1
|
Tian L, Li XH, Zhao YL, Yi HY, Liu XR, Yao R, Hou XM, Zhu X, Huo FQ, Chen T, Liang L. DNMT3a Downregulation Ttriggered Upregulation of GABA A Receptor in the mPFC Promotes Paclitaxel-Induced Pain and Anxiety in Male Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2407387. [PMID: 39679872 DOI: 10.1002/advs.202407387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/25/2024] [Indexed: 12/17/2024]
Abstract
Chemotherapeutic agents, such as paclitaxel (PTX), induce neuroplastic changes and alter gene expression in the prefrontal cortex (PFC), which may be associated with chemotherapy-induced pain and negative emotions. Notably, DNA methylation undergoes adaptive changes in neurological disorders, emerging as a promising target for neuromodulation. In this study, systemic administration of PTX leads to a decrease in the expression of the DNA methyltransferase DNMT3a, while concurrently upregulating the expression of Gabrb1 mRNA and its encoded GABAARβ1 protein in the medial PFC (mPFC) of male mice. Overexpression of DNMT3a in the mPFC alleviates PTX-induced pain hypersensitivity, and anxiety-like behavior in these mice. Additionally, it reverses the PTX-induced increase in inhibitory synaptic transmission in the pyramidal neurons of the mPFC. Mechanistically, the upregulation of GABAARβ1 in the mPFC is linked to the reduced expression of DNMT3a and DNA hypomethylation at the promoter region of the Gabrb1 gene. Furthermore, a long-term diet rich in methyl donors alleviates PTX-induced pain hypersensitivity and anxiety-like behavior in mice. These findings suggest that the DNMT3a-mediated upregulation of GABAARβ1 in the mPFC contributes to PTX-induced neuropathic pain and anxiety, highlighting DNA methylation-dependent epigenetic regulation as a potential therapeutic target for addressing chemotherapy-induced cortical dysfunction.
Collapse
Affiliation(s)
- Lixia Tian
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, P. R. China
| | - Xu-Hui Li
- Center for Neuron and Disease, Frontier Institutes of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P. R. China
| | - Yu-Long Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, P. R. China
| | - Hui-Yuan Yi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, P. R. China
| | - Xue-Ru Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, P. R. China
| | - Rongrong Yao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, P. R. China
| | - Xue-Mei Hou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, P. R. China
| | - Xuan Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, P. R. China
| | - Fu-Quan Huo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, P. R. China
| | - Tao Chen
- Department of Anatomy and K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Lingli Liang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, P. R. China
| |
Collapse
|
2
|
Altamirano FG, Castro-Pascual I, Ponce IT, Coria-Lucero CD, Cargnelutti E, Ferramola ML, Delgado MS, Anzulovich AC, Lacoste MG. Late-Onset Caloric Restriction Improves Cognitive Performance and Restores Circadian Patterns of Neurotrophic, Clock, and Epigenetic Factors in the Hippocampus of Old Male Rats. J Gerontol A Biol Sci Med Sci 2024; 80:glae252. [PMID: 39447038 DOI: 10.1093/gerona/glae252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Indexed: 10/26/2024] Open
Abstract
Aging is a complex multifactorial process that results in a general functional decline, including cognitive impairment. Caloric restriction (CR) can positively influence the aging processes and delay cognitive decline. There is a rhythmic variation in memory and learning processes throughout the day, indicating the involvement of the circadian clock in the regulation of these processes. Despite growing evidence on the efficacy of CR, it has not yet been fully determined whether starting this strategy at an advanced age is beneficial for improving quality of life and eventually, for protection against age-related diseases. Here, we investigated the effect of late-onset CR on the temporal organization of the molecular clock machinery, molecules related to cognitive processes and epigenetic regulation, in the hippocampus of old male rats maintained under constant darkness conditions. Our results evidenced the existence of a highly coordinated temporal organization of Bmal1, Clock, Bdnf, Trkb, Dnmts, Sirt1, and Pgc-1α in the hippocampus of young adult rats. We observed that aging led to cognitive deficits and loss of circadian oscillations of all the above variables. Interestingly, CR restored circadian rhythmicity in all cases and, in addition, improved the cognitive performance of the old animals. This work would highlight the importance of the circadian clock and its synchronization with feeding signals, as the basis of the beneficial effects of CR. Thus, lifestyle modifications, such as CR, might be a powerful intervention to preserve hippocampal circadian organization and cognitive health during aging.
Collapse
Affiliation(s)
- Fernando Gabriel Altamirano
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research - San Luis (IMIBIO-SL), National Council for Scientific and Technical Research (CONICET), San Luis, Argentina
- Faculty of Chemistry, Biochemistry and Pharmacy (FQByF), National University of San Luis (UNSL), San Luis, Argentina
| | - Ivanna Castro-Pascual
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research - San Luis (IMIBIO-SL), National Council for Scientific and Technical Research (CONICET), San Luis, Argentina
- Faculty of Chemistry, Biochemistry and Pharmacy (FQByF), National University of San Luis (UNSL), San Luis, Argentina
| | - Ivana Tamara Ponce
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research - San Luis (IMIBIO-SL), National Council for Scientific and Technical Research (CONICET), San Luis, Argentina
- Faculty of Chemistry, Biochemistry and Pharmacy (FQByF), National University of San Luis (UNSL), San Luis, Argentina
| | - Cinthia Daiana Coria-Lucero
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research - San Luis (IMIBIO-SL), National Council for Scientific and Technical Research (CONICET), San Luis, Argentina
- Faculty of Chemistry, Biochemistry and Pharmacy (FQByF), National University of San Luis (UNSL), San Luis, Argentina
| | - Ethelina Cargnelutti
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research - San Luis (IMIBIO-SL), National Council for Scientific and Technical Research (CONICET), San Luis, Argentina
- Faculty of Chemistry, Biochemistry and Pharmacy (FQByF), National University of San Luis (UNSL), San Luis, Argentina
| | - Mariana Lucila Ferramola
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research - San Luis (IMIBIO-SL), National Council for Scientific and Technical Research (CONICET), San Luis, Argentina
- Faculty of Chemistry, Biochemistry and Pharmacy (FQByF), National University of San Luis (UNSL), San Luis, Argentina
| | - Marcela Silvia Delgado
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research - San Luis (IMIBIO-SL), National Council for Scientific and Technical Research (CONICET), San Luis, Argentina
- Faculty of Chemistry, Biochemistry and Pharmacy (FQByF), National University of San Luis (UNSL), San Luis, Argentina
| | - Ana Cecilia Anzulovich
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research - San Luis (IMIBIO-SL), National Council for Scientific and Technical Research (CONICET), San Luis, Argentina
- Faculty of Chemistry, Biochemistry and Pharmacy (FQByF), National University of San Luis (UNSL), San Luis, Argentina
| | - María Gabriela Lacoste
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research - San Luis (IMIBIO-SL), National Council for Scientific and Technical Research (CONICET), San Luis, Argentina
- Faculty of Chemistry, Biochemistry and Pharmacy (FQByF), National University of San Luis (UNSL), San Luis, Argentina
| |
Collapse
|
3
|
Nayan NM, Husin A, Siran R. The risk of prenatal bisphenol A exposure in early life neurodevelopment: Insights from epigenetic regulation. Early Hum Dev 2024; 198:106120. [PMID: 39293157 DOI: 10.1016/j.earlhumdev.2024.106120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/20/2024]
Abstract
Bisphenols are mainly used as protective coatings for plastics and resin-based materials in various consumer products. Industrial producers have a high demand for bisphenol A (BPA) among all bisphenol substitutes for various consumer products. However, according to reports, prolonged exposure to BPA can cause multiple health issues, including neurodevelopmental disorders in young children. BPA exposure during pregnancy has been considered as the primary cause of increasing the risk of neurological disorders in children as their neural systems are designed to respond to any environmental changes during prenatal life. Recently, there has been an increased focus on the effects of prenatal exposure to BPA, as it has been found to alter gene expression related to epigenetic mechanisms like DNA methylation, histone modification, and microRNA expression. Based on the evidence, frequent interactions can lead to inherited changes in an individual's neural profile. In this review, we delve into the current knowledge regarding the toxicity mechanism of BPA for expecting mothers. Next, we will discuss the possible action of BPA on the epigenetic mechanism during brain development. This is especially important to portray an overview on the role of epigenetic modification caused by prenatal BPA exposure and next, give future directions for improving human health risk assessment caused by BPA exposure.
Collapse
Affiliation(s)
- Norazirah Mat Nayan
- Centre for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA, Malaysia; Institute of Medical Molecular and Biotechnology (IMMB) Faculty of Medicine, Universiti Teknologi MARA, 47000 Sungai Buloh, Selangor, Malaysia
| | - Andrean Husin
- Faculty of Dentistry, Universiti Teknologi MARA, Malaysia; Neuroscience Research Group (NRG), Faculty of Medicine, Universiti Teknologi MARA, Malaysia
| | - Rosfaiizah Siran
- Centre for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA, Malaysia; Neuroscience Research Group (NRG), Faculty of Medicine, Universiti Teknologi MARA, Malaysia.
| |
Collapse
|
4
|
Smolensky I, Zajac-Bakri K, Mallien AS, Gass P, Guzman R, Inta D. Effects of single housing on behavior, corticosterone level and body weight in male and female mice. Lab Anim Res 2024; 40:35. [PMID: 39342357 PMCID: PMC11439328 DOI: 10.1186/s42826-024-00221-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 09/04/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Experimental mice are often single-housed either for an individual analysis (feeding behavior, imaging, calorimetry) or as a stress paradigm (social isolation) in translational biomedical research. Reports of the influence of single housing in rodents are conflicting and may depend on age and duration of isolation. Sex is often not included as a factor. In this study we investigated the effects of 4-week single housing in male and female mice on behavior, body weight, and serum corticosterone levels. RESULTS Behavioral tests showed no effect on anhedonia and stress coping, anxiety and motor exploration. Social avoidance occurred in both males and females. Regarding physiological effects, single housing did not induce changes in serum corticosterone levels, but reduced body weight gain. CONCLUSIONS While some mouse studies of chronic social isolation reported depression-related disturbances, our data suggest that single housing might be not necessarily be too stressful. This is important for animal welfare regulations and experiments in life science research.
Collapse
Affiliation(s)
- Ilya Smolensky
- Department of Community Health, University of Fribourg, Chemin du Musée 4, Fribourg, 1700, Switzerland.
- Department of Biomedicine, University of Basel, Hebelstrasse 20, Basel, 4056, Switzerland.
