1
|
Lin H, Xu Y, Xiong H, Wang L, Shi Y, Wang D, Wang Z, Ren J, Wang S. Mechanism of action of Panax ginseng alcohol extract based on orexin-mediated autophagy in the treatment of sleep and cognition in aged sleep-deprived rats. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118907. [PMID: 39389397 DOI: 10.1016/j.jep.2024.118907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/29/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Panax ginseng (P. ginseng) C. A. Meyer. has been used extensively globally as a medicine. It has a therapeutic effect on sleep and is an attractive alternative for patients with insomnia. The United States Patent of Invention has approved the use of P. ginseng alcohol extract (GAE) in nutraceuticals or food to improve sleep. It has shown promise as an effective therapeutic agent for improving sleep and cognition. However, its mechanism of action is not yet fully understood. AIM OF THE STUDY To investigate the therapeutic benefits of GAE on sleep and cognition and its underlying mechanism in aged sleep-deprived rats, with a focus on orexin-mediated autophagy function. MATERIALS AND METHODS We conducted in vivo tests in an aged sleep-deprivation rat model produced using p-chlorophenylalanine (PCPA) coupled with modified multi-platform method to examine the therapeutic effects and mechanisms of GAE. A pentobarbital sodium-induced sleep test and water maze were used to assess sleep and cognitive performance, respectively. An enzyme-linked immunosorbent assay was used to determine orexin levels and aging and sleep markers in serum and hypothalamic tissues. Hematoxylin-eosin staining and Nissl staining were used to assess histopathological changes, and autophagy levels were assessed using transmission electron microscopy, immunofluorescence. Western blot and immunohistochemical staining were performed to detect the levels of orexin, orexin-receptor proteins, and autophagy-associated proteins to study the effects of GAE on hippocampal neurons, and the underlying mechanisms. RESULTS In aged sleep-deprived rats, GAE treatment prolonged sleep duration, improved cognitive function, prevented hippocampal neuronal damage, increased the number of Nissl bodies, improved aging and sleep markers, and enhanced the LC3A/B expression in autophagosomes and neurons. The amount of orexin in serum and hypothalamic tissue and OX1R, OX2R, and phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) proteins also reduced, which resulted in the inhibition of the PI3K/Akt/mTOR pathway and activation of the autophagy process. CONCLUSIONS GAE may reduce hypothalamic orexin secretion and interact with orexin receptors to inhibit the PI3K/Akt/mTOR signalling network and activate autophagy. This may be a potential mechanism of action of GAE in regulating sleep-related cognitive function.
Collapse
Affiliation(s)
- Haining Lin
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Yunlong Xu
- Prevention and Treatment Center, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Huazhong Xiong
- Prevention and Treatment Center, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Lichao Wang
- Prevention and Treatment Center, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Yuqing Shi
- College of Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Dongyi Wang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Zixu Wang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Jixiang Ren
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China; Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China.
| | - Siming Wang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China; Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China; Key Laboratory of Ginseng Efficacy Substance Base and Biological Mechanism Research, Ministry of Education, Changchun University of Chinese Medicine, Changchun, 130117, China.
| |
Collapse
|
2
|
Osiecka Z, Fausto BA, Gills JL, Sinha N, Malin SK, Gluck MA. Obesity reduces hippocampal structure and function in older African Americans with the APOE-ε4 Alzheimer's disease risk allele. Front Aging Neurosci 2023; 15:1239727. [PMID: 37731955 PMCID: PMC10507275 DOI: 10.3389/fnagi.2023.1239727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/15/2023] [Indexed: 09/22/2023] Open
Abstract
Introduction Excess body weight and Alzheimer's disease (AD) disproportionately affect older African Americans. While mid-life obesity increases risk for AD, few data exist on the relationship between late-life obesity and AD, or how obesity-based and genetic risk for AD interact. Although the APOE-ε4 allele confers a strong genetic risk for AD, it is unclear if late-life obesity poses a greater risk for APOE-ε4 carriers compared to non-carriers. Here we assessed: (1) the influence of body mass index (BMI) (normal; overweight; class 1 obese; ≥ class 2 obese) on cognitive and structural MRI measures of AD risk; and (2) the interaction between BMI and APOE-ε4 in older African Americans. Methods Seventy cognitively normal older African American participants (Mage = 69.50 years; MBMI = 31.01 kg/m2; 39% APOE-ε4 allele carriers; 86% female) completed anthropometric measurements, physical assessments, saliva collection for APOE-ε4 genotyping, cognitive testing, health and lifestyle questionnaires, and structural neuroimaging [volume/surface area (SA) for medial temporal lobe subregions and hippocampal subfields]. Covariates included age, sex, education, literacy, depressive symptomology, and estimated aerobic fitness. Results Using ANCOVAs, we observed that individuals who were overweight demonstrated better hippocampal cognitive function (generalization of learning: a sensitive marker of preclinical AD) than individuals with normal BMI, p = 0.016, ηp2 = 0.18. However, individuals in the obese categories who were APOE-ε4 non-carriers had larger hippocampal subfield cornu Ammonis region 1 (CA1) volumes, while those who were APOE-ε4 carriers had smaller CA1 volumes, p = 0.003, ηp2 = 0.23. Discussion Thus, being overweight by BMI standards may preserve hippocampal function, but obesity reduces hippocampal structure and function in older African Americans with the APOE-ε4 Alzheimer's disease risk allele.
Collapse
Affiliation(s)
- Zuzanna Osiecka
- Aging and Brain Health Alliance, Center for Molecular and Behavioral Neuroscience, Rutgers University–Newark, Newark, NJ, United States
| | - Bernadette A. Fausto
- Aging and Brain Health Alliance, Center for Molecular and Behavioral Neuroscience, Rutgers University–Newark, Newark, NJ, United States
| | - Joshua L. Gills
- Aging and Brain Health Alliance, Center for Molecular and Behavioral Neuroscience, Rutgers University–Newark, Newark, NJ, United States
| | - Neha Sinha
- Aging and Brain Health Alliance, Center for Molecular and Behavioral Neuroscience, Rutgers University–Newark, Newark, NJ, United States
| | - Steven K. Malin
- Department of Kinesiology and Health, School of Arts and Sciences, Rutgers University, New Brunswick, NJ, United States
| | - Mark A. Gluck
- Aging and Brain Health Alliance, Center for Molecular and Behavioral Neuroscience, Rutgers University–Newark, Newark, NJ, United States
| |
Collapse
|
3
|
Chen HL, Tan CT, Wu CC, Liu TC. Effects of Diet and Lifestyle on Audio-Vestibular Dysfunction in the Elderly: A Literature Review. Nutrients 2022; 14:nu14224720. [PMID: 36432406 PMCID: PMC9698578 DOI: 10.3390/nu14224720] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/27/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND The world's age-related health concerns continue to rise. Audio-vestibular disorders, such as hearing loss, tinnitus, and vertigo, are common complaints in the elderly and are associated with social and public health burdens. Various preventative measures can ease their impact, including healthy food consumption, nutritional supplementation, and lifestyle modification. We aim to provide a comprehensive summary of current possible strategies for preventing the age-related audio-vestibular dysfunction. METHODS A PubMed, Embase, and Cochrane review databases search was conducted to identify the relationship between diet, lifestyle, and audio-vestibular dysfunction. "Diet", "nutritional supplement", "lifestyle", "exercise", "physical activity", "tinnitus", "vertigo" and "age-related hearing loss" were used as keywords. RESULTS Audio-vestibular dysfunction develops and progresses as a result of age-related inflammation and oxidative stress. Diets with anti-inflammatory and antioxidant effects have been proposed to alleviate this illness. A high-fat diet may induce oxidative stress and low protein intake is associated with hearing discomfort in the elderly. Increased carbohydrate and sugar intake positively correlate with the incidence of audio-vestibular dysfunction, whereas a Mediterranean-style diet can protect against the disease. Antioxidants in the form of vitamins A, C, and E; physical activity; good sleep quality; smoking cessation; moderate alcohol consumption; and avoiding noise exposure are also beneficial. CONCLUSIONS Adequate diet or nutritional interventions with lifestyle modification may protect against developing audio-vestibular dysfunction in elderly individuals.
Collapse
Affiliation(s)
- Hsin-Lin Chen
- Department of Surgical Oncology, National Taiwan University Cancer Center Hospital, Taipei 100, Taiwan
- Department of Otolaryngology, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Ching-Ting Tan
- Department of Otolaryngology, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Chen-Chi Wu
- Department of Otolaryngology, National Taiwan University Hospital, Taipei 100, Taiwan
- Department of Medical Research, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu 302, Taiwan
- Department of Medical Genetics, National Taiwan University Hospital, Taipei 100, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei 100, Taiwan
- Correspondence: (C.-C.W.); (T.-C.L.)
| | - Tien-Chen Liu
- Department of Otolaryngology, National Taiwan University Hospital, Taipei 100, Taiwan
- Correspondence: (C.-C.W.); (T.-C.L.)
| |
Collapse
|
4
|
Hua Y, Shen J, Fan R, Xiao R, Ma W. High-fat diets containing different types of fatty acids modulate gut-brain axis in obese mice. Nutr Metab (Lond) 2022; 19:40. [PMID: 35739547 PMCID: PMC9219185 DOI: 10.1186/s12986-022-00675-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 05/17/2022] [Indexed: 11/10/2022] Open
Abstract
Background Excessive consumption of high-fat diets is associated with disordered metabolic responses, which may lead to chronic diseases. High-fat diets containing different types of fatty acids lead to distinct alterations in metabolic responses of gut-brain axis. Methods In our study, normal male C57BL/6J mice were fed to multiple high fatty acid diets (long-chain and medium-chain saturated fatty acid, LCSFA and MCSFA group; n-3 and n-6 polyunsaturated fatty acid, n-3 and n-6 PUFA group; monounsaturated fatty acid, MUFA group; trans fatty acid, TFA group) and a basic diet (control, CON group) for 19 weeks. To investigate the effects of high-fat diets on metabolic responses of gut-brain axis in obese mice, blood lipids were detected by fast gas chromatography, and related proteins in brain and intestine were detected using Western blotting, ELISA, and immunochemistry analysis. Results All high-fat diets regardless of their fatty acid composition induced obesity, lipid disorders, intestinal barrier dysfunction, and changes in gut-brain axis related factors except basal diet in mice. For example, the protein expression of zonula occludens-1 (ZO-1) in ileum in the n-3 PUFA group was higher than that in the MCSFA group (P < 0.05). The expressions of insulin in hippocampus and leptin in ileum in the MCSFA group significantly increased, compared with other groups (all Ps < 0.05). Conclusion The high MCSFA diet had the most effect on metabolic disorders in gut-brain axis, but the high n-3 PUFA diet had the least effect on changes in metabolism.
Collapse
Affiliation(s)
- Yinan Hua
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing, 100069, People's Republic of China
| | - Jingyi Shen
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing, 100069, People's Republic of China
| | - Rong Fan
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing, 100069, People's Republic of China
| | - Rong Xiao
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing, 100069, People's Republic of China.
| | - Weiwei Ma
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing, 100069, People's Republic of China.
| |
Collapse
|
5
|
Abstract
The hypocretins (Hcrts), also known as orexins, are two neuropeptides produced exclusively in the lateral hypothalamus. They act on two specific receptors that are widely distributed across the brain and involved in a myriad of neurophysiological functions that include sleep, arousal, feeding, reward, fear, anxiety and cognition. Hcrt cell loss in humans leads to narcolepsy with cataplexy (narcolepsy type 1), a disorder characterized by intrusions of sleep into wakefulness, demonstrating that the Hcrt system is nonredundant and essential for sleep/wake stability. The causal link between Hcrts and arousal/wakefulness stabilisation has led to the development of a new class of drugs, Hcrt receptor antagonists to treat insomnia, based on the assumption that blocking orexin-induced arousal will facilitate sleep. This has been clinically validated: currently, two Hcrt receptor antagonists are approved to treat insomnia (suvorexant and lemborexant), with a New Drug Application recently submitted to the US Food and Drug Administration for a third drug (daridorexant). Other therapeutic applications under investigation include reduction of cravings in substance-use disorders and prevention of neurodegenerative disorders such as Alzheimer's disease, given the apparent bidirectional relationship between poor sleep and worsening of the disease. Circuit neuroscience findings suggest that the Hcrt system is a hub that integrates diverse inputs modulating arousal (e.g., circadian rhythms, metabolic status, positive and negative emotions) and conveys this information to multiple output regions. This neuronal architecture explains the wealth of physiological functions associated with Hcrts and highlights the potential of the Hcrt system as a therapeutic target for a number of disorders. We discuss present and future possible applications of drugs targeting the Hcrt system for the treatment of circuit-related neuropsychiatric and neurodegenerative conditions.
