1
|
Valenza G, Matić Z, Catrambone V. The brain-heart axis: integrative cooperation of neural, mechanical and biochemical pathways. Nat Rev Cardiol 2025:10.1038/s41569-025-01140-3. [PMID: 40033035 DOI: 10.1038/s41569-025-01140-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/10/2025] [Indexed: 03/05/2025]
Abstract
The neural and cardiovascular systems are pivotal in regulating human physiological, cognitive and emotional states, constantly interacting through anatomical and functional connections referred to as the brain-heart axis. When this axis is dysfunctional, neurological conditions can lead to cardiovascular disorders and, conversely, cardiovascular dysfunction can substantially affect brain health. However, the mechanisms and fundamental physiological components of the brain-heart axis remain largely unknown. In this Review, we elucidate these components and identify three primary pathways: neural, mechanical and biochemical. The neural pathway involves the interaction between the autonomic nervous system and the central autonomic network in the brain. The mechanical pathway involves mechanoreceptors, particularly those expressing mechanosensitive Piezo protein channels, which relay crucial information about blood pressure through peripheral and cerebrovascular connections. The biochemical pathway comprises many endogenous compounds that are important mediators of neural and cardiovascular function. This multisystem perspective calls for the development of integrative approaches, leading to new clinical specialties in neurocardiology.
Collapse
Affiliation(s)
- Gaetano Valenza
- Neurocardiovascular Intelligence Lab, Department of Information Engineering & Research Center "E. Piaggio", University of Pisa, Pisa, Italy.
| | - Zoran Matić
- Neurocardiovascular Intelligence Lab, Department of Information Engineering & Research Center "E. Piaggio", University of Pisa, Pisa, Italy
| | - Vincenzo Catrambone
- Neurocardiovascular Intelligence Lab, Department of Information Engineering & Research Center "E. Piaggio", University of Pisa, Pisa, Italy
| |
Collapse
|
2
|
Xu H, Zhang T, Li L, Qu Y, Li L, Yan Y, Wu L, Yan C. Paeoniflorin exerts anti-PTSD effects in adult rats by modulating hippocampus and amygdala histone acetylation modifications in response to early life stress. Chem Biol Interact 2024; 396:111035. [PMID: 38703807 DOI: 10.1016/j.cbi.2024.111035] [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: 10/22/2023] [Revised: 04/12/2024] [Accepted: 05/02/2024] [Indexed: 05/06/2024]
Abstract
Early life stress (ELS) can cause long-term changes by epigenetic factors, especially histone acetylation modification, playing a crucial role, affect normal cognition, mood, and behavior, and increase susceptibility to post-traumatic stress disorder (PTSD) in adulthood. It has been found that paeoniflorin (PF) can cross the blood-brain barrier to exert anti-PTSD effects on adult PTSD rats. However, whether PF can alleviate the harmful effects caused by ELS in adulthood has not yet been reported. Therefore, to explore the relationship between ELS and PTSD susceptibility in adulthood and its mechanism, in this study, SPS was used as a stressor of ELS, and the mathematical tool Z-normalization was employed as an evaluation criterion of behavioral resilience susceptibility. To investigate the regulatory mechanism of PF on histone acetylation in the hippocampus and amygdala of ELS rats in adulthood, using changes in HATs/HDACs as the entry point, meanwhile, the epigenetic marks (H3K9 and H4K12) in the key brain regions of ELS (hippocampus and amygdala) were evaluated, and the effects of PF on behavioral representation and PTSD susceptibility were observed. This study found that ELS lead to a series of PTSD-like behaviors in adulthood and caused imbalance of HATs/HDACs ratio in the hippocampus and amygdala, which confirms that ELS is an important risk factor for the development of PTSD in adulthood. In addition, paeoniflorin may improve ELS-induced PTSD-like behaviors and reduce the susceptibility of ELS rats to develop PTSD in adulthood by modulating the HATs/HDACs ratio in the hippocampus and amygdala.
Collapse
Affiliation(s)
- Hanfang Xu
- Research Center of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, PR China.
| | - Tiange Zhang
- Research Center of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, PR China.
| | - Ling Li
- Research Center of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, PR China
| | - Yue Qu
- Research Center of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, PR China
| | - Lanxin Li
- Research Center of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, PR China
| | - Yuqi Yan
- Research Center of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, PR China
| | - Lili Wu
- Research Center of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, PR China.
| | - Can Yan
- Research Center of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, PR China.
| |
Collapse
|
3
|
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
|
4
|
Damiani F, Cornuti S, Tognini P. The gut-brain connection: Exploring the influence of the gut microbiota on neuroplasticity and neurodevelopmental disorders. Neuropharmacology 2023; 231:109491. [PMID: 36924923 DOI: 10.1016/j.neuropharm.2023.109491] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 02/22/2023] [Accepted: 03/05/2023] [Indexed: 03/17/2023]
Abstract
Neuroplasticity refers to the ability of brain circuits to reorganize and change the properties of the network, resulting in alterations in brain function and behavior. It is traditionally believed that neuroplasticity is influenced by external stimuli, learning, and experience. Intriguingly, there is new evidence suggesting that endogenous signals from the body's periphery may play a role. The gut microbiota, a diverse community of microorganisms living in harmony with their host, may be able to influence plasticity through its modulation of the gut-brain axis. Interestingly, the maturation of the gut microbiota coincides with critical periods of neurodevelopment, during which neural circuits are highly plastic and potentially vulnerable. As such, dysbiosis (an imbalance in the gut microbiota composition) during early life may contribute to the disruption of normal developmental trajectories, leading to neurodevelopmental disorders. This review aims to examine the ways in which the gut microbiota can affect neuroplasticity. It will also discuss recent research linking gastrointestinal issues and bacterial dysbiosis to various neurodevelopmental disorders and their potential impact on neurological outcomes.
Collapse
Affiliation(s)
| | - Sara Cornuti
- Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy
| | - Paola Tognini
- Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy; Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.
| |
Collapse
|
5
|
Castro EM, Lotfipour S, Leslie FM. Nicotine on the developing brain. Pharmacol Res 2023; 190:106716. [PMID: 36868366 PMCID: PMC10392865 DOI: 10.1016/j.phrs.2023.106716] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/23/2023] [Accepted: 02/28/2023] [Indexed: 03/05/2023]
Abstract
Developmental periods such as gestation and adolescence have enhanced plasticity leaving the brain vulnerable to harmful effects from nicotine use. Proper brain maturation and circuit organization is critical for normal physiological and behavioral outcomes. Although cigarette smoking has declined in popularity, noncombustible nicotine products are readily used. The misperceived safety of these alternatives lead to widespread use among vulnerable populations such as pregnant women and adolescents. Nicotine exposure during these sensitive developmental windows is detrimental to cardiorespiratory function, learning and memory, executive function, and reward related circuitry. In this review, we will discuss clinical and preclinical evidence of the adverse alterations in the brain and behavior following nicotine exposure. Time-dependent nicotine-induced changes in reward related brain regions and drug reward behaviors will be discussed and highlight unique sensitivities within a developmental period. We will also review long lasting effects of developmental exposure persisting into adulthood, along with permanent epigenetic changes in the genome which can be passed to future generations. Taken together, it is critical to evaluate the consequences of nicotine exposure during these vulnerable developmental windows due to its direct impact on cognition, potential trajectories for other substance use, and implicated mechanisms for the neurobiology of substance use disorders.
Collapse
Affiliation(s)
- Emily M Castro
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
| | - Shahrdad Lotfipour
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA; Department of Emergency Medicine, School of Medicine, University of California, Irvine, Irvine, CA, USA; Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Frances M Leslie
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
6
|
Environmental enrichment mitigates PTSD-like behaviors in adult male rats exposed to early life stress by regulating histone acetylation in the hippocampus and amygdala. J Psychiatr Res 2022; 155:120-136. [PMID: 36029624 DOI: 10.1016/j.jpsychires.2022.07.067] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/04/2022] [Accepted: 07/25/2022] [Indexed: 11/20/2022]
Abstract
Early life stress (ELS) can cause long-term changes in gene expression, affect cognition, mood, and behavior, and increase susceptibility to post-traumatic stress disorder (PTSD) in adulthood, in which the histone acetylation plays a crucial role. Studies have found that environmental enrichment (EE) mitigated the unfavorable outcomes of ELS. However, the underlying mechanism of the histone acetylation is not yet completely clear. The purpose of this study was to explore the effect of EE on the histone acetylation after ELS. In this study, using single prolonged stress (SPS) paradigm in early adolescent rats explored the long-term effects of ELS on behavior, the activity of histone acetyltransferases (HATs) and histone deacetylases (HDACs), as well as the acetylation levels of the lysine 9 site of histone H3 (H3K9) and lysine 12 site of histone H4 (H4K12) in the hippocampus and amygdala. Meanwhile, the protective effects of EE intervention were examined. We found that adult male rats exposed to ELS showed behavioral changes, including reduced locomotor activity, increased anxiety-like behaviors, impaired spatial learning and memory, enhanced contextual and cued fear memory, and the HATs/HDACs ratio and acetyl H3K9 (Ac-H3K9) and acetyl H4K12 (Ac-H4K12) were increased in the hippocampus and decreased in the amygdala. Furthermore, EE attenuated the behavioral abnormalities from ELS, possibly through down-regulating the activity of HATs in the hippocampus and up-regulating HDACs activities in the amygdala. These finding suggested that EE could ameliorate ELS-induced PTSD-like behaviors by regulating histone acetylation in the hippocampus and amygdala, reducing the susceptibility to PTSD in adulthood.