- Food Research and Innovation Center (FRIC), University of Fribourg, Fribourg, Switzerland.
| | - Kilian Zajac-Bakri
- Department of Community Health, University of Fribourg, Chemin du Musée 4, Fribourg, 1700, Switzerland
- Department of Biomedicine, University of Basel, Hebelstrasse 20, Basel, 4056, Switzerland
- Food Research and Innovation Center (FRIC), University of Fribourg, Fribourg, Switzerland
| | - Anne Stephanie Mallien
- Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, J5, 68159, Mannheim, Germany
| | - Peter Gass
- Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, J5, 68159, Mannheim, Germany
| | - Raphael Guzman
- Department of Biomedicine, University of Basel, Hebelstrasse 20, Basel, 4056, Switzerland
- Department of Neurosurgery, University Hospital Basel, Spitalstrasse 21/Petersgraben 4, Basel, 4031, Switzerland
| | - Dragos Inta
- Department of Community Health, University of Fribourg, Chemin du Musée 4, Fribourg, 1700, Switzerland
- Department of Biomedicine, University of Basel, Hebelstrasse 20, Basel, 4056, Switzerland
- Food Research and Innovation Center (FRIC), University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
5
|
McAtee D, Abdelmoneim A. Effects of developmental exposure to arsenic species on behavioral stress responses in larval zebrafish and implications for stress-related disorders. Toxicol Sci 2024; 201:61-72. [PMID: 38833692 DOI: 10.1093/toxsci/kfae074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024] Open
Abstract
Arsenic (As) is globally detected in drinking water and food products at levels repeatedly surpassing regulatory thresholds. Several neurological and mental health risks linked to arsenic exposure are proposed; however, the nature of these effects and their association with the chemical forms of arsenic are not fully understood. Gaining a clear understanding of the etiologies and characteristics of these effects is crucial, particularly in association with developmental exposures where the nervous system is most vulnerable. In this study, we investigated the effects of early developmental exposure (6- to 120-h postfertilization [hpf]) of larval zebrafish to environmentally relevant concentrations of arsenic species-trivalent/pentavalent, inorganic/organic forms-on developmental, behavioral, and molecular endpoints to determine their effect on stress response and their potential association with stress-related disorders. At 120 hpf, the developing larvae were assessed for a battery of endpoints including survival, developmental malformities, background activity, and behavioral responses to acute visual and acoustic stimuli. Pooled larval samples were analyzed for alterations in the transcript levels of genes associated with developmental neurotoxicity and stress-related disorders. Developmental exposures at target concentrations did not significantly alter survival, overall development, or background activity, and had minor effects on developmental morphology. Sodium arsenate and monomethylarsonic acid exaggerated the behavioral responses of larval zebrafish, whereas sodium arsenite depressed them. Sodium arsenate induced significant effects on molecular biomarkers. This study highlights the effects of developmental exposure to arsenicals on the behavioral stress response, the role chemical formulation plays in exerting toxicological effects, and the possible association with stress-related disorders.
Collapse
Affiliation(s)
- Demetrius McAtee
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, United States
| | - Ahmed Abdelmoneim
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, United States
| |
Collapse
|
6
|
Nayan NM, Kadir SHSA, Husin A, Siran R. Neurodevelopmental effects of prenatal Bisphenol A exposure on the role of microRNA regulating NMDA receptor subunits in the male rat hippocampus. Physiol Behav 2024; 280:114546. [PMID: 38583549 DOI: 10.1016/j.physbeh.2024.114546] [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: 01/28/2024] [Revised: 03/22/2024] [Accepted: 04/05/2024] [Indexed: 04/09/2024]
Abstract
Maternal bisphenol A (BPA) exposure has been reported to cause learning and memory deficits in born offspring. However, little is known that this impairment is potentially caused by epigenetic modulation on the development of NMDA receptor subunits. This study investigates the effect of prenatal BPA exposure on the hippocampal miR-19a and miR-539, which are responsible for regulating NMDA receptor subunits as well as learning and memory functions. Pregnant Sprague Dawley rats were orally administered with 5 mg/kg/day of BPA from pregnancy day 1 (PD1) until gestation day 21 (GD21), while control mothers received no BPA. The mothers were observed daily until GD21 for either a cesarean section or spontaneous delivery. The male offspring were sacrificed when reaching GD21 (fetus), postnatal days 7, 14, 21 (PND7, 14, 21) and adolescent age 35 (AD35) where their hippocampi were dissected from the brain. The expression of targeted miR-19a, miR-539, GRIN2A, and GRIN2B were determined by qRT-PCR while the level of GluN2A and GluN2B were estimated by western blot. At AD35, the rats were assessed with neurobehavioral tests to evaluate their learning and memory function. The findings showed that prenatal BPA exposure at 5 mg/kg/day significantly reduces the expression of miR-19a, miR-539, GRIN2A, and GRIN2B genes in the male rat hippocampus at all ages. The level of GluN2A and GluN2B proteins is also significantly reduced when reaching adolescent age. Consequently, the rats showed spatial and fear memory impairments when reaching AD35. In conclusion, prenatal BPA exposure disrupts the role of miR-19a and miR-539 in regulating the NMDA receptor subunit in the hippocampus which may be one of the causes of memory and learning impairment in adolescent rats.
Collapse
Affiliation(s)
- Norazirah Mat Nayan
- Centre for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA 47000, Sungai Buloh, Selangor, Malaysia; Laboratory Animal Care Unit (LACU), Faculty of Medicine, Universiti Teknologi MARA 47000, Sungai Buloh, Selangor, Malaysia; Institute for Molecular Medicine and Biotechnology (IMMB) Faculty of Medicine, Universiti Teknologi MARA 47000, Sungai Buloh, Selangor, Malaysia
| | - Siti Hamimah Sheikh Abd Kadir
- Institute for Molecular Medicine and Biotechnology (IMMB) Faculty of Medicine, Universiti Teknologi MARA 47000, Sungai Buloh, Selangor, Malaysia
| | - Andrean Husin
- Faculty of Dentistry, Universiti Teknologi MARA 47000, Sungai Buloh, Selangor, Malaysia; Neuroscience Research Group (NRG), Faculty of Medicine, Universiti Teknologi MARA, 47000, Sungai Buloh, Selangor, Malaysia
| | - Rosfaiizah Siran
- Centre for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA 47000, Sungai Buloh, Selangor, Malaysia; Neuroscience Research Group (NRG), Faculty of Medicine, Universiti Teknologi MARA, 47000, Sungai Buloh, Selangor, Malaysia..
| |
Collapse
|
7
|
Zhang G, Zhou X, Feng Q, Ke W, Pan J, Zhang H, Luan Y, Lei B. Nerolidol reduces depression-like behavior in mice and suppresses microglia activation by down-regulating DNA methyltransferase 1. Neuroreport 2024; 35:457-465. [PMID: 38526920 DOI: 10.1097/wnr.0000000000002029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Modern medicine has unveiled that essential oil made from Aquilaria possesses sedative and hypnotic effects. Among the chemical components in Aquilaria, nerolidol, a natural sesquiterpene alcohol, has shown promising effects. This study aimed to unravel the potential of nerolidol in treating depression. Chronic unpredictable mild stress (CUMS) was utilized to induce depression-like behavior in mice, and open field test, sucrose preference, and tail suspension test was conducted. The impacts of nerolidol on the inflammatory response, microglial activation, and DNA methyltransferase 1 (DNMT1) were assessed. To study the regulatory role of DNMT1, lipopolysaccharide (LPS) was used to treat BV2 cells, followed by the evaluation of cell viability and DNMT1 level. Additionally, the influence of DNMT1 overexpression on BV2 cell activation was determined. Behavioral analysis revealed that nerolidol reduced depression-like behavior in mice. Nerolidol reduced the levels of inflammatory factors and microglial activation caused by CUMS. Nerolidol treatment was found to reduce DNMT1 levels in mouse brain tissue and it also decrease the LPS-induced increase in DNMT1 levels in BV2 cells. DNMT1 overexpression reversed the impacts of nerolidol on the inflammation response and cell activation. This study underscores the potential of nerolidol in reducing CUMS-induced depressive-like behavior and inhibiting microglial activation by downregulating DNMT1. These findings offer valuable insights into the potential of nerolidol as a therapeutic option for depression.
Collapse
Affiliation(s)
- Guangcai Zhang
- Rehabilitation Department, Hainan Medical College Affiliated Traditional Chinese Medicine Hospital, Guangzhou University of Traditional Chinese Medicine Affiliated Hainan Traditional Chinese Medicine Hospital, Hainan Traditional Chinese Medicine Hospital, Haikou, Hainan
| | - Xiaohui Zhou
- Rehabilitation Department, Hainan Medical College Affiliated Traditional Chinese Medicine Hospital, Guangzhou University of Traditional Chinese Medicine Affiliated Hainan Traditional Chinese Medicine Hospital, Hainan Traditional Chinese Medicine Hospital, Haikou, Hainan
| | - Qifan Feng
- Rehabilitation Department, Hainan Medical College Affiliated Traditional Chinese Medicine Hospital, Guangzhou University of Traditional Chinese Medicine Affiliated Hainan Traditional Chinese Medicine Hospital, Hainan Traditional Chinese Medicine Hospital, Haikou, Hainan
| | - Weihua Ke
- Rehabilitation Department, Hainan Medical College Affiliated Traditional Chinese Medicine Hospital, Guangzhou University of Traditional Chinese Medicine Affiliated Hainan Traditional Chinese Medicine Hospital, Hainan Traditional Chinese Medicine Hospital, Haikou, Hainan
- Graduate School, Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Jiahui Pan
- Rehabilitation Department, Hainan Medical College Affiliated Traditional Chinese Medicine Hospital, Guangzhou University of Traditional Chinese Medicine Affiliated Hainan Traditional Chinese Medicine Hospital, Hainan Traditional Chinese Medicine Hospital, Haikou, Hainan
| | - Haiying Zhang
- Rehabilitation Department, Hainan Medical College Affiliated Traditional Chinese Medicine Hospital, Guangzhou University of Traditional Chinese Medicine Affiliated Hainan Traditional Chinese Medicine Hospital, Hainan Traditional Chinese Medicine Hospital, Haikou, Hainan
| | - Yixian Luan
- Rehabilitation Department, Hainan Medical College Affiliated Traditional Chinese Medicine Hospital, Guangzhou University of Traditional Chinese Medicine Affiliated Hainan Traditional Chinese Medicine Hospital, Hainan Traditional Chinese Medicine Hospital, Haikou, Hainan
| | - Beibei Lei
- Rehabilitation Department, Hainan Medical College Affiliated Traditional Chinese Medicine Hospital, Guangzhou University of Traditional Chinese Medicine Affiliated Hainan Traditional Chinese Medicine Hospital, Hainan Traditional Chinese Medicine Hospital, Haikou, Hainan
- Graduate School, Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
8
|
Lark AR, Nass SR, Hahn YK, Gao B, Milne GL, Knapp PE, Hauser KF. HIV-1 Tat and morphine interactions dynamically shift striatal monoamine levels and exploratory behaviors over time. J Neurochem 2024; 168:185-204. [PMID: 38308495 PMCID: PMC10922901 DOI: 10.1111/jnc.16057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 12/28/2023] [Accepted: 01/10/2024] [Indexed: 02/04/2024]
Abstract
Despite the advent of combination anti-retroviral therapy (cART), nearly half of people infected with HIV treated with cART still exhibit HIV-associated neurocognitive disorders (HAND). HAND can be worsened by co-morbid opioid use disorder. The basal ganglia are particularly vulnerable to HIV-1 and exhibit higher viral loads and more severe pathology, which can be exacerbated by co-exposure to opioids. Evidence suggests that dopaminergic neurotransmission is disrupted by HIV exposure, however, little is known about whether co-exposure to opioids may alter neurotransmitter levels in the striatum and if this in turn influences behavior. Therefore, we assayed motor, anxiety-like, novelty-seeking, exploratory, and social behaviors, and levels of monoamines and their metabolites following 2 weeks and 2 months of Tat and/or morphine exposure in transgenic mice. Morphine decreased dopamine levels, but significantly elevated norepinephrine, the dopamine metabolites dihydroxyphenylacetic acid (DOPAC) and homovanillic acid (HVA), and the serotonin metabolite 5-hydroxyindoleacetic acid, which typically correlated with increased locomotor behavior. The combination of Tat and morphine altered dopamine, DOPAC, and HVA concentrations differently depending on the neurotransmitter/metabolite and duration of exposure but did not affect the numbers of tyrosine hydroxylase-positive neurons in the mesencephalon. Tat exposure increased the latency to interact with novel conspecifics, but not other novel objects, suggesting the viral protein inhibits exploratory behavior initiation in a context-dependent manner. By contrast, and consistent with prior findings that opioid misuse can increase novelty-seeking behavior, morphine exposure increased the time spent exploring a novel environment. Finally, Tat and morphine interacted to affect locomotor activity in a time-dependent manner, while grip strength and rotarod performance were unaffected. Together, our results provide novel insight into the unique effects of HIV-1 Tat and morphine on monoamine neurochemistry that may underlie their divergent effects on motor and exploratory behavior.
