1
|
Cao X, Wang Q, Zhang L, Sun H, Xu G, Chen X, Wu Z, Liu H, Yuan G, Wu J, Liu T. Oxytocin alleviates high-fat diet-induced anxiety by decreasing glutamatergic synaptic transmission in the ventral dentate gyrus in adolescent mice. Neuropharmacology 2024; 262:110201. [PMID: 39481751 DOI: 10.1016/j.neuropharm.2024.110201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 09/21/2024] [Accepted: 10/27/2024] [Indexed: 11/02/2024]
Abstract
A high-fat diet (HFD)-induced obesity is associated with mental disorders in adolescence. However, the mechanisms underlying these associations remain unclear. In this study, we hypothesized that synaptic remodeling occurs in the ventral hippocampus (vHP) of obese mice. To investigate this, we established a postnatal model of HFD-induced obesity in mice and observed increased body weight, elevated plasma luteinizing hormone and testosterone levels, premature puberty, and enhanced anxiety-like behavior in male subjects. We also examined the effect of HFD on the c-Fos protein expression in the ventral dentate gyrus (vDG) and explored the influence of intracerebroventricular (i.c.v) oxytocin injections on HFD-induced anxiety. Our results indicated an increase in c-Fos-positive cells in the vDG following HFD consumption. Additionally, we recorded the spontaneous synaptic activity of miniature excitatory postsynaptic currents (mEPSCs) in the vDG. Notably, HFD resulted in an elevated mEPSC frequency without affecting mEPSC amplitude. Subsequently, investigations demonstrated that i.c.v oxytocin injections reversed anxiety-like behavior induced by HFD. Moreover, the application of oxytocin in a bath solution reduced the mEPSC frequency in the vDG. These findings suggest that postnatal HFD intake induces synaptic dysfunction in the vDG, associated with the hyperactivity of vDG neurons, potentially contributing to the anxiety-like behavior in juvenile obesity.
Collapse
Affiliation(s)
- Xi Cao
- Department of Pediatrics, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Qiyuan Wang
- Department of Pediatrics, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Lina Zhang
- Department of Pediatrics, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Huichao Sun
- Department of Pediatrics, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Gang Xu
- Jiangxi Provincial Key Laboratory of Trauma, Burn and Pain Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Xiao Chen
- Department of Pediatrics, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Zhihong Wu
- Department of Pediatrics, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Huibao Liu
- Department of Pediatrics, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Gaole Yuan
- Department of Pediatrics, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Jian Wu
- Department of Pediatrics, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Tao Liu
- Department of Pediatrics, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Provincial Key Laboratory of Trauma, Burn and Pain Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
2
|
Goss K, Bueno-Junior LS, Stangis K, Ardoin T, Carmon H, Zhou J, Satapathy R, Baker I, Jones-Tinsley CE, Lim MM, Watson BO, Sueur C, Ferrario CR, Murphy GG, Ye B, Hu Y. Quantifying social roles in multi-animal videos using subject-aware deep-learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.07.602350. [PMID: 39026890 PMCID: PMC11257443 DOI: 10.1101/2024.07.07.602350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Analyzing social behaviors is critical for many fields, including neuroscience, psychology, and ecology. While computational tools have been developed to analyze videos containing animals engaging in limited social interactions under specific experimental conditions, automated identification of the social roles of freely moving individuals in a multi-animal group remains unresolved. Here we describe a deep-learning-based system - named LabGym2 - for identifying and quantifying social roles in multi-animal groups. This system uses a subject-aware approach: it evaluates the behavioral state of every individual in a group of two or more animals while factoring in its social and environmental surroundings. We demonstrate the performance of subject-aware deep-learning in different species and assays, from partner preference in freely-moving insects to primate social interactions in the field. Our subject-aware deep learning approach provides a controllable, interpretable, and efficient framework to enable new experimental paradigms and systematic evaluation of interactive behavior in individuals identified within a group.
Collapse
Affiliation(s)
- Kelly Goss
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
- These authors contributed equally
| | - Lezio S. Bueno-Junior
- Department of Psychiatry, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- These authors contributed equally
| | - Katherine Stangis
- Michigan Neuroscience Institute, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Théo Ardoin
- Master Biodiversité Ecologie et Evolution, Université Paris-Saclay, Orsay, France
- Magistère de Biologie, Université Paris-Saclay, Orsay, France
| | - Hanna Carmon
- Department of Pharmacology and Psychology Department (Biopsychology), University of Michigan, Ann Arbor, MI 48109, USA
| | - Jie Zhou
- Department of Computer Science, Northern Illinois University, DeKalb, IL 60115, USA
| | - Rohan Satapathy
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Isabelle Baker
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Carolyn E. Jones-Tinsley
- Veterans Affairs VISN20 Northwest MIRECC, VA Portland Health Care System, Portland, OR 97239, USA
- Oregon Alzheimer’s Disease Research Center, Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Miranda M. Lim
- Veterans Affairs VISN20 Northwest MIRECC, VA Portland Health Care System, Portland, OR 97239, USA
- Oregon Alzheimer’s Disease Research Center, Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Brendon O. Watson
- Department of Psychiatry, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Cédric Sueur
- Université de Strasbourg, IPHC UMR7178, CNRS, Strasbourg, France
- ANTHROPO-LAB, ETHICS EA 7446, Université Catholique de Lille, Lille, France
| | - Carrie R. Ferrario
- Department of Pharmacology and Psychology Department (Biopsychology), University of Michigan, Ann Arbor, MI 48109, USA
| | - Geoffrey G. Murphy
- Michigan Neuroscience Institute, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Bing Ye
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yujia Hu
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
3
|
Önal D, Korkmaz H, Önal G, Pehlivanoğlu B. Body weight modulates the impact of oxytocin on chronic cold-immobilization stress response. Peptides 2024; 177:171202. [PMID: 38555975 DOI: 10.1016/j.peptides.2024.171202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/21/2024] [Accepted: 03/27/2024] [Indexed: 04/02/2024]
Abstract
By activating the stress system, stress modulates various physiological parameters including food intake, energy consumption, and, consequently, body weight. The role of oxytocin in the regulation of stress and obesity cannot be disregarded. Based on these findings, we aimed to investigate the effect of intranasal oxytocin on stress response in high-fat-diet (HFD)--fed and control-diet-fed rats exposed to chronic stress. Cold-immobilization stress was applied for 5 consecutive days to male Sprague-Dawley rats fed either with a control diet (n=20) or HFD (n=20) for 6 weeks. Half of the animals in each group received oxytocin. Stress response was evaluated via plasma and salivary cortisol levels as well as elevated plus maze scores. Prefrontal cortex and hypothalamic oxytocin receptor (OxtR) expression levels were identified using western blot analysis. The results showed higher stress response in HFD-fed animals than in control animals both under basal and post-stress conditions. Oxytocin application had a prominent anxiolytic effect in the control group but an insignificant effect in the HFD group. While OxtR expression levels in the prefrontal cortex did not vary according to the body weight and oxytocin application, OxtR levels in the hypothalamus were higher in the HFD- and/or oxytocin-treated animals. Our results indicated that the peripheral and central effects of oxytocin vary with body weight. Moreover, obesity masks the anxiolytic effects of oxytocin, probably by reinforcing the stress condition via central OxtRs. In conclusion, elucidating the mechanisms underlying the central effect of oxytocin is important to cope with stress and obesity.