Collapse
|
7
|
Zima L, West R, Smolen P, Kobori N, Hergenroeder G, Choi HA, Moore AN, Redell JB, Dash PK. Epigenetic Modifications and Their Potential Contribution to Traumatic Brain Injury Pathobiology and Outcome. J Neurotrauma 2022; 39:1279-1288. [PMID: 35481812 PMCID: PMC9529317 DOI: 10.1089/neu.2022.0128] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Epigenetic information is not permanently encoded in the DNA sequence, but rather consists of reversible, heritable modifications that regulate the gene expression profile of a cell. Epigenetic modifications can result in cellular changes that can be long lasting and include DNA methylation, histone methylation, histone acetylation, and RNA methylation. As epigenetic modifications are reversible, the enzymes that add (epigenetic writers), the proteins that decode (epigenetic readers), and the enzymes that remove (epigenetic erasers) these modifications can be targeted to alter cellular function and disease biology. While epigenetic modifications and their contributions are intense topics of current research in the context of a number of diseases, including cancer, inflammatory diseases, and Alzheimer disease, the study of epigenetics in the context of traumatic brain injury (TBI) is in its infancy. In this review, we will summarize the experimental and clinical findings demonstrating that TBI triggers epigenetic modifications, with a focus on changes in DNA methylation, histone methylation, and the translational utility of the universal methyl donor S-adenosylmethionine (SAM). Finally, we will review the evidence for using methyl donors as possible treatments for TBI-associated pathology and outcome.
Collapse
Affiliation(s)
- Laura Zima
- Department of Neurological Surgery, University of Texas Health Science Center McGovern Medical School, Houston, Texas, USA
| | - Rebecca West
- Department of Neurobiology and Anatomy, University of Texas Health Science Center McGovern Medical School, Houston, Texas, USA
| | - Paul Smolen
- Department of Neurobiology and Anatomy, University of Texas Health Science Center McGovern Medical School, Houston, Texas, USA
| | - Nobuhide Kobori
- Department of Neurobiology and Anatomy, University of Texas Health Science Center McGovern Medical School, Houston, Texas, USA
| | - Georgene Hergenroeder
- Department of Neurological Surgery, University of Texas Health Science Center McGovern Medical School, Houston, Texas, USA
| | - HuiMahn A. Choi
- Department of Neurological Surgery, University of Texas Health Science Center McGovern Medical School, Houston, Texas, USA
| | - Anthony N. Moore
- Department of Neurobiology and Anatomy, University of Texas Health Science Center McGovern Medical School, Houston, Texas, USA
| | - John B. Redell
- Department of Neurobiology and Anatomy, University of Texas Health Science Center McGovern Medical School, Houston, Texas, USA
| | - Pramod K. Dash
- Department of Neurobiology and Anatomy, University of Texas Health Science Center McGovern Medical School, Houston, Texas, USA
| |
Collapse
|
8
|
Zhou Q, Verne GN. Epigenetic modulation of visceral nociception. Neurogastroenterol Motil 2022; 34:e14443. [PMID: 35950237 PMCID: PMC9787514 DOI: 10.1111/nmo.14443] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 07/29/2022] [Indexed: 12/30/2022]
Abstract
Epigenetics is a process that alters gene activity or phenotype without any changes in the underlying DNA sequence or genotype. These biological changes may have deleterious effects and can lead to various human diseases. Ongoing research is continuing to illuminate the role of epigenetics in a variety of pathophysiologic processes. Several categories of epigenetic mechanisms have been studied including chromatin remodeling, DNA methylation, histone modification, and non-coding RNA mechanisms. These epigenetic changes can have a long-term effect on gene expression without any underlying changes in the DNA sequences. The underlying pathophysiology of disorders of brain-gut interaction and stress-induced visceral pain are not fully understood and the role of epigenetic mechanisms in these disorders are starting to be better understood. Current work is underway to determine how epigenetics plays a role in the neurobiology of patients with chronic visceral pain and heightened visceral nociception. More recently, both animal models and human studies have shown how epigenetic regulation modulates stress-induced visceral pain. While much more work is needed to fully delineate the mechanistic role of epigenetics in the neurobiology of chronic visceral nociception, the current study by Louwies et al., in Neurogastroenterology and Motility provides additional evidence supporting the involvement of epigenetic alterations in the central nucleus of the amygdala in stress-induced visceral hypersensitivity in rodents.
Collapse
Affiliation(s)
- QiQi Zhou
- Department of MedicineUniversity of Tennessee College of MedicineMemphisTennesseeUSA
- Memphis VA Medical CenterResearch ServiceMemphisTennesseeUSA
| | - George Nicholas Verne
- Department of MedicineUniversity of Tennessee College of MedicineMemphisTennesseeUSA
- Memphis VA Medical CenterResearch ServiceMemphisTennesseeUSA
| |
Collapse
|
9
|
Jiang W, Tan XY, Li JM, Yu P, Dong M. DNA Methylation: A Target in Neuropathic Pain. Front Med (Lausanne) 2022; 9:879902. [PMID: 35872752 PMCID: PMC9301322 DOI: 10.3389/fmed.2022.879902] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Neuropathic pain (NP), caused by an injury or a disease affecting the somatosensory nervous system of the central and peripheral nervous systems, has become a global health concern. Recent studies have demonstrated that epigenetic mechanisms are among those that underlie NP; thus, elucidating the molecular mechanism of DNA methylation is crucial to discovering new therapeutic methods for NP. In this review, we first briefly discuss DNA methylation, demethylation, and the associated key enzymes, such as methylases and demethylases. We then discuss the relationship between NP and DNA methylation, focusing on DNA methyltransferases including methyl-CpG-binding domain (MBD) family proteins and ten-eleven translocation (TET) enzymes. Based on experimental results of neuralgia in animal models, the mechanism of DNA methylation-related neuralgia is summarized, and useful targets for early drug intervention in NP are discussed.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Xuan-Yu Tan
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Jia-Ming Li
- Department of Emergency, The First Hospital of Jilin University, Changchun, China
| | - Peng Yu
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Peng Yu
| | - Ming Dong
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
- Ming Dong
| |
Collapse
|
10
|
Dvořáček J, Bednářová A, Krishnan N, Kodrík D. Dopaminergic muhsroom body neurons in Drosophila: flexibility of neuron identity in a model organism? Neurosci Biobehav Rev 2022; 135:104570. [DOI: 10.1016/j.neubiorev.2022.104570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/03/2022] [Accepted: 02/03/2022] [Indexed: 11/28/2022]
|
11
|
Soares-Silva B, Beserra-Filho JIA, Morera PMA, Custódio-Silva AC, Maria-Macêdo A, Silva-Martins S, Alexandre-Silva V, Silva SP, Silva RH, Ribeiro AM. The bee venom active compound melittin protects against bicuculline-induced seizures and hippocampal astrocyte activation in rats. Neuropeptides 2022; 91:102209. [PMID: 34808488 DOI: 10.1016/j.npep.2021.102209] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/26/2021] [Accepted: 11/11/2021] [Indexed: 11/17/2022]
Abstract
Epilepsy is a chronic neuropathology characterized by an abnormal hyperactivity of neurons that generate recurrent, spontaneous, paradoxical and synchronized nerve impulses, leading or not to seizures. This neurological disorder affects around 70 million individuals worldwide. Pharmacoresistance is observed in about 30% of the patients and long-term use of antiepileptics may induce serious side effects. Thus, there is an interest in the study of the therapeutic potential of bioactive substances isolated from natural products in the treatment of epilepsy. Arthropod venoms contain neurotoxins that have high affinity for molecular structures in the neural tissue such as receptors, transporters and ion channels both in glial and neuronal membranes. This study evaluated the potential neuroprotective effect of melittin (MEL), an active compound of bee venom, in the bicuculline-induced seizure model (BIC) in rats. Male Wistar rats (3 months, 250-300 g) were submitted to surgery for the implantation of a unilateral cannula in the lateral ventricle. After the recovery period, rats received a microinjection of saline solution or MEL (0.1 mg per animal). Firstly, rats were evaluated in the open field (20 min) and in the elevated plus maze (5 min) tests after received microinjection of saline or MEL. After, 30 min later animals received BIC (100 mg/ml) or saline, and their behaviors were analyzed for 20 min in the open field according to a seizure scale. At the end, rats were euthanized, brains collected and processed to glial fibrillary acidic protein (GFAP) immunohistochemistry evaluation. No changes were observed in MEL-treated rats in the open field and elevated plus maze. However, 90% of MEL-treated animals were protected against seizures induced by BIC. There was an increase in the latency for the onset of seizures, accompanied by a reduction of GFAP-immunoreactivity cells in the dentate gyrus and CA1. Thus, our study suggests that MEL has an anticonvulsant potential, and further studies are needed to elucidate the mechanisms involved in this action.