Collapse
Affiliation(s)
| | | | | | - Benlian Gao
- Neurochemistry Core, Vanderbilt Brain Institute, Vanderbilt University
| | - Ginger L. Milne
- Neurochemistry Core, Vanderbilt Brain Institute, Vanderbilt University
| | - Pamela E. Knapp
- Department of Pharmacology & Toxicology
- Department of Anatomy and Neurobiology
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University
| | - Kurt F. Hauser
- Department of Pharmacology & Toxicology
- Department of Anatomy and Neurobiology
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University
| |
Collapse
|
9
|
Valvassori SS, Varela RB, Resende WR, Possamai-Della T, Borba LDA, Behenck JP, Réus GZ, Quevedo J. Antidepressant Effect of Sodium Butyrate is Accompanied by Brain Epigenetic Modulation in Rats Subjected to Early or Late Life Stress. Curr Neurovasc Res 2024; 20:586-598. [PMID: 38288841 DOI: 10.2174/0115672026277345240115101852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/15/2023] [Accepted: 10/22/2024] [Indexed: 02/22/2024]
Abstract
BACKGROUND Major depression has a complex and multifactorial etiology constituted by the interaction between genetic and environmental factors in its development. OBJECTIVE The aim of this study was to evaluate the effects of sodium butyrate (SD) on epigenetic enzyme alterations in rats subjected to animal models of depression induced by maternal deprivation (MD) or chronic mild stress (CMS). METHODS To induce MD, male Wistar rats were deprived of maternal care during the first 10 days of life. To induce CMS, rats were subjected to the CMS for 40 days. Adult rats were then treated with daily injections of SD for 7 days. Animals were subjected to the forced swimming test (FST), and then, histone deacetylase (HDAC), histone acetyltransferase (HAT), and DNA methyltransferase (DNMT) activities were evaluated in the brain. RESULTS MD and CMS increased immobility time in FST and increased HDAC and DNMT activity in the animal brains. SD reversed increased immobility induced by both animal models and the alterations in HDAC and DNMT activities. There was a positive correlation between enzyme activities and immobility time for both models. HDAC and DNMT activities also presented a positive correlation between themselves. CONCLUSION These results suggest that epigenetics can play an important role in major depression pathophysiology triggered by early or late life stress and its treatment.
Collapse
Affiliation(s)
- Samira Silva Valvassori
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Roger Bitencourt Varela
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
- Functional Neuromodulation and Novel Therapeutics Laboratory, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Wilson Rodrigues Resende
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Taise Possamai-Della
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Laura de Araujo Borba
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - João Paulo Behenck
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Gislaine Zilli Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX, USA
- Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX, USA
- Faillace Department of Psychiatry and Behavioral Sciences, Center for Interventional Psychiatry, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX, USA
| |
Collapse
|
10
|
Singh M, Saxena S, Mohan KN. DNMT1 downregulation as well as its overexpression distinctly affect mostly overlapping genes implicated in schizophrenia, autism spectrum, epilepsy, and bipolar disorders. Front Mol Neurosci 2023; 16:1275697. [PMID: 38125006 PMCID: PMC10731955 DOI: 10.3389/fnmol.2023.1275697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/15/2023] [Indexed: 12/23/2023] Open
Abstract
Data on schizophrenia (SZ), epilepsy (EPD) and bipolar disorders (BPD) suggested an association of DNMT1 overexpression whereas certain variants of the gene were predicted to result in its increased expression in autism spectrum disorder (ASD). In addition, loss of DNMT1 in frontal cortex resulted in behavioral abnormalities in mice. Here we investigated the effects of increased as well as lack of DNMT1 expression using Dnmt1tet/tet neurons as a model for abnormal neurogenesis and 10,861 genes showing transcript level dysregulation in datasets from the four disorders. In case of overexpression, 3,211 (∼ 30%) genes were dysregulated, affecting pathways involved in neurogenesis, semaphorin signaling, ephrin receptor activity, etc. A disproportionately higher proportion of dysregulated genes were associated with epilepsy. When transcriptome data of Dnmt1tet/tet neurons treated with doxycycline that downregulated DNMT1 was used, 3,356 genes (∼31%) were dysregulated with a significant proportion involved in pathways similar to those in untreated cells. Both conditions resulted in ∼68% of dysregulated genes wherein a majority showed similar patterns of transcript level changes. Among the genes with transcripts returning to normal levels, ribosome assembly/biogenesis was most significant whereas in absence of DNMT1, a new set of 903 genes became dysregulated and are involved in similar pathways as mentioned above. These findings provide support for overexpression of DNMT1 as well as its downregulation as risk factor for the four disorders and that its levels within a tight range are essential for normal neurodevelopment/mental health.
Collapse
Affiliation(s)
- Minali Singh
- Molecular Biology and Genetics Laboratory, Department of Biological Sciences, Birla Institute of Technology and Science, Pilani, Hyderabad, India
| | - Sonal Saxena
- Centre for Human Disease Research, Birla Institute of Technology and Science, Pilani, Hyderabad, India
| | - Kommu Naga Mohan
- Molecular Biology and Genetics Laboratory, Department of Biological Sciences, Birla Institute of Technology and Science, Pilani, Hyderabad, India
- Centre for Human Disease Research, Birla Institute of Technology and Science, Pilani, Hyderabad, India
| |
Collapse
|
11
|
Smolensky I, Zajac-Bakri K, Odermatt TS, Brégère C, Cryan JF, Guzman R, Timper K, Inta D. Sex-specific differences in metabolic hormone and adipose tissue dynamics induced by moderate low-carbohydrate and ketogenic diet. Sci Rep 2023; 13:16465. [PMID: 37777528 PMCID: PMC10542803 DOI: 10.1038/s41598-023-43587-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/26/2023] [Indexed: 10/02/2023] Open
Abstract
Low-carbohydrates diets are increasingly used to treat obesity and metabolic disorders. A very low-carbohydrate, ketogenic diet is hard to follow and, due to the very high fat content, linked to severe side effects, like hyperlipidemia and atherogenesis. Therefore, a less restrictive, unsaturated fat-based low-carbohydrate diet appears as a promising alternative. Since neither sex differences, nor their effect on specific metabolic hormones and adipose tissue compartments have been investigated thoroughly in these diets, we aimed to analyze their dynamics and metabolic factors in mice. We found a significant sexual dimorphism with decreased body weight and subcutaneous fat only in males on ketogenic diet, while diminished insulin, elevated ghrelin and FGF-21 were present with a differential time course in both sexes. The non-ketogenic moderate low-carbohydrate diet increased body weight and perigonadal fat in females, but induced leptin elevation in males. Both diets enhanced transiently TNFɑ only in males and had no impact on behavior. Altogether, these results reveal complex sex-dependent effect of dietary interventions, indicating unexpectedly females as more prone to unfavorable metabolic effects of low-carbohydrate diets.
Collapse
Affiliation(s)
- Ilya Smolensky
- Department of Community Health, University of Fribourg, 1700, Fribourg, Switzerland.
- Department of Biomedicine, University of Basel, 4056, Basel, Switzerland.
| | - Kilian Zajac-Bakri
- Department of Community Health, University of Fribourg, 1700, Fribourg, Switzerland
- Department of Biomedicine, University of Basel, 4056, Basel, Switzerland
| | | | - Catherine Brégère
- Department of Biomedicine, University of Basel, 4056, Basel, Switzerland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, T12TP07, Ireland
- APC Microbiome Ireland, University College Cork, Cork, T12TP07, Ireland
| | - Raphael Guzman
- Department of Biomedicine, University of Basel, 4056, Basel, Switzerland
- Department of Neurosurgery, University Hospital Basel, 4056, Basel, Switzerland
| | - Katharina Timper
- Department of Biomedicine, University of Basel, 4056, Basel, Switzerland
- Department of Endocrinology, Diabetes and Metabolism Clinic, University Hospital Basel, 4056, Basel, Switzerland
| | - Dragos Inta
- Department of Community Health, University of Fribourg, 1700, Fribourg, Switzerland
- Department of Biomedicine, University of Basel, 4056, Basel, Switzerland
| |
Collapse
|
12
|
Tuo LJ, Song XY, Zhu YY, He HN, Song YP, Chen DZ, Zheng XM, Zhang H, Xu DX. Gestational folic acid supplement prevents vitamin D deficiency-induced depression-like behavior by reversing cortical DNA hypomethylation in adult offspring. J Steroid Biochem Mol Biol 2023; 231:106313. [PMID: 37075986 DOI: 10.1016/j.jsbmb.2023.106313] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/27/2023] [Accepted: 04/14/2023] [Indexed: 04/21/2023]
Abstract
Depression is a common mental disorder with an increasing incidence. Several studies have demonstrated that cortical DNA hypomethylation is associated with depression-like behaviors. This study aims to investigate whether maternal vitamin D deficiency (VDD) induces depression-like behaviors and to explore the effects of folic acid supplement on VDD-induced cortical DNA hypomethylation in adult offspring. Female mice were fed with a VDD diet, beginning at 5 weeks of age and throughout pregnancy. Depression-like behaviors were evaluated, and cortical 5-methylcytosine (5mC) content was detected in adult offspring. Results showed that depression-like behaviors were observed in adult offspring of the VDD group. Cortical Ache and Oxtr mRNAs were upregulated in female offspring of the VDD group. Cortical Cpt1a and Htr1b mRNAs were increased in male offspring of the VDD group. Moreover, cortical 5mC content was reduced in offspring of VDD-fed dams. The additional experiment showed that serum folate and cortical S-adenosylmethionine (SAM) contents were decreased in the offspring of the VDD group. Folic acid supplement attenuated VDD-induced SAM depletion and reversed cortical DNA methylation. Moreover, folic acid supplement attenuated VDD-induced upregulation of depression-related genes. In addition, folic acid supplement alleviated maternal VDD-induced depression-like behaviors in adult offspring. These results suggest that maternal VDD induces depression-like behavior in adult offspring by reducing cortical DNA methylation. The gestational folic acid supplement prevents VDD-induced depression-like behavior by reversing cortical DNA hypomethylation in adult offspring.
