1
|
Schneider JS, Williams C, Zafar S, Joo J, Himes BE. Influences of quality of maternal care and environmental enrichment on associative memory function in rats with early life lead exposure. Brain Behav 2024; 14:e70040. [PMID: 39295102 PMCID: PMC11410876 DOI: 10.1002/brb3.70040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 07/11/2024] [Accepted: 08/02/2024] [Indexed: 09/21/2024] Open
Abstract
INTRODUCTION Children in low socioeconomic status (SES) communities are at higher risk of exposure to lead (Pb) and potentially more severe adverse outcomes from Pb exposures. While the factors encompassing SES are complex, low SES households often have less enriching home environments and parent-child interactions. This study investigated the extent to which environmental/behavioral factors (quality of maternal care and richness of the postnatal environment) may modify adverse effects from Pb exposure. METHODS Long-Evans female rats were randomly assigned to Control (no Pb), Early Postnatal (EPN: birth through weaning), or Perinatal (PERI: 14 days pre-mating through weaning) Pb exposure groups. From postnatal days (PNDs) 2-9, maternal care behaviors were observed, and dams were classified as low or high maternal care based on amounts of licking/grooming and arched back nursing. At weaning, pups were randomly assigned to enriched or non-enriched environments. At PND 55, animals began trace fear conditioning and associative memory was tested on days 1, 2, and 10 postconditioning. RESULTS Control offspring showed no significant effects of maternal care or enrichment on task performance. Females with EPN-Pb exposure and males with PERI-Pb exposure living in the non-enriched environment and having an LMC mother had significant memory impairments at days 2 and 10 that were not observed in comparably housed animals with HMC mothers. Enriched animals had no deficits, regardless of maternal care status. CONCLUSION These results show the potential for modulatory influences of maternal care and housing environment on protecting against or reversing at least one aspect of Pb-induced cognitive/behavioral dysfunction.
Collapse
Affiliation(s)
- Jay S. Schneider
- Department of Pathology and Genomic MedicineThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
- Center of Excellence in Environmental Toxicology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Courtney Williams
- Department of Pathology and Genomic MedicineThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Shamaila Zafar
- Department of Pathology and Genomic MedicineThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Jaehyun Joo
- Center of Excellence in Environmental Toxicology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Blanca. E. Himes
- Center of Excellence in Environmental Toxicology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
2
|
Khalil MH. Neurosustainability. Front Hum Neurosci 2024; 18:1436179. [PMID: 39268220 PMCID: PMC11390526 DOI: 10.3389/fnhum.2024.1436179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/07/2024] [Indexed: 09/15/2024] Open
Abstract
While the human brain has evolved extraordinary abilities to dominate nature, modern living has paradoxically trapped it in a contemporary "cage" that stifles neuroplasticity. Within this modern environment lurk unseen natural laws with power to sustain the human brain's adaptive capacities - if consciously orchestrated into the environments we design. For too long our contemporary environments have imposed an unyielding static state, while still neglecting the brain's constant adaptive nature as it evolves to dominate the natural world with increasing sophistication. The theory introduced in this article aims to go back in nature without having to go back in time, introducing and expounding Neurosustainability as a novel paradigm seeing beyond the contemporary confines to architect environments and brains in parallel. Its integrated neuro-evidenced framework proposes four enrichment scopes-spatial, natural, aesthetic, and social-each holding multifaceted attributes promising to sustain regions like the hippocampus, cortex and amygdala. Neurosustainability aims to liberate the quintessential essence of nature to sustain and enhance neuroplastic processes through a cycle that begins with design and extends through epigenetic changes. This paradigm shift aims to foster cognitive health and wellness by addressing issues like stress, depression, anxiety and cognitive decline common in the contemporary era thereby offering a path toward a more neurosustainable era aiming to nurture the evolution of the human brain now and beyond.
Collapse
Affiliation(s)
- Mohamed Hesham Khalil
- Department of Architecture, Faculty of Architecture and History of Art, School of Arts and Humanities, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
3
|
Weiner SP, Vasquez C, Song S, Zhao K, Ali O, Rosenkilde D, Froemke RC, Carr KD. Sex difference in the effect of environmental enrichment on food restriction-induced persistence of cocaine conditioned place preference and mechanistic underpinnings. ADDICTION NEUROSCIENCE 2024; 10:100142. [PMID: 38323217 PMCID: PMC10843874 DOI: 10.1016/j.addicn.2024.100142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Psychosocial and environmental factors, including loss of natural reward, contribute to the risk of drug abuse. Reward loss has been modeled in animals by removal from social or sexual contact, transfer from enriched to impoverished housing, or restriction of food. We previously showed that food restriction increases the unconditioned rewarding effects of abused drugs and the conditioned incentive effects of drug-paired environments. Mechanistic studies provided evidence of decreased basal dopamine (DA) transmission, adaptive upregulation of signaling downstream of D1 DA receptor stimulation, synaptic upscaling and incorporation of calcium-permeable AMPA receptors (CP-AMPARs) in medium spiny neurons (MSNs) of nucleus accumbens (NAc). These findings align with the still evolving 'reward deficiency' hypothesis of drug abuse. The present study tested whether a compound natural reward that is known to increase DA utilization, environmental enrichment, would prevent the persistent expression of cocaine conditioned place preference (CPP) otherwise observed in food restricted rats, along with the mechanistic underpinnings. Because nearly all prior investigations of both food restriction and environmental enrichment effects on cocaine CPP were conducted in male rodents, both sexes were included in the present study. Results indicate that environmental enrichment curtailed the persistence of CPP expression, decreased signaling downstream of the D1R, and decreased the amplitude and frequency of spontaneous excitatory postsynaptic currents (EPSCs) in NAc MSNs of food restricted male, but not female, rats. The failure of environmental enrichment to significantly decrease food restriction-induced synaptic insertion of CP-AMPARs, and how this may accord with previous pharmacological findings that blockade of CP-AMPARs reverses behavioral effects of food restriction is discussed. In addition, it is speculated that estrous cycle-dependent fluctuations in DA release, receptor density and MSN excitability may obscure the effect of increased DA signaling during environmental enrichment, thereby interfering with development of the cellular and behavioral effects that enrichment produced in males.
Collapse
Affiliation(s)
- Sydney P. Weiner
- Department of Psychiatry, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, United States
| | - Carolina Vasquez
- Department of Psychiatry, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, United States
- Diabetes Research Program, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, United States
| | - Soomin Song
- Department of Pathology, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, United States
| | - Kaiyang Zhao
- Department of Psychiatry, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, United States
| | - Omar Ali
- Department of Psychiatry, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, United States
| | - Danielle Rosenkilde
- Department of Psychiatry, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, United States
| | - Robert C. Froemke
- Skirball Institute of Biomolecular Medicine, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, United States
- Department of Otolaryngology, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, United States
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, United States
- Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, United States
| | - Kenneth D. Carr
- Department of Psychiatry, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, United States
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, United States
- Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, United States
| |
Collapse
|
4
|
Voigt MW, Schepers J, Haas J, von Bohlen Und Halbach O. Reduced Levels of Brain-Derived Neurotrophic Factor Affect Body Weight, Brain Weight and Behavior. BIOLOGY 2024; 13:159. [PMID: 38534429 DOI: 10.3390/biology13030159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 03/28/2024]
Abstract
Neurotrophins, which belong to the family of growth factors, not only play crucial roles during development but are also involved in many processes in the postnatal brain. One representative of neurotrophins is brain-derived neurotrophic factor (BDNF). BDNF plays a role in the regulation of body weight and neuronal plasticity and is, therefore, also involved in processes associated with learning and memory formation. Many of the studies on BDNF have been carried out using BDNF-deficient mice. Unfortunately, homozygous deletion of BDNF is lethal in the early postnatal stage, so heterozygous BDNF-deficient mice are often studied. Another possibility is the use of conditional BDNF-deficient mice in which the expression of BDNF is strongly downregulated in some brain cells, for example, in the neurons of the central nervous system, but the expression of BDNF in other cells in the brain is unchanged. To further reduce BDNF expression, we crossed heterozygous BDNF-deficient mice with mice carrying a deletion of BDNF in neurofilament L-positive neurons. These offspring are viable, and the animals with a strong reduction in BDNF in the brain show a strongly increased body weight, which is accompanied by a reduction in brain weight. In addition, these animals show behavioral abnormalities, particularly with regard to locomotion.
Collapse
Affiliation(s)
- Matthias Wilhelm Voigt
- Institut für Anatomie und Zellbiologie, Universitätsmedizin Greifswald, Friedrich-Loeffler Str. 23c, D-17489 Greifswald, Germany
| | - Jens Schepers
- Institut für Anatomie und Zellbiologie, Universitätsmedizin Greifswald, Friedrich-Loeffler Str. 23c, D-17489 Greifswald, Germany
| | - Jacqueline Haas
- Institut für Anatomie und Zellbiologie, Universitätsmedizin Greifswald, Friedrich-Loeffler Str. 23c, D-17489 Greifswald, Germany
| | - Oliver von Bohlen Und Halbach
- Institut für Anatomie und Zellbiologie, Universitätsmedizin Greifswald, Friedrich-Loeffler Str. 23c, D-17489 Greifswald, Germany
| |
Collapse
|
5
|
Dandi Ε, Theotokis P, Petri MC, Sideropoulou V, Spandou E, Tata DA. Environmental enrichment initiated in adolescence restores the reduced expression of synaptophysin and GFAP in the hippocampus of chronically stressed rats in a sex-specific manner. Dev Psychobiol 2023; 65:e22422. [PMID: 37796476 DOI: 10.1002/dev.22422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 08/12/2023] [Accepted: 08/17/2023] [Indexed: 10/06/2023]
Abstract
This study aims at investigating whether environmental enrichment (EE) initiated in adolescence can alter chronic unpredictable stress (CUS)-associated changes in astroglial and synaptic plasticity markers in male and female rats. To this end, we studied possible alterations in hippocampal glial fibrillary acidic protein (GFAP) and synaptophysin (SYN) in CUS rats previously housed in EE. Wistar rats on postnatal day (PND) 23 were housed for 10 weeks in standard housing (SH) or enriched conditions. On PND 66, animals were exposed to CUS for 4 weeks. SYN and GFAP expressions were evaluated in CA1 and CA3 subfields and dentate gyrus (DG). CUS reduced the expression of SYN in all hippocampal areas, whereas lower GFAP expression was evident only in CA1 and CA3. The reduced expression of SYN in DG and CA3 was evident to male SH/CUS rats, whereas the reduced GFAP expression in CA1 and CA3 was limited to SH/CUS females. EE housing increased the hippocampal expression of both markers and protected against CUS-associated decreases. Our findings indicate that the decreases in the expression of SYN and GFAP following CUS are region and sex-specific and underline the neuroprotective role of EE against these CUS-associated changes.
Collapse
Affiliation(s)
- Εvgenia Dandi
- Laboratory of Cognitive Neuroscience, School of Psychology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Maria Christina Petri
- Laboratory of Cognitive Neuroscience, School of Psychology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Vaia Sideropoulou
- Laboratory of Cognitive Neuroscience, School of Psychology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Evangelia Spandou
- Laboratory of Experimental Physiology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Despina A Tata
- Laboratory of Cognitive Neuroscience, School of Psychology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
6
|
Farmer AL, Lewis MH. Reduction of restricted repetitive behavior by environmental enrichment: Potential neurobiological mechanisms. Neurosci Biobehav Rev 2023; 152:105291. [PMID: 37353046 DOI: 10.1016/j.neubiorev.2023.105291] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/04/2023] [Accepted: 06/19/2023] [Indexed: 06/25/2023]
Abstract
Restricted repetitive behaviors (RRB) are one of two diagnostic criteria for autism spectrum disorder and common in other neurodevelopmental and psychiatric disorders. The term restricted repetitive behavior refers to a wide variety of inflexible patterns of behavior including stereotypy, self-injury, restricted interests, insistence on sameness, and ritualistic and compulsive behavior. However, despite their prevalence in clinical populations, their underlying causes remain poorly understood hampering the development of effective treatments. Intriguingly, numerous animal studies have demonstrated that these behaviors are reduced by rearing in enriched environments (EE). Understanding the processes responsible for the attenuation of repetitive behaviors by EE should offer insights into potential therapeutic approaches, as well as shed light on the underlying neurobiology of repetitive behaviors. This review summarizes the current knowledge of the relationship between EE and RRB and discusses potential mechanisms for EE's attenuation of RRB based on the broader EE literature. Existing gaps in the literature and future directions are also discussed.