Collapse
Affiliation(s)
- Laura H Jacobson
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia.,Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia.,Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Daniel Hoyer
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia.,Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia.,Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
6
|
Zhan D, Perrer DA, Decker AM, Langston TL, Mavanji V, Harris DL, Kotz CM, Zhang Y. Discovery of Arylsulfonamides as Dual Orexin Receptor Agonists. J Med Chem 2021; 64:8806-8825. [PMID: 34101446 PMCID: PMC8994207 DOI: 10.1021/acs.jmedchem.1c00841] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Loss of orexin-producing neurons results in narcolepsy with cataplexy, and orexin agonists have been shown to increase wakefulness and alleviate narcolepsy symptoms in animal models. Several OX2R agonists have been reported but with little or no activity at OX1R. We conducted structure-activity relationship studies on the OX2R agonist YNT-185 (2) and discovered dual agonists such as RTOXA-43 (40) with EC50's of 24 nM at both OX2R and OX1R. Computational modeling studies based on the agonist-bound OX2R cryogenic electron microscopy structures showed that 40 bound in the same binding pocket and interactions of the pyridylmethyl group of 40 with OX1R may have contributed to its high OX1R potency. Intraperitoneal injection of 40 increased time awake, decreased time asleep, and increased sleep/wake consolidation in 12-month old mice. This work provides a promising dual small molecule agonist and supports development of orexin agonists as potential treatments for orexin-deficient disorders such as narcolepsy.
Collapse
Affiliation(s)
- Dehui Zhan
- Research Triangle Institute, Research Triangle Park, North Carolina 27709
| | - David A. Perrer
- Research Triangle Institute, Research Triangle Park, North Carolina 27709
| | - Ann M. Decker
- Research Triangle Institute, Research Triangle Park, North Carolina 27709
| | | | - Vijayakumar Mavanji
- Research Service, Veterans Affairs Health Care System, Minneapolis, MN 55417
| | - Danni L. Harris
- Research Triangle Institute, Research Triangle Park, North Carolina 27709
| | - Catherine M. Kotz
- Research Service, Veterans Affairs Health Care System, Minneapolis, MN 55417
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN 55455
- Geriatric, Research, Education and Clinical Center, Minneapolis Veterans Affairs Health Care System, Minneapolis, MN 55417
| | - Yanan Zhang
- Research Triangle Institute, Research Triangle Park, North Carolina 27709
| |
Collapse
|
7
|
Erichsen JM, Calva CB, Reagan LP, Fadel JR. Intranasal insulin and orexins to treat age-related cognitive decline. Physiol Behav 2021; 234:113370. [PMID: 33621561 PMCID: PMC8053680 DOI: 10.1016/j.physbeh.2021.113370] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 02/19/2021] [Indexed: 02/06/2023]
Abstract
The intranasal (IN) administration of neuropeptides, such as insulin and orexins, has been suggested as a treatment strategy for age-related cognitive decline (ARCD). Because dysfunctional neuropeptide signaling is an observed characteristic of ARCD, it has been suggested that IN delivery of insulin and/or orexins may restore endogenous peptide signaling and thereby preserve cognition. IN administration is particularly alluring as it is a relatively non-invasive method that directly targets peptides to the brain. Several laboratories have examined the behavioral effects of IN insulin in young, aged, and cognitively impaired rodents and humans. These studies demonstrated improved performance on various cognitive tasks following IN insulin administration. Fewer laboratories have assessed the effects of IN orexins; however, this peptide also holds promise as an effective treatment for ARCD through the activation of the cholinergic system and/or the reduction of neuroinflammation. Here, we provide a brief overview of the advantages of IN administration and the delivery pathway, then summarize the current literature on IN insulin and orexins. Additional preclinical studies will be useful to ultimately uncover the mechanisms underlying the pro-cognitive effects of IN insulin and orexins, whereas future clinical studies will aid in the determination of the most efficacious dose and dosing paradigm. Eventually, IN insulin and/or orexin administration may be a widely used treatment strategy in the clinic for ARCD.
Collapse
Affiliation(s)
- Jennifer M Erichsen
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC 29208, United States.
| | - Coleman B Calva
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC 29208, United States
| | - Lawrence P Reagan
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC 29208, United States; Columbia VA Health Care System, Columbia, SC, 29208, United States
| | - Jim R Fadel
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC 29208, United States
| |
Collapse
|
8
|
Penna E, Pizzella A, Cimmino F, Trinchese G, Cavaliere G, Catapano A, Allocca I, Chun JT, Campanozzi A, Messina G, Precenzano F, Lanzara V, Messina A, Monda V, Monda M, Perrone-Capano C, Mollica MP, Crispino M. Neurodevelopmental Disorders: Effect of High-Fat Diet on Synaptic Plasticity and Mitochondrial Functions. Brain Sci 2020; 10:brainsci10110805. [PMID: 33142719 PMCID: PMC7694125 DOI: 10.3390/brainsci10110805] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 02/07/2023] Open
Abstract
Neurodevelopmental disorders (NDDs) include diverse neuropathologies characterized by abnormal brain development leading to impaired cognition, communication and social skills. A common feature of NDDs is defective synaptic plasticity, but the underlying molecular mechanisms are only partially known. Several studies have indicated that people’s lifestyles such as diet pattern and physical exercise have significant influence on synaptic plasticity of the brain. Indeed, it has been reported that a high-fat diet (HFD, with 30–50% fat content), which leads to systemic low-grade inflammation, has also a detrimental effect on synaptic efficiency. Interestingly, metabolic alterations associated with obesity in pregnant woman may represent a risk factor for NDDs in the offspring. In this review, we have discussed the potential molecular mechanisms linking the HFD-induced metabolic dysfunctions to altered synaptic plasticity underlying NDDs, with a special emphasis on the roles played by synaptic protein synthesis and mitochondrial functions.
Collapse
Affiliation(s)
- Eduardo Penna
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (F.C.); (G.T.); (G.C.); (A.C.); (I.A.); (M.C.)
| | - Amelia Pizzella
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (F.C.); (G.T.); (G.C.); (A.C.); (I.A.); (M.C.)
| | - Fabiano Cimmino
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (F.C.); (G.T.); (G.C.); (A.C.); (I.A.); (M.C.)
| | - Giovanna Trinchese
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (F.C.); (G.T.); (G.C.); (A.C.); (I.A.); (M.C.)
| | - Gina Cavaliere
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (F.C.); (G.T.); (G.C.); (A.C.); (I.A.); (M.C.)
| | - Angela Catapano
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (F.C.); (G.T.); (G.C.); (A.C.); (I.A.); (M.C.)
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy;
| | - Ivana Allocca
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (F.C.); (G.T.); (G.C.); (A.C.); (I.A.); (M.C.)
| | - Jong Tai Chun
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, 80121 Naples, Italy;
| | - Angelo Campanozzi
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy;
| | - Giovanni Messina
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy;
| | - Francesco Precenzano
- Department of Mental Health, Physical and Preventive Medicine, Clinic of Child and Adolescent Neuropsychiatry, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (F.P.); (V.L.)
| | - Valentina Lanzara
- Department of Mental Health, Physical and Preventive Medicine, Clinic of Child and Adolescent Neuropsychiatry, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (F.P.); (V.L.)
| | - Antonietta Messina
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.M.); (M.M.)
| | - Vincenzo Monda
- Department of Experimental Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 81100 Caserta, Italy;
| | - Marcellino Monda
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.M.); (M.M.)
| | - Carla Perrone-Capano
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy;
- Institute of Genetics and Biophysics “Adriano Buzzati Traverso”, CNR, 80131 Naples, Italy
| | - Maria Pina Mollica
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (F.C.); (G.T.); (G.C.); (A.C.); (I.A.); (M.C.)
- Correspondence: ; Tel.: +39-081-679990; Fax: +39-081-679233
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (F.C.); (G.T.); (G.C.); (A.C.); (I.A.); (M.C.)
| |
Collapse
|
9
|
Um YH, Lim HK. Orexin and Alzheimer's Disease: A New Perspective. Psychiatry Investig 2020; 17:621-626. [PMID: 32517419 PMCID: PMC7385219 DOI: 10.30773/pi.2020.0136] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 05/12/2020] [Indexed: 12/13/2022] Open
Abstract
Orexin's role in human cognition has recently been emphasized and emerging evidences indicate its close relationship with Alzheimer's disease (AD). This review aimed to demonstrate recent research on the relationship between orexin and AD. Orexin's role in stress regulation and memory is discussed, with significant findings related to sexual disparities in stress response, with potential clinical implications pertaining to AD pathology. There are controversies regarding the orexin levels in AD patients, but the role of orexin in the trajectory of AD is still emphasized in recent literatures. Orexin is also accentuated in the context of tau pathology, and orexin as a potential therapeutic target for AD is frequently discussed. Future directions with regard to the relationship between orexin and AD are suggested: 1) consideration for AD trajectory in the measurement of orexin levels, 2) the need for objective measure such as polysomnography and actigraphy, 3) the need for close observation of cognitive profiles of orexin-deficient narcolepsy patients, 4) the need for validation studies by neuroimaging 5) the need for taking account sexual disparities in orexinergic activiation, and 6) consideration for orexin's role as a stress regulator. The aforementioned new perspectives could help unravel the relationship between orexin and AD.
Collapse
Affiliation(s)
- Yoo Hyun Um
- Department of Psychiatry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyun Kook Lim
- Department of Psychiatry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
10
|
Khambadkone SG, Cordner ZA, Tamashiro KLK. Maternal stressors and the developmental origins of neuropsychiatric risk. Front Neuroendocrinol 2020; 57:100834. [PMID: 32084515 PMCID: PMC7243665 DOI: 10.1016/j.yfrne.2020.100834] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 01/23/2020] [Accepted: 02/12/2020] [Indexed: 12/14/2022]
Abstract
The maternal environment during pregnancy is critical for fetal development and perinatal perturbations can prime offspring disease risk. Here, we briefly review evidence linking two well-characterized maternal stressors - psychosocial stress and infection - to increased neuropsychiatric risk in offspring. In the current climate of increasing obesity and globalization of the Western-style diet, maternal overnutrition emerges as a pressing public health concern. We focus our attention on recent epidemiological and animal model evidence showing that, like psychosocial stress and infection, maternal overnutrition can also increase offspring neuropsychiatric risk. Using lessons learned from the psychosocial stress and infection literature, we discuss how altered maternal and placental physiology in the setting of overnutrition may contribute to abnormal fetal development and resulting neuropsychiatric outcomes. A better understanding of converging pathophysiological pathways shared between stressors may enable development of interventions against neuropsychiatric illnesses that may be beneficial across stressors.
Collapse
Affiliation(s)
- Seva G Khambadkone
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular & Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Zachary A Cordner
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kellie L K Tamashiro
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular & Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
11
|
Smith PJ, Mabe SM, Sherwood A, Doraiswamy PM, Welsh-Bohmer KA, Burke JR, Kraus WE, Lin PH, Browndyke JN, Babyak MA, Hinderliter AL, Blumenthal JA. Metabolic and Neurocognitive Changes Following Lifestyle Modification: Examination of Biomarkers from the ENLIGHTEN Randomized Clinical Trial. J Alzheimers Dis 2020; 77:1793-1803. [PMID: 32925039 PMCID: PMC9999371 DOI: 10.3233/jad-200374] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Previous studies have demonstrated that aerobic exercise (AE) and the Dietary Approaches to Stop Hypertension (DASH) diet can improve neurocognition. However, the mechanisms by which lifestyle improves neurocognition have not been widely studied. We examined the associations between changes in metabolic, neurotrophic, and inflammatory biomarkers with executive functioning among participants from the Exercise and Nutritional Interventions for Neurocognitive Health Enhancement (ENLIGHTEN) trial. OBJECTIVE To examine the association between changes in metabolic function and neurocognition among older adults with cognitive impairment, but without dementia (CIND) participating in a comprehensive lifestyle intervention. METHODS ENLIGHTEN participants were randomized using a 2×2 factorial design to receive AE, DASH, both AE+DASH, or a health education control condition (HE) for six months. Metabolic biomarkers included insulin resistance (homeostatic model assessment [HOMA-IR]), leptin, and insulin-like growth factor (IGF-1); neurotrophic biomarkers included brain derived neurotrophic factor (BDNF) and vascular endothelial growth factor (VEGF); and inflammatory biomarkers included interleukin-6 (IL-6) and C-Reactive Protein (CRP). RESULTS Participants included 132 sedentary older adults (mean age = 65 [SD = 7]) with CIND. Results demonstrated that both AE (d = 0.48, p = 0.015) and DASH improved metabolic function (d = 0.37, p = 0.039), without comparable improvements in neurotrophic or inflammatory biomarkers. Greater improvements in metabolic function, including reduced HOMA-IR (B = -2.3 [-4.3, -0.2], p = 0.033) and increased IGF-1 (B = 3.4 [1.2, 5.7], p = 0.004), associated with increases in Executive Function. CONCLUSION Changes in neurocognition after lifestyle modification are associated with improved metabolic function.