Collapse
Affiliation(s)
- Ling-Jin Tuo
- Department of Toxicology, Anhui Medical University, Hefei 230032, China
| | - Xiao-Yue Song
- Department of Toxicology, Anhui Medical University, Hefei 230032, China
| | - Yan-Yan Zhu
- Department of Toxicology, Anhui Medical University, Hefei 230032, China
| | - Hong-Ning He
- Department of Toxicology, Anhui Medical University, Hefei 230032, China
| | - Ya-Ping Song
- Department of Toxicology, Anhui Medical University, Hefei 230032, China
| | - Dao-Zhen Chen
- Department of Clinical Laboratory, Wuxi Maternity and Child Health Care Hospital, Wuxi 214002, China; Laboratory Department of Haidong Second People's Hospital, Haidong, Qinghai 810699, China
| | - Xiao-Min Zheng
- Research Institute for Reproductive Medicine and Genetic Diseases, Wuxi Maternity and Child Health Hospital, Wuxi, 214002, China
| | - Heng Zhang
- Department of Toxicology, Anhui Medical University, Hefei 230032, China; Department of Child Health Care, Wuxi Maternity and Child Health Care Hospital, Wuxi 214002, China.
| | - De-Xiang Xu
- Department of Toxicology, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
13
|
LncRNA XR_351665 Contributes to Chronic Pain-Induced Depression by Upregulating DNMT1 via Sponging miR-152-3p. THE JOURNAL OF PAIN 2023; 24:449-462. [PMID: 36257574 DOI: 10.1016/j.jpain.2022.10.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 10/04/2022] [Accepted: 10/06/2022] [Indexed: 11/29/2022]
Abstract
Chronic pain is frequently comorbid with depression. However, the mechanisms underlying chronic pain-induced depression remain unclear. Here, we found that DNA methyltransferase 1 (DNMT1) was upregulated in the central amygdala (CeA) of spared nerve injury (SNI)-induced chronic pain-depression rats, and knockdown of DNMT1 could improve the depression-like behaviors in SNI rats. Additionally, a panel of differentially expressed lncRNAs, including 38 upregulated and 12 downregulated lncRNAs, were identified by microarray analysis. Bioinformatics analysis suggested that the upregulated lncRNA XR_351665 was the upstream molecule to regulate DNMT1 expression. The knockdown of XR_351665 significantly alleviated the depression-like behaviors in SNI rats, whereas overexpression of XR_351665 induced the depression-like behaviors in naïve rats. Further mechanism-related researches uncovered that XR_351665 functioned as a competing endogenous RNA (ceRNA) to upregulate DNMT1 by competitively sponging miR-152-3p, and subsequently promoted the development of chronic pain-induced depression. Our findings suggest that lncRNA XR_351665 is involved in the development of chronic pain-induced depression by upregulating DNMT1 via sponging miR-152-3p. These data provide novel insight into understanding the pathogenesis of chronic pain-induced depression and identify a potential therapeutic target. PERSPECTIVE: LncRNA XR_351665 in CeA functions as a ceRNA to block the inhibitory effect of miR-152-3p on DNMT1 and contributes to the development of chronic pain-induced depression. These data suggest that manipulation of XR_351665/miR-152-3p/DNMT1 axis may be a potential method to attenuate chronic pain-induced depression.
Collapse
|
14
|
Wang X, Jiang L, Ma W, Zheng X, He E, Zhang B, Vashisth MK, Gong Z. Maternal separation affects Anxiety like behavior begin in adolescence continue through adulthood and related to Dnmt3a expression. J Neurophysiol 2022; 128:611-618. [PMID: 35946792 DOI: 10.1152/jn.00247.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Early life stress, including maternal separation, is among one of the main causes of anxiety in adolescents. DNA methyltransferase 3A (Dnmt3a) is a key molecule that regulates DNA methylation and is found to be associated with anxiety-like behavior. It is not clear whether maternal separation affects anxiety levels in mice at different developmental stages, or whether Dnmt3a plays a role in this process. Here, by using open field test to exploring the effect of maternal separation on anxiety-like behavior in mice of different age, it was found that maternal separation could successfully induce anxiety-like behavior in adolescent mice, and which continued through adulthood. By using western blot, we found the levels of Dnmt3a in the hippocampus and cortex have shown different trends in maternal separation mice on P17. Further, by using immunostaining, we have found that the expression levels of Dnmt3a in the cortex and hippocampus were significantly different, and decreased to varying degrees with the age of mice, which being the reason for different trends. Our results provide an experimental basis for further development of anxiety/depression treatment programs more suitable for adolescence.
Collapse
Affiliation(s)
- Xiaobing Wang
- Department of Anatomy, College of Preclinical Medicine, Dali University, Dali, China
| | - Le Jiang
- Department of Anatomy, College of Preclinical Medicine, Dali University, Dali, China
| | - Wenhao Ma
- Department of Anatomy, College of Preclinical Medicine, Dali University, Dali, China
| | - Xiaoye Zheng
- Department of Anatomy, College of Preclinical Medicine, Dali University, Dali, China
| | - Ershu He
- Department of Anatomy, College of Preclinical Medicine, Dali University, Dali, China
| | - Bensi Zhang
- Department of Anatomy, College of Preclinical Medicine, Dali University, Dali, China
| | - Manoj Kumar Vashisth
- Department of Anatomy, College of Preclinical Medicine, Dali University, Dali, China
| | - Zhiting Gong
- Department of Anatomy, College of Preclinical Medicine, Dali University, Dali, China
| |
Collapse
|
15
|
Tovy A, Reyes JM, Zhang L, Huang YH, Rosas C, Daquinag AC, Guzman A, Ramabadran R, Chen CW, Gu T, Gupta S, Ortinau L, Park D, Cox AR, Rau RE, Hartig SM, Kolonin MG, Goodell MA. Constitutive loss of DNMT3A causes morbid obesity through misregulation of adipogenesis. eLife 2022; 11:e72359. [PMID: 35635747 PMCID: PMC9150890 DOI: 10.7554/elife.72359] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
DNA Methyltransferase 3 A (DNMT3A) is an important facilitator of differentiation of both embryonic and hematopoietic stem cells. Heterozygous germline mutations in DNMT3A lead to Tatton-Brown-Rahman Syndrome (TBRS), characterized by obesity and excessive height. While DNMT3A is known to impact feeding behavior via the hypothalamus, here we investigated a role in adipocyte progenitors utilizing heterozygous knockout mice that recapitulate cardinal TBRS phenotypes. These mice become morbidly obese due to adipocyte enlargement and tissue expansion. Adipose tissue in these mice exhibited defects in preadipocyte maturation and precocious activation of inflammatory gene networks, including interleukin-6 signaling. Adipocyte progenitor cell lines lacking DNMT3A exhibited aberrant differentiation. Furthermore, mice in which Dnmt3a was specifically ablated in adipocyte progenitors showed enlarged fat depots and increased progenitor numbers, partly recapitulating the TBRS obesity phenotypes. Loss of DNMT3A led to constitutive DNA hypomethylation, such that the DNA methylation landscape of young adipocyte progenitors resemble that of older wild-type mice. Together, our results demonstrate that DNMT3A coordinates both the central and local control of energy storage required to maintain normal weight and prevent inflammatory obesity.
Collapse
Affiliation(s)
- Ayala Tovy
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| | - Jaime M Reyes
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Linda Zhang
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of MedicineHoustonUnited States
| | - Yung-Hsin Huang
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
| | - Carina Rosas
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| | - Alexes C Daquinag
- Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science CenterHoustonUnited States
| | - Anna Guzman
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| | - Raghav Ramabadran
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| | - Chun-Wei Chen
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Tianpeng Gu
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| | - Sinjini Gupta
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| | - Laura Ortinau
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
- Center for Metabolic and Degenerative Disease, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at HoustonHoustonUnited States
| | - Dongsu Park
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
- Center for Metabolic and Degenerative Disease, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at HoustonHoustonUnited States
| | - Aaron R Cox
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Baylor College of MedicineHoustonUnited States
| | - Rachel E Rau
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Pediatrics, Baylor College of Medicine and Texas Children's HospitalHoustonUnited States
| | - Sean M Hartig
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Baylor College of MedicineHoustonUnited States
| | - Mikhail G Kolonin
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of MedicineHoustonUnited States
| | - Margaret A Goodell
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
16
|
de Sousa Maciel I, Sales AJ, Casarotto PC, Castrén E, Biojone C, Joca SRL. Nitric Oxide Synthase inhibition counteracts the stress-induced DNA methyltransferase 3b expression in the hippocampus of rats. Eur J Neurosci 2022; 55:2421-2434. [PMID: 33170977 DOI: 10.1111/ejn.15042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/22/2020] [Accepted: 11/02/2020] [Indexed: 11/29/2022]
Abstract
It has been postulated that the activation of NMDA receptors (NMDAr) and nitric oxide (NO) production in the hippocampus is involved in the behavioral consequences of stress. Stress triggers NMDAr-induced calcium influx in limbic areas, such as the hippocampus, which in turn activates neuronal NO synthase (nNOS). Inhibition of nNOS or NMDAr activity can prevent stress-induced effects in animal models, but the molecular mechanisms behind this effect are still unclear. In this study, cultured hippocampal neurons treated with NMDA or dexamethasone showed an increased of DNA methyltransferase 3b (DNMT3b) mRNA expression, which was blocked by pre-treatment with nNOS inhibitor nω -propyl-l-arginine (NPA). In rats submitted to the Learned Helplessness paradigm (LH), we observed that inescapable stress increased DNMT3b mRNA expression at 1h and 24h in the hippocampus. The NOS inhibitors 7-NI and aminoguanidine (AMG) decreased the number of escape failures in LH and counteracted the changes in hippocampal DNMT3b mRNA induced in this behavioral paradigm. Altogether, our data suggest that NO produced in response to NMDAr activation following stress upregulates DNMT3b in the hippocampus.
Collapse
Affiliation(s)
- Izaque de Sousa Maciel
- School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto - SP, Brazil
| | - Amanda J Sales
- School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto - SP, Brazil
| | | | - Eero Castrén
- Neuroscience Center, HiLIFE, University of Helsinki, Finland
| | | | - Sâmia R L Joca
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto -SP, Brazil
| |
Collapse
|
17
|
Lax E. DNA Methylation as a Therapeutic and Diagnostic Target in Major Depressive Disorder. Front Behav Neurosci 2022; 16:759052. [PMID: 35431832 PMCID: PMC9006940 DOI: 10.3389/fnbeh.2022.759052] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 02/10/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Elad Lax
- *Correspondence: Elad Lax ; orcid.org/0000-0002-0310-0520
| |
Collapse
|
18
|
Mavioglu RN, Ramo-Fernandez L, Gumpp AM, Kolassa IT, Karabatsiakis A. A history of childhood maltreatment is associated with altered DNA methylation levels of DNA methyltransferase 1 in maternal but not neonatal mononuclear immune cells. Front Psychiatry 2022; 13:945343. [PMID: 36440389 PMCID: PMC9685310 DOI: 10.3389/fpsyt.2022.945343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022] Open
Abstract
Childhood maltreatment (CM) is associated with alterations in DNA methylation (DNAm) especially in stress response genes. Due to the higher risk of overall health complications of individuals with a parental history of CM, intergenerational transmission of CM-associated DNAm changes has been investigated but remains unclear. In this study, we investigated if different severities of CM have any influence on the DNAm of DNA methyltransferase 1 (DNMT1), an important enzyme of the DNAm machinery, in immune and buccal cells of mother-newborn dyads. DNAm was assessed by mass spectrometry using immune cell DNA from mothers (N = 117) and their newborns (N = 113), and buccal cell DNA of mother-newborn dyads (N = 68 each). Mothers with a history of CM had lower mean methylation of DNMT1 in immune cells compared to the mothers without a CM history. CM status only influenced maternal DNMT1 gene expression when at least moderate CM was reported. Buccal cell DNAm was not associated with CM status. Maternal history of CM was not linked to any alterations in DNMT1 mean DNAm in any of the cell types studied in newborns. We conclude that the CM-associated alterations in DNMT1 DNAm might point to allostatic load and can be physiologically relevant, especially in individuals with more severe CM experiences, resulting in an activated DNA methylation machinery that might influence stress response genes. Our lack of significant findings in buccal cells shows the tissue-specific effects of CM on DNAm. In our sample with low to moderate maternal CM history, there was no intergenerational transmission of DNMT1 DNAm in newborns.