Collapse
Affiliation(s)
- Deniz Önal
- Faculty of Medicine, Department of Physiology, Balıkesir University, Balıkesir, Türkiye.
| | - Hilal Korkmaz
- Faculty of Medicine, Department of Physiology, Hacettepe University, Ankara, Türkiye
| | - Gizem Önal
- Oxford Parkinson's Disease Centre and Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Bilge Pehlivanoğlu
- Faculty of Medicine, Department of Physiology, Hacettepe University, Ankara, Türkiye
| |
Collapse
|
4
|
Lu T, Ding L, Zheng X, Li Y, Wei W, Liu W, Tao J, Xue X. Alisol A Exerts Neuroprotective Effects Against HFD-Induced Pathological Brain Aging via the SIRT3-NF-κB/MAPK Pathway. Mol Neurobiol 2024; 61:753-771. [PMID: 37659035 PMCID: PMC10861652 DOI: 10.1007/s12035-023-03592-5] [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: 01/17/2023] [Accepted: 08/16/2023] [Indexed: 09/05/2023]
Abstract
Chronic consumption of a high-fat diet (HFD) has profound effects on brain aging, which is mainly characterized by cognitive decline, inflammatory responses, and neurovascular damage. Alisol A (AA) is a triterpenoid with therapeutic potential for metabolic diseases, but whether it has a neuroprotective effect against brain aging caused by a HFD has not been investigated. Six-month-old male C57BL6/J mice were exposed to a HFD with or without AA treatment for 12 weeks. Behavioral tasks were used to assess the cognitive abilities of the mice. Neuroinflammation and changes in neurovascular structure in the brains were examined. We further assessed the mechanism by which AA exerts neuroprotective effects against HFD-induced pathological brain aging in vitro and in vivo. Behavioral tests showed that cognitive function was improved in AA-treated animals. AA treatment reduced microglia activation and inflammatory cytokine release induced by a HFD. Furthermore, AA treatment increased the number of hippocampal neurons, the density of dendritic spines, and the expression of tight junction proteins. We also demonstrated that AA attenuated microglial activation by targeting the SIRT3-NF-κB/MAPK pathway and ameliorated microglial activation-induced tight junction degeneration in endothelial cells and apoptosis in hippocampal neurons. The results of this study show that AA may be a promising agent for the treatment of HFD-induced brain aging.
Collapse
Affiliation(s)
- Taotao Lu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350112, China
- Fujian Key Laboratory of Rehabilitation Techniques, Cognitive Rehabilitation, Fuzhou, 350112, China
| | - Linlin Ding
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350112, China
| | - Xiaoqing Zheng
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350112, China
| | - Yongxu Li
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350112, China
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, No 13, Hudongzhi Road, Fuzhou City, 350003, Fujian Province, China
| | - Wei Wei
- Fujian Key Laboratory of Rehabilitation Techniques, Cognitive Rehabilitation, Fuzhou, 350112, China
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, No 13, Hudongzhi Road, Fuzhou City, 350003, Fujian Province, China
| | - Weilin Liu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350112, China
| | - Jing Tao
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350112, China
- Fujian Key Laboratory of Rehabilitation Techniques, Cognitive Rehabilitation, Fuzhou, 350112, China
| | - Xiehua Xue
- Fujian Key Laboratory of Rehabilitation Techniques, Cognitive Rehabilitation, Fuzhou, 350112, China.
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, No 13, Hudongzhi Road, Fuzhou City, 350003, Fujian Province, China.
| |
Collapse
|
5
|
Zhou J, Fan Q, Cai X, Zhang Y, Hou Y, Cao S, Li Z, Feng M, Wang Q, Zhang J, Wang G, Zheng X, Hao H. Ginkgo biloba extract protects against depression-like behavior in mice through regulating gut microbial bile acid metabolism. Chin J Nat Med 2023; 21:745-758. [PMID: 37879793 DOI: 10.1016/s1875-5364(23)60496-0] [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: 07/23/2023] [Indexed: 10/27/2023]
Abstract
Depression is a mental disorder with high morbidity, disability and relapse rates. Ginkgo biloba extract (GBE), a traditional Chinese medicine, has a long history of clinical application in the treatment of cerebral and mental disorders, but the key mechanism remains incompletely understood. Here we showed that GEB exerted anti-depressant effect in mice through regulating gut microbial metabolism. GBE protected against unpredictable mild stress (UMS)-induced despair, anxiety-like and social avoidance behavior in mice without sufficient brain distribution. Fecal microbiome transplantation transmitted, while antibiotic cocktail abrogated the protective effect of GBE. Spatiotemporal bacterial profiling and metabolomics assay revealed a potential involvement of Parasutterella excrementihominis and the bile acid metabolite ursodeoxycholic acid (UDCA) in the effect of GBE. UDCA administration induced depression-like behavior in mice. Together, these findings suggest that GBE acts on gut microbiome-modulated bile acid metabolism to alleviate stress-induced depression.