Collapse
Affiliation(s)
| | - José Ivo Araújo Beserra-Filho
- Departament of Biosciences, Universidade Federal de São Paulo, Santos, Brazil; Department of Pharmacology, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | - Amanda Maria-Macêdo
- Departament of Biosciences, Universidade Federal de São Paulo, Santos, Brazil
| | | | | | - Sara Pereira Silva
- Departament of Biosciences, Universidade Federal de São Paulo, Santos, Brazil
| | - Regina Helena Silva
- Department of Pharmacology, Universidade Federal de São Paulo, São Paulo, Brazil
| | | |
Collapse
|
12
|
Epigenetic correlates of the psychological interventions outcomes: A systematic review and meta-analysis. JOURNAL OF AFFECTIVE DISORDERS REPORTS 2022. [DOI: 10.1016/j.jadr.2022.100310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
13
|
Effects of Thermal Conditioning and Folic Acid on Methylation of the BDNF Promoter Region in Chicks. J Poult Sci 2021; 58:280-285. [PMID: 34899024 PMCID: PMC8630412 DOI: 10.2141/jpsa.0210008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 02/15/2021] [Indexed: 11/21/2022] Open
Abstract
This study aimed to investigate the effects of thermal conditioning and folic acid on the methylation levels of the avian brain-derived neurotrophic factor (BDNF) promoter region at the M3 and M9 positions in the early life of broiler chicks. In Experiment 1, male broiler chicks (day 3 of life) were orally injected with methyl cellulose solution with or without folic acid (25 mg). The chicks in the heat-treatment groups were immediately exposed to a high ambient temperature (40±0.5°C) for 12 h, while chicks in the non-heat treatment groups were left in the thermoneutral zone (30±0.5°C). The groups were as follows: 1) no thermal conditioning group without folic acid (control), 2) thermal conditioning group without folic acid, 3) no thermal conditioning group with folic acid, and 4) thermal conditioning group with folic acid. In Experiment 2, treatments were similar to those in Experiment 1, except for the usage of female chicks. After the treatments, the methylation levels of the BDNF promoter in chicks were determined using semiquantitative PCR. There were no significant differences between groups in the levels of methylation at the M3 position in both males and females as a result of thermal conditioning and folic acid treatment. Interestingly, significant effects of thermal conditioning and folic acid treatment on methylation at the M9 position were found. BDNF methylation levels at M9 significantly decreased following thermal conditioning, while folic acid suppressed demethylation in both male and female chicks. These data suggest that folic acid and thermal conditioning affects DNA methylation patterns in the central nervous system of chicks, regardless of sex.
Collapse
|
14
|
Juruena MF, Gadelrab R, Cleare AJ, Young AH. Epigenetics: A missing link between early life stress and depression. Prog Neuropsychopharmacol Biol Psychiatry 2021; 109:110231. [PMID: 33383101 DOI: 10.1016/j.pnpbp.2020.110231] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/06/2020] [Accepted: 12/21/2020] [Indexed: 02/08/2023]
Abstract
Research has suggested a relationship between early life stress, and depression in particular longer episodes of depression with treatment resistant outcomes. However, the underlying mechanisms for this association remain poorly understood. Molecular studies indicate that, in general, the hereditary character of psychiatric disorders are polygenic, multifactorial and highly complex, with innumerable low-effect genetic variants interacting with each other. In addition, the importance of the environment and its interaction with genes has pointed to a fundamental role of epigenetic mechanisms in psychiatric disorders, such as methylation of deoxyribonucleic acid (DNA), alterations, histone actions and regulation of gene expression by non-coding ribonucleic acids (RNAs). This article provides an overview of the interplay of epigenetics, the HPA axis, early life stress and the development of depression. Advances in our knowledge of epigenetics in the context of early life stress and depression provide a new understanding of the genetic influence on psychopathology and could lead to the identification of new targets for clinical intervention.
Collapse
Affiliation(s)
- Mario F Juruena
- Centre for Affective Disorders, Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, Denmark Hill, London SE5 8AF, UK; South London and Maudsley NHS Foundation Trust (SLaM), UK.
| | | | - Anthony J Cleare
- Centre for Affective Disorders, Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, Denmark Hill, London SE5 8AF, UK; South London and Maudsley NHS Foundation Trust (SLaM), UK
| | - Allan H Young
- Centre for Affective Disorders, Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, Denmark Hill, London SE5 8AF, UK; South London and Maudsley NHS Foundation Trust (SLaM), UK
| |
Collapse
|
15
|
Akbarabadi A, Sadat-Shirazi MS, Kabbaj M, Nouri Zadeh-Tehrani S, Khalifeh S, Pirri F, Zarrindast MR. Effects of Morphine and Maternal Care on Behaviors and Protein Expression of Male Offspring. Neuroscience 2021; 466:58-76. [PMID: 33915201 DOI: 10.1016/j.neuroscience.2021.04.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 01/26/2023]
Abstract
Genes and environment interact during development to alter gene expression and behavior. Parental morphine exposure before conception has devastating effects on the offspring. In the present study, we evaluated the role of maternal care in the intergenerational effect of maternal morphine exposure. Female rats received morphine or saline for ten days and were drugfree for another ten days. Thereafter, they were allowed to mate with drug-naïve male rats. When pups were born, they were cross-fostered to assess the contribution of maternal care versus morphine effects on the offspring. Adult male offspring were examined for anxiety-like behavior, spatial memory, and obsessive-compulsive-like behavior. To determine the mechanisms underlying the observed behavioral changes, protein levels of acetylated histone H3, BDNF, Trk-B, NMDA subunits, p-CREB, and 5-HT3R were measured in the brain. Our results indicate that maternal caregiving is impaired in morphine-abstinent mothers. Interestingly, maternal care behaviors were also affected in drug-naïve mothers that raised offspring of morphine-exposed mothers. In addition, the offspring of morphine abstinent and non-drug dependent mothers, when raised by morphine abstinent mothers, exhibited more anxiety, obsessive-compulsive behaviors and impaired spatial memory. These altered behaviors were associated with alterations in the levels of the above-mentioned proteins. These data illustrate the intergenerational effects of maternal morphine exposure on offspring behaviors. Moreover, exposure to morphine before gestation not only affects maternal care and offspring behavior, but also has negative consequences on behaviors and protein expression in adoptive mothers of affected offspring.
Collapse
Affiliation(s)
- Ardeshir Akbarabadi
- Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mohamed Kabbaj
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306-1270, United States; Program of Neuroscience, Florida State University, Tallahassee, FL 32306-1270, United States
| | | | - Solmaz Khalifeh
- Cognitive and Neuroscience Research Center (CNRC), Tehran Medical Sciences, Amir-Almomenin Hospital, Islamic Azad University, Tehran, Iran
| | - Fardad Pirri
- Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad-Reza Zarrindast
- Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran; Cognitive and Neuroscience Research Center (CNRC), Tehran Medical Sciences, Amir-Almomenin Hospital, Islamic Azad University, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Endocrinology and Metabolism Research Institute, Tehran University of Medical Science, Tehran, Iran.
| |
Collapse
|
16
|
Nicolay-Kritter K, Lassalle J, Guillou JL, Mons N. The histone H3 lysine 9 methyltransferase G9a/GLP complex activity is required for long-term consolidation of spatial memory in mice. Neurobiol Learn Mem 2021; 179:107406. [PMID: 33609736 DOI: 10.1016/j.nlm.2021.107406] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 02/03/2021] [Accepted: 02/11/2021] [Indexed: 02/07/2023]
Abstract
The G9a/G9a-like protein (GLP) histone lysine dimethyltransferase complex and downstream histone H3 lysine 9 dimethylation (H3K9me2) repressive mark have recently emerged as key transcriptional regulators of gene expression programs necessary for long-term memory (LTM) formation in the dorsal hippocampus. However, the role for hippocampal G9a/GLP complex in mediating the consolidation of spatial LTM remains largely unknown. Using a water maze competition task in which both dorsal hippocampus-dependent spatial and striatum-dependent cue navigation strategies are effective to solve the maze, we found that pharmacological inhibition of G9a/GLP activity immediately after learning disrupts long-term consolidation of previously learned spatial information in male mice, hence producing cue bias on the competition test performed 24 h later. Importantly, the inhibition of hippocampal G9a/GLP did not disrupt short-term memory retention. Immunohistochemical analyses revealed increases in global levels of permissive histone H3K9 acetylation in the dorsal hippocampus and dorsal striatum at 1 h post-training, which persisted up to 24 h in the hippocampus. Conversely, H3K9me2 levels were either unchanged in the dorsal hippocampus or transiently decreased at 15 min post-training in the dorsal striatum. Finally, the inhibition of G9a/GLP activity further increased global levels of H3K9 acetylation while decreasing H3K9me2 in the hippocampus at 1 h post-training. However, both marks returned to vehicle control levels at 24 h. Together, these findings support the possibility that G9a/GLP in the dorsal hippocampus is required for the transcriptional switch from short-term to long-term spatial memory formation.
Collapse
Affiliation(s)
- Kyrian Nicolay-Kritter
- Université de Bordeaux, France; Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5287, 33615 Pessac, France
| | - Jordan Lassalle
- Université de Bordeaux, France; Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5287, 33615 Pessac, France
| | - Jean-Louis Guillou
- Université de Bordeaux, France; Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5287, 33615 Pessac, France
| | - Nicole Mons
- Université de Bordeaux, France; Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5287, 33615 Pessac, France.
| |
Collapse
|
17
|
Alcohol as an early life stressor: Epigenetics, metabolic, neuroendocrine and neurobehavioral implications. Neurosci Biobehav Rev 2020; 118:654-668. [PMID: 32976915 DOI: 10.1016/j.neubiorev.2020.08.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/18/2020] [Accepted: 08/25/2020] [Indexed: 12/14/2022]
Abstract
Ethanol exposure during gestation is an early life stressor that profoundly dysregulates structure and functions of the embryonal nervous system, altering the cognitive and behavioral development. Such dysregulation is also achieved by epigenetic mechanisms, which, altering the chromatin structure, redraw the entire pattern of gene expression. In parallel, an oxidative stress response at the cellular level and a global upregulation of neuroendocrine stress response, regulated by the HPA axis, exist and persist in adulthood. This neurobehavioral framework matches those observed in other psychiatric diseases such as mood diseases, depression, autism; those early life stressing events, although probably triggered by specific and different epigenetic mechanisms, give rise to largely overlapping neurobehavioral phenotypes. An early diagnosis of prenatal alcohol exposure, using reliable markers of ethanol intake, together with a deeper understanding of the pathogenic mechanisms, some of them reversible by their nature, can offer a temporal "window" of intervention. Supplementing a mother's diet with protective and antioxidant substances in addition to supportive psychological therapies can protect newborns from being affected.