Collapse
Affiliation(s)
- Rezan Nehir Mavioglu
- Department of Clinical and Biological Psychology, Institute of Psychology and Education, Ulm University, Ulm, Germany
| | - Laura Ramo-Fernandez
- Department of Clinical and Biological Psychology, Institute of Psychology and Education, Ulm University, Ulm, Germany
| | - Anja M Gumpp
- Department of Clinical and Biological Psychology, Institute of Psychology and Education, Ulm University, Ulm, Germany
| | - Iris-Tatjana Kolassa
- Department of Clinical and Biological Psychology, Institute of Psychology and Education, Ulm University, Ulm, Germany
| | - Alexander Karabatsiakis
- Department of Clinical and Biological Psychology, Institute of Psychology and Education, Ulm University, Ulm, Germany.,Department of Psychology, Clinical Psychology II, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
19
|
Yildiz CB, Zimmer-Bensch G. Role of DNMTs in the Brain. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1389:363-394. [DOI: 10.1007/978-3-031-11454-0_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
20
|
Pedrazzi JFC, Sales AJ, Guimarães FS, Joca SRL, Crippa JAS, Del Bel E. Cannabidiol prevents disruptions in sensorimotor gating induced by psychotomimetic drugs that last for 24-h with probable involvement of epigenetic changes in the ventral striatum. Prog Neuropsychopharmacol Biol Psychiatry 2021; 111:110352. [PMID: 34015384 DOI: 10.1016/j.pnpbp.2021.110352] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 05/07/2021] [Accepted: 05/12/2021] [Indexed: 02/06/2023]
Abstract
Cannabidiol (CBD), a major non-psychotomimetic component of the Cannabis sativa plant, shows therapeutic potential in several psychiatric disorders, including schizophrenia. The molecular mechanisms underlying the antipsychotic-like effects of CBD are not fully understood. Schizophrenia and antipsychotic treatment can modulate DNA methylation in the blood and brain, resulting in altered expression of diverse genes associated with this complex disorder. However, to date, the possible involvement of DNA methylation in the antipsychotic-like effects of CBD has not been investigated. Therefore, this study aimed at evaluating in mice submitted to the prepulse inhibition (PPI) model: i) the effects of a single injection of CBD or clozapine followed by AMPH or MK-801 on PPI and global DNA methylation changes in the ventral striatum and prefrontal cortex (PFC); and ii). if the acute antipsychotic-like effects of CBD would last for 24-h. AMPH (5 mg/kg) and MK-801 (0.5 mg/kg) impaired PPI. CBD (30 and 60 mg/kg), similar to clozapine (5 mg/kg), attenuated AMPH- and MK801-induced PPI disruption. AMPH, but not MK-801, increased global DNA methylation in the ventral striatum, an effect prevented by CBD. CBD and clozapine increased, by themselves, DNA methylation in the prefrontal cortex. The acute effects of CBD (30 or 60 mg/kg) on the PPI impairment induced by AMPH or MK-801 was also detectable 24 h later. Altogether, the results show that CBD induces acute antipsychotic-like effects that last for 24-h. It also modulates DNA methylation in the ventral striatum, suggesting a new potential mechanism for its antipsychotic-like effects.
Collapse
Affiliation(s)
- João F C Pedrazzi
- Department of Neurosciences and Behavioral Sciences, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Amanda J Sales
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Francisco S Guimarães
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Sâmia R L Joca
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil; Departament of Biomedicine, Aarhus University, Denmark
| | - José A S Crippa
- Department of Neurosciences and Behavioral Sciences, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Elaine Del Bel
- Department of Morphology, Physiology, and Basic Pathology, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
21
|
Thomas KT, Zakharenko SS. MicroRNAs in the Onset of Schizophrenia. Cells 2021; 10:2679. [PMID: 34685659 PMCID: PMC8534348 DOI: 10.3390/cells10102679] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/30/2021] [Accepted: 10/02/2021] [Indexed: 12/14/2022] Open
Abstract
Mounting evidence implicates microRNAs (miRNAs) in the pathology of schizophrenia. These small noncoding RNAs bind to mRNAs containing complementary sequences and promote their degradation and/or inhibit protein synthesis. A single miRNA may have hundreds of targets, and miRNA targets are overrepresented among schizophrenia-risk genes. Although schizophrenia is a neurodevelopmental disorder, symptoms usually do not appear until adolescence, and most patients do not receive a schizophrenia diagnosis until late adolescence or early adulthood. However, few studies have examined miRNAs during this critical period. First, we examine evidence that the miRNA pathway is dynamic throughout adolescence and adulthood and that miRNAs regulate processes critical to late neurodevelopment that are aberrant in patients with schizophrenia. Next, we examine evidence implicating miRNAs in the conversion to psychosis, including a schizophrenia-associated single nucleotide polymorphism in MIR137HG that is among the strongest known predictors of age of onset in patients with schizophrenia. Finally, we examine how hemizygosity for DGCR8, which encodes an obligate component of the complex that synthesizes miRNA precursors, may contribute to the onset of psychosis in patients with 22q11.2 microdeletions and how animal models of this disorder can help us understand the many roles of miRNAs in the onset of schizophrenia.
Collapse
Affiliation(s)
- Kristen T. Thomas
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Stanislav S. Zakharenko
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
22
|
Abstract
Epigenetic mechanisms such as DNA methylation (DNAm) have been associated with stress responses and increased vulnerability to depression. Abnormal DNAm is observed in stressed animals and depressed individuals. Antidepressant treatment modulates DNAm levels and regulates gene expression in diverse tissues, including the brain and the blood. Therefore, DNAm could be a potential therapeutic target in depression. Here, we reviewed the current knowledge about the involvement of DNAm in the behavioural and molecular changes associated with stress exposure and depression. We also evaluated the possible use of DNAm changes as biomarkers of depression. Finally, we discussed current knowledge limitations and future perspectives.
Collapse
|
23
|
Zhang C, Wu XC, Li S, Dou LJ, Zhou L, Wang FH, Ma K, Huang D, Pan Y, Gu JJ, Cao JY, Wang H, Hao JH. Perinatal low-dose bisphenol AF exposure impairs synaptic plasticity and cognitive function of adult offspring in a sex-dependent manner. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 788:147918. [PMID: 34134381 DOI: 10.1016/j.scitotenv.2021.147918] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/15/2021] [Accepted: 05/16/2021] [Indexed: 06/12/2023]
Abstract
Bisphenol AF (BPAF), a kind of the ideal substitutes of Bisphenol A (BPA), has frequently been detected in environmental media and biological samples. Numerous studies have focused on the reproductive toxicity, cardiotoxicity and endocrine disrupting toxicity of BPAF. However, little evidence is available on neurodevelopmental toxicity of BPAF. Here, our study is to evaluate the effect of perinatal BPAF exposure (0, 0.34, 3.4 and 34 mg/kg body weight/day, correspond to Ctrl, low-, medium- and high-dose groups) on the cognitive function of adult mouse offspring. This study firstly found that perinatal BPAF exposure caused cognitive impairments of mouse offspring, in which male offspring was more sensitive than female offspring in low- and medium-dose BPAF groups. Furthermore, the dendritic arborization and complexity of hippocampal CA1 and DG neurons in male offspring were impaired in all BPAF groups, and these effects were only found in high-dose BPAF group for female offspring. The damage of BPAF to dendritic spines, and the structural basis of learning and memory, was found in male offspring but not in females. Correspondingly, perinatal BPAF exposure significantly downregulated the expressions of hippocampal PSD-95 and Synapsin-1 proteins, and male offspring was more vulnerable than female offspring. Meanwhile, we explored the alteration of hippocampal estrogen receptors (ERs) to explain the sex specific impairment of cognitive function in low- and medium-dose BPAF groups. The results showed that perinatal BPAF exposure significantly decreased the expression of ERα in male offspring in a dose-dependent manner, but not in female offspring. In addition, we found that perinatal BPAF exposure can disordered the balance of oxidation and antioxidation in hippocampus of male offspring. In summary, perinatal low-dose bisphenol AF exposure impairs synaptic plasticity and cognitive function of adult offspring in a sex-dependent manner. The present results provide a pierce of potential mechanism of BPAF-caused neurodevelopmental toxicity.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Xiao-Chang Wu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Sha Li
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Lian-Jie Dou
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Li Zhou
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Feng-Hui Wang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Kai Ma
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Dan Huang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Ying Pan
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Ji-Jun Gu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Ji-Yu Cao
- Teaching Center for Preventive Medicine, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Hua Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China.
| | - Jia-Hu Hao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China.
| |
Collapse
|
24
|
Nass SR, Lark ARS, Hahn YK, McLane VD, Ihrig TM, Contois L, Napier TC, Knapp PE, Hauser KF. HIV-1 Tat and morphine decrease murine inter-male social interactions and associated oxytocin levels in the prefrontal cortex, amygdala, and hypothalamic paraventricular nucleus. Horm Behav 2021; 133:105008. [PMID: 34171549 PMCID: PMC8277758 DOI: 10.1016/j.yhbeh.2021.105008] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/11/2021] [Accepted: 05/22/2021] [Indexed: 12/15/2022]
Abstract
Many persons infected with HIV-1 (PWH) and opioid-dependent individuals experience deficits in sociability that interfere with daily living. Sociability is regulated by the prefrontal cortico-hippocampal-amygdalar circuit. Within this circuit HIV-1 trans-activator of transcription (HIV-1 Tat) and opioids can increase dendritic pathology and alter neuronal firing. Changes in sociability are also associated with dysregulation of hypothalamic neuropeptides such as oxytocin or corticotropin releasing factor (CRF) in the prefrontal cortico-hippocampal-amygdalar circuit. Accordingly, we hypothesized that the interaction of HIV-1 Tat and morphine would impair inter-male social interactions and disrupt oxytocin and CRF within the PFC and associated circuitry. Male mice were exposed to HIV-1 Tat for 8 weeks and administered saline or escalating doses of morphine twice daily (s.c.) during the last 2 weeks of HIV-1 Tat exposure. Tat attenuated aggressive interactions with an unknown intruder, whereas morphine decreased both non-aggressive and aggressive social interactions in the resident-intruder test. However, there was no effect of Tat or morphine on non-reciprocal interactions in the social interaction and novelty tests. Tat, but not morphine, decreased oxytocin levels in the PFC and amygdala, whereas both Tat and morphine decreased the percentage of oxytocin-immunoreactive neurons in the hypothalamic paraventricular nucleus (PVN). In Tat(+) or morphine-exposed mice, regional levels of CRF and oxytocin correlated with alterations in behavior in the social interaction and novelty tests. Overall, decreased expression of oxytocin in the prefrontal cortico-hippocampal-amygdalar circuit is associated with morphine- and HIV-Tat-induced deficits in social behavior.
Collapse
Affiliation(s)
- Sara R Nass
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0613, USA
| | - Arianna R S Lark
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0613, USA
| | - Yun K Hahn
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0709, USA
| | - Virginia D McLane
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0613, USA
| | - Therese M Ihrig
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0613, USA
| | - Liangru Contois
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0613, USA
| | - T Celeste Napier
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, IL 60612-2847, USA; Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL 60612-3818, USA
| | - Pamela E Knapp
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0613, USA; Department of Anatomy and Neurobiology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0709, USA; Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0059, USA
| | - Kurt F Hauser
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0613, USA; Department of Anatomy and Neurobiology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0709, USA; Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0059, USA.
| |
Collapse
|
25
|
Eyolfson E, Bhatt D, Wang M, Lohman AW, Mychasiuk R. Paternal exposure to exercise and/or caffeine and alcohol modify offspring behavioral and pathophysiological recovery from repetitive mild traumatic brain injury in adolescence. GENES, BRAIN, AND BEHAVIOR 2021; 20:egbb12736. [PMID: 33876557 DOI: 10.1111/gbb.12736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/16/2021] [Accepted: 04/17/2021] [Indexed: 11/30/2022]
Abstract
Only recently has the scope of parental research expanded to include the paternal sphere with epidemiological studies implicating stress, nutrition and alcohol consumption in the neurobiological and behavioral characteristics of offspring. This study was designed to determine if paternal exposure to caffeine, alcohol and exercise prior to conception would improve or exacerbate offspring recovery from adolescent repetitive mild traumatic brain injury (RmTBI). Sires received 7 weeks of standard drinking water, or caffeine and ethanol and were housed in regular cages or cages with running wheels, prior to being mated to control females. At postnatal day 40, offspring were administered RmTBI or sham injuries and were assessed for post concussive symptomology. Post-mortem quantitative real-time polymerase chain reaction (qRT-PCR) was used to assess gene expression in the prefrontal cortex (PFC), nucleus accumbens (NAc) and changes in telomere length. Additionally, enzyme-linked immunosorbent assay (ELISA's) were run on serum to detect levels of cytokines, chemokines and sex hormones. Paternal experience did not improve or exacerbate RmTBI behavioral outcomes. However, female and male offspring displayed unique responses to RmTBI and paternal experience, resulting in changes in physical, behavioral and molecular outcomes. Injury and paternal exercise modified changes in female offspring, whereas male offspring were affected by paternal exercise, caffeine and alcohol treatment. Additionally, paternal experience and RmTBI modified expression of many genes in the PFC, NAc, telomere length and levels of sex hormones. Although further exploration is required to understand the heterogeneity that exists in disease risk and resiliency, this study provides corroborating evidence that paternal experiences prior to conception influences offspring development.