Collapse
Affiliation(s)
- Junchi Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qilin Fan
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaoying Cai
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Youying Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yuanlong Hou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Shuqi Cao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ziguang Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Mengzhen Feng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qingqing Wang
- WanBangDe Pharmaceutical Group Co., Ltd., Wenlin 317500, China
| | - Jianbing Zhang
- WanBangDe Pharmaceutical Group Co., Ltd., Wenlin 317500, China
| | - Guangji Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao Zheng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Haiping Hao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Laboratory of Metabolic Regulation and Drug Target Discovery, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
6
|
Tüfekci KK, Bakirhan EG, Terzi F. A Maternal High-Fat Diet Causes Anxiety-Related Behaviors by Altering Neuropeptide Y1 Receptor and Hippocampal Volumes in Rat Offspring: the Potential Effect of N-Acetylcysteine. Mol Neurobiol 2023; 60:1499-1514. [PMID: 36502431 DOI: 10.1007/s12035-022-03158-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/01/2022] [Indexed: 12/14/2022]
Abstract
The children of obese mothers are known to have a high risk of obesity and metabolic disease and are prone to developing cognitive deficits, although the underlying mechanism is not yet fully understood. This study investigated the relationship between neuropeptide Y1 receptor (NPY1R) and anxiety-like behaviors in the hippocampi of male rat offspring exposed to maternal obesity and the potential neuroprotective effects of N-acetylcysteine (NAC). A maternal obesity model was created using a high-fat (60% k/cal) diet. NAC (150 mg/kg) was administered by intragastric gavage for 25 days in both the NAC and obesity + NAC (ObNAC) groups. All male rat offspring were subjected to behavioral testing on postnatal day 28, the end of the experiment. Stereological analysis was performed on hippocampal sections, while NPY1R expression was determined using immunohistochemical methods. Stereological data indicated significant decreases in the total volume of the hippocampus and CA1 and dentate gyrus (DG) regions in the obese (Ob) group (p < 0.01). Decreased NPY1R expression was observed in the Ob group hippocampus (p < 0.01). At behavioral assessments, the Ob group rats exhibited increased anxiety and less social interaction, although the ObNAC group rats exhibited stronger responses than the Ob group (p < 0.01). The study results show that NAC attenuated anxiety-like behaviors and NPY1R expression and also protected hippocampal volume against maternal obesity. The findings indicate that a decrease in NPY1R-positive neurons in the hippocampus of male rats due to maternal conditions may be associated with increased levels of anxiety and a lower hippocampal volume. Additionally, although there is no direct evidence, maintenance of NPY1R expression by NAC may be critical for regulating maternal obesity-induced anxiety-related behaviors and hippocampal structure.
Collapse
Affiliation(s)
- Kıymet Kübra Tüfekci
- Department of Histology and Embryology, Faculty of Medicine, Kastamonu University, Kastamonu, Turkey.
| | - Elfide Gizem Bakirhan
- Department of Histology and Embryology, Faculty of Medicine, Adıyaman University, Adıyaman, Turkey
| | - Funda Terzi
- Department of Pathology, Faculty of Veterinary Medicine, Kastamonu University, Kastamonu, Turkey
| |
Collapse
|
7
|
Tsingotjidou AS. Oxytocin: A Multi-Functional Biomolecule with Potential Actions in Dysfunctional Conditions; From Animal Studies and Beyond. Biomolecules 2022; 12:1603. [PMID: 36358953 PMCID: PMC9687803 DOI: 10.3390/biom12111603] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 10/20/2022] [Accepted: 10/23/2022] [Indexed: 10/13/2023] Open
Abstract
Oxytocin is a hormone secreted from definite neuroendocrine neurons located in specific nuclei in the hypothalamus (mainly from paraventricular and supraoptic nuclei), and its main known function is the contraction of uterine and/or mammary gland cells responsible for parturition and breastfeeding. Among the actions of the peripherally secreted oxytocin is the prevention of different degenerative disorders. These actions have been proven in cell culture and in animal models or have been tested in humans based on hypotheses from previous studies. This review presents the knowledge gained from the previous studies, displays the results from oxytocin intervention and/or treatment and proposes that the well described actions of oxytocin might be connected to other numerous, diverse actions of the biomolecule.
Collapse
Affiliation(s)
- Anastasia S Tsingotjidou
- Laboratory of Anatomy, Histology and Embryology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54 124 Thessaloniki, Greece
| |
Collapse
|
8
|
Kozlova EV, Denys ME, Benedum J, Valdez MC, Enriquez D, Bishay AE, Chinthirla BD, Truong E, Krum JM, DiPatrizio NV, Deol P, Martins-Green M, Curras-Collazo MC. Developmental exposure to indoor flame retardants and hypothalamic molecular signatures: Sex-dependent reprogramming of lipid homeostasis. Front Endocrinol (Lausanne) 2022; 13:997304. [PMID: 36277707 PMCID: PMC9580103 DOI: 10.3389/fendo.2022.997304] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/25/2022] [Indexed: 11/15/2022] Open
Abstract
Polybrominated diphenyl ethers (PBDEs) are a class of flame-retardant organohalogen pollutants that act as endocrine/neuroendocrine disrupting chemicals (EDCs). In humans, exposure to brominated flame retardants (BFR) or other environmentally persistent organic pollutants (POPs) such as polychlorinated biphenyls (PCBs) and novel organophosphate flame retardants has been associated with increasing trends of diabetes and metabolic disease. However, the effects of PBDEs on metabolic processes and their associated sex-dependent features are poorly understood. The metabolic-disrupting effects of perinatal exposure to industrial penta-PBDE mixture, DE-71, on male and female progeny of C57BL/6N mouse dams were examined in adulthood. Dams were exposed to environmentally relevant doses of PBDEs daily for 10 weeks (p.o.): 0.1 (L-DE-71) and 0.4 mg/kg/d (H-DE-71) and offspring parameters were compared to corn oil vehicle controls (VEH/CON). The following lipid metabolism indices were measured: plasma cholesterol, triglycerides, adiponectin, leptin, and liver lipids. L-DE-71 female offspring were particularly affected, showing hypercholesterolemia, elevated liver lipids and fasting plasma leptin as compared to same-sex VEH/CON, while L- and H-DE-71 male F1 only showed reduced plasma adiponectin. Using the quantitative Folch method, we found that mean liver lipid content was significantly elevated in L-DE-71 female offspring compared to controls. Oil Red O staining revealed fatty liver in female offspring and dams. General measures of adiposity, body weight, white and brown adipose tissue (BAT), and lean and fat mass were weighed or measured using EchoMRI. DE-71 did not produce abnormal adiposity, but decreased BAT depots in L-DE-71 females and males relative to same-sex VEH/CON. To begin to address potential central mechanisms of deregulated lipid metabolism, we used RT-qPCR to quantitate expression of hypothalamic genes in energy-regulating circuits that control lipid homeostasis. Both doses of DE-71 sex-dependently downregulated hypothalamic expression of Lepr, Stat3, Mc4r, Agrp, Gshr in female offspring while H-DE-71 downregulated Npy in exposed females relative to VEH/CON. In contrast, exposed male offspring displayed upregulated Stat3 and Mc4r. Intestinal barrier integrity was measured using FITC-dextran since it can lead to systemic inflammation that leads to liver damage and metabolic disease, but was not affected by DE-71 exposure. These findings indicate that maternal transfer of PBDEs disproportionately endangers female offspring to lipid metabolic reprogramming that may exaggerate risk for adult metabolic disease.