Collapse
|
18
|
Environmental enrichment-inspired pharmacological tools for the treatment of addiction. Curr Opin Pharmacol 2020; 56:22-28. [PMID: 32966941 DOI: 10.1016/j.coph.2020.09.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 12/16/2022]
Abstract
Environmental enrichment (EE) has been shown to produce powerful beneficial effects in animal models of addiction. In particular, the ability of EE to promote abstinence and prevent relapse may allow for the identification of brain mechanisms responsible for the recovery from addiction. Indeed, the effects of EE on specific brain mechanisms could be mimicked by old or new molecules, which may become novel medications, called enviromimetics. Here, we review the best known enviromimetics for the treatment of addiction and suggest that, whereas these compounds may be relatively ineffective by themselves, they may be useful complements for existing therapeutic approaches to manage addiction which includes behavioural, environmental and pharmacological interventions.
Collapse
|
19
|
Wagner MA, Erickson KI, Bender CM, Conley YP. The Influence of Physical Activity and Epigenomics On Cognitive Function and Brain Health in Breast Cancer. Front Aging Neurosci 2020; 12:123. [PMID: 32457596 PMCID: PMC7225270 DOI: 10.3389/fnagi.2020.00123] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/14/2020] [Indexed: 12/17/2022] Open
Abstract
The risk of breast cancer increases with age, with the majority of women diagnosed with breast cancer being postmenopausal. It has been estimated that 25-75% of women with breast cancer experience changes in cognitive function (CF) related to disease and treatment, which compromises psychological well-being, decision making, ability to perform daily activities, and adherence to cancer therapy. Unfortunately, the mechanisms that underlie neurocognitive changes in women with breast cancer remain poorly understood, which in turn limits the development of effective treatments and prevention strategies. Exercise has great potential as a non-pharmaceutical intervention to mitigate the decline in CF in women with breast cancer. Evidence suggests that DNA methylation, an epigenetic mechanism for gene regulation, impacts CF and brain health (BH), that exercise influences DNA methylation, and that exercise impacts CF and BH. Although investigating DNA methylation has the potential to uncover the biologic foundations for understanding neurocognitive changes within the context of breast cancer and its treatment as well as the ability to understand how exercise mitigates these changes, there is a dearth of research on this topic. The purpose of this review article is to compile the research in these areas and to recommend potential areas of opportunity for investigation.
Collapse
Affiliation(s)
- Monica A. Wagner
- School of Nursing, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kirk I. Erickson
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States
- Discipline of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, Perth Campus, Murdoch, WA, Australia
| | | | - Yvette P. Conley
- School of Nursing, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
20
|
Stoffel M, Gardini E, Ehrenthal J, Abbruzzese E, Ditzen B. Evaluation of Stress Management and Stress Prevention Using Epigenetic Markers. VERHALTENSTHERAPIE 2020. [DOI: 10.1159/000506323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
21
|
Stoffel M, Gardini E, Ehrenthal J, Abbruzzese E, Ditzen B. Evaluation von Stressprävention und Stressbewältigung mittels epigenetischer Marker. VERHALTENSTHERAPIE 2020. [DOI: 10.1159/000505595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
22
|
Sadat-Shirazi MS, Asgari P, Mahboubi S, Nouri Zadeh-Tehrani S, Ashabi G, Rohbani K, Sabzevari S, Soltani H, Khalifeh S, Zarrindast MR. Effect of morphine exposure on novel object memory of the offspring: The role of histone H3 and ΔFosB. Brain Res Bull 2020; 156:141-149. [PMID: 31958477 DOI: 10.1016/j.brainresbull.2020.01.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 12/19/2019] [Accepted: 01/14/2020] [Indexed: 01/09/2023]
Abstract
It has been demonstrated that alteration in histone acetylation in the regions of the brain involved in the reward which may have an important role in morphine addiction. It is well established that epigenetic changes prior to birth influence the function and development of the brain. The current study was designed to evaluate changes in novel object memory, histone acetylation and ΔFosB in the brain of the offspring of morphine-withdrawn parents. Male and female Wistar rats received morphine orally for 21 following days. After ten days of abstinent, they were prepared for mating. The male offspring of the first parturition were euthanized on postnatal days 5, 21, 30 and 60. The novel object recognition (NOR) test was performed on adult male offspring. The amount of acetylated histone H3 and ΔFosB were evaluated in the prefrontal cortex (PFC) and hippocampus using western blotting. Obtained results indicated that the discrimination index in the NOR test was decreased in the offspring of morphine-withdrawn parents as compared with morphine-naïve offspring. In addition, the level of acetylated histone H3 was decreased in the PFC and hippocampus in the offspring of morphine-withdrawn parents during lifetime (postnatal days 5, 21, 30 and 60). In the case of ΔFosB, it also decreased in these regions in the morphine-withdrawn offspring. These results demonstrated that parental morphine exposure affects NOR memory, and decreased the level of histone H3 acetylation and ΔFosB in the PFC and hippocampus. Taken together, the effect of morphine might be transmitted to the next generation even after stop consuming morphine.
Collapse
Affiliation(s)
| | - Pardis Asgari
- Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran
| | - Sarah Mahboubi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Ghorbangol Ashabi
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kiyana Rohbani
- Department of Molecular and Cellular Sciences, Faculty of Advanced Sciences and Technology, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Saba Sabzevari
- Department of Molecular and Cellular Sciences, Faculty of Advanced Sciences and Technology, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Haniyeh Soltani
- Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran
| | - Solmaz Khalifeh
- Cognitive and Neuroscience Research Center (CNRC), Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad-Reza Zarrindast
- Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran; Cognitive and Neuroscience Research Center (CNRC), Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Endocrinology and Metabolism Research Institute, Tehran University of Medical Science, Tehran, Iran.
| |
Collapse
|
23
|
Shaw K, da Silva RDCO. Look Upstream: Measurement for Innovation on the Upper Rio Negro of the Amazon Basin. Front Pediatr 2020; 8:567257. [PMID: 33585359 PMCID: PMC7875013 DOI: 10.3389/fped.2020.567257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 12/23/2020] [Indexed: 11/22/2022] Open
Abstract
The growth of the randomized controlled trial (RCT) as the "gold standard" for evaluation has justly been praised as an advance in the professionalization of social programs and projects, an "adoption of science" - in the words of the Lancet. None the less, the emphasis on the RCT biases funding for projects that distribute private goods and which focus on "low hanging fruit" in health, nutrition, and sanitation, simply because those areas lend themselves to the sort of measurement that works with RCTs. As a result, many project developers in the government and NGO sectors lament that a hegemonic focus on RCTs impedes creativity or new models that challenge traditional paradigms. This case study of CanalCanoa, a community video coaching project for indigenous parents of young children in the Rio Negro region of the Amazon Basin, offers techniques to measure for innovation. Instead of developing a new RCT for an extremely diverse population (27 ethnic groups) where traditional childcare methods are in historical flux because of urbanization, CanalCanoa measured variables shown by previous RCTs to be causally connected with positive development results. By researching the impact of the intervention on nutrition, language (multilingualism, use of traditional songs and stories), and social network expansion, CanalCanoa measured upstream indicators, thus mixing scientific rigor with an opportunity for innovation and providing important insight and reform of a theory of change.
Collapse
Affiliation(s)
- Kurt Shaw
- Usina da Imaginação, Florianópolis, Brazil
| | | |
Collapse
|
24
|
Arndt DL, Wukitsch TJ, Garcia EJ, Cain M. Histone deacetylase inhibition differentially attenuates cue-induced reinstatement: An interaction of environment and acH3K9 expression in the dorsal striatum. Behav Neurosci 2019; 133:478-488. [PMID: 31343201 DOI: 10.1037/bne0000333] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Substance use disorder is driven by complex gene-environment interactions. Epigenetic histone regulation is a significant contributor to several behavioral phenotypes of drug abuse. The primary epigenetic mechanisms that drive drug taking and drug seeking are still being investigated, and it is unclear how environmental conditions alter epigenetic histone acetylation to change behaviors geared toward drug reward. This study examined the effects of environmental condition on amphetamine self-administration, and whether drug-taking and drug-seeking behaviors could be influenced through inhibition of an epigenetic regulator, histone deacetylase (HDAC). Male rats reared for 30 days in enriched (EC), isolated (IC), or standard conditions (SC) prior to amphetamine (0.03, 0.05, 0.1 mg/kg/infusion, IV) self-administration, extinction, and reinstatement sessions. The HDAC inhibitor, Trichostatin A (TsA; 0.3 mg/kg, IV), was injected 30 min prior to operant sessions. After amphetamine-induced reinstatement (0.25 mg/kg, subcutaneous [s.c.]), tissue was extracted for Western blot analyses of acetylated histone H3 lysine 9 (acH3K9) in the nucleus accumbens (NAc) and dorsal striatum (DSt). While TsA did not significantly affect amphetamine self-administration or extinction, TsA decreased cue-, but not drug-induced reinstatement in IC rats only. In the DSt, but not in the NAc, IC rats exhibited significantly less acH3K9 expression than EC and SC rats, irrespective of TsA treatment. HDAC inhibition decreases cue-induced reinstatement of amphetamine seeking in IC rats. While IC rats exhibit less acH3K9 expression in the DSt, future studies are needed to elucidate the critical epigenetic factors that drive substance abuse, particularly in vulnerable populations. (PsycINFO Database Record (c) 2019 APA, all rights reserved).