Collapse
Affiliation(s)
- Patrick J Smith
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Stephanie M Mabe
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Andrew Sherwood
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - P Murali Doraiswamy
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Kathleen A Welsh-Bohmer
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - James R Burke
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - William E Kraus
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Pao-Hwa Lin
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Jeffrey N Browndyke
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Michael A Babyak
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Alan L Hinderliter
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - James A Blumenthal
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
12
|
Smith PJ. Pathways of Prevention: A Scoping Review of Dietary and Exercise Interventions for Neurocognition. Brain Plast 2019; 5:3-38. [PMID: 31970058 PMCID: PMC6971820 DOI: 10.3233/bpl-190083] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease and related dementias (ADRD) represent an increasingly urgent public health concern, with an increasing number of baby boomers now at risk. Due to a lack of efficacious therapies among symptomatic older adults, an increasing emphasis has been placed on preventive measures that can curb or even prevent ADRD development among middle-aged adults. Lifestyle modification using aerobic exercise and dietary modification represents one of the primary treatment modalities used to mitigate ADRD risk, with an increasing number of trials demonstrating that exercise and dietary change, individually and together, improve neurocognitive performance among middle-aged and older adults. Despite several optimistic findings, examination of treatment changes across lifestyle interventions reveals a variable pattern of improvements, with large individual differences across trials. The present review attempts to synthesize available literature linking lifestyle modification to neurocognitive changes, outline putative mechanisms of treatment improvement, and discuss discrepant trial findings. In addition, previous mechanistic assumptions linking lifestyle to neurocognition are discussed, with a focus on potential solutions to improve our understanding of individual neurocognitive differences in response to lifestyle modification. Specific recommendations include integration of contemporary causal inference approaches for analyzing parallel mechanistic pathways and treatment-exposure interactions. Methodological recommendations include trial multiphase optimization strategy (MOST) design approaches that leverage individual differences for improved treatment outcomes.
Collapse
Affiliation(s)
- Patrick J. Smith
- Department of Psychiatry and Behavioral Sciences (Primary), Duke University Medical Center, NC, USA
- Department of Medicine (Secondary), Duke University Medical Center, NC, USA
- Department of Population Health Sciences (Secondary), Duke University, NC, USA
| |
Collapse
|
13
|
Nyberg CK, Fjell AM, Walhovd KB. Level of body fat relates to memory decline and interacts with age in its association with hippocampal and subcortical atrophy. Neurobiol Aging 2019; 91:112-124. [PMID: 32224068 DOI: 10.1016/j.neurobiolaging.2019.10.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 09/28/2019] [Accepted: 10/08/2019] [Indexed: 12/28/2022]
Abstract
Higher levels of body fat have shown adverse effects on multiple aspects of health, including cognitive and neuroanatomical changes. We tested the relationships of body fat levels and cholesterol to longitudinal age trajectories of subcortical gray matter volume (SCV), hippocampal volume (HCV), and episodic memory. Body fat was indexed by a concerted factor of BMI, visceral adipose tissue, percentage body fat, and total fat mass and was included in the analyses as a cross-sectional measure. We hypothesized that higher level of body fat would be related to steeper age trajectories of SCV, HCV, and memory. The sample consisted of 581 participants (20-83 years) with 942 magnetic resonance imaging and 945 memory examinations. Using generalized additive mixed models, a negative effect of body fat was found on SCV, HCV, and memory. Age and body fat interacted in their association with brain volume change. The results suggest that among cognitively healthy adults, there is a negative effect of higher body fat on SCV, HCV, and memory decline, an effect that increased with age for the neuroanatomical volumes.
Collapse
Affiliation(s)
- Claudia Kim Nyberg
- Department of Psychology, Center for Lifespan Changes in Brain and Cognition, University of Oslo, Oslo, Norway; Research Department, Sunnaas Rehabilitation Hospital, Nesoddtangen, Norway.
| | - Anders M Fjell
- Department of Psychology, Center for Lifespan Changes in Brain and Cognition, University of Oslo, Oslo, Norway; Department of radiology and nuclear medicine, Oslo University Hospital, Oslo, Norway
| | - Kristine B Walhovd
- Department of Psychology, Center for Lifespan Changes in Brain and Cognition, University of Oslo, Oslo, Norway; Department of radiology and nuclear medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
14
|
Nam GE, Park YG, Han K, Kim MK, Koh ES, Kim ES, Lee MK, Kim B, Hong OK, Kwon HS. BMI, Weight Change, and Dementia Risk in Patients With New-Onset Type 2 Diabetes: A Nationwide Cohort Study. Diabetes Care 2019; 42:1217-1224. [PMID: 31177182 DOI: 10.2337/dc18-1667] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 03/31/2019] [Indexed: 02/03/2023]
Abstract
OBJECTIVE This study examined the association between baseline BMI, percentage weight change, and the risk of dementia in patients newly diagnosed with type 2 diabetes. RESEARCH DESIGN AND METHODS Using the South Korean National Health Insurance Service-National Health Screening Cohort database, we identified 167,876 subjects aged ≥40 years diagnosed with new-onset type 2 diabetes between 2007 and 2012. Their weight changes were monitored for ∼2 years after diagnosis, with follow-up assessments occurring for an average of 3.5 years. The hazard ratios (HRs) and Bonferroni-adjusted 95% CIs of all-cause dementia, Alzheimer disease (AD), and vascular dementia were estimated using multivariable Cox proportional hazards regression models. RESULTS We identified 2,563 incident dementia cases during follow-up. Baseline BMI among patients with new-onset type 2 diabetes was inversely associated with the risk of all-cause dementia and AD, independent of confounding variables (P for trend <0.001). The percentage weight change during the 2 years after a diagnosis of type 2 diabetes showed significant U-shaped associations with the risk of all-cause dementia development (P < 0.001); the HRs of the disease increased significantly when weight loss or gain was >10% (1.34 [95% CI 1.11-1.63] and 1.38 [1.08-1.76], respectively). Additionally, weight loss >10% was associated with an increased risk of AD (HR 1.26 [95% CI 1.01-1.59]). CONCLUSIONS A lower baseline BMI was associated with increased risks of all-cause dementia and AD in patients with new-onset type 2 diabetes. Weight loss or weight gain after the diagnosis of diabetes was associated with an increased risk of all-cause dementia. Weight loss was associated with an increased risk of AD.
Collapse
Affiliation(s)
- Ga Eun Nam
- Department of Family Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Yong Gyu Park
- Department of Biostatistics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kyungdo Han
- Department of Biostatistics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mee Kyoung Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Sil Koh
- Division of Nephrology, Department of Internal Medicine, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Sook Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Republic of Korea
| | - Min-Kyung Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Myongji Hospital, Hanyang University Medical Center, Gyeonggi-do, Republic of Korea
| | - Bongsung Kim
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Republic of Korea
| | - Oak-Kee Hong
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyuk-Sang Kwon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
15
|
Steward T, Mestre-Bach G, Granero R, Sánchez I, Riesco N, Vintró-Alcaraz C, Sauchelli S, Jiménez-Murcia S, Agüera Z, Fernández-García JC, Garrido-Sánchez L, Tinahones FJ, Casanueva FF, Baños RM, Botella C, Crujeiras AB, Torre RDL, Fernández-Real JM, Frühbeck G, Ortega FJ, Rodríguez A, Menchón JM, Fernández-Aranda F. Reduced Plasma Orexin-A Concentrations are Associated with Cognitive Deficits in Anorexia Nervosa. Sci Rep 2019; 9:7910. [PMID: 31133733 PMCID: PMC6536521 DOI: 10.1038/s41598-019-44450-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 05/07/2019] [Indexed: 01/06/2023] Open
Abstract
Orexins/hypocretins are neuropeptides implicated in numerous processes, including food intake and cognition. The role of these peptides in the psychopathology of anorexia nervosa (AN) remains poorly understood. The aim of the current study was to evaluate the associations between plasma orexin-A (OXA) concentrations and neuropsychological functioning in adult women with AN, and a matched control group. Fasting plasma OXA concentrations were taken in 51 females with AN and in 51 matched healthy controls. Set-shifting was assessed using the Wisconsin Card Sorting Test (WCST), whereas decision making was measured using the Iowa Gambling Task (IGT). The AN group exhibited lower plasma OXA levels than the HC group. Lower mean scores were obtained on the IGT in AN patients. WCST perseverative errors were significantly higher in the AN group compared to HC. In both the AN and HC group, OXA levels were negatively correlated with WCST non-perseverative errors. Reduced plasma OXA concentrations were found to be associated with set-shifting impairments in AN. Taking into consideration the function of orexins in promoting arousal and cognitive flexibility, future studies should explore whether orexin partly underpins the cognitive impairments found in AN.