Collapse
Affiliation(s)
- Elena V. Kozlova
- Department of Molecular, Cell & Systems Biology, University of California, Riverside, Riverside, CA, United States
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States
| | - Maximillian E. Denys
- Department of Molecular, Cell & Systems Biology, University of California, Riverside, Riverside, CA, United States
| | - Jonathan Benedum
- Department of Molecular, Cell & Systems Biology, University of California, Riverside, Riverside, CA, United States
| | - Matthew C. Valdez
- Department of Molecular, Cell & Systems Biology, University of California, Riverside, Riverside, CA, United States
| | - Dave Enriquez
- Department of Molecular, Cell & Systems Biology, University of California, Riverside, Riverside, CA, United States
| | - Anthony E. Bishay
- Department of Molecular, Cell & Systems Biology, University of California, Riverside, Riverside, CA, United States
| | - Bhuvaneswari D. Chinthirla
- Department of Molecular, Cell & Systems Biology, University of California, Riverside, Riverside, CA, United States
| | - Edward Truong
- Department of Molecular, Cell & Systems Biology, University of California, Riverside, Riverside, CA, United States
| | - Julia M. Krum
- Department of Molecular, Cell & Systems Biology, University of California, Riverside, Riverside, CA, United States
| | - Nicholas V. DiPatrizio
- Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Poonamjot Deol
- Department of Molecular, Cell & Systems Biology, University of California, Riverside, Riverside, CA, United States
| | - Manuela Martins-Green
- Department of Molecular, Cell & Systems Biology, University of California, Riverside, Riverside, CA, United States
| | - Margarita C. Curras-Collazo
- Department of Molecular, Cell & Systems Biology, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
9
|
Khazen T, Narattil NR, Ferreira G, Maroun M. Hippocampal oxytocin is involved in spatial memory and synaptic plasticity deficits following acute high-fat diet intake in juvenile rats. Cereb Cortex 2022; 33:3934-3943. [PMID: 35989314 DOI: 10.1093/cercor/bhac317] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 07/22/2022] [Accepted: 07/23/2022] [Indexed: 11/12/2022] Open
Abstract
The hippocampus undergoes maturation during juvenility, a period of increased vulnerability to environmental challenges. We recently found that acute high-fat diet (HFD) impaired hippocampal long-term potentiation (LTP) and hippocampal-dependent spatial memory. We also recently reported that similar HFD exposure affected prefrontal plasticity and social memory through decreased oxytocin levels in the prefrontal cortex. In the present study, we therefore evaluated whether hippocampal oxytocin levels are also affected by juvenile HFD and could mediate deficits of hippocampal LTP and spatial memory. We found that postweaning HFD decreased oxytocin levels in the CA1 of the dorsal hippocampus. Interestingly, systemic injection of high, but not low, dose of oxytocin rescued HFD-induced LTP impairment in CA1. Moreover, deficits in long-term object location memory (OLM) were prevented by systemic injection of both high and low dose of oxytocin as well as by intra-CA1 infusion of oxytocin receptor agonist. Finally, we found that blocking oxytocin receptors in CA1 impaired long-term OLM in control-fed juvenile rats. These results suggest that acute HFD intake lowers oxytocin levels in the CA1 that lead to CA1 plasticity impairment and spatial memory deficits in juveniles. Further, these results provide the first evidence for the regulatory role of oxytocin in spatial memory.
Collapse
Affiliation(s)
- Tala Khazen
- Sagol Department of Neurobiology, Faculty of Natural Sciences, and The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa 3498838, Israel
| | - Nisha Rajan Narattil
- Sagol Department of Neurobiology, Faculty of Natural Sciences, and The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa 3498838, Israel
| | - Guillaume Ferreira
- University of Bordeaux, INRAE, Nutrition and Integrative Neurobiology, UMR 1286, Bordeaux, 33076, France
| | - Mouna Maroun
- Sagol Department of Neurobiology, Faculty of Natural Sciences, and The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa 3498838, Israel
| |
Collapse
|
10
|
Zhao Y, Tan DC, Peng B, Yang L, Zhang SY, Shi RP, Chong CM, Zhong ZF, Wang SP, Liang QL, Wang YT. Neuroendocrine-Immune Regulatory Network of Eucommia ulmoides Oliver. Molecules 2022; 27:molecules27123697. [PMID: 35744822 PMCID: PMC9229650 DOI: 10.3390/molecules27123697] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/23/2022] [Accepted: 05/31/2022] [Indexed: 02/04/2023] Open
Abstract
Eucommia ulmoides Oliver (E. ulmoides) is a popular medicinal herb and health supplement in China, Japan, and Korea, and has a variety of pharmaceutical properties. The neuroendocrine-immune (NEI) network is crucial in maintaining homeostasis and physical or psychological functions at a holistic level, consistent with the regulatory theory of natural medicine. This review aims to systematically summarize the chemical compositions, biological roles, and pharmacological properties of E. ulmoides to build a bridge between it and NEI-associated diseases and to provide a perspective for the development of its new clinical applications. After a review of the literature, we found that E. ulmoides has effects on NEI-related diseases including cancer, neurodegenerative disease, hyperlipidemia, osteoporosis, insomnia, hypertension, diabetes mellitus, and obesity. However, clinical studies on E. ulmoides were scarce. In addition, E. ulmoides derivatives are diverse in China, and they are mainly used to enhance immunity, improve hepatic damage, strengthen bones, and lower blood pressure. Through network pharmacological analysis, we uncovered the possibility that E. ulmoides is involved in functional interactions with cancer development, insulin resistance, NAFLD, and various inflammatory pathways associated with NEI diseases. Overall, this review suggests that E. ulmoides has a wide range of applications for NEI-related diseases and provides a direction for its future research and development.