Collapse
Affiliation(s)
- Anna L Farmer
- Department of Psychology, University of Florida, Gainesville, FL, USA.
| | - Mark H Lewis
- Department of Psychology, University of Florida, Gainesville, FL, USA; Department of Psychiatry, University of Florida, Gainesville, FL, USA
| |
Collapse
|
7
|
Valero-Aracama MJ, Zheng F, Alzheimer C. Dorsal-Ventral Gradient of Activin Regulates Strength of GABAergic Inhibition along Longitudinal Axis of Mouse Hippocampus in an Activity-Dependent Fashion. Int J Mol Sci 2023; 24:13145. [PMID: 37685952 PMCID: PMC10487617 DOI: 10.3390/ijms241713145] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/20/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
The functional and neurophysiological distinction between the dorsal and ventral hippocampus affects also GABAergic inhibition. In line with this notion, ventral CA1 pyramidal cells displayed a more dynamic and effective response to inhibitory input compared to their dorsal counterparts. We posit that this difference is effected by the dorsal-ventral gradient of activin A, a member of the transforming growth factor-β family, which is increasingly recognized for its modulatory role in brain regions involved in cognitive functions and affective behavior. Lending credence to this hypothesis, we found that in slices from transgenic mice expressing a dominant-negative mutant of activin receptor IB (dnActRIB), inhibitory transmission was enhanced only in CA1 neurons of the dorsal hippocampus, where the basal activin A level is much higher than in the ventral hippocampus. We next asked how a rise in endogenous activin A would affect GABAergic inhibition along the longitudinal axis of the hippocampus. We performed ex vivo recordings in wild-type and dnActRIB mice after overnight exposure to an enriched environment (EE), which engenders a robust increase in activin A levels in both dorsal and ventral hippocampi. Compared to control mice from standard cages, the behaviorally induced surge in activin A produced a decline in ventral inhibition, an effect that was absent in slices from dnActRIB mice. Underscoring the essential role of activin in the EE-associated modulation of ventral inhibition, this effect was mimicked by acute application of recombinant activin A in control slices. In summary, both genetic and behavioral manipulations of activin receptor signaling affected the dorsal-ventral difference in synaptic inhibition, suggesting that activin A regulates the strength of GABAergic inhibition in a region-specific fashion.
Collapse
|
8
|
Costa GA, de Gusmão Taveiros Silva NK, Marianno P, Chivers P, Bailey A, Camarini R. Environmental Enrichment Increased Bdnf Transcripts in the Prefrontal Cortex: Implications for an Epigenetically Controlled Mechanism. Neuroscience 2023; 526:277-289. [PMID: 37419403 DOI: 10.1016/j.neuroscience.2023.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 06/26/2023] [Accepted: 07/01/2023] [Indexed: 07/09/2023]
Abstract
Environmental enrichment (EE) is a condition characterized by its complexity regarding social contact, exposure to novelty, tactile stimuli and voluntary exercise, also is considered as a eustress model. The impact of EE on brain physiology and behavioral outcomes may be at least partly underpinned by mechanisms involving the modulation of the brain-derived neurotrophic factor (BDNF), but the connection between specific Bdnf exon expression and their epigenetic regulation remain poorly understood. This study aimed to dissect the transcriptional and epigenetic regulatory effect of 54-day exposure to EE on BDNF by analysing individual BDNF exons mRNA expression and the DNA methylation profile of a key transcriptional regulator of the Bdnf gene, exon IV, in the prefrontal cortex (PFC) of C57BL/6 male mice (sample size = 33). Bdnf exons II, IV, VI and IX mRNA expression were upregulated and methylation levels at two CpG sites of exon IV were reduced in the PFC of EE mice. As deficit in exon IV expression has also been causally implicated in stress-related psychopathologies, we also assessed anxiety-like behavior and plasma corticosterone levels in these mice to determine any potential correlation. However, no changes were observed in EE mice. The findings may suggest an EE-induced epigenetic control of BDNF exon expression via a mechanism involving exon IV methylation. The findings of this study contribute to the current literature by dissecting the Bdnf gene topology in the PFC where transcriptional and epigenetic regulatory effect of EE takes place.
Collapse
Affiliation(s)
- Gabriel Araújo Costa
- Pharmacology Department, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Priscila Marianno
- Pharmacology Department, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Priti Chivers
- School of Biosciences & Medicine, Faculty of Health & Medical Sciences, University of Surrey, Guildford, UK
| | - Alexis Bailey
- Pharmacology Section, Institute of Medical and Biomedical Education, St George's University of London, London, UK.
| | - Rosana Camarini
- Pharmacology Department, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
9
|
Ke T, Tinkov AA, Skalny AV, Santamaria A, Rocha JBT, Bowman AB, Chen W, Aschner M. Epigenetics and Methylmercury-Induced Neurotoxicity, Evidence from Experimental Studies. TOXICS 2023; 11:toxics11010072. [PMID: 36668798 PMCID: PMC9860901 DOI: 10.3390/toxics11010072] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/08/2023] [Accepted: 01/10/2023] [Indexed: 05/14/2023]
Abstract
MeHg is an environmental neurotoxin that can adversely affect the development of the nervous system. The molecular integrity of chromatin in the nucleus is an important target of MeHg. Low levels of MeHg trigger epigenetic mechanisms that may be involved in long-lasting and transgenerational neurotoxicity after exposure. Emerging evidence has shown that these mechanisms include histone modification, siRNA, and DNA methylation. The MeHg-induced inhibition of neurodifferentiation and neurogenesis are mechanistically associated with epigenetic alterations in critical genes, such as neurotrophin brain-derived neurotrophic factor (BDNF). Further, MeHg exposure has been shown to alter the activity and/or expression of the upstream regulators of chromatin structure, including histone deacetylases (HDACs) and DNA methyltransferase (DNMTs), which may trigger permanent alterations in histone modifications and DNA methylation. MeHg-exposure also alters several species of miRNA that are associated with neurodevelopment. Genetic studies in the C. elegans model of MeHg-induced toxicity proposes a potential interplay between exogenous RNAi and antioxidant defense. In this review, we discuss the molecular basis for MeHg exposure-induced alterations in chromatin structure and the roles of histone modifications, siRNA, and DNA methylation in MeHg-induced neurotoxic effects.
Collapse
Affiliation(s)
- Tao Ke
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Correspondence: (T.K.); (M.A.)
| | - Alexey A. Tinkov
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, 150003 Yaroslavl, Russia
- Department of Medical Elementology, RUDN University, 117198 Moscow, Russia
| | - Anatoly V. Skalny
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
- Department of Medical Elementology, RUDN University, 117198 Moscow, Russia
| | - Abel Santamaria
- Laboratorio de Aminoácidos Excitadores/Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía, Mexico City 14269, Mexico
| | - Joao B. T. Rocha
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil
| | - Aaron B. Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Wen Chen
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Correspondence: (T.K.); (M.A.)
| |
Collapse
|
10
|
Kurowska-Rucińska E, Ruciński J, Myślińska D, Grembecka B, Wrona D, Majkutewicz I. Dimethyl Fumarate Alleviates Adult Neurogenesis Disruption in Hippocampus and Olfactory Bulb and Spatial Cognitive Deficits Induced by Intracerebroventricular Streptozotocin Injection in Young and Aged Rats. Int J Mol Sci 2022; 23:ijms232415449. [PMID: 36555093 PMCID: PMC9779626 DOI: 10.3390/ijms232415449] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
The disorder of adult neurogenesis is considered an important mechanism underlying the learning and memory impairment observed in Alzheimer's disease (AD). The sporadic nonhereditary form of AD (sAD) affects over 95% of AD patients and is related to interactions between genetic and environmental factors. An intracerebroventricular injection of streptozotocin (STZ-ICV) is a representative and well-established method to induce sAD-like pathology. Dimethyl fumarate (DMF) has antioxidant and anti-inflammatory properties and is used for multiple sclerosis treatment. The present study determines whether a 26-day DMF therapy ameliorates the disruption of adult neurogenesis and BDNF-related neuroprotection in the hippocampus and olfactory bulb (OB) in an STZ-ICV rat model of sAD. Considering age as an important risk factor for developing AD, this study was performed using 3-month-old (the young group) and 22-month-old (the aged group) male Wistar rats. Spatial cognitive functions were evaluated with the Morris water maze task. Immunofluorescent labelling was used to assess the parameters of adult neurogenesis and BDNF-related neuroprotection in the hippocampus and OB. Our results showed that the STZ-ICV evoked spatial learning and memory impairment and disturbances in adult neurogenesis and BDNF expression in both examined brain structures. In the aged animals, the deficits were more severe. We found that the DMF treatment significantly alleviated STZ-ICV-induced behavioural and neuronal disorders in both age groups of the rats. Our findings suggest that DMF, due to its beneficial effect on the formation of new neurons and BDNF-related neuroprotection, may be considered as a promising new therapeutic agent in human sAD.
Collapse
|
11
|
Manosso LM, Broseghini LDR, Campos JMB, Padilha APZ, Botelho MEM, da Costa MA, Abelaira HM, Gonçalves CL, Réus GZ. Beneficial effects and neurobiological aspects of environmental enrichment associated to major depressive disorder and autism spectrum disorder. Brain Res Bull 2022; 190:152-167. [PMID: 36191730 DOI: 10.1016/j.brainresbull.2022.09.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/15/2022]
Abstract
A suitable enriched environment favors development but can also influence behavior and neuronal circuits throughout development. Studies have shown that environmental enrichment (EE) can be used as an essential tool or combined with conventional treatments to improve psychiatric and neurological symptoms, including major depressive disorder (MDD) and autism spectrum disorder (ASD). Both disorders affect a significant percentage of the world's population and have complex pathophysiology. Moreover, the available treatments for MDD and ASD are still inadequate for many affected individuals. Experimental models demonstrate that EE has significant positive effects on behavioral modulation. In addition, EE has effects on neurobiology, including improvement in synaptic connections and neuroplasticity, modulation of neurotransmissions, a decrease in inflammation and oxidative stress, and other neurobiology effects that can be involved in the pathophysiology of MDD and ASD. Thus, this review aims to describe the leading behavioral and neurobiological effects associated with EE in MDD and ASD.
Collapse
Affiliation(s)
- Luana M Manosso
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Lia D R Broseghini
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - José Marcelo B Campos
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Alex Paulo Z Padilha
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Maria Eduarda M Botelho
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Maiara A da Costa
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Helena M Abelaira
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Cinara L Gonçalves
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Gislaine Z Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
12
|
Keloglan Musuroglu S, Ozturk DM, Sahin L, Cevik OS, Cevik K. Environmental enrichment as a strategy: Attenuates the anxiety and memory impairment in social isolation stress. Int J Dev Neurosci 2022; 82:499-512. [PMID: 35724417 DOI: 10.1002/jdn.10205] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/12/2022] [Accepted: 06/07/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Social isolation (SI) early in life produces behavioral and cognitive abnormalities. On the contrary, environmental enrichment (EE) offers beneficial effects on brain plasticity and development. This study was designed to examine how EE affects memory functions, anxiety level, and the expression levels of memory/anxiety-related genes such as NR2A, NR2B, BDNF, and cFos in the hippocampus of socially isolated rats. MATERIALS AND METHODS Wistar albino male rats (n = 40) were separated into the five groups: Standard cage (SC), SI, EE, SI + SC, and SI + EE group. For each group, eight rats were housed, either grouped or isolated, in a standard or 3-week EE, respectively. Morris water maze test (MWMT) was used for measuring the learning and memory function. Elevated plus maze (EPM) and open field (OF) were used for the evaluation of anxiety behavior. Blood corticosterone level was evaluated by the ELISA method. The expression levels of genes were measured by the RT-PCR method. RESULTS Results showed that EE increased memory performance in the SI group (p < 0.05). SI caused anxiety while EE improved anxiety behavior (p < 0.05). There was no significant difference between the groups in the OF test. Corticosterone levels did not change between groups. BDNF expression level was downregulated in EE and SI + SC compared with the SC group (respectively; p = 0.012; p = 0.011). NR2A, NR2B, and cFos expression levels did not change between groups significantly. CONCLUSIONS SI impaired memory performance while EE has beneficial effects on memory in socially isolated rats. EE alone was insufficient to cause alterations in the memory performance. The therapeutic effects of EE became strengthened while applied together with stress protocol. Together with improving the effectiveness of memory function, EE has the potential to decrease anxiety behavior. EE seemed to be the reason for decreasing in BDNF.