Collapse
Affiliation(s)
- Trevor Steward
- Department of Psychiatry, Bellvitge University Hospital-IDIBELL, Barcelona, Spain.,Ciber Fisiopatologia Obesidad y Nutrición, Instituto Salud Carlos III (Spain), Madrid, Spain
| | - Gemma Mestre-Bach
- Department of Psychiatry, Bellvitge University Hospital-IDIBELL, Barcelona, Spain.,Ciber Fisiopatologia Obesidad y Nutrición, Instituto Salud Carlos III (Spain), Madrid, Spain
| | - Roser Granero
- Ciber Fisiopatologia Obesidad y Nutrición, Instituto Salud Carlos III (Spain), Madrid, Spain.,Departament de Psicobiologia i Metodologia, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Isabel Sánchez
- Department of Psychiatry, Bellvitge University Hospital-IDIBELL, Barcelona, Spain.,Ciber Fisiopatologia Obesidad y Nutrición, Instituto Salud Carlos III (Spain), Madrid, Spain
| | - Nadine Riesco
- Department of Psychiatry, Bellvitge University Hospital-IDIBELL, Barcelona, Spain.,Ciber Fisiopatologia Obesidad y Nutrición, Instituto Salud Carlos III (Spain), Madrid, Spain
| | - Cristina Vintró-Alcaraz
- Department of Psychiatry, Bellvitge University Hospital-IDIBELL, Barcelona, Spain.,Ciber Fisiopatologia Obesidad y Nutrición, Instituto Salud Carlos III (Spain), Madrid, Spain
| | - Sarah Sauchelli
- Department of Psychiatry, Bellvitge University Hospital-IDIBELL, Barcelona, Spain
| | - Susana Jiménez-Murcia
- Department of Psychiatry, Bellvitge University Hospital-IDIBELL, Barcelona, Spain.,Ciber Fisiopatologia Obesidad y Nutrición, Instituto Salud Carlos III (Spain), Madrid, Spain.,Department of Clinical Sciences, School of Medicine, University of Barcelona, Barcelona, Spain
| | - Zaida Agüera
- Department of Psychiatry, Bellvitge University Hospital-IDIBELL, Barcelona, Spain.,Ciber Fisiopatologia Obesidad y Nutrición, Instituto Salud Carlos III (Spain), Madrid, Spain
| | - Jose C Fernández-García
- Ciber Fisiopatologia Obesidad y Nutrición, Instituto Salud Carlos III (Spain), Madrid, Spain.,Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Hospital Clínico Virgen de la Victoria, Málaga, Spain
| | - Lourdes Garrido-Sánchez
- Ciber Fisiopatologia Obesidad y Nutrición, Instituto Salud Carlos III (Spain), Madrid, Spain.,Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Hospital Clínico Virgen de la Victoria, Málaga, Spain
| | - Francisco J Tinahones
- Ciber Fisiopatologia Obesidad y Nutrición, Instituto Salud Carlos III (Spain), Madrid, Spain.,Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Hospital Clínico Virgen de la Victoria, Málaga, Spain
| | - Felipe F Casanueva
- Ciber Fisiopatologia Obesidad y Nutrición, Instituto Salud Carlos III (Spain), Madrid, Spain.,Molecular and Celular Endocrinology, Instituto de Investigacion Sanitaria (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS) and Santiago de Compostela University (USC), Santiago de Compostela, Spain
| | - Rosa M Baños
- Ciber Fisiopatologia Obesidad y Nutrición, Instituto Salud Carlos III (Spain), Madrid, Spain.,Department of Psychological, Personality, Evaluation and Treatment of the University of Valencia, Valencia, Spain
| | - Cristina Botella
- Ciber Fisiopatologia Obesidad y Nutrición, Instituto Salud Carlos III (Spain), Madrid, Spain
| | - Ana B Crujeiras
- Ciber Fisiopatologia Obesidad y Nutrición, Instituto Salud Carlos III (Spain), Madrid, Spain.,Molecular and Celular Endocrinology, Instituto de Investigacion Sanitaria (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS) and Santiago de Compostela University (USC), Santiago de Compostela, Spain
| | - Rafael de la Torre
- Ciber Fisiopatologia Obesidad y Nutrición, Instituto Salud Carlos III (Spain), Madrid, Spain.,Integrated Pharmacology and Systems Neurosciences Research Group, Neuroscience Research Program Organization IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Department of Health and Experimental Sciences, Universitat Pompeu Fabra Barcelona, Barcelona, Spain
| | - Jose M Fernández-Real
- Ciber Fisiopatologia Obesidad y Nutrición, Instituto Salud Carlos III (Spain), Madrid, Spain.,Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació, Biomèdica de Girona (IdIBGi), Hospital Dr Josep Trueta, Girona, Spain
| | - Gema Frühbeck
- Ciber Fisiopatologia Obesidad y Nutrición, Instituto Salud Carlos III (Spain), Madrid, Spain.,Metabolic Research Laboratory, Clínica Universidad de Navarra, University of Navarra-IdiSNA, Pamplona, Spain
| | - Francisco J Ortega
- Ciber Fisiopatologia Obesidad y Nutrición, Instituto Salud Carlos III (Spain), Madrid, Spain.,Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació, Biomèdica de Girona (IdIBGi), Hospital Dr Josep Trueta, Girona, Spain
| | - Amaia Rodríguez
- Ciber Fisiopatologia Obesidad y Nutrición, Instituto Salud Carlos III (Spain), Madrid, Spain.,Metabolic Research Laboratory, Clínica Universidad de Navarra, University of Navarra-IdiSNA, Pamplona, Spain
| | - José M Menchón
- Department of Psychiatry, Bellvitge University Hospital-IDIBELL, Barcelona, Spain.,Department of Clinical Sciences, School of Medicine, University of Barcelona, Barcelona, Spain.,CIBER Salud Mental, Instituto Salud Carlos III (Spain), Madrid, Spain
| | - Fernando Fernández-Aranda
- Department of Psychiatry, Bellvitge University Hospital-IDIBELL, Barcelona, Spain. .,Ciber Fisiopatologia Obesidad y Nutrición, Instituto Salud Carlos III (Spain), Madrid, Spain. .,Department of Clinical Sciences, School of Medicine, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
16
|
Cordner ZA, Khambadkone SG, Boersma GJ, Song L, Summers TN, Moran TH, Tamashiro KLK. Maternal high-fat diet results in cognitive impairment and hippocampal gene expression changes in rat offspring. Exp Neurol 2019; 318:92-100. [PMID: 31051155 DOI: 10.1016/j.expneurol.2019.04.018] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/13/2019] [Accepted: 04/29/2019] [Indexed: 01/01/2023]
Abstract
Consumption of a high-fat diet has long been known to increase risk for obesity, diabetes, and the metabolic syndrome. Further evidence strongly suggests that these same metabolic disorders are associated with an increased risk of cognitive impairment later in life. Now faced with an expanding global burden of obesity and increasing prevalence of dementia due to an aging population, understanding the effects of high-fat diet consumption on cognition is of increasingly critical importance. Further, the developmental origins of many adult onset neuropsychiatric disorders have become increasingly clear, indicating a need to investigate effects of various risk factors, including diet, across the lifespan. Here, we use a rat model to assess the effects of maternal diet during pregnancy and lactation on cognition and hippocampal gene expression of offspring. Behaviorally, adult male offspring of high-fat fed dams had impaired object recognition memory and impaired spatial memory compared to offspring of chow-fed dams. In hippocampus, we found decreased expression of Insr, Lepr, and Slc2a1 (GLUT1) among offspring of high-fat fed dams at postnatal day 21. The decreased expression of Insr and Lepr persisted at postnatal day 150. Together, these data provide additional evidence to suggest that maternal exposure to high-fat diet during pregnancy and lactation can have lasting effects on the brain, behavior, and cognition on adult offspring.
Collapse
Affiliation(s)
- Zachary A Cordner
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA
| | - Seva G Khambadkone
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA; Cellular & Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA
| | - Gretha J Boersma
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA
| | - Lin Song
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA
| | - Tyler N Summers
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA
| | - Timothy H Moran
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA; Cellular & Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA
| | - Kellie L K Tamashiro
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA; Cellular & Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA.
| |
Collapse
|
17
|
Suarez AN, Noble EE, Kanoski SE. Regulation of Memory Function by Feeding-Relevant Biological Systems: Following the Breadcrumbs to the Hippocampus. Front Mol Neurosci 2019; 12:101. [PMID: 31057368 PMCID: PMC6482164 DOI: 10.3389/fnmol.2019.00101] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 04/03/2019] [Indexed: 12/15/2022] Open
Abstract
The hippocampus (HPC) controls fundamental learning and memory processes, including memory for visuospatial navigation (spatial memory) and flexible memory for facts and autobiographical events (declarative memory). Emerging evidence reveals that hippocampal-dependent memory function is regulated by various peripheral biological systems that are traditionally known for their roles in appetite and body weight regulation. Here, we argue that these effects are consistent with a framework that it is evolutionarily advantageous to encode and recall critical features surrounding feeding behavior, including the spatial location of a food source, social factors, post-absorptive processing, and other episodic elements of a meal. We review evidence that gut-to-brain communication from the vagus nerve and from feeding-relevant endocrine systems, including ghrelin, insulin, leptin, and glucagon-like peptide-1 (GLP-1), promote hippocampal-dependent spatial and declarative memory via neurotrophic and neurogenic mechanisms. The collective literature reviewed herein supports a model in which various stages of feeding behavior and hippocampal-dependent memory function are closely linked.
Collapse
Affiliation(s)
| | | | - Scott E. Kanoski
- Human and Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
18
|
Maletínská L, Popelová A, Železná B, Bencze M, Kuneš J. The impact of anorexigenic peptides in experimental models of Alzheimer's disease pathology. J Endocrinol 2019; 240:R47-R72. [PMID: 30475219 DOI: 10.1530/joe-18-0532] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 11/20/2018] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder in the elderly population. Numerous epidemiological and experimental studies have demonstrated that patients who suffer from obesity or type 2 diabetes mellitus have a higher risk of cognitive dysfunction and AD. Several recent studies demonstrated that food intake-lowering (anorexigenic) peptides have the potential to improve metabolic disorders and that they may also potentially be useful in the treatment of neurodegenerative diseases. In this review, the neuroprotective effects of anorexigenic peptides of both peripheral and central origins are discussed. Moreover, the role of leptin as a key modulator of energy homeostasis is discussed in relation to its interaction with anorexigenic peptides and their analogs in AD-like pathology. Although there is no perfect experimental model of human AD pathology, animal studies have already proven that anorexigenic peptides exhibit neuroprotective properties. This phenomenon is extremely important for the potential development of new drugs in view of the aging of the human population and of the significantly increasing incidence of AD.
Collapse
Affiliation(s)
- Lenka Maletínská
- Institute of Organic Chemistry and Biochemistry AS CR, Prague, Czech Republic
| | - Andrea Popelová
- Institute of Organic Chemistry and Biochemistry AS CR, Prague, Czech Republic
| | - Blanka Železná
- Institute of Organic Chemistry and Biochemistry AS CR, Prague, Czech Republic
| | - Michal Bencze
- Institute of Organic Chemistry and Biochemistry AS CR, Prague, Czech Republic
- Institute of Physiology AS CR, Prague, Czech Republic
| | - Jaroslav Kuneš
- Institute of Organic Chemistry and Biochemistry AS CR, Prague, Czech Republic
- Institute of Physiology AS CR, Prague, Czech Republic
| |
Collapse
|
19
|
Baykal S, Albayrak Y, Durankuş F, Güzel S, Abbak Ö, Potas N, Beyazyüz M, Karabekiroğlu K, Donma MM. Decreased serum orexin A levels in drug-naive children with attention deficit and hyperactivity disorder. Neurol Sci 2019; 40:593-602. [PMID: 30617449 DOI: 10.1007/s10072-018-3692-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 12/19/2018] [Indexed: 11/25/2022]
Affiliation(s)
- Saliha Baykal
- Faculty of Medicine, Department of Child and Adolescent Psychiatry, Tekirdağ Namık Kemal University, Tekirdag, Turkey.
| | - Yakup Albayrak
- Faculty of Medicine, Department of Psychiatry, Tekirdağ Namık Kemal University, Tekirdag, Turkey
| | - Ferit Durankuş
- Faculty of Medicine, Department of Pediatrics, Okan University, Istanbul, Turkey
| | - Savaş Güzel
- Faculty of Medicine, Department of Biochemistry, Tekirdağ Namık Kemal University, Tekirdag, Turkey
| | - Özlem Abbak
- Faculty of Medicine, Department of Psychiatry, Tekirdağ Namık Kemal University, Tekirdag, Turkey
| | - Nihan Potas
- Faculty of Economics and Administrative Science, Department of Healthcare Management, Gazi University, Ankara, Turkey
| | - Murat Beyazyüz
- Faculty of Medicine, Department of Psychiatry, Tekirdağ Namık Kemal University, Tekirdag, Turkey
| | - Koray Karabekiroğlu
- Faculty of Medicine, Department of Child and Adolescent Psychiatry, Ondokuz Mayıs University, Samsun, Turkey
| | - Mustafa Metin Donma
- Faculty of Medicine, Department of Pediatrics, Tekirdağ Namık Kemal University, Tekirdag, Turkey
| |
Collapse
|
20
|
Smith PJ, Mabe S, Sherwood A, Babyak MA, Murali Doraiswamy P, Welsh-Bohmer KA, Kraus W, Burke J, Hinderliter A, Blumenthal JA. Association Between Insulin Resistance, Plasma Leptin, and Neurocognition in Vascular Cognitive Impairment. J Alzheimers Dis 2019; 71:921-929. [PMID: 31476159 PMCID: PMC10840083 DOI: 10.3233/jad-190569] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Greater body weight has been associated impairments in neurocognition and greater dementia risk, although the mechanisms linking weight and neurocognition have yet to be adequately delineated. OBJECTIVE To examine metabolic mechanisms underlying the association between obesity and neurocognition. METHODS We conducted a secondary analysis of weight, neurocognition, and the potentially mediating role of metabolic and inflammatory biomarkers among 160 participants from the ENLIGHTEN trial of vascular cognitive impairment, no dementia (CIND). Neurocognition was assessed using a 45-minute assessment battery assessing Executive Function, Verbal and Visual Memory. We considered three metabolic biomarkers: insulin resistance (homeostatic model assessment [HOMA-IR]), plasma leptin, and insulin-like growth factor (IGF-1). Inflammation was assessed using C-reactive protein. Multiple regression analyses were used. RESULTS Participants included 160 sedentary older adults with CIND. Participants tended to be overweight or obese (mean BMI = 32.5 [SD = 4.8]). Women exhibited higher BMI (p = 0.043), CRP (p < 0.001), and leptin (p < 0.001) compared with men. Higher BMI levels were associated with worse performance on measures of Executive Function (β= -0.16, p = 0.024) and Verbal Memory (β= -0.16, p = 0.030), but not Visual Memory (β= 0.05, p = 0.500). Worse metabolic biomarker profiles also were associated with lower Executive Function (β= -0.12, p = 0.050). Mediation analyses suggested leptin was a plausible candidate as a mediator between BMI and Executive Function. CONCLUSIONS In overweight and obese adults with vascular CIND, the association between greater weight and poorer executive function may be mediated by higher leptin resistance.