Collapse
Affiliation(s)
- Yi Zhao
- Macau Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China; (Y.Z.); (D.-C.T.); (B.P.); (L.Y.); (S.-Y.Z.); (R.-P.S.); (C.-M.C.); (Z.-F.Z.); (S.-P.W.)
| | - De-Chao Tan
- Macau Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China; (Y.Z.); (D.-C.T.); (B.P.); (L.Y.); (S.-Y.Z.); (R.-P.S.); (C.-M.C.); (Z.-F.Z.); (S.-P.W.)
| | - Bo Peng
- Macau Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China; (Y.Z.); (D.-C.T.); (B.P.); (L.Y.); (S.-Y.Z.); (R.-P.S.); (C.-M.C.); (Z.-F.Z.); (S.-P.W.)
| | - Lin Yang
- Macau Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China; (Y.Z.); (D.-C.T.); (B.P.); (L.Y.); (S.-Y.Z.); (R.-P.S.); (C.-M.C.); (Z.-F.Z.); (S.-P.W.)
| | - Si-Yuan Zhang
- Macau Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China; (Y.Z.); (D.-C.T.); (B.P.); (L.Y.); (S.-Y.Z.); (R.-P.S.); (C.-M.C.); (Z.-F.Z.); (S.-P.W.)
| | - Rui-Peng Shi
- Macau Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China; (Y.Z.); (D.-C.T.); (B.P.); (L.Y.); (S.-Y.Z.); (R.-P.S.); (C.-M.C.); (Z.-F.Z.); (S.-P.W.)
| | - Cheong-Meng Chong
- Macau Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China; (Y.Z.); (D.-C.T.); (B.P.); (L.Y.); (S.-Y.Z.); (R.-P.S.); (C.-M.C.); (Z.-F.Z.); (S.-P.W.)
| | - Zhang-Feng Zhong
- Macau Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China; (Y.Z.); (D.-C.T.); (B.P.); (L.Y.); (S.-Y.Z.); (R.-P.S.); (C.-M.C.); (Z.-F.Z.); (S.-P.W.)
| | - Sheng-Peng Wang
- Macau Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China; (Y.Z.); (D.-C.T.); (B.P.); (L.Y.); (S.-Y.Z.); (R.-P.S.); (C.-M.C.); (Z.-F.Z.); (S.-P.W.)
| | - Qiong-Lin Liang
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Beijing Key Lab of Microanalytical Methods & Instrumentation, Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing 100084, China
- Correspondence: (Q.-L.L.); (Y.-T.W.); Tel.: +86-010-6277-2263 (Q.-L.L.); +853-8822-4691 (Y.-T.W.); Fax: +86-010-6277-2263 (Q.-L.L.); +853-2884-1358 (Y.-T.W.)
| | - Yi-Tao Wang
- Macau Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China; (Y.Z.); (D.-C.T.); (B.P.); (L.Y.); (S.-Y.Z.); (R.-P.S.); (C.-M.C.); (Z.-F.Z.); (S.-P.W.)
- Correspondence: (Q.-L.L.); (Y.-T.W.); Tel.: +86-010-6277-2263 (Q.-L.L.); +853-8822-4691 (Y.-T.W.); Fax: +86-010-6277-2263 (Q.-L.L.); +853-2884-1358 (Y.-T.W.)
| |
Collapse
|
11
|
Kozlova EV, Valdez MC, Denys ME, Bishay AE, Krum JM, Rabbani KM, Carrillo V, Gonzalez GM, Lampel G, Tran JD, Vazquez BM, Anchondo LM, Uddin SA, Huffman NM, Monarrez E, Olomi DS, Chinthirla BD, Hartman RE, Kodavanti PRS, Chompre G, Phillips AL, Stapleton HM, Henkelmann B, Schramm KW, Curras-Collazo MC. Persistent autism-relevant behavioral phenotype and social neuropeptide alterations in female mice offspring induced by maternal transfer of PBDE congeners in the commercial mixture DE-71. Arch Toxicol 2022; 96:335-365. [PMID: 34687351 PMCID: PMC8536480 DOI: 10.1007/s00204-021-03163-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/16/2021] [Indexed: 12/30/2022]
Abstract
Polybrominated diphenyl ethers (PBDEs) are ubiquitous persistent organic pollutants (POPs) that are known neuroendocrine disrupting chemicals with adverse neurodevelopmental effects. PBDEs may act as risk factors for autism spectrum disorders (ASD), characterized by abnormal psychosocial functioning, although direct evidence is currently lacking. Using a translational exposure model, we tested the hypothesis that maternal transfer of a commercial mixture of PBDEs, DE-71, produces ASD-relevant behavioral and neurochemical deficits in female offspring. C57Bl6/N mouse dams (F0) were exposed to DE-71 via oral administration of 0 (VEH/CON), 0.1 (L-DE-71) or 0.4 (H-DE-71) mg/kg bw/d from 3 wk prior to gestation through end of lactation. Mass spectrometry analysis indicated in utero and lactational transfer of PBDEs (in ppb) to F1 female offspring brain tissue at postnatal day (PND) 15 which was reduced by PND 110. Neurobehavioral testing of social novelty preference (SNP) and social recognition memory (SRM) revealed that adult L-DE-71 F1 offspring display deficient short- and long-term SRM, in the absence of reduced sociability, and increased repetitive behavior. These effects were concomitant with reduced olfactory discrimination of social odors. Additionally, L-DE-71 exposure also altered short-term novel object recognition memory but not anxiety or depressive-like behavior. Moreover, F1 L-DE-71 displayed downregulated mRNA transcripts for oxytocin (Oxt) in the bed nucleus of the stria terminalis (BNST) and supraoptic nucleus, and vasopressin (Avp) in the BNST and upregulated Avp1ar in BNST, and Oxtr in the paraventricular nucleus. Our work demonstrates that developmental PBDE exposure produces ASD-relevant neurochemical, olfactory processing and behavioral phenotypes that may result from early neurodevelopmental reprogramming within central social and memory networks.