Collapse
Affiliation(s)
| | - Duygu Murat Ozturk
- Midwifery Department, Faculty of Health Sciences, Amasya University, Amasya, Turkey
| | - Leyla Sahin
- Physiology Department, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Ozge Selin Cevik
- Physiology Department, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Kenan Cevik
- Health Sciences Institute, Faculty of Medicine, Mersin University, Mersin, Turkey
| |
Collapse
|
13
|
Zou J, Walter TJ, Barnett A, Rohlman A, Crews FT, Coleman LG. Ethanol Induces Secretion of Proinflammatory Extracellular Vesicles That Inhibit Adult Hippocampal Neurogenesis Through G9a/GLP-Epigenetic Signaling. Front Immunol 2022; 13:866073. [PMID: 35634322 PMCID: PMC9136051 DOI: 10.3389/fimmu.2022.866073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 04/07/2022] [Indexed: 12/15/2022] Open
Abstract
Adult hippocampal neurogenesis (AHN) is involved in learning and memory as well as regulation of mood. Binge ethanol reduces AHN, though the mechanism is unknown. Microglia in the neurogenic niche are important regulators of AHN, and ethanol promotes proinflammatory microglia activation. We recently reported that extracellular vesicles (EVs) mediate ethanol-induced inflammatory signaling in microglia. Therefore, we investigated the role of EVs in ethanol-induced loss of adult hippocampal neurogenesis. At rest, microglia promoted neurogenesis through the secretion of pro-neurogenic extracellular vesicles (pn-EVs). Depletion of microglia using colony-stimulating factor 1 receptor (CSFR1) inhibition in vivo or using ex vivo organotypic brain slice cultures (OBSCs) caused a 30% and 56% loss of neurogenesis in the dentate, respectively, as measured by immunohistochemistry for doublecortin (DCX). Likewise, chemogenetic inhibition of microglia using a CD68.hM4di construct caused a 77% loss in OBSC, indicating a pro-neurogenic resting microglial phenotype. EVs from control OBSC were pro-neurogenic (pn-EVs), enhancing neurogenesis when transferred to other naive OBSC and restoring neurogenesis in microglia-depleted cultures. Ethanol inhibited neurogenesis and caused secretion of proinflammatory EVs (EtOH-EVs). EtOH-EVs reduced hippocampal neurogenesis in naïve OBSC by levels similar to ethanol. Neurogenesis involves complex regulation of chromatin structure that could involve EV signaling. Accordingly, EtOH-EVs were found to be enriched with mRNA for the euchromatin histone lysine methyltransferase (Ehm2t/G9a), an enzyme that reduces chromatin accessibility through histone-3 lysine-9 di-methylation (H3K9me2). EtOH-EVs induced G9a and H3K9me2 by 2-fold relative to pn-EVs in naïve OBSCs. Pharmacological inhibition of G9a with either BIX-01294 or UNC0642 prevented loss of neurogenesis caused by both EtOH and EtOH-EVs. Thus, this work finds that proinflammatory EtOH-EVs promote the loss of adult hippocampal neurogenesis through G9a-mediated epigenetic modification of chromatin structure.
Collapse
Affiliation(s)
- Jian Zou
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States
| | - T. Jordan Walter
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States
| | - Alexandra Barnett
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States
| | - Aaron Rohlman
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States
| | - Fulton T. Crews
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States
- Department of Psychiatry, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States
| | - Leon G. Coleman
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States
| |
Collapse
|
14
|
Gutiérrez-Vera B, Rivera-Olvera A, Escobar ML. ENVIRONMENTAL ENRICHMENT ATTENUATES CONDITIONED TASTE AVERSION THROUGH THE RESTORATION OF BDNF LEVELS IN THE INSULAR CORTEX. Behav Brain Res 2022; 430:113947. [DOI: 10.1016/j.bbr.2022.113947] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 05/07/2022] [Accepted: 05/24/2022] [Indexed: 12/20/2022]
|
15
|
Cutuli D, Landolfo E, Petrosini L, Gelfo F. Environmental Enrichment Effects on the Brain-Derived Neurotrophic Factor Expression in Healthy Condition, Alzheimer's Disease, and Other Neurodegenerative Disorders. J Alzheimers Dis 2021; 85:975-992. [PMID: 34897089 DOI: 10.3233/jad-215193] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Brain-derived neurotrophic factor (BDNF), a protein belonging to the neurotrophin family, is known to be heavily involved in synaptic plasticity processes that support brain development, post-lesion regeneration, and cognitive performances, such as learning and memory. Evidence indicates that BDNF expression can be epigenetically regulated by environmental stimuli and thus can mediate the experience-dependent brain plasticity. Environmental enrichment (EE), an experimental paradigm based on the exposure to complex stimulations, constitutes an efficient means to investigate the effects of high-level experience on behavior, cognitive processes, and neurobiological correlates, as the BDNF expression. In fact, BDNF exerts a key role in mediating and promoting EE-induced plastic changes and functional improvements in healthy and pathological conditions. This review is specifically aimed at providing an updated framework of the available evidence on the EE effects on brain and serum BDNF levels, by taking into account both changes in protein expression and regulation of gene expression. A further purpose of the present review is analyzing the potential of BDNF regulation in coping with neurodegenerative processes characterizing Alzheimer's disease (AD), given BDNF expression alterations are described in AD patients. Moreover, attention is also paid to EE effects on BDNF expression in other neurodegenerative disease. To investigate such a topic, evidence provided by experimental studies is considered. A deeper understanding of environmental ability in modulating BDNF expression in the brain may be fundamental in designing more tuned and effective applications of complex environmental stimulations as managing approaches to AD.
Collapse
Affiliation(s)
- Debora Cutuli
- IRCCS Fondazione Santa Lucia, Rome, Italy.,Department of Psychology, University Sapienza of Rome, Rome, Italy
| | - Eugenia Landolfo
- IRCCS Fondazione Santa Lucia, Rome, Italy.,Department of Psychology, University Sapienza of Rome, Rome, Italy
| | | | - Francesca Gelfo
- IRCCS Fondazione Santa Lucia, Rome, Italy.,Department of Human Sciences, Guglielmo Marconi University, Rome, Italy
| |
Collapse
|
16
|
Effects of two different early socialization models on social behavior and physiology of suckling piglets. Appl Anim Behav Sci 2021. [DOI: 10.1016/j.applanim.2021.105436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
17
|
Bayat M, Kohlmeier KA, Haghani M, Haghighi AB, Khalili A, Bayat G, Hooshmandi E, Shabani M. Co-treatment of vitamin D supplementation with enriched environment improves synaptic plasticity and spatial learning and memory in aged rats. Psychopharmacology (Berl) 2021; 238:2297-2312. [PMID: 33991198 DOI: 10.1007/s00213-021-05853-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 04/15/2021] [Indexed: 11/26/2022]
Abstract
RATIONALE AND OBJECTIVE Environmental enrichment (EE) has been shown in old rats to improve learning and memory. Vitamin D (VitD) has also been shown to modulate age-related, cognitive dysfunction. As both EE and VitD could work to improve cognition via enhancement of neurotrophic factors, their effects might occlude one another. Therefore, a clinically relevant question is whether noted cognition-promoting effects of EE and VitD can co-occur. METHODS Aged rats were housed for 6 weeks in one of three housing conditions: environmentally enriched (EE), socially enriched (SE), or standard condition (SC). Further, a 4th group was co-treated with VitD supplementation (400 IU kg-1 daily, 6 weeks) under EE conditions (EE + VitD). RESULTS Treatment with VitD and EE housing were associated with higher score on measures of learning and memory and exhibited lower anxiety scores compared to EE alone, SE or SC as assayed in the elevated plus maze, Morris water maze, passive avoidance, and open field tasks. Additionally, in the EE + VitD group, mRNA expression levels of NGF, TrkA, BDNF, Nrf2, and IGF-1 were significantly higher compared to expression seen in the EE group. Furthermore, field potential recordings showed that EE + VitD resulted in a greater enhancement of hippocampal LTP and neuronal excitability when compared to EE alone. CONCLUSIONS These findings demonstrate that in aged rats exposure to EE and VitD results in effects on hippocampal cognitive dysfunction and molecular mechanisms which are greater than effects of EE alone, suggesting potential for synergistic therapeutic effects for management of age-related cognitive decline.
Collapse
Affiliation(s)
- Mahnaz Bayat
- Clinical Neurology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Kristi A Kohlmeier
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Masoud Haghani
- Histomorphometry and Stereology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Azadeh Khalili
- Evidence-Based Phytotherapy and Complementary Medicine Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Gholamreza Bayat
- Department of Physiology and Pharmacology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Etrat Hooshmandi
- Clinical Neurology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Shabani
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
18
|
Seo MK, Choi AJ, Seog DH, Lee JG, Park SW. Early Enriched Environment Prevents Epigenetic p11 Gene Changes Induced by Adulthood Stress in Mice. Int J Mol Sci 2021; 22:ijms22041928. [PMID: 33672075 PMCID: PMC7919643 DOI: 10.3390/ijms22041928] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/09/2021] [Accepted: 02/12/2021] [Indexed: 12/21/2022] Open
Abstract
Positive experiences in early life may improve the capacity to cope with adulthood stress through epigenetic modification. We investigated whether an enriched environment (EE) in the postnatal period affected epigenetic changes in the p11 gene induced by chronic unpredictable stress (CUS) in adult C57BL/6J mice. EE was introduced for 5 weeks during postnatal days 21–55. After EE, the mice were subjected to CUS for 4 weeks. EE prevented depression-like behavior induced by adult CUS. EE prevented a decrease in p11 mRNA and histone H3 acetylation induced by CUS, with changes in the expression of histone deacetylase 5. Moreover, EE prevented changes in trimethylation of histone H3 lysine 4 (H3K4) and H3K27 induced by CUS. Furthermore, EE had positive effects on behavior and epigenetic alterations in adult mice without CUS. These results suggest that one of the underlying mechanisms of early-life EE may involve epigenetic modification of the hippocampal p11 gene promoter.
Collapse
Affiliation(s)
- Mi Kyoung Seo
- Paik Institute for Clinical Research, Inje University, Busan 47392, Korea;
| | | | - Dae-Hyun Seog
- Department of Biochemistry, College of Medicine, Inje University, Busan 47392, Korea;
- Dementia and Neurodegenerative Disease Research Center, Inje University, Busan 47392, Korea
| | - Jung Goo Lee
- Paik Institute for Clinical Research, Inje University, Busan 47392, Korea;
- Department of Psychiatry, College of Medicine, Haeundae Paik Hospital, Inje University, Busan 48108, Korea
- Department of Health Science and Technology, Graduate School, Inje University, Busan 47392, Korea
- Correspondence: (J.G.L.); (S.W.P.); Tel.: +82-51-797-3300 (J.G.L.); +82-51-890-6071 (S.W.P.); Fax: +82-51-894-6709 (J.G.L. & S.W.P.)
| | - Sung Woo Park
- Paik Institute for Clinical Research, Inje University, Busan 47392, Korea;
- Department of Health Science and Technology, Graduate School, Inje University, Busan 47392, Korea
- Department of Convergence Biomedical Science, College of Medicine, Inje University, Busan 47392, Korea
- Correspondence: (J.G.L.); (S.W.P.); Tel.: +82-51-797-3300 (J.G.L.); +82-51-890-6071 (S.W.P.); Fax: +82-51-894-6709 (J.G.L. & S.W.P.)
| |
Collapse
|
19
|
Intervention of Brain-Derived Neurotrophic Factor and Other Neurotrophins in Adult Neurogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1331:95-115. [PMID: 34453295 DOI: 10.1007/978-3-030-74046-7_8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cell survival during adult neurogenesis and the modulation of each step, namely, proliferation, lineage differentiation, migration, maturation, and functional integration of the newborn cells into the existing circuitry, is regulated by intrinsic and extrinsic factors. Transduction of extracellular niche signals triggers the activation of intracellular mechanisms that regulate adult neurogenesis by affecting gene expression. While the intrinsic factors include transcription factors and epigenetic regulators, the extrinsic factors are molecular signals that are present in the neurogenic niche microenvironment. These include morphogens, growth factors, neurotransmitters, and signaling molecules secreted as soluble factors or associated to the extracellular matrix. Among these molecular mechanisms are neurotrophins and neurotrophin receptors which have been implicated in the regulation of adult neurogenesis at different levels, with brain-derived neurotrophic factor (BDNF) being the most studied neurotrophin. In this chapter, we review the current knowledge about the role of neurotrophins in the regulation of adult neurogenesis in both the subventricular zone (SVZ) and the hippocampal subgranular zone (SGZ).