Collapse
Affiliation(s)
| | | | | | - Mary Cain
- Department of Psychological Sciences
| |
Collapse
|
25
|
Hsieh MT, Lin CC, Lee CT, Huang TL. Abnormal Brain-Derived Neurotrophic Factor Exon IX Promoter Methylation, Protein, and mRNA Levels in Patients with Major Depressive Disorder. J Clin Med 2019; 8:jcm8050568. [PMID: 31027379 PMCID: PMC6571872 DOI: 10.3390/jcm8050568] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/24/2019] [Accepted: 04/24/2019] [Indexed: 12/15/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) exon IX promoter methylation levels, serum BDNF protein levels, and serum mRNA levels were investigated in patients with major depressive disorder (MDD) and healthy controls. Over two years, 51 patients with MDD and 62 healthy controls were recruited. Peripheral blood was drawn from all participants to analyze the BDNF exon IX promoter methylation levels as well as serum BDNF protein and mRNA levels, at baseline and after four weeks of antidepressant treatment. Methylation sequential analysis showed that patients with MDD (n = 39) had a higher methylation level at CpG site 217 and lower methylation levels at CpG site 327 and CpG site 362. Drug responders (n = 25) had a higher methylation level at CpG site 24 and CpG site 324 than the non-responders (n = 11). Patients with MDD had a lower serum BDNF protein and mRNA levels than the healthy controls. In conclusion, these results showed that BDNF exon IX promoter methylation levels, serum BDNF protein level, and serum BDNF mRNA level could contribute to the pathophysiology of a major depressive disorder.
Collapse
Affiliation(s)
- Men-Ting Hsieh
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan.
| | - Chin-Chuen Lin
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan.
| | - Chien-Te Lee
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan.
| | - Tiao-Lai Huang
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan.
| |
Collapse
|
26
|
Rahul DR, Ponniah RJ. Decoding the biology of language and its implications in language acquisition. J Biosci 2019; 44:25. [PMID: 30837376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Associating human genetic makeup with the faculty of language has long been a goal for biolinguistics. This stimulated the idea that language is attributed to genes and language disabilities are caused by genetic mutations. However, application of genetic knowledge on language intervention is still a gap in the existing literature. In an effort to bridge this gap, this article presents an account of genetic and neural associations of language and synthesizes the genetic, neural, epigenetic and environmental facets involved in language. In addition to describing the association of genes with language, the neural and epigenetic aspects of language are also explored. Further, the environmental aspects of language such as language input, emotion and cognition are also traced back to gene expressions. Therefore, effective language intervention for language learning difficulties must offer genetics-informed solutions, both linguistic and medical.
Collapse
Affiliation(s)
- D R Rahul
- National Institute of Technology, Tiruchirappalli, Tamil Nadu, India
| | | |
Collapse
|
27
|
|
28
|
Faggi L, Porrini V, Lanzillotta A, Benarese M, Mota M, Tsoukalas D, Parrella E, Pizzi M. A Polyphenol-Enriched Supplement Exerts Potent Epigenetic-Protective Activity in a Cell-Based Model of Brain Ischemia. Nutrients 2019; 11:nu11020345. [PMID: 30736313 PMCID: PMC6412333 DOI: 10.3390/nu11020345] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/27/2019] [Accepted: 02/03/2019] [Indexed: 12/12/2022] Open
Abstract
Bioactive components, due in part to their epigenetic properties, are beneficial for preventing several human diseases including cerebrovascular pathologies. However, no clear demonstration supports the idea that these molecules still conserve their epigenetic effects when acting at very low concentrations reproducing the brain levels achieved after oral administration of a micronutrient supplement. In the present study, we used a cellular model of brain ischemia to investigate the neuroprotective and epigenetic activities of a commercially available micronutrient mixture (polyphenol-enriched micronutrient mixture, PMM) enriched in polyphenols ((-)-epigallocatechin-3-gallate, quercetin, resveratrol), α-lipoic acid, vitamins, amino acids and other micronutrients. Mimicking the suggested dietary supplementation, primary cultures of mouse cortical neurons were pre-treated with PMM and then subjected to oxygen glucose deprivation (OGD). Pre-treatment with PMM amounts to provide bioactive components in the medium in the nanomolar range potently prevented neuronal cell death. The protection was associated with the deacetylation of the lysin 310 (K310) on NF-κB/RelA as well as the deacetylation of H3 histones at the promoter of Bim, a pro-apoptotic target of ac-RelA(K310) in brain ischemia. Epigenetic regulators known to shape the acetylation state of ac-RelA(K310) moiety are the histone acetyl transferase CBP/p300 and the class III histone deacetylase sirtuin-1. In view of that evidence, the protection we here report unveils the efficacy of bioactive components endowed with either inhibitory activity on CBP/p300 or stimulating activity on the AMP-activated protein kinase–sirtuin 1 pathway. Our results support a potential synergistic effect of micronutrients in the PMM, suggesting that the intake of a polyphenol-based micronutrient mixture can reduce neuronal vulnerability to stressful conditions at concentrations compatible with the predicted brain levels reached by a single constituent after an oral dose of PMM.
Collapse
Affiliation(s)
- Lara Faggi
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
| | - Vanessa Porrini
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
| | - Annamaria Lanzillotta
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
| | - Marina Benarese
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
| | - Mariana Mota
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
| | - Dimitris Tsoukalas
- European Institute of Nutritional Medicine, E.I.Nu.M., Viale Liegi 44, 00198 Rome, Italy.
| | - Edoardo Parrella
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
| | - Marina Pizzi
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
| |
Collapse
|
29
|
Richter-Levin G, Stork O, Schmidt MV. Animal models of PTSD: a challenge to be met. Mol Psychiatry 2019; 24:1135-1156. [PMID: 30816289 PMCID: PMC6756084 DOI: 10.1038/s41380-018-0272-5] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 08/13/2018] [Accepted: 09/11/2018] [Indexed: 02/07/2023]
Abstract
Recent years have seen increased interest in psychopathologies related to trauma exposure. Specifically, there has been a growing awareness to posttraumatic stress disorder (PTSD) in part due to terrorism, climate change-associated natural disasters, the global refugee crisis, and increased violence in overpopulated urban areas. However, notwithstanding the increased awareness to the disorder, the increasing number of patients, and the devastating impact on the lives of patients and their families, the efficacy of available treatments remains limited and highly unsatisfactory. A major scientific effort is therefore devoted to unravel the neural mechanisms underlying PTSD with the aim of paving the way to developing novel or improved treatment approaches and drugs to treat PTSD. One of the major scientific tools used to gain insight into understanding physiological and neuronal mechanisms underlying diseases and for treatment development is the use of animal models of human diseases. While much progress has been made using these models in understanding mechanisms of conditioned fear and fear memory, the gained knowledge has not yet led to better treatment options for PTSD patients. This poor translational outcome has already led some scientists and pharmaceutical companies, who do not in general hold opinions against animal models, to propose that those models should be abandoned. Here, we critically examine aspects of animal models of PTSD that may have contributed to the relative lack of translatability, including the focus on the exposure to trauma, overlooking individual and sex differences, and the contribution of risk factors. Based on findings from recent years, we propose research-based modifications that we believe are required in order to overcome some of the shortcomings of previous practice. These modifications include the usage of animal models of PTSD which incorporate risk factors and of the behavioral profiling analysis of individuals in a sample. These modifications are aimed to address factors such as individual predisposition and resilience, thus taking into consideration the fact that only a fraction of individuals exposed to trauma develop PTSD. We suggest that with an appropriate shift of practice, animal models are not only a valuable tool to enhance our understanding of fear and memory processes, but could serve as effective platforms for understanding PTSD, for PTSD drug development and drug testing.
Collapse
Affiliation(s)
- Gal Richter-Levin
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel. .,The Integrated Brain and Behavior Research Center (IBBR), University of Haifa, Haifa, Israel. .,Psychology Department, University of Haifa, Haifa, Israel.
| | - Oliver Stork
- 0000 0001 1018 4307grid.5807.aDepartment of Genetics & Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany ,grid.452320.2Center for Behavioral Brain Sciences, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Mathias V. Schmidt
- 0000 0000 9497 5095grid.419548.5Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
30
|
Kilvitis HJ, Ardia DR, Thiam M, Martin LB. Corticosterone is correlated to mediators of neural plasticity and epigenetic potential in the hippocampus of Senegalese house sparrows (Passer domesticus). Gen Comp Endocrinol 2018; 269:177-183. [PMID: 30257180 DOI: 10.1016/j.ygcen.2018.09.014] [Citation(s) in RCA: 5] [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: 03/13/2018] [Revised: 09/19/2018] [Accepted: 09/21/2018] [Indexed: 10/28/2022]
Abstract
Our previous research on range-expanding house sparrows in Kenya revealed that (i) range-edge birds released more corticosterone (CORT) in response to a stressor than range-core birds, ii) that range-edge birds were more exploratory than range-core birds, and that (iii) all birds exhibited extensive variation in genome-wide DNA methylation among individuals, regardless of their position along the range expansion. Within the hippocampus, mediators of neural plasticity such as brain-derived neurotrophic factor (BDNF), can influence and be influenced by CORT, hippocampus-associated behaviors and regulatory epigenetic modification enzymes. Here, we investigated whether individuals and populations colonizing a new geographic range, Senegal, vary in the expression of BDNF and DNA methyltransferases (DNMTs) within the hippocampus and the release of CORT in response to a stressor. DNMT expression is an important mediator of epigenetic potential, the propensity of a genome to capacitate phenotypic variation via mechanisms such as DNA methylation. We surveyed three populations across Senegal, predicting that hippocampal BDNF and DNMT expression would be highest at the range-edge, and that BDNF and DNMT would be inversely related to one another, but would each positively covary with CORT within individuals. We found a nonlinear relationship between CORT and BDNF expression within individuals. Moreover, we found that CORT positively covaried with DNMT1 expression in a more recently established population, while the reverse was true in the oldest population (i.e. at the range-core). Our study is among the first to explore whether and how variation in CORT regulation contributes to variation in mediators of neural plasticity and epigenetic potential within the hippocampus of a range-expanding vertebrate.