Collapse
Affiliation(s)
- Eric Eyolfson
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
| | - Dhyey Bhatt
- Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Melinda Wang
- Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
| | - Alexander W Lohman
- Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Richelle Mychasiuk
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
26
|
Sales AJ, Maciel IS, Suavinha ACDR, Joca SRL. Modulation of DNA Methylation and Gene Expression in Rodent Cortical Neuroplasticity Pathways Exerts Rapid Antidepressant-Like Effects. Mol Neurobiol 2021; 58:777-794. [PMID: 33025509 DOI: 10.1007/s12035-020-02145-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Stress increases DNA methylation, primarily a suppressive epigenetic mechanism catalyzed by DNA methyltransferases (DNMT), and decreases the expression of genes involved in neuronal plasticity and mood regulation. Despite chronic antidepressant treatment decreases stress-induced DNA methylation, it is not known whether inhibition of DNMT would convey rapid antidepressant-like effects. AIM This work tested such a hypothesis and evaluated whether a behavioral effect induced by DNMT inhibitors (DNMTi) corresponds with changes in DNA methylation and transcript levels in genes consistently associated with the neurobiology of depression and synaptic plasticity (BDNF, TrkB, 5-HT1A, NMDA, and AMPA). METHODS Male Wistar rats received intraperitoneal (i.p.) injection of two pharmacologically different DNMTi (5-AzaD 0.2 and 0.6 mg/kg or RG108 0.6 mg/kg) or vehicle (1 ml/kg), 1 h or 7 days before the learned helplessness test (LH). DNA methylation in target genes and the correspondent transcript levels were measured in the hippocampus (HPC) and prefrontal cortex (PFC) using meDIP-qPCR. In parallel separate groups, the antidepressant-like effect of 5-AzaD and RG108 was investigated in the forced swimming test (FST). The involvement of cortical BDNF-TrkB-mTOR pathways was assessed by intra-ventral medial PFC (vmPFC) injections of rapamycin (mTOR inhibitor), K252a (TrkB receptor antagonist), or vehicle (0.2 μl/side). RESULTS We found that both 5-AzaD and RG108 acutely and 7 days before the test decreased escape failures in the LH. LH stress increased DNA methylation and decreased transcript levels of BDNF IV and TrkB in the PFC, effects that were not significantly attenuated by RG108 treatment. The systemic administration of 5-AzaD (0.2 mg/kg) and RG108 (0.2 mg/kg) induced an antidepressant-like effect in FST, which was, however, attenuated by TrkB and mTOR inhibition into the vmPFC. CONCLUSION These findings suggest that acute inhibition of stress-induced DNA methylation promotes rapid and sustained antidepressant effects associated with increased BDNF-TrkB-mTOR signaling in the PFC.
Collapse
Affiliation(s)
- Amanda J Sales
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
- FMRP-USP, Av Bandeirantes, 3900, Ribeirão Preto, SP, 14049-900, Brazil.
| | - Izaque S Maciel
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Angélica C D R Suavinha
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Sâmia R L Joca
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
- FCFRP-USP, Av Café, sn, Monte Alegre, Ribeirão Preto, SP, 14040-903, Brazil.
| |
Collapse
|
27
|
Yang M, Barrios J, Yan J, Zhao W, Yuan S, Dong E, Ai X. Causal roles of stress kinase JNK2 in DNA methylation and binge alcohol withdrawal-evoked behavioral deficits. Pharmacol Res 2021; 164:105375. [PMID: 33316384 PMCID: PMC7867628 DOI: 10.1016/j.phrs.2020.105375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/20/2020] [Accepted: 12/06/2020] [Indexed: 11/27/2022]
Abstract
Excessive binge alcohol intake is a common drinking pattern in humans, especially during holidays. Cessation of the binge drinking often leads to aberrant withdrawal behaviors, as well as serious heart rhythm abnormalities (clinically diagnosed as Holiday Heart Syndrome (HHS)). In our HHS mouse model with well-characterized binge alcohol withdrawal (BAW)-induced heart phenotypes, BAW leads to anxiety-like behaviors and cognitive impairment. We have previously reported that stress-activated c-Jun NH(2)-terminal kinase (JNK) plays a causal role in BAW-induced heart phenotypes. In the HHS brain, we found that activation of JNK2 (but not JNK1 and JNK3) in the prefrontal cortex (PFC), but not hippocampus and amygdala, led to anxiety-like behaviors and impaired cognition. DNA methylation mediated by a crucial DNA methylation enzyme, DNA methyltransferase1 (DNMT1), is known to be critical in alcohol-associated behavioral deficits. In HHS mice, JNK2 in the PFC (but not hippocampus and amygdala) causally enhanced total genomic DNA methylation via increased DNMT1 expression, which was regulated by enhanced binding of JNK downstream transcriptional factor c-JUN to the DNMT1 promoter. JNK2-specific inhibition either by an inhibitor JNK2I or JNK2 knockout completely offset c-JUN-regulated DNMT1 upregulation and restored the level of DNA methylation in HHS PFC to the baseline levels seen in sham controls. Strikingly, either JNK2-specific inhibition or genetic JNK2 depletion or DNMT1 inhibition (by an inhibitor 5-Azacytidine) completely abolished BAW-evoked behavioral deficits. In conclusion, our studies revealed a novel mechanism by which JNK2 drives BAW-evoked behavioral deficits through a DNMT1-regulated DNA hypermethylation. JNK2 could be a novel therapeutic target for alcohol withdrawal treatment and/or prevention.
Collapse
Affiliation(s)
- Mei Yang
- Department of Physiology & Biophysics, Rush University Medical Center, Chicago, IL, 60607, USA
| | - Jasson Barrios
- Department of Physiology & Biophysics, Rush University Medical Center, Chicago, IL, 60607, USA
| | - Jiajie Yan
- Department of Physiology & Biophysics, Rush University Medical Center, Chicago, IL, 60607, USA
| | - Weiwei Zhao
- Department of Physiology & Biophysics, Rush University Medical Center, Chicago, IL, 60607, USA
| | - Shengtao Yuan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, 210009, China
| | - Erbo Dong
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, IL, 60612, USA.
| | - Xun Ai
- Department of Physiology & Biophysics, Rush University Medical Center, Chicago, IL, 60607, USA.
| |
Collapse
|
28
|
Ang MJ, Lee S, Kim JC, Kim SH, Moon C. Behavioral Tasks Evaluating Schizophrenia-like Symptoms in Animal Models: A Recent Update. Curr Neuropharmacol 2021; 19:641-664. [PMID: 32798374 PMCID: PMC8573744 DOI: 10.2174/1570159x18666200814175114] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 07/23/2020] [Accepted: 07/31/2020] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Schizophrenia is a serious mental illness that affects more than 21 million people worldwide. Both genetics and the environment play a role in its etiology and pathogenesis. Symptoms of schizophrenia are mainly categorized into positive, negative, and cognitive. One major approach to identify and understand these diverse symptoms in humans has been to study behavioral phenotypes in a range of animal models of schizophrenia. OBJECTIVE We aimed to provide a comprehensive review of the behavioral tasks commonly used for measuring schizophrenia-like behaviors in rodents together with an update of the recent study findings. METHODS Articles describing phenotypes of schizophrenia-like behaviors in various animal models were collected through a literature search in Google Scholar, PubMed, Web of Science, and Scopus, with a focus on advances over the last 10 years. RESULTS Numerous studies have used a range of animal models and behavioral paradigms of schizophrenia to develop antipsychotic drugs for improved therapeutics. In establishing animal models of schizophrenia, the candidate models were evaluated for schizophrenia-like behaviors using several behavioral tasks for positive, negative, and cognitive symptoms designed to verify human symptoms of schizophrenia. Such validated animal models were provided as rapid preclinical avenues for drug testing and mechanistic studies. CONCLUSION Based on the most recent advances in the field, it is apparent that a myriad of behavior tests are needed to confirm and evaluate the congruency of animal models with the numerous behaviors and clinical signs exhibited by patients with schizophrenia.
Collapse
Affiliation(s)
| | | | | | | | - Changjong Moon
- Address correspondence to this author at the Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, South Korea, Tel: +82-62-530-2838; E-mail:
| |
Collapse
|
29
|
Lai YH, Audira G, Liang ST, Siregar P, Suryanto ME, Lin HC, Villalobos O, Villaflores OB, Hao E, Lim KH, Hsiao CD. Duplicated dnmt3aa and dnmt3ab DNA Methyltransferase Genes Play Essential and Non-Overlapped Functions on Modulating Behavioral Control in Zebrafish. Genes (Basel) 2020; 11:genes11111322. [PMID: 33171840 PMCID: PMC7695179 DOI: 10.3390/genes11111322] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 12/22/2022] Open
Abstract
DNA methylation plays several roles in regulating neuronal proliferation, differentiation, and physiological functions. The major de novo methyltransferase, DNMT3, controls the DNA methylation pattern in neurons according to environmental stimulations and behavioral regulations. Previous studies demonstrated that knockout of Dnmt3 induced mouse anxiety; however, controversial results showed that activation of Dnmt3 causes anxiolytic behavior. Thus, an alternative animal model to clarify Dnmt3 on modulating behavior is crucial. Therefore, we aimed to establish a zebrafish (Danio rerio) model to clarify the function of dnmt3 on fish behavior by behavioral endpoint analyses. We evaluated the behaviors of the wild type, dnmt3aa, and dnmt3ab knockout (KO) fish by the novel tank, mirror biting, predator avoidance, social interaction, shoaling, circadian rhythm locomotor activity, color preference, and short-term memory tests. The results indicated that the dnmt3aa KO fish possessed abnormal exploratory behaviors and less fear response to the predator. On the other hand, dnmt3ab KO fish displayed less aggression, fear response to the predator, and interests to interact with their conspecifics, loosen shoaling formation, and dysregulated color preference index ranking. Furthermore, both knockout fishes showed higher locomotion activity during the night cycle, which is a sign of anxiety. However, changes in some neurotransmitter levels were observed in the mutant fishes. Lastly, whole-genome DNA methylation sequencing demonstrates a potential network of Dnmt3a proteins that is responsive to behavioral alterations. To sum up, the results suggested that the dnmt3aa KO or dnmt3ab KO fish display anxiety symptoms, which supported the idea that Dnmt3 modulates the function involved in emotional control, social interaction, and cognition.
Collapse
Affiliation(s)
- Yu-Heng Lai
- Department of Chemistry, Chinese Culture University, Taipei 11114, Taiwan;
| | - Gilbert Audira
- Department of Chemistry, Chung Yuan Christian University, Chung-Li 320314, Taiwan; (G.A.); (P.S.)
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li 320314, Taiwan; (S.-T.L.); (M.E.S.)
| | - Sung-Tzu Liang
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li 320314, Taiwan; (S.-T.L.); (M.E.S.)
| | - Petrus Siregar
- Department of Chemistry, Chung Yuan Christian University, Chung-Li 320314, Taiwan; (G.A.); (P.S.)