Collapse
Affiliation(s)
- Patrick J. Smith
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Stephanie Mabe
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Andrew Sherwood
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Michael A. Babyak
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - P. Murali Doraiswamy
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Kathleen A. Welsh-Bohmer
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - William Kraus
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - James Burke
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Alan Hinderliter
- Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - James A. Blumenthal
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
21
|
Li N, Arbuckle TE, Muckle G, Lanphear BP, Boivin M, Chen A, Dodds L, Fraser WD, Ouellet E, Séguin JR, Velez MP, Yolton K, Braun JM. Associations of cord blood leptin and adiponectin with children's cognitive abilities. Psychoneuroendocrinology 2019; 99:257-264. [PMID: 30390444 PMCID: PMC6239208 DOI: 10.1016/j.psyneuen.2018.10.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/22/2018] [Accepted: 10/23/2018] [Indexed: 12/11/2022]
Abstract
Background Adipocytokines may play a role in fetal programming of neurodevelopment. We aimed to investigate the associations between cord blood adipocytokine concentrations and children's intelligence test scores. Methods We used data from two ongoing pregnancy cohorts in North America: the Maternal-Infant Research on Environmental Chemicals (MIREC, n = 429) and Health Outcomes and Measures of the Environment (HOME, n = 183) Studies. Umbilical cord blood adipocytokine concentrations were measured using enzyme-linked immunosorbent assays. We assessed children's Intelligence Quotient (IQ) and its components using the Wechsler Preschool and Primary Scales of Intelligence-III or Wechsler Intelligence Scale for Children-IV. We used linear regression and linear mixed models to estimate associations between log2-transformed adipocytokine concentrations and children's IQ after adjusting for sociodemographic, perinatal, and child factors. Results After adjusting for covariates, cord blood adiponectin was positively associated with children's full-scale IQ scores at age 3 years in the MIREC Study (β = 1.4, 95% confidence interval [CI]: 0.2, 2.5) and at ages 5 and 8 years in the HOME Study (β = 1.7, CI: -0.1, 3.5). Adiponectin was positively associated with performance IQ in both studies (MIREC: β = 2.0, CI: 0.7, 3.3; HOME: β = 2.2, CI: 0.5, 3.9). Adiponectin was positively associated with working memory composite scores at age 8 in the HOME Study (β = 3.1, CI: 1.0, 5.2). Leptin was not associated with children's IQ in either study. Conclusions Cord blood adiponectin was associated with higher full-scale and performance IQ and working memory composite scores in children. Future studies are needed to explore the mechanisms underlying these associations.
Collapse
Affiliation(s)
- Nan Li
- Department of Epidemiology, Brown University, Providence, RI, United States.
| | - Tye E Arbuckle
- Population Studies Division, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, Ontario, Canada
| | - Gina Muckle
- School of Psychology, Laval University, Ville de Québec, Québec, Canada
| | - Bruce P Lanphear
- Faculty of Health Sciences, Simon Fraser University, British Columbia, Canada; Child and Family Research Institute, BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Michel Boivin
- School of Psychology, Laval University, Ville de Québec, Québec, Canada
| | - Aimin Chen
- Division of Epidemiology, Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Linda Dodds
- Perinatal Epidemiology Research Unit, IWK Health Center, Halifax, Canada
| | - William D Fraser
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Mother and Child University Hospital Center, Montreal, Québec, Canada; Centre de recherche du CHUS (CHU de Sherbrooke), University of Sherbrooke, Sherbrooke, Québec, Canada
| | - Emmanuel Ouellet
- CHU de Québec-Université Laval Research Center, Ville de Québec, Québec, Canada
| | - Jean R Séguin
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Mother and Child University Hospital Center, Montreal, Québec, Canada; Department of Psychiatry, University of Montréal, Montréal, Québec, Canada
| | - Maria P Velez
- Department of Obstetrics and Gynecology, Queen's University, Kingston, Ontario, Canada
| | - Kimberly Yolton
- Department of Pediatrics, Division of General and Community Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Joseph M Braun
- Department of Epidemiology, Brown University, Providence, RI, United States
| |
Collapse
|
22
|
Leptin in depression: a potential therapeutic target. Cell Death Dis 2018; 9:1096. [PMID: 30367065 PMCID: PMC6203758 DOI: 10.1038/s41419-018-1129-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 06/02/2018] [Accepted: 07/04/2018] [Indexed: 12/15/2022]
Abstract
Leptin, produced and secreted by white adipose tissue, plays a critical role in regulating body weight, food intake, and energy metabolism. Recently, several studies have identified an underlying role for leptin in regulation of mood and cognition via regulation of synaptic changes in the brain that have been associated with antidepressant-like actions. Brain neural plasticity occurs in response to a range of intrinsic and extrinsic stimuli, including those that may mediate the effects of antidepressants. Neural plasticity theories of depression are thought to explain multiple aspects of depression and the effects of antidepressants. It is also well documented that leptin has effects on neural plasticity. This review summarizes the recent literature on the role of leptin in neural plasticity in order to elaborate the possible mechanism of leptin’s antidepressant-like effects. Recent findings provide new insights into the underlying mechanisms of neural plasticity in depression. Leptin may influence these mechanisms and consequently constitute a possible target for novel therapeutic approaches to the treatment of depression.
Collapse
|
23
|
A cafeteria diet alters the decision making strategy and metabolic markers in Sprague-Dawley male rats. Appl Anim Behav Sci 2018. [DOI: 10.1016/j.applanim.2017.10.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
24
|
Hsu TM, Noble EE, Reiner DJ, Liu CM, Suarez AN, Konanur VR, Hayes MR, Kanoski SE. Hippocampus ghrelin receptor signaling promotes socially-mediated learned food preference. Neuropharmacology 2017; 131:487-496. [PMID: 29191751 DOI: 10.1016/j.neuropharm.2017.11.039] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/12/2017] [Accepted: 11/23/2017] [Indexed: 12/24/2022]
Abstract
Social cues are potent regulators of feeding behavior, yet the neurobiological mechanisms through which social cues influence food intake are poorly understood. Here we investigate the hypothesis that the appetite-promoting gut-derived hormone, ghrelin, signals in the hippocampus to promote learned social aspects of feeding behavior. We utilized a procedure known as 'social transmission of food preference' (STFP) in which rats ('Observers') experience a social interaction with another rat ('Demonstrators') that recently consumed flavored/scented chow. STFP learning in Observer rats is indicated by a significant preference for the Demonstrator paired flavor of chow vs. a novel unpaired flavor of chow in a subsequent consumption choice test. Our results show that relative to vehicle treatment, ghrelin targeted to the ventral CA1 subregion of the hippocampus (vHP) enhanced STFP learning in rats. Additionally, STFP was impaired following peripheral injections of l-cysteine that reduce circulating ghrelin levels, suggesting that vHP ghrelin-mediated effects on STFP require peripheral ghrelin release. Finally, the endogenous relevance of vHP ghrelin receptor (GHSR-1A) signaling in STFP is supported by our data showing that STFP learning was eliminated following targeted viral vector RNA interference-mediated knockdown of vHP GHSR-1A mRNA. Control experiments indicate that vHP ghrelin-mediated STFP effects are not secondary to altered social exploration and food intake, nor to altered food preference learning based on nonsocial olfactory cues. Overall these data reveal a novel neurobiological system that promotes conditioned, social aspects of feeding behavior.
Collapse
Affiliation(s)
- Ted M Hsu
- Human and Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA; Neuroscience Program, University of Southern California, Los Angeles, CA, USA
| | - Emily E Noble
- Human and Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - David J Reiner
- Translational Neuroscience Program, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Clarissa M Liu
- Human and Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA; Neuroscience Program, University of Southern California, Los Angeles, CA, USA
| | - Andrea N Suarez
- Human and Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Vaibhav R Konanur
- Graduate Program in Neuroscience, University of Illinois at Chicago, Chicago, IL, USA
| | - Matthew R Hayes
- Translational Neuroscience Program, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Scott E Kanoski
- Human and Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA; Neuroscience Program, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
25
|
McGregor G, Harvey J. Food for thought: Leptin regulation of hippocampal function and its role in Alzheimer's disease. Neuropharmacology 2017; 136:298-306. [PMID: 28987937 DOI: 10.1016/j.neuropharm.2017.09.038] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 09/27/2017] [Accepted: 09/30/2017] [Indexed: 01/08/2023]
Abstract
Accumulating evidence indicates that diet and body weight are important factors associated with Alzheimer's disease (AD), with a significant increase in AD risk linked to mid-life obesity, and weight loss frequently occurring in the early stages of AD. This has fuelled interest in the hormone leptin, as it is an important hypothalamic regulator of food intake and body weight, but leptin also markedly influences the functioning of the hippocampus; a key brain region that degenerates in AD. Increasing evidence indicates that leptin has cognitive enhancing properties as it facilitates the cellular events that underlie hippocampal-dependent learning and memory. However, significant reductions in leptin's capacity to regulate hippocampal synaptic function occurs with age and dysfunctions in the leptin system are associated with an increased risk of AD. Moreover, leptin is a potential novel target in AD as leptin treatment has beneficial effects in various models of AD. Here we summarise recent advances in leptin neurobiology with particular focus on regulation of hippocampal synaptic function by leptin and the implications of this for neurodegenerative disorders like AD. This article is part of the Special Issue entitled 'Metabolic Impairment as Risk Factors for Neurodegenerative Disorders.'
Collapse
Affiliation(s)
- Gemma McGregor
- Division of Neuroscience, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, United Kingdom
| | - Jenni Harvey
- Division of Neuroscience, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, United Kingdom.
| |
Collapse
|
26
|
Zanini P, Arbo BD, Niches G, Czarnabay D, Benetti F, Ribeiro MF, Cecconello AL. Diet-induced obesity alters memory consolidation in female rats. Physiol Behav 2017; 180:91-97. [PMID: 28821446 DOI: 10.1016/j.physbeh.2017.08.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 08/14/2017] [Accepted: 08/14/2017] [Indexed: 11/29/2022]
Abstract
Obesity is a multifactorial disease characterized by the abnormal or excessive fat accumulation, which is caused by an energy imbalance between consumed and expended calories. Obesity leads to an inflammatory response that may result in peripheral and central metabolic changes, including insulin and leptin resistance. Insulin and leptin resistance have been associated with metabolic and cognitive dysfunctions. Obesity and some neurodegenerative diseases that lead to dementia affect mainly women. However, the effects of diet-induced obesity on memory consolidation in female rats are poorly understood. Therefore, the aim of this study was to evaluate the effect of a hypercaloric diet on the object recognition memory of female rats and on possible related metabolic changes. The animals submitted to the hypercaloric diet presented a higher food intake in grams and in calories, resulting in increased weight gain and liposomatic index in comparison with the animals exposed to the control diet. These animals presented a memory deficit in the object recognition test and increased serum levels of glucose and leptin. However, no significant differences were found in the serum levels of insulin, TNF-α and IL-1β, in the index of insulin resistance (HOMA), in the hippocampal levels of insulin, TNF-α and IL-1β, as well as on Akt expression or activation in the hippocampus. Our findings indicate that adult female rats submitted to a hypercaloric diet present memory consolidation impairment, which could be associated with diet-induced weight gain and leptin resistance, even without the development of insulin resistance.
Collapse
Affiliation(s)
- P Zanini
- Laboratório de Interação Neuro-Humoral, Department of Physiology, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Sarmento Leite, 500, 90050-170 Porto Alegre, RS, Brazil
| | - B D Arbo
- Laboratório de Interação Neuro-Humoral, Department of Physiology, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Sarmento Leite, 500, 90050-170 Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Farmacologia e Terapêutica, Department of Pharmacology, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Sarmento Leite, 500, 90050-170 Porto Alegre, RS, Brazil
| | - G Niches
- Laboratório de Interação Neuro-Humoral, Department of Physiology, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Sarmento Leite, 500, 90050-170 Porto Alegre, RS, Brazil
| | - D Czarnabay
- Laboratório de Neurofisiologia Cognitiva e do Desenvolvimento, Department of Physiology, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Sarmento Leite, 500, 90050-170 Porto Alegre, RS, Brazil
| | - F Benetti
- Laboratório de Neurofisiologia Cognitiva e do Desenvolvimento, Department of Physiology, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Sarmento Leite, 500, 90050-170 Porto Alegre, RS, Brazil
| | - M F Ribeiro
- Laboratório de Interação Neuro-Humoral, Department of Physiology, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Sarmento Leite, 500, 90050-170 Porto Alegre, RS, Brazil
| | - A L Cecconello
- Laboratório de Interação Neuro-Humoral, Department of Physiology, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Sarmento Leite, 500, 90050-170 Porto Alegre, RS, Brazil.
| |
Collapse
|
27
|
Van Doorn C, Macht VA, Grillo CA, Reagan LP. Leptin resistance and hippocampal behavioral deficits. Physiol Behav 2017; 176:207-213. [PMID: 28267584 PMCID: PMC10538552 DOI: 10.1016/j.physbeh.2017.03.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 03/02/2017] [Accepted: 03/02/2017] [Indexed: 01/05/2023]
Abstract
The adipocyte-derived hormone leptin is an important regulator of body weight and metabolism through activation of brain leptin receptors expressed in regions such as the hypothalamus. Beyond these well described and characterized activities of leptin in the hypothalamus, it is becoming increasingly clear that the central activities of leptin extend to the hippocampus. Indeed, leptin receptors are expressed in the hippocampus where these receptors are proposed to mediate various aspects of hippocampal synaptic plasticity that ultimately impact cognitive function. This concept is supported by studies demonstrating that leptin promotes hippocampal-dependent learning and memory, as well as studies indicating that leptin resistance is associated with deficits in hippocampal-dependent behaviors and in the induction of depressive-like behaviors. The effects of leptin on cognitive/behavioral plasticity in the hippocampus may be regulated by direct activation of leptin receptors expressed in the hippocampus; additionally, leptin-mediated activation of synaptic networks that project to the hippocampus may also impact hippocampal-mediated behaviors. In view of these previous observations, the goal of this review will be to discuss the mechanisms through which leptin facilitates cognition and behavior, as well as to dissect the loci at which leptin resistance leads to impairments in hippocampal synaptic plasticity, including the development of cognitive deficits and increased risk of depressive illness in metabolic disorders such as obesity and type 2 diabetes mellitus (T2DM).