Collapse
|
20
|
Veschsanit N, Yang JL, Ngampramuan S, Viwatpinyo K, Pinyomahakul J, Lwin T, Chancharoen P, Rungruang S, Govitrapong P, Mukda S. Melatonin reverts methamphetamine-induced learning and memory impairments and hippocampal alterations in mice. Life Sci 2020; 265:118844. [PMID: 33278389 DOI: 10.1016/j.lfs.2020.118844] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 11/26/2020] [Accepted: 11/27/2020] [Indexed: 02/07/2023]
Abstract
AIMS Methamphetamine (METH) has become a major public health problem because of its abuse and profound neurotoxic effects, causing alterations in brain structure and function, and impairing cognitive functions, including attention, decision making, emotional memory, and working memory. This study aimed to determine whether melatonin (MEL), the circadian-control hormone, which has roles beyond circadian rhythm regulation, could restore METH-induced cognitive and neuronal impairment. MAIN METHODS Mice were treated with either METH (1 mg/kg) or saline for 7 days, followed by MEL (10 mg/kg) or saline for another 14 days. The Morris water maze (MWM) test was performed one day after the last saline or MEL injection. The hippocampal neuronal density, synaptic density, and receptors involved in learning and memory, along with downstream signaling molecules (NMDA receptor subunits GluN2A, GluN2B, and CaMKII) were investigated by immunoblotting. KEY FINDINGS METH administration significantly extended escape latency in learning phase and reduced the number of target crossings in memory test-phase as well as decreased the expression of BDNF, NMDA receptors, TrkB receptors, CaMKII, βIII tubulin, and synaptophysin. MEL treatment significantly ameliorated METH-induced increased escape latency, decreased the number of target crossings and decreased expression of BDNF, NMDA receptors, TrkB receptors, CaMKII, βIII tubulin and synaptophysin. SIGNIFICANCE METH administration impairs learning and memory in mice, and MEL administration restores METH-induced neuronal impairments which is probably through the changes in BDNF, NMDA receptors, TrkB receptors, CaMKII, βIII tubulin and synaptophysin. Therefore, MEL is potentially an innovative and promising treatment for learning and memory impairment of humans.
Collapse
Affiliation(s)
- Nisarath Veschsanit
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand
| | - Jenq-Lin Yang
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Sukonthar Ngampramuan
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand
| | - Kittikun Viwatpinyo
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand
| | - Jitrapa Pinyomahakul
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand
| | - Thit Lwin
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand; Department of Anatomy, Defence Services Medical Academy, Mingalardon, Yangon 11021, Myanmar
| | - Pongrung Chancharoen
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand; Faculty of Allied Health Sciences, Burapha University, Seansuk, Chonburi 20131, Thailand
| | - Saowalak Rungruang
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Piyarat Govitrapong
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand; Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok 10210, Thailand
| | - Sujira Mukda
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand.
| |
Collapse
|
21
|
Jiang YG, Wang YH, Zhang H, Wang ZY, Liu YQ. Effects of early-life zinc deficiency on learning and memory in offspring and the changes in DNA methylation patterns. Nutr Neurosci 2020; 25:1001-1010. [PMID: 33078688 DOI: 10.1080/1028415x.2020.1831259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To investigate the effect of maternal zinc deficiency on learning and memory in offspring and the changes in DNA methylation patterns. METHODS Pregnant rats were divided into zinc adequate (ZA), zinc deficient (ZD), and paired fed (PF) groups. Serum zinc contents and AKP activity in mother rats and offspring at P21 (end of lactation) and P60 (weaned, adult) were detected. Cognitive ability of offspring at P21 and P60 were determined by Morris water maze. The expression of proteins including DNMT3a, DNMT1, GADD45β, MeCP2 and BDNF in the offspring hippocampus were detected by Western-blot. The methylation status of BDNF promoter region in hippocampus of offspring rats was detected by MS-qPCR. RESULTS Compared with the ZA and PF groups, pups in the ZD group had lower zinc levels and AKP activity in the serum, spent more time finding the platform and spent less time going through the platform area. Protein expression of DNMT1 and GADD45b were downregulated in the ZD group during P0 and P21 but not P60 compared with the ZA and PF group, these results were consistent with a reduction in BDNF protein at P0 (neonate), P21. However, when pups of rats in the ZD group were supplemented with zinc ion from P21 to P60, MeCP2 and GADD45b expression were significantly downregulated compared with the ZA and PF group. CONCLUSION Post-weaning zinc supplementation may improve cognitive impairment induced by early life zinc deficiency, whereas it may not completely reverse the abnormal expression of particular genes that are involved in DNA methylation, binding to methylated DNA and neurogenesis.
Collapse
Affiliation(s)
- Yu-Gang Jiang
- Department of Nutrition, Tianjin Institute of Environmental & Operational Medicine, Tianjin, People's Republic of China
| | - Yong-Hui Wang
- Department of Nutrition, Tianjin Institute of Environmental & Operational Medicine, Tianjin, People's Republic of China
| | - Han Zhang
- Department of Nutrition, Tianjin Institute of Environmental & Operational Medicine, Tianjin, People's Republic of China.,College of Public Health, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Zi-Yu Wang
- Department of Nutrition, Tianjin Institute of Environmental & Operational Medicine, Tianjin, People's Republic of China
| | - Yan-Qiang Liu
- College of Life Sciences, Nan Kai University, Tianjin, People's Republic of China
| |
Collapse
|
22
|
Minutillo A, Panza G, Mauri MC. Musical practice and BDNF plasma levels as a potential marker of synaptic plasticity: an instrument of rehabilitative processes. Neurol Sci 2020; 42:1861-1867. [PMID: 32940801 PMCID: PMC8043880 DOI: 10.1007/s10072-020-04715-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/08/2020] [Indexed: 01/06/2023]
Abstract
Background and objectives The aim of the study was to investigate the influence of musical practice on brain plasticity. BDNF (brain-derived neurotrophic factor) is a neurotrophin involved in neuroplasticity and synaptic function. Materials and methods We recruited 48 healthy subjects of equal age and sex (21 musicians and 27 non-musicians). All subjects were administered the AQ (Autism-Spectrum Questionnaire) and plasma levels (PLs) of BDNF, oxytocin (OT), and vasopressin (VP) were measured in the blood sample of every participant. Results. The difference between BDNF PLs in the two groups was found to be statistically significant (t = − 2.214, p = 0.03). Furthermore, oxytocin (OT) PLs and musical practice were found to be independent positive predictors of BDNF PLs (p < 0.04). We also found a negative correlation between BDNF PLs and AD (attention to detail) sub-scale score of AQ throughout the whole sample. Assuming BDNF PLs to be a marker of synaptic plasticity, higher PLs could be associated with the activation of alternative neural pathways: a lower score in the “attention to detail” sub-scale could imply greater flexibility of higher cerebral functions among musicians. Further researches should be conducted to assess the rehabilitative usefulness of these findings among patients affected by psychiatric disorders.
Collapse
Affiliation(s)
- Alessandro Minutillo
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy. .,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.
| | - Gabriele Panza
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Massimo Carlo Mauri
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
23
|
Pan B, Zhou Y, Li H, Li Y, Xue X, Li L, Liu Q, Zhao X, Niu Q. Relationship between occupational aluminium exposure and histone lysine modification through methylation. J Trace Elem Med Biol 2020; 61:126551. [PMID: 32470791 DOI: 10.1016/j.jtemb.2020.126551] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 04/26/2020] [Accepted: 05/11/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND Aluminium is an environmental neurotoxin to which human beings are extensively exposed. However, the molecular mechanism of aluminium toxicity remains unclear. METHODS The changes in cognitive function of aluminum exposed workers under long-term occupational exposure were evaluated, and the relationship between cognitive changes, plasma memory related BDNF and EGR1 protein expression, and variations of epigenetic markers H3K4me3, H3K9me2, H3K27me3 expression levels in blood was explored. RESULTS MMSE, DSFT, DST scores in cognitive function and the levels of plasma BDNF and EGR1 protein expression decreased with the increase of blood aluminum level. H3K4me3, H3K9me2, H3K27me3 expression levels in peripheral blood lymphocytes of aluminum exposed workers were statistically different (all P<0.05). H3K4me3, H3K9me2 and H3K27me3 expression levels in lymphocytes were correlated with blood aluminum level. BDNF, EGR1 protein level and H3K4me3, H3K9me2, H3K27me3 expression levels have different degrees of correlation. There was a linear regression relationship between plasma BDNF, H3K4me3 and H3K9me2. H3K9me2 had a greater effect on BDNF than H3K4me3. There is a linear regression relationship between EGR1, H3K4me3 and H3K27me3, and the influence of H3K4me3 on EGR1 is greater than that of H3K27me3 on EGR1. CONCLUSION Alummnum may regulate the expression of BDNF and EGR1 by regulating H3K4me3, H3K27me3 and H3K9me2, and affect the cognitive function of workers by affecting the expression of BDNF and EGR1.
Collapse
Affiliation(s)
- Baolong Pan
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Sixth Hospital of Shanxi Medical University (General Hospital of Tisco), China
| | - Yue Zhou
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China
| | - Huan Li
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China; Key Lab of Cellular Physiology of Education Ministry, Shanxi Medical University, China
| | - Yaqin Li
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China
| | - Xingli Xue
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China
| | - Liang Li
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China
| | - Qun Liu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China
| | - Xiaoyan Zhao
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China
| | - Qiao Niu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China; Key Lab of Cellular Physiology of Education Ministry, Shanxi Medical University, China.
| |
Collapse
|
24
|
Poon CH, Heng BC, Lim LW. New insights on brain-derived neurotrophic factor epigenetics: from depression to memory extinction. Ann N Y Acad Sci 2020; 1484:9-31. [PMID: 32808327 DOI: 10.1111/nyas.14458] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/21/2020] [Accepted: 07/14/2020] [Indexed: 12/14/2022]
Abstract
Advances in characterizing molecular profiles provide valuable insights and opportunities for deciphering the neuropathology of depression. Although abnormal brain-derived neurotrophic factor (BDNF) expression in depression has gained much support from preclinical and clinical research, how it mediates behavioral alterations in the depressed state remains largely obscure. Environmental factors contribute significantly to the onset of depression and produce robust epigenetic changes. Epigenetic regulation of BDNF, as one of the most characterized gene loci in epigenetics, has recently emerged as a target in research on memory and psychiatric disorders. Specifically, epigenetic alterations of BDNF exons are heavily involved in mediating memory functions and antidepressant effects. In this review, we discuss key research on stress-induced depression from both preclinical and clinical studies, which revealed that differential epigenetic regulation of specific BDNF exons is associated with depression pathophysiology. Considering that BDNF has a central role in depression, we argue that memory extinction, an adaptive response to fear exposure, is dependent on BDNF modulation and holds promise as a prospective target for alleviating or treating depression and anxiety disorders.