Collapse
Affiliation(s)
- Elena V Kozlova
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, 92521, USA
- Neuroscience Graduate Program, University of California, Riverside, CA, 92521, USA
| | - Matthew C Valdez
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, 92521, USA
- Neuroscience Graduate Program, University of California, Riverside, CA, 92521, USA
- Neurological and Endocrine Toxicology Branch, Public Health and Integrated Toxicology Division, CPHEA/ORD, U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC, 27711, USA
| | - Maximillian E Denys
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, 92521, USA
| | - Anthony E Bishay
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, 92521, USA
| | - Julia M Krum
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, 92521, USA
| | - Kayhon M Rabbani
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, 92521, USA
| | - Valeria Carrillo
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, 92521, USA
| | - Gwendolyn M Gonzalez
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, 92521, USA
| | - Gregory Lampel
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, 92521, USA
| | - Jasmin D Tran
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, 92521, USA
| | - Brigitte M Vazquez
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, 92521, USA
| | - Laura M Anchondo
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, 92521, USA
| | - Syed A Uddin
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, 92521, USA
| | - Nicole M Huffman
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, 92521, USA
| | - Eduardo Monarrez
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, 92521, USA
| | - Duraan S Olomi
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, 92521, USA
| | - Bhuvaneswari D Chinthirla
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, 92521, USA
| | - Richard E Hartman
- Department of Psychology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Prasada Rao S Kodavanti
- Neurological and Endocrine Toxicology Branch, Public Health and Integrated Toxicology Division, CPHEA/ORD, U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC, 27711, USA
| | - Gladys Chompre
- Biotechnology Department, Pontifical Catholic University of Puerto Rico, Ponce, Puerto Rico, 00717-9997, USA
| | - Allison L Phillips
- Duke University, Nicholas School of the Environment, Durham, NC, 27710, USA
| | | | - Bernhard Henkelmann
- Helmholtz Zentrum Munchen, Molecular EXposomics (MEX), German National Research Center for Environmental Health (GmbH), Ingolstaedter Landstrasse 1, Neuherberg, Munich, Germany
| | - Karl-Werner Schramm
- Helmholtz Zentrum Munchen, Molecular EXposomics (MEX), German National Research Center for Environmental Health (GmbH), Ingolstaedter Landstrasse 1, Neuherberg, Munich, Germany
- Department Für Biowissenschaftliche Grundlagen, TUM, Wissenschaftszentrum Weihenstephan für Ernährung, Landnutzung Und Umwelt, Weihenstephaner Steig 23, 85350, Freising, Germany
| | | |
Collapse
|
12
|
Niu J, Tong J, Blevins JE. Oxytocin as an Anti-obesity Treatment. Front Neurosci 2021; 15:743546. [PMID: 34720864 PMCID: PMC8549820 DOI: 10.3389/fnins.2021.743546] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 09/16/2021] [Indexed: 12/19/2022] Open
Abstract
Obesity is a growing health concern, as it increases risk for heart disease, hypertension, type 2 diabetes, cancer, COVID-19 related hospitalizations and mortality. However, current weight loss therapies are often associated with psychiatric or cardiovascular side effects or poor tolerability that limit their long-term use. The hypothalamic neuropeptide, oxytocin (OT), mediates a wide range of physiologic actions, which include reproductive behavior, formation of prosocial behaviors and control of body weight. We and others have shown that OT circumvents leptin resistance and elicits weight loss in diet-induced obese rodents and non-human primates by reducing both food intake and increasing energy expenditure (EE). Chronic intranasal OT also elicits promising effects on weight loss in obese humans. This review evaluates the potential use of OT as a therapeutic strategy to treat obesity in rodents, non-human primates, and humans, and identifies potential mechanisms that mediate this effect.
Collapse
Affiliation(s)
- JingJing Niu
- VA Puget Sound Health Care System, Office of Research and Development, Medical Research Service, Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States.,Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States
| | - Jenny Tong
- VA Puget Sound Health Care System, Office of Research and Development, Medical Research Service, Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States.,Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States
| | - James E Blevins
- VA Puget Sound Health Care System, Office of Research and Development, Medical Research Service, Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States.,Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
13
|
Anekonda VT, Thompson BW, Ho JM, Roberts ZS, Edwards MM, Nguyen HK, Dodson AD, Wolden-Hanson T, Chukri DW, Herbertson AJ, Graham JL, Havel PJ, Wietecha TA, O’Brien KD, Blevins JE. Hindbrain Administration of Oxytocin Reduces Food Intake, Weight Gain and Activates Catecholamine Neurons in the Hindbrain Nucleus of the Solitary Tract in Rats. J Clin Med 2021; 10:5078. [PMID: 34768597 PMCID: PMC8584350 DOI: 10.3390/jcm10215078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 11/16/2022] Open
Abstract
Existing studies show that CNS oxytocin (OT) signaling is important in the control of energy balance, but it is unclear which neurons may contribute to these effects. Our goals were to examine (1) the dose-response effects of acute OT administration into the third (3V; forebrain) and fourth (4V; hindbrain) ventricles to assess sensitivity to OT in forebrain and hindbrain sites, (2) the extent to which chronic 4V administration of OT reduces weight gain associated with the progression of diet-induced obesity, and (3) whether nucleus tractus solitarius (NTS) catecholamine neurons are downstream targets of 4V OT. Initially, we examined the dose-response effects of 3V and 4V OT (0.04, 0.2, 1, or 5 μg). 3V and 4V OT (5 μg) suppressed 0.5-h food intake by 71.7 ± 6.0% and 60 ± 12.9%, respectively. 4V OT (0.04, 0.2, 1 μg) reduced food intake by 30.9 ± 12.9, 42.1 ± 9.4, and 56.4 ± 9.0%, respectively, whereas 3V administration of OT (1 μg) was only effective at reducing 0.5-h food intake by 38.3 ± 10.9%. We subsequently found that chronic 4V OT infusion, as with chronic 3V infusion, reduced body weight gain (specific to fat mass) and tended to reduce plasma leptin in high-fat diet (HFD)-fed rats, in part, through a reduction in energy intake. Lastly, we determined that 4V OT increased the number of hindbrain caudal NTS Fos (+) neurons (156 ± 25) relative to vehicle (12 ± 3). The 4V OT also induced Fos in tyrosine hydroxylase (TH; marker of catecholamine neurons) (+) neurons (25 ± 7%) relative to vehicle (0.8 ± 0.3%). Collectively, these findings support the hypothesis that OT within the hindbrain is effective at reducing food intake, weight gain, and adiposity and that NTS catecholamine neurons in addition to non-catecholaminergic neurons are downstream targets of CNS OT.