Collapse
Affiliation(s)
- Catherine Van Doorn
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29208, United States
| | - Victoria A Macht
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29208, United States
| | - Claudia A Grillo
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29208, United States
| | - Lawrence P Reagan
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29208, United States; W.J.B. Dorn VA Medical Center, Columbia, SC 29208, United States.
| |
Collapse
|
28
|
Hagar JM, Macht VA, Wilson SP, Fadel JR. Upregulation of orexin/hypocretin expression in aged rats: Effects on feeding latency and neurotransmission in the insular cortex. Neuroscience 2017; 350:124-132. [PMID: 28344067 DOI: 10.1016/j.neuroscience.2017.03.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 03/14/2017] [Accepted: 03/15/2017] [Indexed: 10/19/2022]
Abstract
Aging is associated with changes in numerous homeostatic functions, such as food intake, that are thought to be mediated by the hypothalamus. Orexin/hypocretin neurons of the hypothalamus regulate several physiological functions, including feeding, sleep and wakefulness. Evidence from both clinical and animal studies supports the notion that aging is associated with loss or dysregulation of the orexin system. Here, we used virus-mediated gene transfer to manipulate expression of orexin peptides in young and aged rats and examined behavioral and neurochemical correlates of food intake in these animals. Aged rats showed slower feeding latencies when presented with palatable food compared to young control rats, and these deficits were ameliorated by upregulation of orexin expression. Similarly, young animals treated with a virus designed to decrease preproorexin expression showed longer feeding latencies reminiscent of aged control rats. Feeding was also associated with increased acetylcholine, glutamate and GABA efflux in insular cortex of young control animals. Orexin upregulation did not restore deficits in feeding-elicited release of these neurotransmitters in aged rats, but did enhance basal neurotransmitter levels which may have contributed to the behavioral correlates of these genetic manipulations. These studies demonstrate that age-related deficits in behavioral and neurochemical measures of feeding are likely to be mediated, in part, by the orexin system. Because these same neurotransmitter systems have been shown to underlie orexin effects on cognition, treatments which increase orexin function may have potential for improving both physiological and cognitive manifestations of certain age-related disorders.
Collapse
Affiliation(s)
- Janel M Hagar
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - Victoria A Macht
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29208, USA; Department of Psychology, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - Steven P Wilson
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - James R Fadel
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29208, USA.
| |
Collapse
|
29
|
Macht VA, Vazquez M, Petyak CE, Grillo CA, Kaigler K, Enos RT, McClellan JL, Cranford TL, Murphy EA, Nyland JF, Solomon G, Gertler A, Wilson MA, Reagan LP. Leptin resistance elicits depressive-like behaviors in rats. Brain Behav Immun 2017; 60:151-160. [PMID: 27743935 DOI: 10.1016/j.bbi.2016.10.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 09/22/2016] [Accepted: 10/10/2016] [Indexed: 12/13/2022] Open
Abstract
There is a growing appreciation that the complications of obesity extend to the central nervous system (CNS) and include increased risk for development of neuropsychiatric co-morbidities such as depressive illness. The neurological consequences of obesity may develop as a continuum and involve a progression of pathological features which is initiated by leptin resistance. Leptin resistance is a hallmark feature of obesity, but it is unknown whether leptin resistance or blockage of leptin action is casually linked to the neurological changes which underlie depressive-like phenotypes. Accordingly, the aim of the current study was to examine whether chronic administration of a pegylated leptin receptor antagonist (Peg-LRA) elicits depressive-like behaviors in adult male rats. Peg-LRA administration resulted in endocrine and metabolic features that are characteristic of an obesity phenotype. Peg-LRA rats also exhibited increased immobility in the forced swim test, depressive-like behaviors that were accompanied by indices of peripheral inflammation. These results demonstrate that leptin resistance elicits an obesity phenotype that is characterized by peripheral immune changes and depressive-like behaviors in rats, supporting the concept that co-morbid obesity and depressive illness develop as a continuum resulting from changes in the peripheral endocrine and metabolic milieu.
Collapse
Affiliation(s)
- V A Macht
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29208, USA; Department of Psychology, University of South Carolina, Columbia, SC 29208, USA
| | - M Vazquez
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - C E Petyak
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - C A Grillo
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - K Kaigler
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - R T Enos
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - J L McClellan
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - T L Cranford
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - E A Murphy
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - J F Nyland
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - G Solomon
- Institute of Biochemistry, Food Science and Nutrition, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - A Gertler
- Institute of Biochemistry, Food Science and Nutrition, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - M A Wilson
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29208, USA; W.J.B. Dorn VA Medical Center, Columbia, SC 29208, USA
| | - L P Reagan
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29208, USA; W.J.B. Dorn VA Medical Center, Columbia, SC 29208, USA.
| |
Collapse
|
30
|
Grote CW, Wright DE. A Role for Insulin in Diabetic Neuropathy. Front Neurosci 2016; 10:581. [PMID: 28066166 PMCID: PMC5179551 DOI: 10.3389/fnins.2016.00581] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 12/06/2016] [Indexed: 12/13/2022] Open
Abstract
The peripheral nervous system is one of several organ systems that are profoundly affected in diabetes. The longstanding view is that insulin does not have a major role in modulating neuronal function in both central and peripheral nervous systems is now being challenged. In the setting of insulin deficiency or excess insulin, it is logical to propose that insulin dysregulation can contribute to neuropathic changes in sensory neurons. This is particularly important as sensory nerve damage associated with prediabetes, type 1 and type 2 diabetes is so prevalent. Here, we discuss the current experimental literature related to insulin's role as a potential neurotrophic factor in peripheral nerve function, as well as the possibility that insulin deficiency plays a role in diabetic neuropathy. In addition, we discuss how sensory neurons in the peripheral nervous system respond to insulin similar to other insulin-sensitive tissues. Moreover, studies now suggest that sensory neurons can also become insulin resistant like other tissues. Collectively, emerging studies are revealing that insulin signaling pathways are active contributors to sensory nerve modulation, and this review highlights this novel activity and should provide new insight into insulin's role in both peripheral and central nervous system diseases.
Collapse
Affiliation(s)
- Caleb W Grote
- Department of Anatomy and Cell Biology, University of Kansas Medical Center Kansas City, KS, USA
| | - Douglas E Wright
- Department of Anatomy and Cell Biology, University of Kansas Medical Center Kansas City, KS, USA
| |
Collapse
|
31
|
Ferrario CR, Labouèbe G, Liu S, Nieh EH, Routh VH, Xu S, O'Connor EC. Homeostasis Meets Motivation in the Battle to Control Food Intake. J Neurosci 2016; 36:11469-11481. [PMID: 27911750 PMCID: PMC5125214 DOI: 10.1523/jneurosci.2338-16.2016] [Citation(s) in RCA: 163] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 08/30/2016] [Accepted: 09/05/2016] [Indexed: 01/09/2023] Open
Abstract
Signals of energy homeostasis interact closely with neural circuits of motivation to control food intake. An emerging hypothesis is that the transition to maladaptive feeding behavior seen in eating disorders or obesity may arise from dysregulation of these interactions. Focusing on key brain regions involved in the control of food intake (ventral tegmental area, striatum, hypothalamus, and thalamus), we describe how activity of specific cell types embedded within these regions can influence distinct components of motivated feeding behavior. We review how signals of energy homeostasis interact with these regions to influence motivated behavioral output and present evidence that experience-dependent neural adaptations in key feeding circuits may represent cellular correlates of impaired food intake control. Future research into mechanisms that restore the balance of control between signals of homeostasis and motivated feeding behavior may inspire new treatment options for eating disorders and obesity.
Collapse
Affiliation(s)
- Carrie R Ferrario
- University of Michigan Medical School, Department of Pharmacology, Ann Arbor, Michigan 48109-5632
| | - Gwenaël Labouèbe
- University of Lausanne, Center for Integrative Genomics, Lausanne, CH1015, Switzerland
| | - Shuai Liu
- University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Edward H Nieh
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | | | - Shengjin Xu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia 20147, and
| | - Eoin C O'Connor
- University of Geneva, Department of Basic Neuroscience, Geneva, CH1211, Switzerland
| |
Collapse
|
32
|
Witte AV, Köbe T, Graunke A, Schuchardt JP, Hahn A, Tesky VA, Pantel J, Flöel A. Impact of leptin on memory function and hippocampal structure in mild cognitive impairment. Hum Brain Mapp 2016; 37:4539-4549. [PMID: 27511061 DOI: 10.1002/hbm.23327] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 06/08/2016] [Accepted: 07/18/2016] [Indexed: 12/12/2022] Open
Abstract
Metabolic changes have been suggested to contribute to dementia and its precursor mild cognitive impairment (MCI), yet previous results particularly for the "satiety hormone" leptin are mixed. Therefore, we aimed to determine if MCI patients show systematic differences in leptin, independent of sex, adipose mass, age, and glucose and lipid metabolism, and whether leptin levels correlated with memory performance and hippocampal integrity. Forty MCI patients (20 females, aged 67 years ± 7 SD) were compared to 40 healthy controls (HC) that were pair-wise matched for sex, age, and body fat. Memory performance was assessed using the auditory verbal learning test. Volume and microstructure of the hippocampus were determined using 3T-neuroimaging. Fasting serum markers of leptin, glucose and lipid metabolism, and other confounding factors were assayed. MCI patients, compared with HC, showed lower serum leptin, independent of sex, age, and body fat (P < 0.001). Glucose and lipid markers did not attenuate these results. Moreover, MCI patients exhibited poorer memory and lower volume and microstructural integrity within hippocampal subfields. While leptin and memory were not significantly correlated, mediation analyses indicated that lower leptin contributed to poorer memory through its negative effect on right hippocampus volume and left hippocampus microstructure. We demonstrated that MCI is associated with lower serum leptin independent of sex, age, body fat, glucose, and lipid metabolism. Our data further suggest that inefficient leptin signaling could partly contribute to decreases in memory performance through changes in hippocampus structure, a hypothesis that should now be verified in longitudinal studies. Hum Brain Mapp 37:4539-4549, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- A Veronica Witte
- Department of Neurology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.,Max-Planck-Institute of Human Cognitive and Brain Sciences, Stephanstraße 1A, 04103, Leipzig, Germany
| | - Theresa Köbe
- Department of Neurology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Anders Graunke
- Department of Neurology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Jan Philipp Schuchardt
- Department of Nutrition Physiology and Human Nutrition, Gottfried Wilhelm Leibniz University, Am Kleinen Felde 30, 30167, Hannover, Germany
| | - Andreas Hahn
- Department of Nutrition Physiology and Human Nutrition, Gottfried Wilhelm Leibniz University, Am Kleinen Felde 30, 30167, Hannover, Germany
| | - Valentina A Tesky
- Institute of General Practice, Goethe-University, Frankfurt Am Main, 60590, Germany
| | - Johannes Pantel
- Institute of General Practice, Goethe-University, Frankfurt Am Main, 60590, Germany
| | - Agnes Flöel
- Department of Neurology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.,Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| |
Collapse
|
33
|
Abstract
Accounts of altered eating behavior in semantic dementia generally emphasize gluttony and abnormal food preferences. Here we describe two female patients with no past history of eating disorders who developed early prominent aversion to food in the context of an otherwise typical semantic dementia syndrome. One patient (aged 57) presented features in line with anorexia nervosa while the second patient (aged 58) presented with a syndrome more suggestive of bulimia nervosa. These cases add to the growing spectrum of apparently dichotomous behavior patterns in the frontotemporal dementias and illustrate a potentially under-recognized cause of eating disorders presenting in later life.