Collapse
Affiliation(s)
- Chi Him Poon
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Boon Chin Heng
- Peking University School of Stomatology, Beijing, China.,Department of Biological Sciences, Sunway University, Selangor, Malaysia
| | - Lee Wei Lim
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,Department of Biological Sciences, Sunway University, Selangor, Malaysia
| |
Collapse
|
25
|
Luo L, Reimert I, Middelkoop A, Kemp B, Bolhuis JE. Effects of Early and Current Environmental Enrichment on Behavior and Growth in Pigs. Front Vet Sci 2020; 7:268. [PMID: 32582773 PMCID: PMC7287207 DOI: 10.3389/fvets.2020.00268] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 04/21/2020] [Indexed: 02/06/2023] Open
Abstract
Enriched environments are known to beneficially affect the behavior of pigs, as compared with barren pens. The influence of enrichment may, however, depend on pigs' early life housing experiences. The aim of this study was to investigate the long-term effects of early and later life environmental enrichment on behavior and growth in pigs with different coping styles. Pigs were housed in either barren pens or in larger pens enriched with rooting substrates from birth, and half of them experienced a housing switch, i.e., a loss or gain of enrichment, at 7 weeks of age, creating four treatment groups. Home pen behavior and body weight were recorded until 19 weeks of age. Pigs were classified as reactive or proactive based on a backtest at 2 weeks of age. Enrichment increased time spent exploring, chewing, and play and decreased oral manipulation of penmates and pen-directed exploring and chewing. Behavior of pigs that switched from barren to enriched pens or vice versa reflected not only their actual environment, but also their early life housing. As early and later life enrichment affected most behaviors in opposite directions, effects of enrichment, or lack thereof, after the switch were more pronounced in pigs that had experienced a different early life condition. For instance, pigs experiencing an upgrade from barren to enriched pens seemed to "catch-up" by showing more exploration and play. Conversely, pigs exposed to a downgrade displayed more oral manipulation of penmates than ones kept barren throughout, which particularly held for pigs with a reactive coping style. Effects of early life and current housing on several other behaviors depended on coping style too. Pigs housed in enriched conditions appeared better able to cope with weaning than barren housed pigs, as they gained more weight and had higher feed intake post-weaning. Barren housed pigs had a lower body weight than enriched pigs just before the switch, after which growth was mainly determined by actual housing, with enriched kept pigs having a higher feed intake and body weight. Thus, not only current housing conditions, but also a (mis)match with the early life environment may affect behavior and growth of pigs.
Collapse
Affiliation(s)
- Lu Luo
- Adaptation Physiology Group, Department of Animal Sciences, Wageningen University & Research, Wageningen, Netherlands
| | - Inonge Reimert
- Adaptation Physiology Group, Department of Animal Sciences, Wageningen University & Research, Wageningen, Netherlands
| | - Anouschka Middelkoop
- Adaptation Physiology Group, Department of Animal Sciences, Wageningen University & Research, Wageningen, Netherlands
| | - Bas Kemp
- Adaptation Physiology Group, Department of Animal Sciences, Wageningen University & Research, Wageningen, Netherlands
| | - J Elizabeth Bolhuis
- Adaptation Physiology Group, Department of Animal Sciences, Wageningen University & Research, Wageningen, Netherlands
| |
Collapse
|
26
|
How the enriched get richer? Experience-dependent modulation of microRNAs and the therapeutic effects of environmental enrichment. Pharmacol Biochem Behav 2020; 195:172940. [PMID: 32413435 DOI: 10.1016/j.pbb.2020.172940] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/23/2020] [Accepted: 05/01/2020] [Indexed: 11/20/2022]
Abstract
Environmental enrichment and physical exercise have many well-established health benefits. Although these environmental manipulations are known to delay symptom onset and progression in a variety of neurological and psychiatric conditions, the mechanisms underlying these effects remain poorly understood. A notable candidate molecular mechanism is that of microRNA, a family of small noncoding RNAs that are important regulators of gene expression. Research investigating the many diverse roles of microRNAs has greatly expanded over the past decade, with several promising preclinical and clinical studies highlighting the role of dysregulated microRNA expression (in the brain, blood and other peripheral systems) in understanding the aetiology of disease. Altered microRNA levels have also been described following environmental interventions such as exercise and environmental enrichment in non-clinical populations and wild-type animals, as well as in some brain disorders and associated preclinical models. Recent studies exploring the effects of stimulating environments on microRNA levels in the brain have revealed an array of changes that are likely to have important downstream effects on gene expression, and thus may regulate a variety of cellular processes. Here we review literature that explores the differential expression of microRNAs in rodents following environmental enrichment and exercise, in both healthy control animals and preclinical models of relevance to neurological and psychiatric disorders.
Collapse
|
27
|
Rojas-Carvajal M, Sequeira-Cordero A, Brenes JC. Neurobehavioral Effects of Restricted and Unpredictable Environmental Enrichment in Rats. Front Pharmacol 2020; 11:674. [PMID: 32477137 PMCID: PMC7235364 DOI: 10.3389/fphar.2020.00674] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 04/24/2020] [Indexed: 12/21/2022] Open
Abstract
To study how motivational factors modulate experience-dependent neurobehavioral plasticity, we modify a protocol of environmental enrichment (EE) in rats. We assumed that the benefits derived from EE might vary according to the level of incentive salience attributed to it. To enhance the rewarding properties of EE, access to the EE cage varied randomly from 2 to 48 h for 30 days (REE). The REE group was enriched only 50% of the time and was compared to standard housing and continuous EE (CEE) groups. As behavioral readout, we analyzed the spontaneous activity and the ultrasonic vocalizations (USVs) within the EE cage weekly, and in the open field test at the end of the experiment. In the cage, REE increased the utilization of materials, physical activity, and the rate of appetitive USVs. In the OF, the CEE-induced enhancements in novelty habituation and social signaling were equaled by the REE. At the neural level, we measured the expression of genes related to neural plasticity and epigenetic regulations in different brain regions. In the dorsal striatum and hippocampus, REE upregulated the expression of the brain-derived neurotrophic factor, its tropomyosin kinase B receptor, and the DNA methyltransferase 3A. Altogether, our results suggest that the higher activity within the cage and the augmented incentive motivation provoked by the REE boosted its neurobehavioral effects equaling or surpassing those observed in the CEE condition. As constant exposures to treatments or stimulating environments are virtually impossible for humans, restricted EE protocols would have greater translational value than traditional ones.
Collapse
Affiliation(s)
- Mijail Rojas-Carvajal
- Neuroscience Research Center, University of Costa Rica, San Pedro, Costa Rica.,Institute for Psychological Research, University of Costa Rica, San Pedro, Costa Rica
| | - Andrey Sequeira-Cordero
- Neuroscience Research Center, University of Costa Rica, San Pedro, Costa Rica.,Institute for Health Research, University of Costa Rica, San Pedro, Costa Rica
| | - Juan C Brenes
- Neuroscience Research Center, University of Costa Rica, San Pedro, Costa Rica.,Institute for Psychological Research, University of Costa Rica, San Pedro, Costa Rica
| |
Collapse
|
28
|
Wu T, Sun XY, Yang X, Liu L, Tong K, Gao Y, Hao JR, Cao J, Gao C. Histone H3K9 Trimethylation Downregulates the Expression of Brain-Derived Neurotrophic Factor in the Dorsal Hippocampus and Impairs Memory Formation During Anaesthesia and Surgery. Front Mol Neurosci 2019; 12:246. [PMID: 31708739 PMCID: PMC6823536 DOI: 10.3389/fnmol.2019.00246] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 09/24/2019] [Indexed: 01/01/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is essential for cognitive and memory functions. Abnormal BDNF expression in the central nervous system may impair these functions. Anaesthesia and surgery can induce perioperative neurocognitive disorders (PND). Clinical studies show that BDNF expression is decreased in patients presenting with cognitive impairment after anaesthesia and surgery. However, the molecular mechanism is still unclear. Epigenetic regulation plays an important role in cognition. The hypermethylation of H3K9 is crucial for transcriptional silencing and the onset of cognitive disorders. Here, we hypothesised that H3K9 trimethylation repressed BDNF expression and impaired memory formation or recall during anaesthesia and surgery. Laparotomy under isoflurane inhalation anaesthesia, behavioural tests, Western blotting, quantitative real-time reverse-transcription polymerase chain reaction (qRT-PCR), chromatin immunoprecipitation (ChIP), and immunohistochemistry were used in this study. BDNF expression was decreased in the hippocampus after anaesthesia and surgery. Cognitive impairment affected memory formation but not recall. The trimethylation of H3K9 downregulated BDNF expression. The overexpression of BDNF or use of exogenous BDNF improved the impairment of memory formation caused by anaesthesia and surgery. Therefore, inhibiting H3K9 trimethylation and increasing the expression of BDNF may help prevent PND in the clinical setting.
Collapse
Affiliation(s)
- Tong Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, Xuzhou, China.,Department of Anesthesia, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiao-Yu Sun
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, Xuzhou, China
| | - Xiu Yang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, Xuzhou, China
| | - Le Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, Xuzhou, China
| | - Kun Tong
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, Xuzhou, China
| | - Ya Gao
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, Xuzhou, China
| | - Jing-Ru Hao
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, Xuzhou, China
| | - Jing Cao
- Department of Anesthesia, Xuzhou Central Hospital, The Xuzhou Clinical College of Xuzhou Medical University, Xuzhou, China
| | - Can Gao
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
29
|
Fusco S, Spinelli M, Cocco S, Ripoli C, Mastrodonato A, Natale F, Rinaudo M, Livrizzi G, Grassi C. Maternal insulin resistance multigenerationally impairs synaptic plasticity and memory via gametic mechanisms. Nat Commun 2019; 10:4799. [PMID: 31641124 PMCID: PMC6805915 DOI: 10.1038/s41467-019-12793-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 09/27/2019] [Indexed: 12/22/2022] Open
Abstract
Metabolic diseases harm brain health and cognitive functions, but whether maternal metabolic unbalance may affect brain plasticity of next generations is still unclear. Here, we demonstrate that maternal high fat diet (HFD)-dependent insulin resistance multigenerationally impairs synaptic plasticity, learning and memory. HFD downregulates BDNF and insulin signaling in maternal tissues and epigenetically inhibits BDNF expression in both germline and hippocampus of progeny. Notably, exposure of the HFD offspring to novel enriched environment restores Bdnf epigenetic activation in the male germline and counteracts the transmission of cognitive impairment to the next generations. BDNF administration to HFD-fed mothers or preserved insulin sensitivity in HFD-fed p66Shc KO mice also prevents the intergenerational transmission of brain damage to the progeny. Collectively, our data suggest that maternal diet multigenerationally impacts on descendants' brain health via gametic mechanisms susceptible to lifestyle.
Collapse
Affiliation(s)
- Salvatore Fusco
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy. .,Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy.
| | - Matteo Spinelli
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Sara Cocco
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Cristian Ripoli
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy.,Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Alessia Mastrodonato
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Francesca Natale
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Marco Rinaudo
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Giulia Livrizzi
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Claudio Grassi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy. .,Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy.
| |
Collapse
|
30
|
Keller SM, Doherty TS, Roth TL. Pharmacological manipulation of DNA methylation normalizes maternal behavior, DNA methylation, and gene expression in dams with a history of maltreatment. Sci Rep 2019; 9:10253. [PMID: 31311968 PMCID: PMC6635500 DOI: 10.1038/s41598-019-46539-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 06/25/2019] [Indexed: 01/02/2023] Open
Abstract
The quality of parental care received during development profoundly influences an individual's phenotype, including that of maternal behavior. We previously found that female rats with a history of maltreatment during infancy mistreat their own offspring. One proposed mechanism through which early-life experiences influence behavior is via epigenetic modifications. Indeed, our lab has identified a number of brain epigenetic alterations in female rats with a history of maltreatment. Here we sought to investigate the role of DNA methylation in aberrant maternal behavior. We administered zebularine, a drug known to alter DNA methylation, to dams exposed during infancy to the scarcity-adversity model of low nesting resources, and then characterized the quality of their care towards their offspring. First, we replicate that dams with a history of maltreatment mistreat their own offspring. Second, we show that maltreated-dams treated with zebularine exhibit lower levels of adverse care toward their offspring. Third, we show that administration of zebularine in control dams (history of nurturing care) enhances levels of adverse care. Lastly, we show altered methylation and gene expression in maltreated dams normalized by zebularine. These findings lend support to the hypothesis that epigenetic alterations resulting from maltreatment causally relate to behavioral outcomes.
