 2 months of age were exposed to COFs at 32 mg/m3 for 0, 0.5, 1, 2, or 4 h/day for 56 days. The estrous cycle in rats was studied twice at 7:00 a.m. and 7:00 p.m. on the 43rd treatment day until the current estrous cycle was complete. The rat body weight was measured before the experiment and at day 56 post-exposure. At the end of the experiment, rat blood was collected for gonadal hormone assay, and ovaries were collected for histology and mRNA isolation. The mRNA levels of GPR30, EGFR, STAT3, and ERK were determined by quantitative RT-PCR.
Results
At a concentration of 32.21 ± 5.11 mg/m3, COF exposure extended the estrous cycle in rats, and ovary coefficient decreased. COFs showed various effects on the sex hormone levels and follicles, depending on its exposure level. Exposure to COFs led to the changes in mRNA levels of the G-protein-coupled receptor 30 (GPR30), epidermal growth factor receptor (EGFR), signal transducer and activator of transcription 3 (STAT3), and extracellular signal-regulated kinase (ERK).
Conclusion
This study indicated that cooking oil fume exposure disrupted the estrous cycle, sex hormone patterns, and follicle development in female rats in a dose-dependent manner. These adverse effects of cooking oil fumes on female reproductive health were correlated with the G-protein-coupled receptor 30-mediated signaling pathway.
Highlights
Subchronic exposure to COFs for 56 days had gonadal toxicity in female rats, that disrupted the estrous cycle, sex hormone patterns, and follicle development in a dose-dependent manner.
Reproductive endocrine disruption might be one of the female gonadotoxicity mechanisms of COFs.
These adverse effects of COFs on female reproductive health were correlated with the GPR30-mediated signaling pathway.
Collapse
|
48
|
Liu CM, Davis EA, Suarez AN, Wood RI, Noble EE, Kanoski SE. Sex Differences and Estrous Influences on Oxytocin Control of Food Intake. Neuroscience 2019; 447:63-73. [PMID: 31738883 DOI: 10.1016/j.neuroscience.2019.10.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/08/2019] [Accepted: 10/10/2019] [Indexed: 10/25/2022]
Abstract
Central oxytocin potently reduces food intake and is being pursued as a clinical treatment for obesity. While sexually dimorphic effects have been described for the effects of oxytocin on several behavioral outcomes, the role of sex in central oxytocin modulation of feeding behavior is poorly understood. Here we investigated the effects of sex, estrous cycle stage, and female sex hormones (estrogen, progesterone) on central oxytocin-mediated reduction of food intake in rats. Results show that while intracerebroventricular (ICV) oxytocin potently reduces chow intake in both male and female rats, these effects were more pronounced in males than in females. We next examined whether estrous cycle stage affects oxytocin's food intake-reducing effects in females. Results show that ICV oxytocin administration significantly reduces food intake during all estrous cycle stages except proestrous, suggesting that female sex hormones may modulate the feeding effects of oxytocin. Indeed, additional results reveal that estrogen, but not progesterone replacement, in ovariectomized rats abolishes oxytocin-mediated reductions in chow intake. Lastly, oxytocin receptor mRNA (Oxtr) quantification (via quantitative PCR) and anatomical localization (via fluorescent in situ hybridization) in previously established sites of action for oxytocin control of food intake revealed comparable Oxtr expression between male and female rats, suggesting that observed sex and estrous differences may be based on variations in ligand availability and/or binding. Overall, these data show that estrogen reduces the effectiveness of central oxytocin to inhibit food intake, suggesting that sex hormones and estrous cycle should be considered in clinical investigations of oxytocin for obesity treatment.
Collapse
Affiliation(s)
- Clarissa M Liu
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, United States; Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, United States
| | - Elizabeth A Davis
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, United States
| | - Andrea N Suarez
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, United States
| | - Ruth I Wood
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, United States; Department of Integrative Anatomical Sciences, Keck School of Medicine of University of Southern California, Los Angeles, CA, United States
| | - Emily E Noble
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, United States; Department of Foods and Nutrition, University of Georgia, Athens, GA, United States.
| | - Scott E Kanoski
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, United States; Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, United States.
| |
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW Females experience eating disorders at substantially higher rates than males. Although sociocultural factors have traditionally been thought to underlie this sex disparity, accumulating evidence implicates differential exposure to gonadal hormones early in life. Gonadal hormones also impact within-sex variability in disordered eating, helping to explain why not all women develop an eating disorder, and some men do. We review recent findings regarding these gonadal hormone effects and their implications for the etiology of eating disorders. RECENT FINDINGS Males are exposed to significantly higher testosterone levels than females perinatally, and this exposure appears to protect against later binge eating in males relative to females. Within-sex, higher estradiol levels among females and higher testosterone levels among males appear to be protective. Progesterone exhibits minimal direct phenotypic effects on disordered eating but appears to counteract the protective effects of estrogen in adult females. Importantly, gonadal hormone effects may be moderated by psychosocial factors. SUMMARY Evidence suggests that gonadal hormones play a critical role in the etiology of disordered eating. Overall, higher testosterone and estrogen appear to be protective across development. Additional research is needed to identify mechanisms underlying these effects and further explore interactions between hormonal and psychosocial risk.
Collapse
|
50
|
Alonso-Caraballo Y, Ferrario CR. Effects of the estrous cycle and ovarian hormones on cue-triggered motivation and intrinsic excitability of medium spiny neurons in the Nucleus Accumbens core of female rats. Horm Behav 2019; 116:104583. [PMID: 31454509 PMCID: PMC7256930 DOI: 10.1016/j.yhbeh.2019.104583] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/01/2019] [Accepted: 08/20/2019] [Indexed: 11/08/2022]
Abstract
Naturally occurring alterations in estradiol influence food intake in females. However, how motivational responses to food cues are affected by the estrous cycle or ovarian hormones is unknown. In addition, while individual susceptibility to obesity is accompanied by enhanced incentive motivational responses to food cues and increased NAc intrinsic excitability in males, studies in females are absent. Therefore, we examined basal differences in intrinsic NAc excitability of obesity-prone vs. obesity-resistant females and determined how conditioned approach (a measure of cue-triggered motivation), food intake, and motivation for food vary with the cycle in naturally cycling female obesity-prone, obesity-resistant, and outbred Sprague-Dawley rats. Finally, we used ovariectomy followed by hormone treatment to determine the role of ovarian hormones in cue-triggered motivation in selectively-bred and outbred female rats. We found that intrinsic excitability of NAc MSNs and conditioned approach are enhanced in female obesity-prone vs. obesity-resistant rats. These effects were driven by greater MSN excitability and conditioned approach behavior during metestrus/diestrus vs. proestrus/estrus in obesity-prone but not obesity-resistant rats, despite similar regulation of food intake and food motivation by the cycle in these groups. Furthermore, estradiol and progesterone treatment reduced conditioned approach behavior in obesity-prone and outbred Sprague-Dawley females. To our knowledge, these data are the first to demonstrate cycle- and hormone-dependent effects on the motivational response to a food cue, and the only studies to date to determine how individual susceptibility to obesity influences NAc excitability, cue-triggered food-seeking, and differences in the regulation of these neurobehavioral responses by the estrous cycle.
Collapse
Affiliation(s)
| | - Carrie R Ferrario
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, United States of America; Department of Pharmacology, University of Michigan, Ann Arbor, MI, United States of America.
| |
Collapse
|