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li 320314, Taiwan; (S.-T.L.); (M.E.S.)
| | - Michael Edbert Suryanto
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li 320314, Taiwan; (S.-T.L.); (M.E.S.)
| | - Huan-Chau Lin
- Division of Hematology and Oncology, Department of Internal Medicine, Mackay Memorial Hospital, Number 92, Section 2, Chungshan North Road, Taipei 10449, Taiwan;
| | - Omar Villalobos
- Department of Pharmacy, Faculty of Pharmacy, University of Santo Tomas, Manila 1015, Philippines;
| | - Oliver B. Villaflores
- Department of Biochemistry, Faculty of Pharmacy, University of Santo Tomas, Manila 1015, Philippines;
| | - Erwei Hao
- Guangxi Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning 530200, Guangxi, China
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning 530200, Guangxi, China
- Correspondence: (E.H.); (K.-H.L.); (C.-D.H.)
| | - Ken-Hong Lim
- Division of Hematology and Oncology, Department of Internal Medicine, Mackay Memorial Hospital, Number 92, Section 2, Chungshan North Road, Taipei 10449, Taiwan;
- Department of Medicine, MacKay Medical College, Sanzhi Dist., New Taipei City 252, Taiwan
- Correspondence: (E.H.); (K.-H.L.); (C.-D.H.)
| | - Chung-Der Hsiao
- Department of Chemistry, Chung Yuan Christian University, Chung-Li 320314, Taiwan; (G.A.); (P.S.)
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li 320314, Taiwan; (S.-T.L.); (M.E.S.)
- Center of Nanotechnology, Chung Yuan Christian University, Chung-Li 320314, Taiwan
- Correspondence: (E.H.); (K.-H.L.); (C.-D.H.)
| |
Collapse
|
30
|
Linde J, Zimmer-Bensch G. DNA Methylation-Dependent Dysregulation of GABAergic Interneuron Functionality in Neuropsychiatric Diseases. Front Neurosci 2020; 14:586133. [PMID: 33041771 PMCID: PMC7525021 DOI: 10.3389/fnins.2020.586133] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 08/25/2020] [Indexed: 12/30/2022] Open
Abstract
Neuropsychiatric diseases, such as mood disorders, schizophrenia, and autism, represent multifactorial disorders, differing in causes, disease onset, severity, and symptoms. A common feature of numerous neuropsychiatric conditions are defects in the cortical inhibitory GABAergic system. The balance of excitation and inhibition is fundamental for proper and efficient information processing in the cerebral cortex. Thus, altered inhibition is suggested to account for pathological symptoms like cognitive impairments and dysfunctional multisensory integration. While it became apparent that most of these diseases have a clear genetic component, environmental influences emerged as an impact of disease manifestation, onset, and severity. Epigenetic mechanisms of transcriptional control, such as DNA methylation, are known to be responsive to external stimuli, and are suspected to be implicated in the functional impairments of GABAergic interneurons, and hence, the pathophysiology of neuropsychiatric diseases. Here, we provide an overview about the multifaceted functional implications of DNA methylation and DNA methyltransferases in cortical interneuron development and function in health and disease. Apart from the regulation of gamma-aminobutyric acid-related genes and genes relevant for interneuron development, we discuss the role of DNA methylation-dependent regulation of synaptic transmission by the modulation of endocytosis-related genes as potential pathophysiological mechanisms underlying neuropsychiatric conditions. Deciphering the hierarchy and mechanisms of changes in epigenetic signatures is crucial to develop effective strategies for treatment and prevention.
Collapse
Affiliation(s)
- Jenice Linde
- Division of Functional Epigenetics in the Animal Model, Institute for Biology II, RWTH Aachen University, Aachen, Germany.,Research Training Group 2416 MultiSenses - MultiScales, RWTH Aachen University, Aachen, Germany
| | - Geraldine Zimmer-Bensch
- Division of Functional Epigenetics in the Animal Model, Institute for Biology II, RWTH Aachen University, Aachen, Germany.,Research Training Group 2416 MultiSenses - MultiScales, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
31
|
Raja GL, Lite C, Subhashree KD, Santosh W, Barathi S. Prenatal bisphenol-A exposure altered exploratory and anxiety-like behaviour and induced non-monotonic, sex-specific changes in the cortical expression of CYP19A1, BDNF and intracellular signaling proteins in F1 rats. Food Chem Toxicol 2020; 142:111442. [DOI: 10.1016/j.fct.2020.111442] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 05/16/2020] [Accepted: 05/17/2020] [Indexed: 12/18/2022]
|
32
|
Nass SR, Hahn YK, McLane VD, Varshneya NB, Damaj MI, Knapp PE, Hauser KF. Chronic HIV-1 Tat exposure alters anterior cingulate cortico-basal ganglia-thalamocortical synaptic circuitry, associated behavioral control, and immune regulation in male mice. Brain Behav Immun Health 2020; 5:100077. [PMID: 33083793 PMCID: PMC7571616 DOI: 10.1016/j.bbih.2020.100077] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 04/25/2020] [Indexed: 12/28/2022] Open
Abstract
HIV-1 selectively disrupts neuronal integrity within specific brain regions, reflecting differences in viral tropism and/or the regional differences in the vulnerability of distinct neuronal subpopulations within the CNS. Deficits in prefrontal cortex (PFC)-mediated executive function and the resultant loss of behavioral control are a particularly debilitating consequence of neuroHIV. To explore how HIV-1 disrupts executive function, we investigated the effects of 48 h, 2 and/or 8 weeks of HIV-1 Tat exposure on behavioral control, synaptic connectivity, and neuroimmune function in the anterior cingulate cortex (ACC) and associated cortico-basal ganglia (BG)-thalamocortical circuitry in adult, Tat transgenic male mice. HIV-1 Tat exposure increased novelty-exploration in response to novel food, flavor, and environmental stimuli, suggesting that Tat triggers increased novelty-exploration in situations of competing motivation (e.g., drive to feed or explore vs. fear of novel, brightly lit open areas). Furthermore, Tat induced adaptability in response to an environmental stressor and pre-attentive filtering deficits. The behavioral insufficiencies coincided with decreases in the inhibitory pre- and post-synaptic proteins, synaptotagmin 2 and gephyrin, respectively, in the ACC, and alterations in specific pro- and anti-inflammatory cytokines out of 23 assayed. The interaction of Tat exposure and the resultant time-dependent, selective alterations in CCL4, CXCL1, IL-12p40, and IL-17A levels in the PFC predicted significant decreases in adaptability. Tat decreased dendritic spine density and cortical VGLUT1 inputs, while increasing IL-1β, IL-6, CCL5, and CCL11 in the striatum. Alternatively, IL-1α, CCL5, and IL-13 were decreased in the mediodorsal thalamus despite the absence of synaptic changes. Thus, HIV-1 Tat appears to uniquely and systematically disrupt immune regulation and the inhibitory and excitatory synaptic balance throughout the ACC-BG-thalamocortical circuitry resulting in a loss of behavioral control.
Collapse
Affiliation(s)
- Sara R. Nass
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA, 23298-0613, USA
| | - Yun K. Hahn
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, P.O. Box 980709, Richmond, VA, 23298-0709, USA
| | - Virginia D. McLane
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA, 23298-0613, USA
| | - Neil B. Varshneya
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA, 23298-0613, USA
| | - M. Imad Damaj
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA, 23298-0613, USA
| | - Pamela E. Knapp
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA, 23298-0613, USA
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, P.O. Box 980709, Richmond, VA, 23298-0709, USA
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, P.O. Box 980059, Richmond, VA, 23298-0059, USA
| | - Kurt F. Hauser
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA, 23298-0613, USA
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, P.O. Box 980709, Richmond, VA, 23298-0709, USA
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, P.O. Box 980059, Richmond, VA, 23298-0059, USA
| |
Collapse
|
33
|
Pensold D, Reichard J, Van Loo KMJ, Ciganok N, Hahn A, Bayer C, Liebmann L, Groß J, Tittelmeier J, Lingner T, Salinas-Riester G, Symmank J, Halfmann C, González-Bermúdez L, Urbach A, Gehrmann J, Costa I, Pieler T, Hübner CA, Vatter H, Kampa B, Becker AJ, Zimmer-Bensch G. DNA Methylation-Mediated Modulation of Endocytosis as Potential Mechanism for Synaptic Function Regulation in Murine Inhibitory Cortical Interneurons. Cereb Cortex 2020; 30:3921-3937. [PMID: 32147726 PMCID: PMC7264686 DOI: 10.1093/cercor/bhaa009] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 12/14/2019] [Accepted: 01/10/2020] [Indexed: 12/25/2022] Open
Abstract
The balance of excitation and inhibition is essential for cortical information processing, relying on the tight orchestration of the underlying subcellular processes. Dynamic transcriptional control by DNA methylation, catalyzed by DNA methyltransferases (DNMTs), and DNA demethylation, achieved by ten–eleven translocation (TET)-dependent mechanisms, is proposed to regulate synaptic function in the adult brain with implications for learning and memory. However, focus so far is laid on excitatory neurons. Given the crucial role of inhibitory cortical interneurons in cortical information processing and in disease, deciphering the cellular and molecular mechanisms of GABAergic transmission is fundamental. The emerging relevance of DNMT and TET-mediated functions for synaptic regulation irrevocably raises the question for the targeted subcellular processes and mechanisms. In this study, we analyzed the role dynamic DNA methylation has in regulating cortical interneuron function. We found that DNMT1 and TET1/TET3 contrarily modulate clathrin-mediated endocytosis. Moreover, we provide evidence that DNMT1 influences synaptic vesicle replenishment and GABAergic transmission, presumably through the DNA methylation-dependent transcriptional control over endocytosis-related genes. The relevance of our findings is supported by human brain sample analysis, pointing to a potential implication of DNA methylation-dependent endocytosis regulation in the pathophysiology of temporal lobe epilepsy, a disease characterized by disturbed synaptic transmission.