Collapse
Affiliation(s)
- Alexandra E Thompson
- a Dementia Research Centre, UCL Institute of Neurology , University College London , London , UK.,b Medical School , University of Adelaide , Adelaide , Australia
| | - Camilla N Clark
- a Dementia Research Centre, UCL Institute of Neurology , University College London , London , UK
| | - Christopher J Hardy
- a Dementia Research Centre, UCL Institute of Neurology , University College London , London , UK
| | - Phillip D Fletcher
- a Dementia Research Centre, UCL Institute of Neurology , University College London , London , UK
| | | | - Jonathan D Rohrer
- a Dementia Research Centre, UCL Institute of Neurology , University College London , London , UK
| | - Jason D Warren
- a Dementia Research Centre, UCL Institute of Neurology , University College London , London , UK
| |
Collapse
|
34
|
Role of Leptin and Orexin-A Within the Suprachiasmatic Nucleus on Anxiety-Like Behaviors in Hamsters. Mol Neurobiol 2016; 54:2674-2684. [DOI: 10.1007/s12035-016-9847-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 03/09/2016] [Indexed: 01/09/2023]
|
35
|
Zajo KN, Fadel JR, Burk JA. Orexin A-induced enhancement of attentional processing in rats: role of basal forebrain neurons. Psychopharmacology (Berl) 2016; 233:639-47. [PMID: 26534765 PMCID: PMC4729649 DOI: 10.1007/s00213-015-4139-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 10/26/2015] [Indexed: 12/28/2022]
Abstract
RATIONALE Orexins are neuropeptides released in multiple brain regions from neurons that originate within the lateral hypothalamus and contiguous perfornical area. The basal forebrain, a structure implicated in attentional processing, receives orexinergic inputs. Our previous work demonstrated that administration of an orexin-1 receptor antagonist, SB-334867, systemically or via infusion directly into the basal forebrain, can disrupt performance in a task that places explicit demands on attentional processing. OBJECTIVES Given that the orexin-1 receptor binds orexin A with high affinity, we tested whether orexin A could enhance attention in rats. METHODS Attentional performance was assessed using a task that required discrimination of variable duration visual signals from trials when no signal was presented. We also tested whether infusions of orexin A into the lateral ventricle could attenuate deficits following lesions of medial prefrontal cortical cholinergic projections that arise from the basal forebrain. RESULTS Infusions of orexin A into the basal forebrain attenuated distracter-induced decreases in attentional performance. Orexin A attenuated deficits in lesioned animals when a visual distracter was presented. CONCLUSION The present results support the view that orexin A can enhance attentional performance via actions in the basal forebrain and may be beneficial for some conditions characterized by attentional dysfunction due to disruption of cortical cholinergic inputs.
Collapse
Affiliation(s)
- Kristin N. Zajo
- Department of Psychology, College of William & Mary, Williamsburg, Virginia 23187, USA
| | - Jim R. Fadel
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina 29208, USA
| | - Joshua A. Burk
- Department of Psychology, College of William & Mary, Williamsburg, Virginia 23187, USA
| |
Collapse
|
36
|
Abstract
Sleep and energy balance are essential for health. The two processes act in concert to regulate central and peripheral homeostasis. During sleep, energy is conserved due to suspended activity, movement, and sensory responses, and is redirected to restore and replenish proteins and their assemblies into cellular structures. During wakefulness, various energy-demanding activities lead to hunger. Thus, hunger promotes arousal, and subsequent feeding, followed by satiety that promotes sleep via changes in neuroendocrine or neuropeptide signals. These signals overlap with circuits of sleep-wakefulness, feeding, and energy expenditure. Here, we will briefly review the literature that describes the interplay between the circadian system, sleep-wake, and feeding-fasting cycles that are needed to maintain energy balance and a healthy metabolic profile. In doing so, we describe the neuroendocrine, hormonal/peptide signals that integrate sleep and feeding behavior with energy metabolism.
Collapse
Affiliation(s)
- Charu Shukla
- Department of Psychiatry, VA Boston Healthcare System, Harvard Medical School, West Roxbury, MA, USA
| | - Radhika Basheer
- Department of Psychiatry, VA Boston Healthcare System, Harvard Medical School, West Roxbury, MA, USA
| |
Collapse
|
37
|
Cordner ZA, Tamashiro KLK. Effects of high-fat diet exposure on learning & memory. Physiol Behav 2015; 152:363-71. [PMID: 26066731 PMCID: PMC5729745 DOI: 10.1016/j.physbeh.2015.06.008] [Citation(s) in RCA: 201] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 05/14/2015] [Accepted: 06/05/2015] [Indexed: 01/13/2023]
Abstract
The associations between consumption of a high-fat or 'Western' diet and metabolic disorders such as obesity, diabetes, and cardiovascular disease have long been recognized and a great deal of evidence now suggests that diets high in fat can also have a profound impact on the brain, behavior, and cognition. Here, we will review the techniques most often used to assess learning and memory in rodent models and discuss findings from studies assessing the cognitive effects of high-fat diet consumption. The review will then consider potential underlying mechanisms in the brain and conclude by reviewing emerging literature suggesting that maternal consumption of a high-fat diet may have effects on the learning and memory of offspring.
Collapse
Affiliation(s)
- Zachary A Cordner
- Cellular & Molecular Medicine Graduate Program, Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Kellie L K Tamashiro
- Cellular & Molecular Medicine Graduate Program, Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States.
| |
Collapse
|
38
|
Abstract
Clinical studies suggest a link between type 2 diabetes mellitus (T2DM) and insulin resistance (IR) and cognitive dysfunction, but there are significant gaps in our knowledge of the mechanisms underlying this relationship. Animal models of IR help to bridge these gaps and point to hippocampal IR as a potential mediator of cognitive dysfunction in T2DM, as well as in Alzheimer disease (AD). This Review highlights these observations and discusses intervention studies which suggest that the restoration of insulin activity in the hippocampus may be an effective strategy to alleviate the cognitive decline associated with T2DM and AD.
Collapse
|
39
|
Scheinert RB, Asokan A, Rani A, Kumar A, Foster TC, Ormerod BK. Some hormone, cytokine and chemokine levels that change across lifespan vary by cognitive status in male Fischer 344 rats. Brain Behav Immun 2015; 49:216-32. [PMID: 26093306 PMCID: PMC4567443 DOI: 10.1016/j.bbi.2015.06.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Revised: 05/26/2015] [Accepted: 06/08/2015] [Indexed: 12/21/2022] Open
Abstract
We trained and tested young (6-8months; n=13), middle-aged (12-14months; n=41), and aged (22-24months; n=24) male Fischer 344 rats in a rapid acquisition water maze task and then quantified 27 stress hormones, cytokines and chemokines in their serum, hippocampi and frontal cortices using bead assay kits and xMAP technology. Middle-aged and aged rats learned the location of the hidden platform over training trials more slowly than their young counterparts. After training, young rats outperformed middle-aged and aged rats on both immediate and 24h retention probe trials and about half of the middle-aged and aged (aging) rats exhibited impaired performances when tested on the retention probe trial 24h later. The concentrations of many serum, hippocampal and cortical analytes changed with age often in networks that may represent age-sensitive signaling pathways and the concentrations of some of these analytes correlated with water maze learning and/or memory scores. Serum GRO/KC and RANTES levels, hippocampal GM-CSF levels and cortical IL-9 and RANTES levels were significantly higher in rats categorized as memory-impaired versus elite agers based upon their 24h probe trial performances. Our data add to the emerging picture of how age-related changes in immune and neuroimmune system signaling impacts cognition.
Collapse
Affiliation(s)
- Rachel B Scheinert
- National Institute of Mental Health, NIH, Bethesda, MD, USA; J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Aditya Asokan
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Asha Rani
- Department of Neuroscience, University of Florida, Gainesville, FL, USA; McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Ashok Kumar
- Department of Neuroscience, University of Florida, Gainesville, FL, USA; McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Thomas C Foster
- Department of Neuroscience, University of Florida, Gainesville, FL, USA; McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
| | - Brandi K Ormerod
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA; Department of Neuroscience, University of Florida, Gainesville, FL, USA; McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
40
|
Low-fat and low-protein diets are associated with hearing discomfort among the elderly of Korea. Br J Nutr 2015; 114:1711-7. [PMID: 26388267 DOI: 10.1017/s0007114515003463] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Research on the effect of low-fat intake on hearing is limited. This study aimed to elucidate the relationship between fat, carbohydrate and protein intake and the presence of hearing discomfort among the elderly. The Korean National Health and Nutrition Examination Survey was conducted from 2009 through to 2012. A total of 4615 participants ranging in age from 60 to 80 years underwent a pure-tone audiometric evaluation, a physical examination and a nutritional survey. The associations between the participants' hearing thresholds and their protein/fat/carbohydrate intake/total energy intake were analysed using simple and multiple regression models with complex sampling adjusted for confounding factors, such as BMI, income level, smoking status and a history of hypertension, hyperlipidaemia and diabetes. Low fat and protein intakes were associated with hearing discomfort (OR 0·82, 95 % CI 0·71, 0·96, P=0·011; OR 0·81, 95 % CI 0·67, 0·96, P=0·017, respectively). This study revealed that low fat and protein intakes are associated with hearing discomfort in the elderly Korean population.
Collapse
|
41
|
Magalhães CA, Carvalho MG, Sousa LP, Caramelli P, Gomes KB. Leptin in Alzheimer's disease. Clin Chim Acta 2015; 450:162-8. [PMID: 26279362 DOI: 10.1016/j.cca.2015.08.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 08/08/2015] [Accepted: 08/12/2015] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of progressive dementia in the elderly population. AD is histologically characterized by accumulation of amyloid-β protein (Aβ) on extracellular plaques and deposition of hyperphosphorylated tau protein in intracellular neurofibrillary tangles. Several studies have shown that obesity may precede dementia and that lifestyle factors play a critical role in the onset of AD. Furthermore, accumulating evidence indicates that obesity is an independent risk factor for developing AD. In this scenario, the understanding of the role of adipose tissue in brain health is essential to clarify the establishment of demential processes. The objective of this work was to review studies regarding leptin, an anorexigenic peptide hormone synthesized in adipocytes, in the context of dementia. Some authors proposed that leptin evaluation might be a better predictor of dementia than traditional anthropometric measures. Leptin, once established as a biomarker, could enhance the understanding of late-onset AD risk over the life course, as well as the clinical progression of prodromal state to manifested AD. Other studies have proposed that leptin presents neuroprotective activities, which could be explained by inhibiting the amyloidogenic process, reducing the levels of tau protein phosphorylation and improving the cognitive function.
Collapse
Affiliation(s)
- C A Magalhães
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - M G Carvalho
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - L P Sousa
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - P Caramelli
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - K B Gomes
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil..
| |
Collapse
|
42
|
Xue Y, Li J, Yan L, Lu L, Liao FF. Genetic variability to diet-induced hippocampal dysfunction in BXD recombinant inbred (RI) mouse strains. Behav Brain Res 2015; 292:83-94. [PMID: 26092713 DOI: 10.1016/j.bbr.2015.06.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 06/11/2015] [Accepted: 06/13/2015] [Indexed: 11/26/2022]
Abstract
Evidence has emerged suggesting that diet-induced obesity can have a negative effect on cognitive function. Here, we exploited a mouse genetic reference population to look for the linkage between these two processes on a genome-wide scale. The focus of this report is to determine whether the various BXD RI strains exhibited different behavioral performance and hippocampal function under high fat dietary (HFD) condition. We quantified genetic variation in body weight gain and consequent influences on behavioral tests in a cohort of 14 BXD strains of mice (8-12 mice/strain, n = 153), for which we have matched data on gene expression and neuroanatomical changes in the hippocampus. It showed that BXD66 was the most susceptible, whereas BXD77 was the least susceptible strain to dietary influences. The performance of spatial reference memory tasks was strongly correlated with body weight gain (P < 0.05). The obesity-prone strains displayed more pronounced spatial memory defects compared to the obesity-resistant strains. These abnormalities were associated with neuroinflammation, synaptic dysfunction, and neuronal loss in the hippocampus. The biological relevance of DSCAM gene polymorphism was assessed using the trait correlation analysis tool in Genenetwork. Furthermore, a significant strain-dependent gene expression difference of DSCAM was detected in the hippocampus of obese BXD strains by real-time quantitative PCR. In conclusion, a variety of across-strain hippocampal alterations and genetic predispositions to diet-induced obesity were found in a set of BXD strains. The obesity-prone and obesity-resistant lines we have identified should be highly useful to study the molecular genetics of diet-induced cognitive decline.