Collapse
Affiliation(s)
- Samantha M Keller
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, 19716, USA
| | - Tiffany S Doherty
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, 19716, USA
| | - Tania L Roth
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, 19716, USA.
| |
Collapse
|
31
|
Giacobbo BL, de Freitas BS, Vedovelli K, Schlemmer LM, Pires VN, Antoniazzi V, Santos CDSD, Paludo L, Borges JV, de Lima DB, Schröder N, de Vries EFJ, Bromberg E. Long-term environmental modifications affect BDNF concentrations in rat hippocampus, but not in serum. Behav Brain Res 2019; 372:111965. [PMID: 31125621 DOI: 10.1016/j.bbr.2019.111965] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/19/2019] [Accepted: 05/20/2019] [Indexed: 01/01/2023]
Abstract
The role of mBDNF on the beneficial effects of cognitive stimulation on the brain remains controversial, as well as the potential of peripheral mBDNF as a biomarker of environmental effects on its central status. We investigated the effect of different environmental conditions on recognition memory, proBDNF, mBDNF and synaptophysin levels in the hippocampus, and on mBDNF levels in blood. Male Wistar rats (6 and 17 months-old) were assigned to cognitively enriched (EE), standard (SE) and impoverished (IE) environmental conditions for twelve weeks. Novel object recognition was performed at week 10. When the animals were 9 and 20-months old, hippocampus was collected for mBDNF, proBDNF and synaptophysin analysis; serum was analyzed for mBDNF levels. The cognitively EE improved recognition memory, resulted in a trend to increased hippocampal mBDNF and augmented synaptophysin levels. Accordingly, hippocampal mBDNF, proBDNF and synaptophysin were significantly higher in EE than IE animals. Hippocampal mBDNF was positively correlated to proBDNF, cellular and behavioral plasticity markers. No effect of age was seen on the studied variables. Moreover, no significant effects of EE or IE on serum mBDNF were observed. Serum mBDNF also failed to correlate with hippocampal mBDNF, proBDNF and with the cellular and behavioral plasticity markers. These findings indicate that mBDNF is involved in neuronal and behavioral plasticity mechanisms induced by cognitively enriched environments, and that peripheral mBDNF may not always be a reliable biomarker of the effects of environmental settings on central mBDNF and plasticity, which is of special interest from a translational research perspective.
Collapse
Affiliation(s)
- Bruno Lima Giacobbo
- Laboratory of Biology and Development of the Nervous System, School of Sciences, Pontifical Catholic University of Rio Grande do Sul, Ipiranga Av. 6681, 90619-900, Porto Alegre, Brazil; Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 31.001, 9713 GZ, Groningen, the Netherlands
| | - Betânia Souza de Freitas
- Laboratory of Biology and Development of the Nervous System, School of Sciences, Pontifical Catholic University of Rio Grande do Sul, Ipiranga Av. 6681, 90619-900, Porto Alegre, Brazil
| | - Kelem Vedovelli
- Laboratory of Biology and Development of the Nervous System, School of Sciences, Pontifical Catholic University of Rio Grande do Sul, Ipiranga Av. 6681, 90619-900, Porto Alegre, Brazil; Institute of Geriatrics and Gerontology, School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Ipiranga Av. 6690, 90610-000, Porto Alegre, Brazil
| | - Lívia Machado Schlemmer
- Laboratory of Biology and Development of the Nervous System, School of Sciences, Pontifical Catholic University of Rio Grande do Sul, Ipiranga Av. 6681, 90619-900, Porto Alegre, Brazil
| | - Vivian Naziaseno Pires
- Laboratory of Biology and Development of the Nervous System, School of Sciences, Pontifical Catholic University of Rio Grande do Sul, Ipiranga Av. 6681, 90619-900, Porto Alegre, Brazil
| | - Vinicius Antoniazzi
- Laboratory of Biology and Development of the Nervous System, School of Sciences, Pontifical Catholic University of Rio Grande do Sul, Ipiranga Av. 6681, 90619-900, Porto Alegre, Brazil
| | - Cristophod de Souza Dos Santos
- Laboratory of Biology and Development of the Nervous System, School of Sciences, Pontifical Catholic University of Rio Grande do Sul, Ipiranga Av. 6681, 90619-900, Porto Alegre, Brazil
| | - Leticia Paludo
- Laboratory of Biology and Development of the Nervous System, School of Sciences, Pontifical Catholic University of Rio Grande do Sul, Ipiranga Av. 6681, 90619-900, Porto Alegre, Brazil
| | - Juliano Viana Borges
- Laboratory of Biology and Development of the Nervous System, School of Sciences, Pontifical Catholic University of Rio Grande do Sul, Ipiranga Av. 6681, 90619-900, Porto Alegre, Brazil
| | - Daiane Borba de Lima
- Laboratory of Biology and Development of the Nervous System, School of Sciences, Pontifical Catholic University of Rio Grande do Sul, Ipiranga Av. 6681, 90619-900, Porto Alegre, Brazil
| | - Nadja Schröder
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil; Department of Physiology, Institute for Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Sarmento Leite, 500, 90050-170 Porto Alegre, Brazil
| | - Erik F J de Vries
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 31.001, 9713 GZ, Groningen, the Netherlands
| | - Elke Bromberg
- Laboratory of Biology and Development of the Nervous System, School of Sciences, Pontifical Catholic University of Rio Grande do Sul, Ipiranga Av. 6681, 90619-900, Porto Alegre, Brazil; Institute of Geriatrics and Gerontology, School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Ipiranga Av. 6690, 90610-000, Porto Alegre, Brazil; National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil.
| |
Collapse
|
32
|
Kathpalia P, Nag TC, Chattopadhyay P, Sharma A, Bhat MA, Roy TS, Wadhwa S. In ovo Sound Stimulation Mediated Regulation of BDNF in the Auditory Cortex and Hippocampus of Neonatal Chicks. Neuroscience 2019; 408:293-307. [PMID: 31026564 DOI: 10.1016/j.neuroscience.2019.04.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/15/2019] [Accepted: 04/07/2019] [Indexed: 12/22/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is known to mediate activity-dependent changes in the developing auditory system. Its expression in the brainstem auditory nuclei, auditory cortex and hippocampus of neonatal chicks (Gallus gallus domesticus) in response to in ovo high intensity sound exposure at 110 dB (arrhythmic sound: recorded traffic noise, 30-3000 Hz with peak at 2700 Hz, rhythmic sound: sitar music, 100-4000 Hz) was examined to understand the previously reported altered volume and neuronal number in these regions. In the brainstem auditory nuclei, no mature BDNF, but proBDNF at the protein level was detected, and no change in its levels was observed after in ovo sound stimulation (music and noise). Increased ProBDNF protein levels were found in the auditory cortex in response to arrhythmic sound, along with decreased levels of one of the BDNF mRNA transcripts, in response to both rhythmic and arrhythmic sound stimulation. In the hippocampus, increased levels of mature BDNF were found in response to music. Expression microarray analysis was performed to understand changes in gene expression in the hippocampus in response to music and noise, followed by gene ontology analysis showing enrichment of probable signaling pathways. Differentially expressed genes like CAMK1 and STAT1 were found to be involved in downstream signaling on comparing music versus noise-exposed chicks. In conclusion, we report that BDNF is differentially regulated in the auditory cortex at the transcriptional and post-translational level, and in the hippocampus at the post-translational level in response to in ovo sound stimulation.
Collapse
Affiliation(s)
- Poorti Kathpalia
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Tapas Chandra Nag
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India.
| | | | - Arundhati Sharma
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Muzaffer Ahmed Bhat
- Department of Physiology, All India Institute of Medical Sciences, New Delhi, India
| | - Tara Sankar Roy
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Shashi Wadhwa
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India; Department of Anatomy, North Delhi Municipal Medical College, New Delhi, India
| |
Collapse
|
33
|
Rossetti MF, Schumacher R, Lazzarino GP, Gomez AL, Varayoud J, Ramos JG. The impact of sensory and motor enrichment on the epigenetic control of steroidogenic-related genes in rat hippocampus. Mol Cell Endocrinol 2019; 485:44-53. [PMID: 30721712 DOI: 10.1016/j.mce.2019.01.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 01/15/2019] [Accepted: 01/29/2019] [Indexed: 02/07/2023]
Abstract
In the present study, we analyzed the effects of a short-term environmental enrichment on the mRNA expression and DNA methylation of steroidogenic enzymes in the hippocampus. Thus, young adult (80-day-old) and middle-aged (350-day-old) Wistar female rats were exposed to sensory (SE) or motor (ME) enrichment during 10 days and compared to animals housed under standard conditions. SE was provided by an assortment of objects that included plastic tubes and toys; for ME, rodent wheels were provided. In young adult animals, SE and ME increased the mRNA expression of cytochrome P450 17α-hydroxylase/c17,20-lyase, steroid 5α-reductase type 1 (5αR-1) and 3α-hydroxysteroid dehydrogenase and decreased the methylation levels of 5αR-1 gene. In middle-aged rats, ME and SE upregulated the gene expression of aldosterone synthase and decreased the methylation state of its promoter. These results propose that SE and ME differentially regulate the transcription of neurosteroidogenic enzymes through epigenetic mechanisms in young and aged rats.
Collapse
Affiliation(s)
- Maria Florencia Rossetti
- Departamento de Bioquímica Clínica y Cuantitativa, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Instituto de Salud y Ambiente del Litoral(ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral-CONICET, Santa Fe, Argentina.
| | - Rocio Schumacher
- Instituto de Salud y Ambiente del Litoral(ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral-CONICET, Santa Fe, Argentina.
| | - Gisela Paola Lazzarino
- Instituto de Salud y Ambiente del Litoral(ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral-CONICET, Santa Fe, Argentina.
| | - Ayelen Luciana Gomez
- Instituto de Salud y Ambiente del Litoral(ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral-CONICET, Santa Fe, Argentina; Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina.
| | - Jorgelina Varayoud
- Instituto de Salud y Ambiente del Litoral(ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral-CONICET, Santa Fe, Argentina; Cátedra de Fisiología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina.
| | - Jorge Guillermo Ramos
- Departamento de Bioquímica Clínica y Cuantitativa, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Instituto de Salud y Ambiente del Litoral(ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral-CONICET, Santa Fe, Argentina.
| |
Collapse
|
34
|
Abstract
Our social environment, from the microscopic to the macro-social, affects us for the entirety of our lives. One integral line of research to examine how interpersonal and societal environments can get "under the skin" is through the lens of epigenetics. Epigenetic mechanisms are adaptations made to our genome in response to our environment which include tags placed on and removed from the DNA itself to how our DNA is packaged, affecting how our genes are read, transcribed, and interact. These tags are affected by social environments and can persist over time; this may aid us in responding to experiences and exposures, both the enriched and the disadvantageous. From memory formation to immune function, the experience-dependent plasticity of epigenetic modifications to micro- and macro-social environments may contribute to the process of learning from comfort, pain, and stress to better survive in whatever circumstances life has in store.
Collapse
Affiliation(s)
- Sarah M Merrill
- Centre for Molecular Medicine and Therapeutics, British Columbia Children's Hospital, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Nicole Gladish
- Centre for Molecular Medicine and Therapeutics, British Columbia Children's Hospital, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Michael S Kobor
- Centre for Molecular Medicine and Therapeutics, British Columbia Children's Hospital, Vancouver, BC, Canada.
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.