Collapse
Affiliation(s)
- Vishwanath T. Anekonda
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (V.T.A.); (B.W.T.); (J.M.H.); (Z.S.R.); (M.M.E.); (H.K.N.); (A.D.D.); (T.W.-H.); (D.W.C.); (A.J.H.)
| | - Benjamin W. Thompson
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (V.T.A.); (B.W.T.); (J.M.H.); (Z.S.R.); (M.M.E.); (H.K.N.); (A.D.D.); (T.W.-H.); (D.W.C.); (A.J.H.)
| | - Jacqueline M. Ho
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (V.T.A.); (B.W.T.); (J.M.H.); (Z.S.R.); (M.M.E.); (H.K.N.); (A.D.D.); (T.W.-H.); (D.W.C.); (A.J.H.)
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA;
| | - Zachary S. Roberts
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (V.T.A.); (B.W.T.); (J.M.H.); (Z.S.R.); (M.M.E.); (H.K.N.); (A.D.D.); (T.W.-H.); (D.W.C.); (A.J.H.)
| | - Melise M. Edwards
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (V.T.A.); (B.W.T.); (J.M.H.); (Z.S.R.); (M.M.E.); (H.K.N.); (A.D.D.); (T.W.-H.); (D.W.C.); (A.J.H.)
| | - Ha K. Nguyen
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (V.T.A.); (B.W.T.); (J.M.H.); (Z.S.R.); (M.M.E.); (H.K.N.); (A.D.D.); (T.W.-H.); (D.W.C.); (A.J.H.)
| | - Andrew D. Dodson
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (V.T.A.); (B.W.T.); (J.M.H.); (Z.S.R.); (M.M.E.); (H.K.N.); (A.D.D.); (T.W.-H.); (D.W.C.); (A.J.H.)
| | - Tami Wolden-Hanson
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (V.T.A.); (B.W.T.); (J.M.H.); (Z.S.R.); (M.M.E.); (H.K.N.); (A.D.D.); (T.W.-H.); (D.W.C.); (A.J.H.)
| | - Daniel W. Chukri
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (V.T.A.); (B.W.T.); (J.M.H.); (Z.S.R.); (M.M.E.); (H.K.N.); (A.D.D.); (T.W.-H.); (D.W.C.); (A.J.H.)
| | - Adam J. Herbertson
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (V.T.A.); (B.W.T.); (J.M.H.); (Z.S.R.); (M.M.E.); (H.K.N.); (A.D.D.); (T.W.-H.); (D.W.C.); (A.J.H.)
| | - James L. Graham
- Department of Nutrition and Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (J.L.G.); (P.J.H.)
| | - Peter J. Havel
- Department of Nutrition and Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (J.L.G.); (P.J.H.)
| | - Tomasz A. Wietecha
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA;
- UW Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, WA 98109, USA;
| | - Kevin D. O’Brien
- UW Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, WA 98109, USA;
- Division of Cardiology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - James E. Blevins
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; (V.T.A.); (B.W.T.); (J.M.H.); (Z.S.R.); (M.M.E.); (H.K.N.); (A.D.D.); (T.W.-H.); (D.W.C.); (A.J.H.)
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA;
| |
Collapse
|
14
|
Duan C, Huang L, Zhang C, Zhang L, Xia X, Zhong Z, Wang B, Wang Y, Man Hoi MP, Ding W, Yang Y. Gut commensal-derived butyrate reverses obesity-induced social deficits and anxiety-like behaviors via regulation of microglial homeostasis. Eur J Pharmacol 2021; 908:174338. [PMID: 34270984 DOI: 10.1016/j.ejphar.2021.174338] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 06/17/2021] [Accepted: 07/11/2021] [Indexed: 12/15/2022]
Abstract
Neuropsychiatric dysfunction and reactive microglia are hallmarks of high-fat diet (HFD)-induced obesity, yet whether these reactive microglia contribute to HFD-induced obesity-related behavioral abnormalities and the underlying mechanisms remain unclear. Here, we show that HFD feeding causes social deficits and anxiety-like behaviors with impaired neuronal activity and alters the gut microbiota, particularly by depleting Lactobacillus reuteri (L. reuteri), in mice. The profiles of microbiome and metabolome in HFD-fed mice predict that specific microbial taxa and their metabolites regulate HFD-induced obesity-related behavioral abnormalities. Oral treatment with the L. reuteri reduces microglial activation and increases dendritic spine density, thus ameliorates social deficits and anxiety in HFD-fed mice. HFD-fed mice that are administered L. reuteri are also found to accumulate butyrate in their gut, sera and brain. Moreover, supplementation of butyrate improves behavioral abnormalities and modulates microglial homeostasis in HFD-fed mice. In addition, selectively removal of microglia through a pharmacologic approach can rescue dendritic spine loss and increase neuronal activity that profoundly alleviates social deficits and anxiety arising from HFD-induced obesity. Overall, this study reveals an unexpected pivotal role of gut commensal-derived butyrate in HFD-induced social deficits and anxiety-like behaviors through regulation of microglial homeostasis and identifies a potential probiotic treatment for HFD-induced obesity-related behavioral abnormalities.
Collapse
Affiliation(s)
- Chengxing Duan
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ling Huang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Chi Zhang
- College of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Lu Zhang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xiuwen Xia
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Zhanqiong Zhong
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Baojia Wang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yili Wang
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Maggie Pui Man Hoi
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China
| | - Weijun Ding
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Youjun Yang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China.
| |
Collapse
|
15
|
Ortiz-Valladares M, Pedraza-Medina R, Pinto-González MF, Muñiz JG, Gonzalez-Perez O, Moy-López NA. Neurobiological approaches of high-fat diet intake in early development and their impact on mood disorders in adulthood: A systematic review. Neurosci Biobehav Rev 2021; 129:218-230. [PMID: 34324919 DOI: 10.1016/j.neubiorev.2021.07.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/14/2021] [Accepted: 07/25/2021] [Indexed: 01/21/2023]
Abstract
The early stage of development is a vulnerable period for progeny neurodevelopment, altering cytogenetic and correct cerebral functionality. The exposure High-Fat Diet (HFD) is a factor that impacts the future mental health of individuals. This review analyzes possible mechanisms involved in the development of mood disorders in adulthood because of maternal HFD intake during gestation and lactation, considering previously reported findings in the last five years, both in humans and animal models. Maternal HFD could induce alterations in mood regulation, reported as increased stress response, anxiety-like behavior, and depressive-like behavior. These changes were mostly related to HPA axis dysregulations and neuroinflammatory responses. In conclusion, there could be a relationship between HFD consumption during the early stages of life and the development of psychopathologies during adulthood. These findings provide guidelines for the understanding of possible mechanisms involved in mood disorders, however, there is still a need for more human clinical studies that provide evidence to improve the understanding of maternal nutrition and future mental health outcomes in the offspring.