Collapse
Affiliation(s)
| | | | - Lei Yan
- Department of Genetics, Genomics & Informatics, University Tennessee Health Science Center, 874 Union Avenue, Memphis, TN 38163, USA
| | - Lu Lu
- Department of Genetics, Genomics & Informatics, University Tennessee Health Science Center, 874 Union Avenue, Memphis, TN 38163, USA; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong 226000, China.
| | | |
Collapse
|
43
|
Nixon JP, Mavanji V, Butterick TA, Billington CJ, Kotz CM, Teske JA. Sleep disorders, obesity, and aging: the role of orexin. Ageing Res Rev 2015; 20:63-73. [PMID: 25462194 DOI: 10.1016/j.arr.2014.11.001] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 09/19/2014] [Accepted: 11/14/2014] [Indexed: 02/03/2023]
Abstract
The hypothalamic neuropeptides orexin A and B (hypocretin 1 and 2) are important homeostatic mediators of central control of energy metabolism and maintenance of sleep/wake states. Dysregulation or loss of orexin signaling has been linked to narcolepsy, obesity, and age-related disorders. In this review, we present an overview of our current understanding of orexin function, focusing on sleep disorders, energy balance, and aging, in both rodents and humans. We first discuss animal models used in studies of obesity and sleep, including loss of function using transgenic or viral-mediated approaches, gain of function models using exogenous delivery of orexin receptor agonist, and naturally-occurring models in which orexin responsiveness varies by individual. We next explore rodent models of orexin in aging, presenting evidence that orexin loss contributes to age-related changes in sleep and energy balance. In the next section, we focus on clinical importance of orexin in human obesity, sleep, and aging. We include discussion of orexin loss in narcolepsy and potential importance of orexin in insomnia, correlations between animal and human studies of age-related decline, and evidence for orexin involvement in age-related changes in cognitive performance. Finally, we present a summary of recent studies of orexin in neurodegenerative disease. We conclude that orexin acts as an integrative homeostatic signal influencing numerous brain regions, and that this pivotal role results in potential dysregulation of multiple physiological processes when orexin signaling is disrupted or lost.
Collapse
|
44
|
Oania R, McEvoy LK. Plasma leptin levels are not predictive of dementia in patients with mild cognitive impairment. Age Ageing 2015; 44:53-8. [PMID: 25349150 DOI: 10.1093/ageing/afu160] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND animal studies suggest a neuroprotective role for leptin, but human studies have shown mixed results. We examined whether plasma leptin levels in individuals with mild cognitive impairment (MCI) were related to cognitive function at baseline and whether higher leptin levels were associated with reduced risk of dementia. METHODS we categorised 352 MCI participants into sex-specific tertiles based on log-transformed fasting plasma leptin levels. In sex-stratified analyses, we investigated whether cognitive ability differed by leptin tertile. We also examined whether the risk of dementia over a 3-year follow-up period differed by leptin level. Analyses controlled for numerous potential confounding variables, including body mass index, hypertension and levels of blood insulin and C-reactive protein. RESULTS baseline cognitive ability did not differ as a function of leptin level, nor were higher leptin levels associated with reduced hazard of developing dementia. Controlling for related co-variates did not reveal any significant associations between leptin and dementia risk. CONCLUSION in this cohort of older adults with MCI, plasma leptin level was not associated with cognitive function at baseline, nor did it predict risk of dementia. Other biological measures, such as volumetric MRI and cerebrospinal fluid protein levels, have demonstrated robust dementia prediction in this cohort. Thus, the current negative findings suggest that plasma leptin, on its own, is unlikely to become a useful clinical biomarker for Alzheimer's disease. Efforts to develop other blood-based biomarkers are needed.
Collapse
Affiliation(s)
- Rafael Oania
- John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Linda K McEvoy
- Department of Radiology, University of California, San Diego, 9500 Gilman Dr MC 0841, La Jolla, CA 92037, USA
| |
Collapse
|
45
|
Zink AN, Perez-Leighton CE, Kotz CM. The orexin neuropeptide system: physical activity and hypothalamic function throughout the aging process. Front Syst Neurosci 2014; 8:211. [PMID: 25408639 PMCID: PMC4219460 DOI: 10.3389/fnsys.2014.00211] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 10/07/2014] [Indexed: 12/18/2022] Open
Abstract
There is a rising medical need for novel therapeutic targets of physical activity. Physical activity spans from spontaneous, low intensity movements to voluntary, high-intensity exercise. Regulation of spontaneous and voluntary movement is distributed over many brain areas and neural substrates, but the specific cellular and molecular mechanisms responsible for mediating overall activity levels are not well understood. The hypothalamus plays a central role in the control of physical activity, which is executed through coordination of multiple signaling systems, including the orexin neuropeptides. Orexin producing neurons integrate physiological and metabolic information to coordinate multiple behavioral states and modulate physical activity in response to the environment. This review is organized around three questions: (1) How do orexin peptides modulate physical activity? (2) What are the effects of aging and lifestyle choices on physical activity? (3) What are the effects of aging on hypothalamic function and the orexin peptides? Discussion of these questions will provide a summary of the current state of knowledge regarding hypothalamic orexin regulation of physical activity during aging and provide a platform on which to develop improved clinical outcomes in age-associated obesity and metabolic syndromes.
Collapse
Affiliation(s)
- Anastasia N Zink
- Graduate Program in Neuroscience, School of Medicine, University of Minnesota Minneapolis, MN, USA
| | | | - Catherine M Kotz
- Graduate Program in Neuroscience, School of Medicine, University of Minnesota Minneapolis, MN, USA ; GRECC (11G), Minneapolis VA Healthcare System Minneapolis, MN, USA ; Department of Food Science and Nutrition, University of Minnesota Saint Paul, MN, USA
| |
Collapse
|
46
|
Sobów T, Fendler W, Magierski R. Body mass index and mild cognitive impairment-to-dementia progression in 24 months: a prospective study. Eur J Clin Nutr 2014; 68:1216-9. [DOI: 10.1038/ejcn.2014.167] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 07/12/2014] [Indexed: 02/03/2023]
|
47
|
Ménard C, Quirion R, Bouchard S, Ferland G, Gaudreau P. Glutamatergic signaling and low prodynorphin expression are associated with intact memory and reduced anxiety in rat models of healthy aging. Front Aging Neurosci 2014; 6:81. [PMID: 24847259 PMCID: PMC4019859 DOI: 10.3389/fnagi.2014.00081] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 04/16/2014] [Indexed: 11/13/2022] Open
Abstract
The LOU/C/Jall (LOU) rat strain is considered a model of healthy aging due to its increased longevity, maintenance of stable body weight (BW) throughout life and low incidence of age-related diseases. However, aging LOU rat cognitive and anxiety status has yet to be investigated. In the present study, male and female LOU rat cognitive performances (6-42 months) were assessed using novel object recognition and Morris Water Maze tasks. Recognition memory remained intact in all LOU rats up to 42 months of age. As for spatial memory, old LOU rat performed similarly as young animals for learning acquisition, reversal learning, and retention. While LOU rat BW remained stable despite aging, 20-month-old ad-libitum-fed (OAL) male Sprague Dawley rats become obese. We determined if long-term caloric restriction (LTCR) prevents age-related BW increase and cognitive deficits in this rat strain, as observed in the obesity-resistant LOU rats. Compared to young animals, recognition memory was impaired in OAL but intact in 20-month-old calorie-restricted (OCR) rats. Similarly, OAL spatial learning acquisition was impaired but LTCR prevented the deficits. Exacerbated stress responses may favor age-related cognitive decline. In the elevated plus maze and open field tasks, LOU and OCR rats exhibited high levels of exploratory activity whereas OAL rats displayed anxious behaviors. Expression of prodynorphin (Pdyn), an endogenous peptide involved in stress-related memory impairments, was increased in the hippocampus of OAL rats. Group 1 metabotropic glutamate receptor 5 and immediate early genes Homer 1a and Arc expression, both associated with successful cognitive aging, were unaltered in aging LOU rats but lower in OAL than OCR rats. Altogether, our results, supported by principal component analysis and correlation matrix, suggest that intact memory and low anxiety are associated with glutamatergic signaling and low Pdyn expression in the hippocampus of non-obese aging rats.
Collapse
Affiliation(s)
- Caroline Ménard
- Neuroscience Division, Douglas Mental Health University Institute Research Center Montreal, QC, Canada ; Department of Psychiatry, McGill University Montreal, QC, Canada ; Laboratory of Neuroendocrinology of Aging, Centre Hospitalier de l'Université de Montréal Research Center Montreal, QC, Canada ; Department of Medicine, University of Montreal Montreal, QC, Canada
| | - Rémi Quirion
- Neuroscience Division, Douglas Mental Health University Institute Research Center Montreal, QC, Canada ; Department of Psychiatry, McGill University Montreal, QC, Canada
| | - Sylvain Bouchard
- Faculty of Medicine, University of Montreal Montreal, QC, Canada
| | - Guylaine Ferland
- Hôpital du Sacré-Coeur de Montréal Research Center Montreal, QC, Canada ; Department of Nutrition, University of Montreal Montreal, QC, Canada
| | - Pierrette Gaudreau
- Laboratory of Neuroendocrinology of Aging, Centre Hospitalier de l'Université de Montréal Research Center Montreal, QC, Canada ; Department of Medicine, University of Montreal Montreal, QC, Canada
| |
Collapse
|
48
|
Abstract
Adipose tissue is a known source of proinflammatory cytokines in obese humans and animal models, including the db/db mouse, in which obesity arises as a result of leptin receptor insensitivity. Inflammatory cytokines induce cognitive deficits across numerous conditions, but no studies have determined whether obesity-induced inflammation mediates synaptic dysfunction. To address this question, we used a treadmill training paradigm in which mice were exposed to daily training sessions or an immobile belt, with motivation achieved by delivery of compressed air on noncompliance. Treadmill training prevented hippocampal microgliosis, abolished expression of microglial activation markers, and also blocked the functional sensitization observed in isolated cells after ex vivo exposure to lipopolysaccharide. Reduced microglial reactivity with exercise was associated with reinstatement of hippocampus-dependent memory, reversal of deficits in long-term potentiation, and normalization of hippocampal dendritic spine density. Because treadmill training evokes broad responses not limited to the immune system, we next assessed whether directly manipulating adiposity through lipectomy and fat transplantation influences inflammation, cognition, and synaptic plasticity. Lipectomy prevents and fat transplantation promotes systemic and central inflammation, with associated alterations in cognitive and synaptic function. Levels of interleukin 1β (IL1β) emerged as a correlate of adiposity and cognitive impairment across both the treadmill and lipectomy studies, so we manipulated hippocampal IL1 signaling using intrahippocampal delivery of IL1 receptor antagonist (IL1ra). Intrahippocampal IL1ra prevented synaptic dysfunction, proinflammatory priming, and cognitive impairment. This pattern supports a central role for IL1-mediated neuroinflammation as a mechanism for cognitive deficits in obesity and diabetes.
Collapse
|
49
|
Perianes-Cachero A, Burgos-Ramos E, Puebla-Jiménez L, Canelles S, Frago L, Hervás-Aguilar A, de Frutos S, Toledo-Lobo M, Mela V, Viveros M, Argente J, Chowen J, Arilla-Ferreiro E, Barrios V. Acute up-regulation of the rat brain somatostatin receptor-effector system by leptin is related to activation of insulin signaling and may counteract central leptin actions. Neuroscience 2013; 252:289-301. [DOI: 10.1016/j.neuroscience.2013.08.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 08/07/2013] [Accepted: 08/12/2013] [Indexed: 11/28/2022]
|
50
|
Daulatzai MA. Neurotoxic Saboteurs: Straws that Break the Hippo’s (Hippocampus) Back Drive Cognitive Impairment and Alzheimer’s Disease. Neurotox Res 2013; 24:407-59. [DOI: 10.1007/s12640-013-9407-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 06/06/2013] [Accepted: 06/17/2013] [Indexed: 12/29/2022]
|