- Human Early Learning Partnership, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
35
|
Wearick-Silva LE, Orso R, Martins LA, Creutzberg KC, Centeno-Silva A, Xavier LL, Grassi-Oliveira R, Mestriner RG. Dual influences of early life stress induced by limited bedding on walking adaptability and Bdnf/TrkB and Drd1/Drd2 gene expression in different mouse brain regions. Behav Brain Res 2018; 359:66-72. [PMID: 30347225 DOI: 10.1016/j.bbr.2018.10.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/10/2018] [Accepted: 10/17/2018] [Indexed: 12/30/2022]
Abstract
Introduction Evidence suggests early life stress impairs development, quality of life and increases vulnerability to disease. One important aspect of the stress experience is its impact on cognitive-motor performance, which includes the ability to adapt walking according to the environmental conditions. This study aimed to investigate how early-life stress affects walking adaptability of mice, while investigating BDNF/TrkB and Drd1/Drd2 expression in different brain regions. Methods Briefly, we exposed male C56BL/6 to the limited bedding protocol (LB) from post-natal day (PND) 2 to PND9 and then tested animals in the ladder walking task at PND60. RT-qPCR was used to investigate gene expression in the mPFC, hippocampus, motor cortex and cerebellum 2 h after the task Results LB induced a wide range of variability and therefore two distinct subgroups of animals within the LB group were established: a) superior performance (LB-SP); and b) inferior performance (LB-IP), compared to controls. Additionally, Drd1 gene expression was increased in the mPFC of LB-IP animals and in the cerebellum of LB-SP animals, while Drd2 expression was reduced in the hippocampus of the LB-IP group. BDNF exon IV gene expression in the mPFC and motor cortex was increased in both the LB-IP and LB-SP subgroups. TrkB gene expression in the hippocampus was reduced in the LB-IP group. A strong negative correlation was found between walking adaptability performance and BDNF exon IV gene expression in the motor cortex. Conversely, a positive correlation was found between walking adaptability performance and TrkB expression in the mPFC and a negative correlation in the hippocampus. Both Drd1 and Drd2 gene expression were negatively correlated with the ability to adapt walking. Conclusions Overall, our findings suggest exposure to early life stress leads to distinct walking adaptability phenotypes, which may be related to Drd1, Drd2, Bdnf exon IV and TrkB gene expression in brain regions that influence walking adaptability.
Collapse
Affiliation(s)
- L E Wearick-Silva
- Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil; Brain Institute of Rio Grande do Sul, Porto Alegre, RS, Brazil; Developmental Cognitive Neuroscience Laboratory, Porto Alegre, RS, Brazil
| | - R Orso
- Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil; Brain Institute of Rio Grande do Sul, Porto Alegre, RS, Brazil; Developmental Cognitive Neuroscience Laboratory, Porto Alegre, RS, Brazil
| | - L A Martins
- Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil; Neurorehabilitation and Neural Repair Research Group, Porto Alegre, RS, Brazil
| | - K C Creutzberg
- Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil; Brain Institute of Rio Grande do Sul, Porto Alegre, RS, Brazil; Developmental Cognitive Neuroscience Laboratory, Porto Alegre, RS, Brazil
| | - A Centeno-Silva
- Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil; Brain Institute of Rio Grande do Sul, Porto Alegre, RS, Brazil; Developmental Cognitive Neuroscience Laboratory, Porto Alegre, RS, Brazil
| | - L L Xavier
- Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil; Neurorehabilitation and Neural Repair Research Group, Porto Alegre, RS, Brazil
| | - R Grassi-Oliveira
- Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil; Brain Institute of Rio Grande do Sul, Porto Alegre, RS, Brazil; Developmental Cognitive Neuroscience Laboratory, Porto Alegre, RS, Brazil
| | - R G Mestriner
- Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil; Neurorehabilitation and Neural Repair Research Group, Porto Alegre, RS, Brazil.
| |
Collapse
|
36
|
How early media exposure may affect cognitive function: A review of results from observations in humans and experiments in mice. Proc Natl Acad Sci U S A 2018; 115:9851-9858. [PMID: 30275319 DOI: 10.1073/pnas.1711548115] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) is now among the most commonly diagnosed chronic psychological dysfunctions of childhood. By varying estimates, it has increased by 30% in the past 20 years. Environmental factors that might explain this increase have been explored. One such factor may be audiovisual media exposure during early childhood. Observational studies in humans have linked exposure to fast-paced television in the first 3 years of life with subsequent attentional deficits in later childhood. Although longitudinal and well controlled, the observational nature of these studies precludes definitive conclusions regarding a causal relationship. As experimental studies in humans are neither ethical nor practical, mouse models of excessive sensory stimulation (ESS) during childhood, akin to the enrichment studies that have previously shown benefits of stimulation in rodents, have been developed. Experimental studies using this model have corroborated that ESS leads to cognitive and behavioral deficits, some of which may be potentially detrimental. Given the ubiquity of media during childhood, these findings in humansand rodents perhaps have important implications for public health.
Collapse
|
37
|
Dong BE, Xue Y, Sakata K. The effect of enriched environment across ages: A study of anhedonia and BDNF gene induction. GENES BRAIN AND BEHAVIOR 2018; 17:e12485. [PMID: 29717802 DOI: 10.1111/gbb.12485] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 04/26/2018] [Accepted: 04/26/2018] [Indexed: 12/19/2022]
Abstract
Enriched environment treatment (EET) is a potential intervention for depression by inducing brain-derived neurotrophic factor (BDNF). However, its age dependency remains unclear. We recently found that EET during early-life development (ED) was effective in increasing exploratory activity and anti-despair behavior, particularly in promoter IV-driven BDNF deficient mice (KIV), with the largest BDNF protein induction in the hippocampus and frontal cortex. Here, we further determined age dependency of EET effects on anhedonia and promoter-specific BDNF transcription, by using the sucrose preference test and qRT-PCR. Wild-type (WT) and KIV mice received 2 months of EET during ED, young-adulthood and old-adulthood (0-2, 2-4 and 12-14 months, respectively). All KIV groups showed reduced sucrose preference, which EET equally reversed regardless of age. EET increased hippocampal BDNF mRNA levels for all ages and genotypes, but increased frontal cortex BDNF mRNA levels only in ED KIV and old WT mice. Transcription by promoters I and IV was age-dependent in the hippocampus of WT mice: more effective induction of exon IV or I during ED or old-adulthood, respectively. Transcription by almost all 9 promoters was age-specific in the frontal cortex, mostly observed in ED KIV mice. After discontinuance of EET, the EET effects on anti-anhedonia and BDNF transcription in both regions persisted only in ED KIV mice. These results suggested that EET was equally effective in reversing anhedonia and inducing hippocampal BDNF transcription, but was more effective during ED in inducing frontal cortex BDNF transcription and for lasting anti-anhedonic and BDNF effects particularly in promoter IV-BDNF deficiency.
Collapse
Affiliation(s)
- B E Dong
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Y Xue
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee
| | - K Sakata
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
38
|
Burns SB, Szyszkowicz JK, Luheshi GN, Lutz PE, Turecki G. Plasticity of the epigenome during early-life stress. Semin Cell Dev Biol 2018; 77:115-132. [DOI: 10.1016/j.semcdb.2017.09.033] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 09/08/2017] [Accepted: 09/22/2017] [Indexed: 12/22/2022]
|
39
|
Effect of Maternal Administration of Edible Bird's Nest on the Learning and Memory Abilities of Suckling Offspring in Mice. Neural Plast 2018; 2018:7697261. [PMID: 29765403 PMCID: PMC5885349 DOI: 10.1155/2018/7697261] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 12/04/2017] [Indexed: 11/17/2022] Open
Abstract
Although human brains continue developing throughout the underage developmental stages, the infancy period is considered the most important one for the whole life. It has been reported that sialic acid from edible bird's nest (EBN) can facilitate the development of brain and intelligence. In this study, by oral administration of EBN to female mice during the pregnancy or lactation period, the effects of EBN on the levels of sialic acid in mouse milk were determined using high-performance liquid chromatography (HPLC). Furthermore, the spatial learning performances of their offspring were assessed using the Morris water maze test. Additionally, cerebral malondialdehyde (MDA), superoxide dismutase (SOD), choline acetyltransferase (ChAT), and acetylcholinesterase (AChE) in cubs nursed by the female mice given the EBN homogenate were examined, while BDNF immunohistochemical staining and neuron count in hippocampi were investigated as well. These results showed that administration with EBN in maternal mice during pregnancy or lactation period can improve the learning and memory functions in their offspring, possibly by increasing the activities of SOD and ChAT and, at the meantime, decreasing the levels of MDA and activities of AChE. Moreover, BDNF levels for CA1, CA2, and CA3 regions in hippocampi and the numbers of dyed neurons in CA1, CA2, CA3, and DG regions among the offspring were significantly enhanced due to the intake of EBN by the maternal mice. We concluded that maternal administration of EBN during the pregnancy and lactation periods can improve the spatial learning performances in the offspring.
Collapse
|
40
|
Zhou H, Wang B, Sun H, Xu X, Wang Y. Epigenetic Regulations in Neural Stem Cells and Neurological Diseases. Stem Cells Int 2018; 2018:6087143. [PMID: 29743892 PMCID: PMC5878882 DOI: 10.1155/2018/6087143] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 01/08/2018] [Indexed: 12/17/2022] Open
Abstract
Among the regulatory mechanisms of the renewal and differentiation of neural stem cells, recent evidences support that epigenetic modifications such as DNA methylation, histone modification, and noncoding RNAs play critical roles in the regulation on the proliferation and differentiation of neural stem cells. In this review, we discussed recent advances of DNA modifications on the regulative mechanisms of neural stem cells. Among these epigenetic modifications, DNA 5-hydroxymethylcytosine (5hmC) modification is emerging as an important modulator on the proliferation and differentiation of neural stem cells. At the same time, Ten-eleven translocation (Tet) methylcytosine dioxygenases, the rate-limiting enzyme for the 5-hydroxymethylation reaction from 5-methylcytosine to 5-hydroxymethylcytosine, play a critical role in the tumorigenesis and the proliferation and differentiation of stem cells. The functions of 5hmC and TET proteins on neural stem cells and their roles in neurological diseases are discussed.
Collapse
Affiliation(s)
- Hang Zhou
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Bin Wang
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Hao Sun
- Department of Orthopedics, Clinical Medical School, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou 225001, China
| | - Xingshun Xu
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Yongxiang Wang
- Department of Orthopedics, Clinical Medical School, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou 225001, China
| |
Collapse
|
41
|
Testing experience and environmental enrichment potentiated open-field habituation and grooming behaviour in rats. Anim Behav 2018. [DOI: 10.1016/j.anbehav.2018.01.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
42
|
Zhang TY, Keown CL, Wen X, Li J, Vousden DA, Anacker C, Bhattacharyya U, Ryan R, Diorio J, O'Toole N, Lerch JP, Mukamel EA, Meaney MJ. Environmental enrichment increases transcriptional and epigenetic differentiation between mouse dorsal and ventral dentate gyrus. Nat Commun 2018; 9:298. [PMID: 29352183 PMCID: PMC5775256 DOI: 10.1038/s41467-017-02748-x] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 12/19/2017] [Indexed: 01/01/2023] Open
Abstract
Early life experience influences stress reactivity and mental health through effects on cognitive-emotional functions that are, in part, linked to gene expression in the dorsal and ventral hippocampus. The hippocampal dentate gyrus (DG) is a major site for experience-dependent plasticity associated with sustained transcriptional alterations, potentially mediated by epigenetic modifications. Here, we report comprehensive DNA methylome, hydroxymethylome and transcriptome data sets from mouse dorsal and ventral DG. We find genome-wide transcriptional and methylation differences between dorsal and ventral DG, including at key developmental transcriptional factors. Peripubertal environmental enrichment increases hippocampal volume and enhances dorsal DG-specific differences in gene expression. Enrichment also enhances dorsal-ventral differences in DNA methylation, including at binding sites of the transcription factor NeuroD1, a regulator of adult neurogenesis. These results indicate a dorsal-ventral asymmetry in transcription and methylation that parallels well-known functional and anatomical differences, and that may be enhanced by environmental enrichment.
Collapse
Affiliation(s)
- Tie-Yuan Zhang
- Sackler Program for Epigenetics and Psychobiology, McGill University, Montréal, H4H 1R3, Canada.
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, H4H 1R3, Canada.