Collapse
Affiliation(s)
- Holly J Kilvitis
- University of South Florida, Department of Integrative Biology, Tampa, FL, USA.
| | - Daniel R Ardia
- Franklin & Marshall College, Department of Biology, Lancaster, PA, USA
| | - Massamba Thiam
- Universite Cheikh Anta Diop, Department of Biology, Dakar, Senegal
| | - Lynn B Martin
- University of South Florida, Department of Global Health, Tampa, FL, USA
| |
Collapse
|
31
|
Abstract
This commentary reviews the concept of experience-dependent epigenetic modifications in the CNS as a core mechanism underlying individuality and individuation at the behavioral level. I use the term individuation to refer to the underlying neurobiological processes that result in individuality, with the discussion focusing on individuality of cognitive, emotional, and behavioral repertoire. The review describes recent work supporting the concept of neuroepigenetic mechanisms underlying individuation, possible roles of transgenerational effects, and implications for precision medicine.
Collapse
Affiliation(s)
- J David Sweatt
- Department of Pharmacology, Vanderbilt University, Nashville, TN
| |
Collapse
|
32
|
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]
|
33
|
Memory Function in Feeding Habit Transformation of Mandarin Fish ( Siniperca chuatsi). Int J Mol Sci 2018; 19:ijms19041254. [PMID: 29690543 PMCID: PMC5979507 DOI: 10.3390/ijms19041254] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 04/08/2018] [Accepted: 04/10/2018] [Indexed: 12/03/2022] Open
Abstract
Mandarin fish refuse dead prey fish or artificial diets and can be trained to transform their inborn feeding habit. To investigate the effect of memory on feeding habit transformation, we compared the reaction time to dead prey fish and the success rate of feeding habit transformation to dead prey fish with training of mandarin fish in the 1st experimental group (trained once) and the 2nd experimental group (trained twice). The mandarin fish in the 2nd group had higher success rate of feeding habit transformation (100%) than those in the 1st group (67%), and shorter reaction time to dead prey fish (<1 s) than those in the 1st group (>1 s). Gene expression of cAMP responsive element binding protein I (Creb I), brain-derived neurotrophic factor (Bdnf), CCAAT enhancer binding protein delta (C/EBPD), fos-related antigen 2 (Fra2), and proto-oncogenes c-fos (c-fos) involved in long-term memory formation were significantly increased in the 2nd group after repeated training, and taste 1 receptor member 1 (T1R1), involved in feeding habit formation, was significantly increased in brains of the 2nd group after repeated training. DNA methylation levels at five candidate CpG (cytosine–guanine) sites contained in the predicted CpG island in the 5′-flanking region of T1R1 were significantly decreased in brains of the 2nd group compared with that of the 1st group. These results indicated that the repeated training can improve the feeding habit transformation through the memory formation of accepting dead prey fish. DNA methylation of the T1R1 might be a regulatory factor for feeding habit transformation from live prey fish to dead prey fish in mandarin fish.
Collapse
|
34
|
Kilvitis HJ, Hanson H, Schrey AW, Martin LB. Epigenetic Potential as a Mechanism of Phenotypic Plasticity in Vertebrate Range Expansions. Integr Comp Biol 2018; 57:385-395. [PMID: 28859411 DOI: 10.1093/icb/icx082] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
SYNOPSIS During range expansions, organisms are often exposed to multiple pressures, including novel enemies (i.e., predators, competitors and/or parasites) and unfamiliar or limited resources. Additionally, small propagule sizes at range edges can result in genetic founder effects and bottlenecks, which can affect phenotypic diversity and thus selection. Despite these obstacles, individuals in expanding populations often thrive at the periphery of a range, and this success may be mediated by phenotypic plasticity. Increasing evidence suggests that epigenetic mechanisms may underlie such plasticity because they allow for more rapid phenotypic responses to novel environments than are possible via the accumulation of genetic variation. Here, we review how molecular epigenetic mechanisms could facilitate plasticity in range-expanding organisms, emphasizing the roles of DNA methylation and other epigenetic marks in the physiological regulatory networks that drive whole-organism performance. We focus on the hypothalamic-pituitary-adrenal (HPA) axis, arguing that epigenetically-mediated plasticity in the regulation of glucocorticoids in particular might strongly impact range expansions. We hypothesize that novel environments release and/or select for epigenetic potential in HPA variation and hence organismal performance and ultimately fitness.
Collapse
Affiliation(s)
- Holly J Kilvitis
- Department of Integrative Biology, University of South Florida, Tampa, FL 33620, USA
| | - Haley Hanson
- Department of Integrative Biology, University of South Florida, Tampa, FL 33620, USA
| | - Aaron W Schrey
- Department of Biology, Armstrong State University, Savannah, GA 31419, USA
| | - Lynn B Martin
- Department of Integrative Biology, University of South Florida, Tampa, FL 33620, USA
| |
Collapse
|
35
|
Bales KL, Ianov L, Kennedy AJ, Sweatt JD, Gross AK. Autosomal dominant retinitis pigmentosa rhodopsin mutant Q344X drives specific alterations in chromatin complex gene transcription. Mol Vis 2018; 24:153-164. [PMID: 29463953 PMCID: PMC5815338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 02/13/2018] [Indexed: 10/25/2022] Open
Abstract
Purpose Epigenetic and transcriptional mechanisms have been shown to contribute to long-lasting functional changes in adult neurons. The purpose of this study was to identify any such modifications in diseased retinal tissues from a mouse model of rhodopsin mutation-associated autosomal dominant retinitis pigmentosa (ADRP), Q344X, relative to age-matched wild-type (WT) controls. Methods We performed RNA sequencing (RNA-seq) at poly(A) selected RNA to profile the transcriptional patterns in 3-week-old ADRP mouse model rhodopsin Q344X compared to WT controls. Differentially expressed genes were determined by DESeq2 using the Benjamini & Hochberg p value adjustment and an absolute log2 fold change cutoff. Quantitative western blots were conducted to evaluate protein expression levels of histone H3 phosphorylated at serine 10 and histone H4. qRT-PCR was performed to validate the expression patterns of differentially expressed genes. Results We observed significant differential expression in 2151 genes in the retina of Q344X mice compared to WT controls, including downregulation in the potassium channel gene, Kcnv2, and differential expression of histone genes, including the H1 family histone member, H1foo; the H3 histone family 3B, H3f3b; and the histone deacetylase 9, Hdac9. Quantitative western blots revealed statistically significant decreased protein expression of both histone H3 phosphorylated at serine 10 and histone H4 in 3-week-old Q344X retinas. Furthermore, qRT-PCR performed on select differentially expressed genes based on our RNA-seq results revealed matched expression patterns of up or downregulation. Conclusions These findings provide evidence that transcriptomic alterations occur in the ADRP mouse model rhodopsin Q344X retina and that these processes may contribute to the dysfunction and neurodegeneration seen in this animal model.
Collapse
Affiliation(s)
- Katie L. Bales
- School of Optometry, Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL
| | - Lara Ianov
- School of Medicine, Civitan International Research Center, University of Alabama at Birmingham, AL
| | - Andrew J. Kennedy
- Department of Chemistry and Biochemistry, Bates College, Lewiston, ME
| | - J. David Sweatt
- School of Medicine, Department of Pharmacology, Vanderbilt University, TN
| | - Alecia K. Gross
- School of Optometry, Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL
- School of Medicine, Civitan International Research Center, University of Alabama at Birmingham, AL
| |
Collapse
|
36
|
Synergistic Association of Valproate and Resveratrol Reduces Brain Injury in Ischemic Stroke. Int J Mol Sci 2018; 19:ijms19010172. [PMID: 29316653 PMCID: PMC5796121 DOI: 10.3390/ijms19010172] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 12/15/2017] [Accepted: 01/02/2018] [Indexed: 11/16/2022] Open
Abstract
Histone deacetylation, together with altered acetylation of NF-κB/RelA, encompassing the K310 residue acetylation, occur during brain ischemia. By restoring the normal acetylation condition, we previously reported that sub-threshold doses of resveratrol and entinostat (MS-275), respectively, an activator of the AMP-activated kinase (AMPK)-sirtuin 1 pathway and an inhibitor of class I histone deacetylases (HDACs), synergistically elicited neuroprotection in a mouse model of ischemic stroke. To improve the translational power of this approach, we investigated the efficacy of MS-275 replacement with valproate, the antiepileptic drug also reported to be a class I HDAC blocker. In cortical neurons previously exposed to oxygen glucose deprivation (OGD), valproate elicited neuroprotection at 100 nmol/mL concentration when used alone and at 1 nmol/mL concentration when associated with resveratrol (3 nmol/mL). Resveratrol and valproate restored the acetylation of histone H3 (K9/18), and they reduced the RelA(K310) acetylation and the Bim level in neurons exposed to OGD. Chromatin immunoprecipitation analysis showed that the synergistic drug association impaired the RelA binding to the Bim promoter, as well as the promoter-specific H3 (K9/18) acetylation. In mice subjected to 60 min of middle cerebral artery occlusion (MCAO), the association of resveratrol 680 µg/kg and valproate 200 µg/kg significantly reduced the infarct volume as well as the neurological deficits. The present study suggests that valproate and resveratrol may represent a promising ready-to-use strategy to treat post-ischemic brain damage.