Collapse
Affiliation(s)
| | - Ricardo Pedraza-Medina
- Medical Science Postgraduate Program, School of Medicine, University of Colima, Colima, Mexico
| | | | - Jorge Guzmán Muñiz
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima, Mexico
| | - Oscar Gonzalez-Perez
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima, Mexico
| | | |
Collapse
|
16
|
Komleva Y, Chernykh A, Lopatina O, Gorina Y, Lokteva I, Salmina A, Gollasch M. Inflamm-Aging and Brain Insulin Resistance: New Insights and Role of Life-style Strategies on Cognitive and Social Determinants in Aging and Neurodegeneration. Front Neurosci 2021; 14:618395. [PMID: 33519369 PMCID: PMC7841337 DOI: 10.3389/fnins.2020.618395] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
Over the past decades, the human life span has dramatically increased, and therefore, a steady increase in diseases associated with age (such as Alzheimer's disease and Parkinson's disease) is expected. In these neurodegenerative diseases, there is a cognitive decline and memory loss, which accompany increased systemic inflammation, the inflamm-aging, and the insulin resistance. Despite numerous studies of age-related pathologies, data on the contribution of brain insulin resistance and innate immunity components to aging are insufficient. Recently, much research has been focused on the consequences of nutrients and adiposity- and nutrient-related signals in brain aging and cognitive decline. Moreover, given the role of metainflammation in neurodegeneration, lifestyle interventions such as calorie restriction may be an effective way to break the vicious cycle of metainflammation and have a role in social behavior. The various effects of calorie restriction on metainflammation, insulin resistance, and neurodegeneration have been described. Less attention has been paid to the social determinants of aging and the possible mechanism by which calorie restriction might influence social behavior. The purpose of this review is to discuss current knowledge in the interdisciplinary field of geroscience-immunosenescence, inflamm-aging, and metainflammation-which makes a significant contribution to aging. A substantial part of the review is devoted to frontiers in the brain insulin resistance in relation to neuroinflammation. In addition, we summarize new data on potential mechanisms of calorie restriction that influence as a lifestyle intervention on the social brain. This knowledge can be used to initiate successful aging and slow the onset of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yulia Komleva
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Biochemistry, Medical, Pharmaceutical & Toxicological Chemistry, Krasnoyarsk State Medical University named after Professor V.F. Voyno-Yasenetsky, Ministry of Health of the Russian Federation, Krasnoyarsk, Russia
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Professor V.F. Voyno-Yasenetsky, Ministry of Health of the Russian Federation, Krasnoyarsk, Russia
| | - Anatoly Chernykh
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Professor V.F. Voyno-Yasenetsky, Ministry of Health of the Russian Federation, Krasnoyarsk, Russia
| | - Olga Lopatina
- Department of Biochemistry, Medical, Pharmaceutical & Toxicological Chemistry, Krasnoyarsk State Medical University named after Professor V.F. Voyno-Yasenetsky, Ministry of Health of the Russian Federation, Krasnoyarsk, Russia
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Professor V.F. Voyno-Yasenetsky, Ministry of Health of the Russian Federation, Krasnoyarsk, Russia
| | - Yana Gorina
- Department of Biochemistry, Medical, Pharmaceutical & Toxicological Chemistry, Krasnoyarsk State Medical University named after Professor V.F. Voyno-Yasenetsky, Ministry of Health of the Russian Federation, Krasnoyarsk, Russia
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Professor V.F. Voyno-Yasenetsky, Ministry of Health of the Russian Federation, Krasnoyarsk, Russia
| | - Irina Lokteva
- Medical Center “Private Practice”, Krasnoyarsk, Russia
| | - Alla Salmina
- Experimental and Clinical Research Center (ECRC), Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Biochemistry, Medical, Pharmaceutical & Toxicological Chemistry, Krasnoyarsk State Medical University named after Professor V.F. Voyno-Yasenetsky, Ministry of Health of the Russian Federation, Krasnoyarsk, Russia
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Professor V.F. Voyno-Yasenetsky, Ministry of Health of the Russian Federation, Krasnoyarsk, Russia
| | - Maik Gollasch
- Experimental and Clinical Research Center (ECRC), Charité - Universitätsmedizin Berlin, Berlin, Germany
- Greifswald Medical School, University of Greifswald, Greifswald, Germany
- Geriatric Medicine Center, Wolgast Hospital, Wolgast, Germany
| |
Collapse
|
17
|
Wei J, Ma L, Ju P, Yang B, Wang YX, Chen J. Involvement of Oxytocin Receptor/Erk/MAPK Signaling in the mPFC in Early Life Stress-Induced Autistic-Like Behaviors. Front Cell Dev Biol 2020; 8:564485. [PMID: 33134294 PMCID: PMC7561716 DOI: 10.3389/fcell.2020.564485] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 09/10/2020] [Indexed: 12/13/2022] Open
Abstract
The neonatal or infant period is a critical stage for the development of brain neuroplasticity. Early life stresses in the neonatal period, including neonatal maternal separation (NMS), have adverse effects on an increased risk of psychiatric disorders in juveniles and adults. However, the underlying molecular mechanisms are not largely understood. Here, we found that juvenile rats subjected to 4 h daily NMS during postnatal days 1 to 20 exhibited autistic-like behavioral deficits without impairments in learning and memory functions. Molecular mechanism studies showed that oxytocin receptor (OXTR) in the medial prefrontal cortex of NMS rats was evidently downregulated when compared with control pups, especially in neurons. Erk/MAPK signaling, the downstream coupling signaling of OTXR, was also inhibited in NMS juvenile rats. Treatment with oxytocin could relieve NMS-induced social deficit behaviors and activated phosphorylation of Erk/MAPK signaling. Furthermore, medication with the inhibitor of H3K4 demethylase alleviated the abnormal behaviors in NMS rats and increased the expression of OXTR in the medial prefrontal cortex, which showed an epigenetic mechanism underlying social deficits induced by NMS. Taken together, these findings identified a molecular mechanism by which disruptions of mother-infant interactions influenced later displays of typical social behaviors and suggested the potential for NMS-driven epigenetic tuning of OXTR expression.
Collapse
Affiliation(s)
- Jinbao Wei
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,King's Lab, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China.,Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Le Ma
- King's Lab, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Peijun Ju
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Beibei Yang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yong-Xiang Wang
- King's Lab, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Jinghong Chen
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|