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, 6875 boul. Lasalle, Montréal, H4H 1R3, Canada.
| | - Christopher L Keown
- Department of Cognitive Science, University of California, 9500 Gilman Dr., La Jolla, San Diego, 92093, CA, USA
| | - Xianglan Wen
- Sackler Program for Epigenetics and Psychobiology, McGill University, Montréal, H4H 1R3, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, H4H 1R3, Canada
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, 6875 boul. Lasalle, Montréal, H4H 1R3, Canada
| | - Junhao Li
- Department of Cognitive Science, University of California, 9500 Gilman Dr., La Jolla, San Diego, 92093, CA, USA
| | - Dulcie A Vousden
- Department of Medical Biophysics, The Hospital for Sick Children, University of Toronto, Toronto, M5G 1X8, Canada
| | - Christoph Anacker
- Sackler Program for Epigenetics and Psychobiology, McGill University, Montréal, H4H 1R3, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, H4H 1R3, Canada
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, 6875 boul. Lasalle, Montréal, H4H 1R3, Canada
| | - Urvashi Bhattacharyya
- Department of Cognitive Science, University of California, 9500 Gilman Dr., La Jolla, San Diego, 92093, CA, USA
| | - Richard Ryan
- Sackler Program for Epigenetics and Psychobiology, McGill University, Montréal, H4H 1R3, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, H4H 1R3, Canada
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, 6875 boul. Lasalle, Montréal, H4H 1R3, Canada
| | - Josie Diorio
- Sackler Program for Epigenetics and Psychobiology, McGill University, Montréal, H4H 1R3, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, H4H 1R3, Canada
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, 6875 boul. Lasalle, Montréal, H4H 1R3, Canada
| | - Nicholas O'Toole
- Sackler Program for Epigenetics and Psychobiology, McGill University, Montréal, H4H 1R3, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, H4H 1R3, Canada
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, 6875 boul. Lasalle, Montréal, H4H 1R3, Canada
| | - Jason P Lerch
- Department of Medical Biophysics, The Hospital for Sick Children, University of Toronto, Toronto, M5G 1X8, Canada
| | - Eran A Mukamel
- Department of Cognitive Science, University of California, 9500 Gilman Dr., La Jolla, San Diego, 92093, CA, USA.
| | - Michael J Meaney
- Sackler Program for Epigenetics and Psychobiology, McGill University, Montréal, H4H 1R3, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, H4H 1R3, Canada
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, 6875 boul. Lasalle, Montréal, H4H 1R3, Canada
- Singapore Institute for Clinical Sciences, Singapore, 117609, Singapore
| |
Collapse
|
43
|
Hsiao YH, Chang CH, Gean PW. Impact of social relationships on Alzheimer's memory impairment: mechanistic studies. J Biomed Sci 2018; 25:3. [PMID: 29325565 PMCID: PMC5764000 DOI: 10.1186/s12929-018-0404-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 01/02/2018] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by progressive memory and neuronal loss culminating in cognitive impairment that not only affects a person's living ability but also becomes a society's as well as a family's economic burden. AD is the most common form of dementia in older persons. It is expected that the number of people with AD dementia will increase dramatically in the next 30 years, projecting to 75 million in 2030 and 131.5 million in 2050 worldwide. So far, no sufficient evidence is available to support that any medicine is able to prevent or reverse the progression of the disease. Early studies have shown that social environment, particularly social relationships, can affect one's behavior and mental health. A study analyzing the correlation between loneliness and risk of developing AD revealed that lonely persons had higher risk of AD compared with persons who were not lonely. On the other hand, it has been reported that we can prevent cognitive decline and delay the onset of AD if we keep mentally active and frequently participate in social activities. In this review, we focus on the impact of social behaviors on the progression of cognitive deficit in animal models of AD with a particular emphasis on a mechanistic scheme that explains how social isolation exacerbates cognitive impairment and how social interaction with conspecifics rescues AD patients' memory deficit.
Collapse
Affiliation(s)
- Ya-Hsin Hsiao
- Department of Pharmacology, College of Medicine, National Cheng Kung University, No.1, Ta-Shieh Rd, Tainan City, 701, Taiwan
| | - Chih-Hua Chang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, No.1, Ta-Shieh Rd, Tainan City, 701, Taiwan
| | - Po-Wu Gean
- Department of Pharmacology, College of Medicine, National Cheng Kung University, No.1, Ta-Shieh Rd, Tainan City, 701, Taiwan.
| |
Collapse
|
44
|
Barnett Burns S, Almeida D, Turecki G. The Epigenetics of Early Life Adversity: Current Limitations and Possible Solutions. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 157:343-425. [DOI: 10.1016/bs.pmbts.2018.01.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
45
|
Bator E, Latusz J, Wędzony K, Maćkowiak M. Adolescent environmental enrichment prevents the emergence of schizophrenia-like abnormalities in a neurodevelopmental model of schizophrenia. Eur Neuropsychopharmacol 2018; 28:97-108. [PMID: 29174863 DOI: 10.1016/j.euroneuro.2017.11.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 10/25/2017] [Accepted: 11/09/2017] [Indexed: 01/15/2023]
Abstract
In the present study, we investigated whether exposure to an enriched environment (EE) during adolescence might affect the behavioural dysfunction (sensorimotor gating deficit, memory and social interaction impairments) and neurochemical changes (GAD67 expression, histone methylation) induced by methylazoxymethanol (MAM) in the MAM-E17 rat model of schizophrenia. EE was introduced for 7 days in early adolescence (days 23-29), and behavioural and biochemical studies were performed on adult rats at postnatal day 70. The results showed that exposure to EE prevented the development of adult behavioural deficits induced by prenatal MAM administration. EE also prevented the decrease in GAD67 mRNA and protein levels induced by MAM in the medial prefrontal cortex (mPFC). Moreover, EE inhibited the reductions in the amount of Gad1 bound to H3K4me3 and in the total H3K4me3 protein level induced by prenatal MAM administration in the adult mPFC. However, there was no effect of EE on behaviour or levels of the various neurochemical markers in adult rats prenatally treated with vehicle. Thus, these results indicate that EE exposure during early adolescence may inhibit the development of schizophrenia related symptoms through epigenetic mechanisms that regulate the expression of genes (e.g., Gad1) that are impaired in schizophrenia.
Collapse
Affiliation(s)
- Ewelina Bator
- Institute of Pharmacology, Polish Academy of Sciences, Laboratory of Pharmacology and Brain Biostructure, Smętna Street 12, 31-343 Kraków, Poland
| | - Joachim Latusz
- Institute of Pharmacology, Polish Academy of Sciences, Laboratory of Pharmacology and Brain Biostructure, Smętna Street 12, 31-343 Kraków, Poland
| | - Krzysztof Wędzony
- Institute of Pharmacology, Polish Academy of Sciences, Laboratory of Pharmacology and Brain Biostructure, Smętna Street 12, 31-343 Kraków, Poland
| | - Marzena Maćkowiak
- Institute of Pharmacology, Polish Academy of Sciences, Laboratory of Pharmacology and Brain Biostructure, Smętna Street 12, 31-343 Kraków, Poland.
| |
Collapse
|
46
|
Chan CB, Ye K. Sex differences in brain-derived neurotrophic factor signaling and functions. J Neurosci Res 2017; 95:328-335. [PMID: 27870419 DOI: 10.1002/jnr.23863] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 05/27/2016] [Accepted: 07/11/2016] [Indexed: 01/12/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) is a member of the neurotrophin family that plays a critical role in numerous neuronal activities. Recent studies have indicated that some functions or action mechanisms of BDNF vary in a sex-dependent manner. In particular, BDNF content in some brain parts and the tendency to develop BDNF deficiency-related diseases such as depression are greater in female animals. With the support of relevant studies, it has been suggested that sex hormones or steroids can modulate the activities of BDNF, which may account for its functional discrepancy in different sexes. Indeed, the cross-talk between BDNF and sex steroids has been detected for decades, and some sex steroids, such as estrogen, have a positive regulatory effect on BDNF expression and signaling. Thus, the sex of animal models that are used in studying the functions of BDNF is critical. This Mini-Review summarizes our current findings on the differences in expression, signaling, and functions of BDNF between sexes. We also discuss the potential mechanisms for mediating these differential responses, with a specific emphasis on sex steroids. By presenting and discussing these findings, we seek to encourage researchers to take sex influences into consideration when designing experiments, interpreting results, and drawing conclusions. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Chi Bun Chan
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, USA
| |
Collapse
|
47
|
Birkel L. Decreased use of spatial pattern separation in contemporary lifestyles may contribute to hippocampal atrophy and diminish mental health. Med Hypotheses 2017; 107:55-63. [DOI: 10.1016/j.mehy.2017.07.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 07/14/2017] [Indexed: 10/19/2022]
|
48
|
Kieffer DA, Medici V. Wilson disease: At the crossroads between genetics and epigenetics-A review of the evidence. LIVER RESEARCH 2017; 1:121-130. [PMID: 29270329 PMCID: PMC5734098 DOI: 10.1016/j.livres.2017.08.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Environmental factors, including diet, exercise, stress, and toxins, profoundly impact disease phenotypes. This review examines how Wilson disease (WD), an autosomal recessive genetic disorder, is influenced by genetic and environmental inputs. WD is caused by mutations in the copper-transporter gene ATP7B, leading to the accumulation of copper in the liver and brain, resulting in hepatic, neurological, and psychiatric symptoms. These symptoms range in severity and can first appear anytime between early childhood and old age. Over 300 disease-causing mutations in ATP7B have been identified, but attempts to link genotype to the phenotypic presentation have yielded little insight, prompting investigators to identify alternative mechanisms, such as epigenetics, to explain the highly varied clinical presentation. Further, WD is accompanied by structural and functional abnormalities in mitochondria, potentially altering the production of metabolites that are required for epigenetic regulation of gene expression. Notably, environmental exposure affects the regulation of gene expression and mitochondrial function. We present the "multi-hit" hypothesis of WD progression, which posits that the initial hit is an environmental factor that affects fetal gene expression and epigenetic mechanisms and subsequent "hits" are environmental exposures that occur in the offspring after birth. These environmental hits and subsequent changes in epigenetic regulation may impact copper accumulation and ultimately WD phenotype. Lifestyle changes, including diet, increased physical activity, stress reduction, and toxin avoidance, might influence the presentation and course of WD, and therefore may serve as potential adjunctive or replacement therapies.
Collapse
|
49
|
Effects of environmental enrichment upon ethanol-induced conditioned place preference and pre-frontal BDNF levels in adolescent and adult mice. Sci Rep 2017; 7:8574. [PMID: 28819238 PMCID: PMC5561235 DOI: 10.1038/s41598-017-08795-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 07/18/2017] [Indexed: 01/04/2023] Open
Abstract
Environmental enrichment (EE) provides a non-pharmacological tool to alter drug-induced reward, yet its effects on ethanol-induced reward remain controversial. We analyzed adolescent vs. adult (mice) differences in the influence of EE on ethanol-induced conditioned place preference (CPP). The effects of these treatments on brain-derived neurotrophic factor (BDNF) levels in the prefrontal cortex were examined in a separate group of animals. Ethanol-induced CPP was found in adults, and it was similar in EE and in animals reared under standard housing conditions (SC). Adolescents kept under EE, but not those in SC, exhibited CPP. Among SC, but not among EE, adolescents, BDNF levels were significantly lower in those treated with ethanol than in those given vehicle. These results indicate that, compared to adults, adolescent exhibited reduced sensitivity to ethanol’s rewarding effects, yet the youth but not the adults exhibited sensitivity to the promoting effect of EE upon CPP by ethanol. Ethanol significantly reduced BDNF levels in adolescents reared under standard housing conditions, but not in adult mice nor in adolescents given EE housing conditions. The present results add to the plethora of adolescent-specific responses to ethanol or to environmental stimuli that may put the youth at risk for escalation of ethanol intake.
Collapse
|
50
|
Abstract
More and more people are living into the 90s or becoming centenarians. But, the gift of increased ‘age span’ seldom equates with an improved ‘health-span’. Governments across the world are expressing concern about the epidemic of chronic disease, and have responded by initiating policies that make prevention, reduction and treatment of chronic disease, a public health priority. But understanding, how to age long and well, with the avoidance of chronic disease and later life complex disease morbidity is challenging. While inherited genes have an undoubted role to play in the chance of maintaining good health or conversely a predilection to developing disease and chronic ill health, there is increasing evidence that behavioural and environmental life-style choices may contribute up to 50% of the variability of human lifespan. Physical exercise is readily available to everyone, and is a simple cheap and effective form of life-style intervention. Exercise appears to help maintain good health and to reduce the risk of developing chronic disease and ill health. Evidence suggests that physical activity improves well-being across many health domains through out life, continues to offer important health benefits in older age groups and tracks with a ‘healthy ageing’ profile. Although many of the molecular pathways remain to be fully identified, here we discuss how physical activity and exercise is understood to produce changes in the human epigenome, which have the potential to enhance cognitive and psychological health, improve muscular fitness, and lead to better ageing with improved quality of life in older age.
Collapse
|