Collapse
|
37
|
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]
|
38
|
Abstract
Studies of the mammalian nervous system have revealed widespread epigenetic regulation underlying gene expression intrinsic to basic neurobiological function as well as neurological disease. Over the past decade, a critical role has emerged for the neural regulation of chromatin-modifying enzymes during both development and adulthood, and in response to external stimuli. These biochemical data are complemented by numerous next generation sequencing (NGS) studies that quantify the extent of chromatin and DNA modifications in neurons. Neuroepigenetic editing tools can be applied to distinguish between the mere presence and functional relevance of such modifications to neural transcription and animal behavior. This review discusses current advances in neuroepigenetic editing, highlighting methodological considerations pertinent to neuroscience, such as delivery methods and the spatiotemporal specificity of editing. Although neuroepigenetic editing is a nascent field, the studies presented in this review demonstrate the enormous potential of this approach for basic neurobiological research and therapeutic application.
Collapse
Affiliation(s)
- Peter J Hamilton
- The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY, New York, USA
| | - Carissa J Lim
- Department of Pharmacology, The University of Pennsylvania, Philadelphia, PA, USA
| | - Eric J Nestler
- The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY, New York, USA
| | - Elizabeth A Heller
- Department of Pharmacology, The University of Pennsylvania, Philadelphia, PA, USA. .,Penn Epigenetics Institute, The University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
39
|
Cross-talk between the epigenome and neural circuits in drug addiction. PROGRESS IN BRAIN RESEARCH 2017; 235:19-63. [PMID: 29054289 DOI: 10.1016/bs.pbr.2017.08.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Drug addiction is a behavioral disorder characterized by dysregulated learning about drugs and associated cues that result in compulsive drug seeking and relapse. Learning about drug rewards and predictive cues is a complex process controlled by a computational network of neural connections interacting with transcriptional and molecular mechanisms within each cell to precisely guide behavior. The interplay between rapid, temporally specific neuronal activation, and longer-term changes in transcription is of critical importance in the expression of appropriate, or in the case of drug addiction, inappropriate behaviors. Thus, these factors and their interactions must be considered together, especially in the context of treatment. Understanding the complex interplay between epigenetic gene regulation and circuit connectivity will allow us to formulate novel therapies to normalize maladaptive reward behaviors, with a goal of modulating addictive behaviors, while leaving natural reward-associated behavior unaffected.
Collapse
|
40
|
Sweatt JD, Tamminga CA. An epigenomics approach to individual differences and its translation to neuropsychiatric conditions. DIALOGUES IN CLINICAL NEUROSCIENCE 2017. [PMID: 27757063 PMCID: PMC5067146 DOI: 10.31887/dcns.2016.18.3/dsweatt] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
This review concerns epigenetic mechanisms and their roles in conferring interindividual differences, especially as related to experientially acquired and genetically driven changes in central nervous system (CNS) function. In addition, the review contains commentary regarding the possible ways in which epigenomic changes may contribute to neuropsychiatric conditions and disorders and ways in which epigenotyping might be cross-correlated with clinical phenotyping in the context of precision medicine. The review begins with a basic description of epigenetic marking in the CNS and how these changes are powerful regulators of gene readout. Means for characterizing the individual epigenotype are briefly described, with a focus on DNA cytosine methylation as a readily measurable, stable epigenetic mark. This background enables a discussion of how “epigenotyping” might be integrated along with genotyping and deep phenotyping as a means of implementing advanced precision medicine. Finally, the commentary addresses two exemplars when considering how epigenotype may correlate with and modulate cognitive and behavioral phenotype: schizophrenia and Rett syndrome. These two disorders provide an interesting compare-and-contrast example regarding possible epigenotypic regulation of behavior: whereas Rett syndrome is clearly established as being caused by disruption of the function of an epigenetic “reader” of the DNA cytosine methylome—methyl-CpG-binding protein 2 (MeCP2)—the case for a role for epigenetic mechanisms in schizophrenia is still quite speculative.
Collapse
Affiliation(s)
- J David Sweatt
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Carol A Tamminga
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
41
|
Lancaster K, Morris JP, Connelly JJ. Neuroimaging Epigenetics: Challenges and Recommendations for Best Practices. Neuroscience 2017; 370:88-100. [PMID: 28801185 DOI: 10.1016/j.neuroscience.2017.08.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 07/31/2017] [Accepted: 08/01/2017] [Indexed: 12/14/2022]
Abstract
Neuroimaging epigenetics is an interdisciplinary application of epigenetics to cognitive neuroscience that seeks to identify molecular and neural predictors of human behavior. This approach can be sensitive to the dynamic interaction between biological predisposition and environmental influences, and is potentially more informative than an approach using static genetic code. Recent work in this field has generated considerable enthusiasm, yet caution is warranted since any novel cross-disciplinary approach lacks a set of established conventions or standards. In this paper we review existing research in the field of imaging epigenetics, outline important caveats and considerations, and suggest a set of guidelines for researchers conducting this work.
Collapse
|
42
|
Lo SQ, Sng JCG, Augustine GJ. Defining a critical period for inhibitory circuits within the somatosensory cortex. Sci Rep 2017; 7:7271. [PMID: 28779074 PMCID: PMC5544762 DOI: 10.1038/s41598-017-07400-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 06/28/2017] [Indexed: 11/21/2022] Open
Abstract
Although experience-dependent changes in brain inhibitory circuits are thought to play a key role during the “critical period” of brain development, the nature and timing of these changes are poorly understood. We examined the role of sensory experience in sculpting an inhibitory circuit in the primary somatosensory cortex (S1) of mice by using optogenetics to map the connections between parvalbumin (PV) expressing interneurons and layer 2/3 pyramidal cells. Unilateral whisker deprivation decreased the strength and spatial range of inhibitory input provided to pyramidal neurons by PV interneurons in layers 2/3, 4 and 5. By varying the time when sensory input was removed, we determined that the critical period closes around postnatal day 14. This yields the first precise time course of critical period plasticity for an inhibitory circuit.
Collapse
Affiliation(s)
- Shun Qiang Lo
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.,Institute of Molecular and Cell Biology (A*STAR), Singapore, Singapore.,Marine Biological Laboratory, Woods Hole, USA
| | - Judy C G Sng
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Singapore Institute of Clinical Sciences, Agency for Science and Technology (A*STAR), Singapore, Singapore
| | - George J Augustine
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore. .,Institute of Molecular and Cell Biology (A*STAR), Singapore, Singapore. .,Marine Biological Laboratory, Woods Hole, USA.
| |
Collapse
|
43
|
Ranjan V, Singh S, Siddiqui SA, Tripathi S, Khan MY, Prakash A. Differential Histone Acetylation in Sub-Regions of Bed Nucleus of the Stria Terminalis Underlies Fear Consolidation and Extinction. Psychiatry Investig 2017; 14:350-359. [PMID: 28539954 PMCID: PMC5440438 DOI: 10.4306/pi.2017.14.3.350] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 05/31/2016] [Accepted: 07/06/2016] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE The hallmark of anxiety disorders is excessive fear. Previous studies have suggested that selective neural projections from Basal nucleus of stria terminalis (BNST) to amygdala and vice-versa precisely control the fear learning process. However the exact mechanism how the BNST controls fear consolidation and its extinction is largely unknown. In the present study we observed the changes in the BNST sub-regions following fear conditioning and its extinction. METHODS The change in the number of positive neurons was determined by immunohistochemistry for Acetyl H3 (Histone 3), Acetyl H4 (Histone 4), cAMP response element binding Protein (CBP) and c-fos in three sub-regions of the BNST namely the anterio-lateral BNST (STLP) and anterio-medial BNST (STMA), and lateral-ventral BNST (STLV) of rats subjected to auditory fear conditioning and extinction. RESULTS We found significant increase in the number of CBP, acetyl H3 and acetyl H4 positive neurons in the STMA and STLV but not in the STLP after fear conditioning. However, following fear extinction the number of CBP, acetyl H3 and acetyl H4 positive neurons increased significantly in the STLP but not in the STMA and STLV. Similar changes were observed in the number of c-fos positive neurons after fear consolidation and extinction. CONCLUSION The results from this study suggest that the differential histone acetylation in the different sub-regions of the BNST following fear learning and its extinction may be responsible for changes in the neuronal activation patterns resulting in either fear or less fear.
Collapse
Affiliation(s)
- Vandana Ranjan
- Department of Biochemistry, Dr. R M L Avadh University, Lucknow, India
| | - Sanjay Singh
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | | | - Sukanya Tripathi
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Mohd Yahiya Khan
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Anand Prakash
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| |
Collapse
|
44
|
Phillips C. Physical Activity Modulates Common Neuroplasticity Substrates in Major Depressive and Bipolar Disorder. Neural Plast 2017; 2017:7014146. [PMID: 28529805 PMCID: PMC5424494 DOI: 10.1155/2017/7014146] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 02/10/2017] [Accepted: 03/16/2017] [Indexed: 12/13/2022] Open
Abstract
Mood disorders (MDs) are chronic, recurrent mental diseases that affect millions of individuals worldwide. Although the biogenic amine model has provided some clinical utility, a need remains to better understand the interrelated mechanisms that contribute to neuroplasticity deficits in MDs and the means by which various therapeutics mitigate them. Of those therapeutics being investigated, physical activity (PA) has shown clear and consistent promise. Accordingly, the aims of this review are to (1) explicate key modulators, processes, and interactions that impinge upon multiple susceptibility points to effectuate neuroplasticity deficits in MDs; (2) explore the putative mechanisms by which PA mitigates these features; (3) review protocols used to induce the positive effects of PA in MDs; and (4) highlight implications for clinicians and researchers.
Collapse
|
45
|
Shpigler HY, Saul MC, Murdoch EE, Cash-Ahmed AC, Seward CH, Sloofman L, Chandrasekaran S, Sinha S, Stubbs LJ, Robinson GE. Behavioral, transcriptomic and epigenetic responses to social challenge in honey bees. GENES BRAIN AND BEHAVIOR 2017; 16:579-591. [DOI: 10.1111/gbb.12379] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 03/03/2017] [Accepted: 03/14/2017] [Indexed: 01/06/2023]
Affiliation(s)
- H. Y. Shpigler
- Carl R. Woese Institute for Genomic Biology; University of Illinois at Urbana-Champaign (UIUC); Urbana IL USA
| | - M. C. Saul
- Carl R. Woese Institute for Genomic Biology; University of Illinois at Urbana-Champaign (UIUC); Urbana IL USA
| | - E. E. Murdoch
- Carl R. Woese Institute for Genomic Biology; University of Illinois at Urbana-Champaign (UIUC); Urbana IL USA
| | - A. C. Cash-Ahmed
- Carl R. Woese Institute for Genomic Biology; University of Illinois at Urbana-Champaign (UIUC); Urbana IL USA
| | - C. H. Seward
- Carl R. Woese Institute for Genomic Biology; University of Illinois at Urbana-Champaign (UIUC); Urbana IL USA
- Department of Cell and Developmental Biology; University of Illinois at Urbana-Champaign (UIUC); Urbana IL USA
| | - L. Sloofman
- Carl R. Woese Institute for Genomic Biology; University of Illinois at Urbana-Champaign (UIUC); Urbana IL USA
- Center for Biophysics and Quantitative Biology; University of Illinois at Urbana-Champaign (UIUC); Urbana IL USA
| | - S. Chandrasekaran
- Harvard Society of Fellows; Harvard University; Cambridge MA USA
- Faculty of Arts and Sciences; Harvard University; Cambridge MA USA
- Broad Institute of MIT and Harvard; Cambridge MA USA
- Department of Biomedical Engineering; University of Michigan; Ann Arbor MI USA
| | - S. Sinha
- Carl R. Woese Institute for Genomic Biology; University of Illinois at Urbana-Champaign (UIUC); Urbana IL USA
- Center for Biophysics and Quantitative Biology; University of Illinois at Urbana-Champaign (UIUC); Urbana IL USA
- Department of Computer Science; University of Illinois at Urbana-Champaign (UIUC); Urbana IL USA
- Department of Entomology; University of Illinois at Urbana-Champaign (UIUC); Urbana IL USA
| | - L. J. Stubbs
- Carl R. Woese Institute for Genomic Biology; University of Illinois at Urbana-Champaign (UIUC); Urbana IL USA
- Department of Cell and Developmental Biology; University of Illinois at Urbana-Champaign (UIUC); Urbana IL USA
- Neuroscience Program; University of Illinois at Urbana-Champaign (UIUC); Urbana IL USA
| | - G. E. Robinson
- Carl R. Woese Institute for Genomic Biology; University of Illinois at Urbana-Champaign (UIUC); Urbana IL USA
- Department of Entomology; University of Illinois at Urbana-Champaign (UIUC); Urbana IL USA
- Neuroscience Program; University of Illinois at Urbana-Champaign (UIUC); Urbana IL USA
| |
Collapse
|
46
|
MacDuffie KE, Strauman TJ. Understanding Our Own Biology: The Relevance of Auto-Biological Attributions for Mental Health. CLINICAL PSYCHOLOGY-SCIENCE AND PRACTICE 2017. [DOI: 10.1111/cpsp.12188] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
47
|
Hemstedt TJ, Lattal KM, Wood MA. Reconsolidation and extinction: Using epigenetic signatures to challenge conventional wisdom. Neurobiol Learn Mem 2017; 142:55-65. [PMID: 28119018 DOI: 10.1016/j.nlm.2017.01.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 01/15/2017] [Accepted: 01/16/2017] [Indexed: 12/17/2022]
Abstract
Epigenetic mechanisms have the potential to give rise to lasting changes in cell function that ultimately can affect behavior persistently. This concept is especially interesting with respect to fear reconsolidation and fear memory extinction. These two behavioral approaches are used in the laboratory to investigate how fear memory can be attenuated, which becomes important when searching for therapeutic intervention to treat anxiety disorders and post-traumatic stress disorder. Here we review the role of several key epigenetic mechanisms in reconsolidation and extinction of learned fear and their potential to persistently alter behavioral responses to conditioned cues. We also briefly discuss how epigenetic mechanisms may establish persistent behaviors that challenge our definitions of extinction and reconsolidation.
Collapse
Affiliation(s)
- Thekla J Hemstedt
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA; Center for the Neurobiology of Learning and Memory, Irvine, CA, USA
| | - K Matthew Lattal
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Marcelo A Wood
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA; Center for the Neurobiology of Learning and Memory, Irvine, CA, USA.
| |
Collapse
|
48
|
Labouesse MA, Dong E, Grayson DR, Guidotti A, Meyer U. Maternal immune activation induces GAD1 and GAD2 promoter remodeling in the offspring prefrontal cortex. Epigenetics 2016; 10:1143-55. [PMID: 26575259 DOI: 10.1080/15592294.2015.1114202] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Maternal infection during pregnancy increases the risk of neurodevelopmental disorders in the offspring. In addition to its influence on other neuronal systems, this early-life environmental adversity has been shown to negatively affect cortical γ-aminobutyric acid (GABA) functions in adult life, including impaired prefrontal expression of enzymes required for GABA synthesis. The underlying molecular processes, however, remain largely unknown. In the present study, we explored whether epigenetic modifications represent a mechanism whereby maternal infection during pregnancy can induce such GABAergic impairments in the offspring. We used an established mouse model of prenatal immune challenge that is based on maternal treatment with the viral mimetic poly(I:C). We found that prenatal immune activation increased prefrontal levels of 5-methylated cytosines (5mC) and 5-hydroxymethylated cytosines (5hmC) in the promoter region of GAD1, which encodes the 67-kDa isoform of the GABA-synthesising enzyme glutamic acid decarboxylase (GAD67). The early-life challenge also increased 5mC levels at the promoter region of GAD2, which encodes the 65-kDa GAD isoform (GAD65). These effects were accompanied by elevated GAD1 and GAD2 promoter binding of methyl CpG-binding protein 2 (MeCP2) and by reduced GAD67 and GAD65 mRNA expression. Moreover, the epigenetic modifications at the GAD1 promoter correlated with prenatal infection-induced impairments in working memory and social interaction. Our study thus highlights that hypermethylation of GAD1 and GAD2 promoters may be an important molecular mechanism linking prenatal infection to presynaptic GABAergic impairments and associated behavioral and cognitive abnormalities in the offspring.
Collapse
Affiliation(s)
| | - Erbo Dong
- b Psychiatric Institute, Department of Psychiatry; College of Medicine; University of Illinois at Chicago ; Chicago , Illinois , USA
| | - Dennis Robert Grayson
- b Psychiatric Institute, Department of Psychiatry; College of Medicine; University of Illinois at Chicago ; Chicago , Illinois , USA
| | - Alessandro Guidotti
- b Psychiatric Institute, Department of Psychiatry; College of Medicine; University of Illinois at Chicago ; Chicago , Illinois , USA
| | - Urs Meyer
- a Physiology and Behavior Laboratory; ETH Zurich ; Schwerzenbach , Switzerland.,c Institute of Pharmacology and Toxicology; University of Zurich-Vetsuisse ; Zurich , Switzerland
| |
Collapse
|
49
|
Rosales-Reynoso M, Ochoa-Hernández A, Juárez-Vázquez C, Barros-Núñez P. Epigenetic mechanisms in the development of memory and their involvement in certain neurological diseases. NEUROLOGÍA (ENGLISH EDITION) 2016. [DOI: 10.1016/j.nrleng.2014.02.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
50
|
Abstract
The last decade has been marked by an increased interest in relating epigenetic mechanisms to complex human behaviors, although this interest has not been balanced, accentuating various types of affective and primarily ignoring cognitive functioning. Recent animal model data support the view that epigenetic processes play a role in learning and memory consolidation and help transmit acquired memories even across generations. In this review, we provide an overview of various types of epigenetic mechanisms in the brain (DNA methylation, histone modification, and noncoding RNA action) and discuss their impact proximally on gene transcription, protein synthesis, and synaptic plasticity and distally on learning, memory, and other cognitive functions. Of particular importance are observations that neuronal activation regulates the dynamics of the epigenome's functioning under precise timing, with subsequent alterations in the gene expression profile. In turn, epigenetic regulation impacts neuronal action, closing the circle and substantiating the signaling pathways that underlie, at least partially, learning, memory, and other cognitive processes.
Collapse
|