Collapse
Affiliation(s)
- Daniel Pensold
- Institute of Human Genetics, University Hospital Jena, 07743 Jena, Germany.,Division of Functional Epigenetics, Institute of Zoology (Biology 2), RWTH Aachen University, 52074 Aachen, Germany
| | - Julia Reichard
- Institute of Human Genetics, University Hospital Jena, 07743 Jena, Germany.,Division of Functional Epigenetics, Institute of Zoology (Biology 2), RWTH Aachen University, 52074 Aachen, Germany.,Research Training Group 2416 Multi Senses-Multi Scales, RWTH Aachen University, 52074 Aachen, Germany
| | - Karen M J Van Loo
- Department of Neuropathology, Section for Translational Epilepsy Research, University of Bonn Medical Center, 53105 Bonn, Germany
| | - Natalja Ciganok
- Division of Systems Neurophysiology, Institute of Zoology (Biology 2), RWTH Aachen University, 52074 Aachen, Germany
| | - Anne Hahn
- Institute of Human Genetics, University Hospital Jena, 07743 Jena, Germany
| | - Cathrin Bayer
- Institute of Human Genetics, University Hospital Jena, 07743 Jena, Germany.,Division of Functional Epigenetics, Institute of Zoology (Biology 2), RWTH Aachen University, 52074 Aachen, Germany
| | - Lutz Liebmann
- Institute of Human Genetics, University Hospital Jena, 07743 Jena, Germany
| | - Jonas Groß
- Institute of Human Genetics, University Hospital Jena, 07743 Jena, Germany
| | | | - Thomas Lingner
- Department of Developmental Biochemistry, Transcriptome and Genome Analysis Laboratory (TAL), University of Goettingen, 37077 Goettingen, Germany
| | - Gabriela Salinas-Riester
- Department of Developmental Biochemistry, Transcriptome and Genome Analysis Laboratory (TAL), University of Goettingen, 37077 Goettingen, Germany
| | - Judit Symmank
- Institute of Human Genetics, University Hospital Jena, 07743 Jena, Germany
| | - Claas Halfmann
- Division of Systems Neurophysiology, Institute of Zoology (Biology 2), RWTH Aachen University, 52074 Aachen, Germany
| | | | - Anja Urbach
- Clinic for Neurology, University Hospital Jena, 07743 Jena, Germany
| | - Julia Gehrmann
- Institute for Computational Genomics, University Hospital Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Ivan Costa
- Institute for Computational Genomics, University Hospital Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Tomas Pieler
- Department of Developmental Biochemistry, Centre for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University of Goettingen, 37077 Goettingen, Germany
| | - Christian A Hübner
- Institute of Human Genetics, University Hospital Jena, 07743 Jena, Germany
| | - Hartmut Vatter
- Clinic for Neurosurgery, University of Bonn Medical Center, 53105 Bonn, Germany
| | - Björn Kampa
- Division of Systems Neurophysiology, Institute of Zoology (Biology 2), RWTH Aachen University, 52074 Aachen, Germany.,JARA BRAIN, Institute for Neuroscience and Medicine, Forschungszentrum Jülich, 52425, Germany
| | - Albert J Becker
- Department of Neuropathology, Section for Translational Epilepsy Research, University of Bonn Medical Center, 53105 Bonn, Germany
| | - Geraldine Zimmer-Bensch
- Institute of Human Genetics, University Hospital Jena, 07743 Jena, Germany.,Division of Functional Epigenetics, Institute of Zoology (Biology 2), RWTH Aachen University, 52074 Aachen, Germany.,Research Training Group 2416 Multi Senses-Multi Scales, RWTH Aachen University, 52074 Aachen, Germany
| |
Collapse
|
34
|
Bacosides Encapsulated in Lactoferrin Conjugated PEG-PLA-PCL-OH Based Polymersomes Act as Epigenetic Modulator in Chemically Induced Amnesia. Neurochem Res 2020; 45:796-808. [DOI: 10.1007/s11064-020-02953-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/04/2019] [Accepted: 01/02/2020] [Indexed: 12/13/2022]
|
35
|
Abstract
Depression is one of the most common psychiatric disorders affecting public health. Studies over the past years suggest that the methylations of some specific genes such as BDNF, SLC6A4, and NR3C1 play an important role in the development of depression. Recently, epigenetic evidences suggest that the expression levels of DNA methyltransferases differ in several brain areas including the prefrontal cortex, hippocampus, amygdala, and nucleus accumbens in depression patients and animal models, but the potential link between the expression levels of DNA methylatransferases and the methylations of specific genes needs further investigation to clarify the pathogenesis of depression.
Collapse
Affiliation(s)
- Zhenghao Duan
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China.,Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jie Lu
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| |
Collapse
|
36
|
de Nijs L, Choe K, Steinbusch H, Schijns OEMG, Dings J, van den Hove DLA, Rutten BPF, Hoogland G. DNA methyltransferase isoforms expression in the temporal lobe of epilepsy patients with a history of febrile seizures. Clin Epigenetics 2019; 11:118. [PMID: 31426844 PMCID: PMC6701147 DOI: 10.1186/s13148-019-0721-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 08/02/2019] [Indexed: 11/10/2022] Open
Abstract
Background Temporal lobe epilepsy (TLE) with hippocampal sclerosis (HS) is a common pharmaco-resistant epilepsy referred for adult epilepsy surgery. Though associated with prolonged febrile seizures (FS) in childhood, the neurobiological basis for this relationship is not fully understood and currently no preventive or curative therapies are available. DNA methylation, an epigenetic mechanism catalyzed by DNA methyltransferases (DNMTs), potentially plays a pivotal role in epileptogenesis associated with FS. In an attempt to start exploring this notion, the present cross-sectional pilot study investigated whether global DNA methylation levels (5-mC and 5-hmC markers) and DNMT isoforms (DNMT1, DNMT3a1, and DNMT3a2) expression would be different in hippocampal and neocortical tissues between controls and TLE patients with or without a history of FS. Results We found that global DNA methylation levels and DNMT3a2 isoform expression were lower in the hippocampus for all TLE groups when compared to control patients, with a more significant decrease amongst the TLE groups with a history of FS. Interestingly, we showed that DNMT3a1 expression was severely diminished in the hippocampus of TLE patients with a history of FS in comparison with control and other TLE groups. In the neocortex, we found a higher expression of DNMT1 and DNMT3a1 as well as increased levels of global DNA methylation for all TLE patients compared to controls. Conclusion Together, the findings of this descriptive cross-sectional pilot study demonstrated brain region-specific changes in DNMT1 and DNMT3a isoform expression as well as global DNA methylation levels in human TLE with or without a history of FS. They highlighted a specific implication of DNMT3a isoforms in TLE after FS. Therefore, longitudinal studies that aim at targeting DNMT3a isoforms to evaluate the potential causal relationship between FS and TLE or treatment of FS-induced epileptogenesis seem warranted. Electronic supplementary material The online version of this article (10.1186/s13148-019-0721-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Laurence de Nijs
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 50, 6229, ER, Maastricht, The Netherlands. .,GIGA-Neurosciences, University of Liège, Liège, Belgium.
| | - Kyonghwan Choe
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 50, 6229, ER, Maastricht, The Netherlands
| | - Hellen Steinbusch
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 50, 6229, ER, Maastricht, The Netherlands
| | - Olaf E M G Schijns
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 50, 6229, ER, Maastricht, The Netherlands.,Department of Neurosurgery, Maastricht University Medical Center, Maastricht, The Netherlands.,Academic Center for Epileptology (ACE), Maastricht University Medical Center, Maastricht, The Netherlands
| | - Jim Dings
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 50, 6229, ER, Maastricht, The Netherlands.,Department of Neurosurgery, Maastricht University Medical Center, Maastricht, The Netherlands.,Academic Center for Epileptology (ACE), Maastricht University Medical Center, Maastricht, The Netherlands
| | - Daniel L A van den Hove
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 50, 6229, ER, Maastricht, The Netherlands.,Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany
| | - Bart P F Rutten
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 50, 6229, ER, Maastricht, The Netherlands
| | - Govert Hoogland
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 50, 6229, ER, Maastricht, The Netherlands.,Department of Neurosurgery, Maastricht University Medical Center, Maastricht, The Netherlands.,Academic Center for Epileptology (ACE), Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
37
|
Melcangi RC, Casarini L, Marino M, Santi D, Sperduti S, Giatti S, Diviccaro S, Grimoldi M, Caruso D, Cavaletti G, Simoni M. Altered methylation pattern of the SRD5A2 gene in the cerebrospinal fluid of post-finasteride patients: a pilot study. Endocr Connect 2019; 8:1118-1125. [PMID: 31272082 PMCID: PMC6652249 DOI: 10.1530/ec-19-0199] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 07/04/2019] [Indexed: 12/16/2022]
Abstract
CONTEXT Post-finasteride syndrome (PFS) occurs in patients with androgenic alopecia after suspension of the finasteride treatment, leading to a large variety of persistent side effects. Despite the severity of the clinical picture, the mechanism underlying the PFS symptoms onset and persistence is still unclear. OBJECTIVE To study whether epigenetic modifications occur in PFS patients. METHODS Retrospective analysis of a multicentric, prospective, longitudinal, case-control clinical trial, enrolling 16 PFS patients, compared to 20 age-matched healthy men. Main outcomes were methylation pattern of SRD5A1 and SRD5A2 promoters and concentration of 11 neuroactive steroids, measured by liquid chromatography-tandem mass spectrometry, in blood and cerebrospinal fluid (CSF) samples. RESULTS SRD5A1 and SRD5A2 methylation analysis was performed in all blood samples (n = 16 PFS patients and n = 20 controls), in 16 CSF samples from PFS patients and in 13 CSF samples from controls. The SRD5A2 promoter was more frequently methylated in CSF of PFS patients compared to controls (56.3 vs 7.7%). No promoter methylation was detected in blood samples in both groups. No methylation occurred in the SRD5A1 promoter of both groups. Unmethylated controls compared to unmethylated SRD5A2 patients showed higher pregnenolone, dihydrotestosterone and dihydroprogesterone, together with lower testosterone CSF levels. Andrological and neurological assessments did not differ between methylated and unmethylated subjects. CONCLUSIONS For the first time, we demonstrate a tissue-specific methylation pattern of SRD5A2 promoter in PFS patients. Although we cannot conclude whether this pattern is prenatally established or induced by finasteride treatment, it could represent an important mechanism of neuroactive steroid levels and behavioural disturbances previously described in PFS.
Collapse
Affiliation(s)
- Roberto Cosimo Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Marco Marino
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Daniele Santi
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Department of Medical Specialties, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Samantha Sperduti
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Silvia Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Silvia Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Maria Grimoldi
- Neurology Division, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Donatella Caruso
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Guido Cavaletti
- Experimental Neurology Unit and Milan Center for Neuroscience, School of Medicine and Surgery, University of Milano Bicocca, Monza, Italy
| | - Manuela Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, Modena, Italy
- Department of Medical Specialties, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
- Correspondence should be addressed to M Simoni:
| |
Collapse
|
38
|
Todorov G, Cunha C. Hypothesis: Regulation of neuroplasticity may involve I-motif and G-quadruplex DNA formation modulated by epigenetic mechanisms. Med Hypotheses 2019; 127:129-135. [PMID: 31088636 DOI: 10.1016/j.mehy.2019.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 01/12/2023]
Abstract
Recent studies demonstrated the existence in vivo of various functional DNA structures that differ from the double helix. The G-quadruplex (G4) and intercalated motif (I-motif or IM) DNA structures are formed as knots where, correspondingly, guanines or cytosines on the same strand of DNA bind to each other. There are grounds to believe that G4 and IM sequences play a significant role in regulating gene expression considering their tendency to be found in or near regulatory sites (such as promoters, enhancers, and telomeres) as well as the correlation between the prevalence of G4 or IM conformations and specific phases of cell cycle. Notably, G4 and IM capable sequences tend to be found on the opposite strands of the same DNA site with at most one of the two structures formed at any given time. The recent evidence that K+, Mg2+ concentrations directly affect IM formation (and likely G4 formation indirectly) lead us to believe that these structures may play a major role in synaptic plasticity of neurons, and, therefore, in a variety of central nervous system (CNS) functions including memory, learning, habitual behaviors, pain perception and others. Furthermore, epigenetic mechanisms, which have an important role in synaptic plasticity and memory formation, were also shown to influence formation and stability of G4s and IMs. Our hypothesis is that non-canonical DNA and RNA structures could be an integral part of neuroplasticity control via gene expression regulation at the level of transcription, translation and splicing. We propose that the regulatory activity of DNA IM and G4 structures is modulated by DNA methylation/demethylation of the IM and/or G4 sequences, which facilitates the switch between canonical and non-canonical conformation. Other neuronal mechanisms interacting with the formation and regulatory activity of non-canonical DNA and RNA structures, particularly G4, IM and triplexes, may involve microRNAs as well as ion and proton fluxes. We are proposing experiments in acute brain slices and in vivo to test our hypothesis. The proposed studies would provide new insights into fundamental neuronal mechanisms in health and disease and potentially open new avenues for treating mental health disorders.
Collapse
Affiliation(s)
- German Todorov
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, NY, USA
| | - Catarina Cunha
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, NY, USA.
| |
Collapse
|
39
|
Walton E, Relton CL, Caramaschi D. Using Openly Accessible Resources to Strengthen Causal Inference in Epigenetic Epidemiology of Neurodevelopment and Mental Health. Genes (Basel) 2019; 10:E193. [PMID: 30832291 PMCID: PMC6470715 DOI: 10.3390/genes10030193] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 01/25/2019] [Indexed: 12/18/2022] Open
Abstract
The recent focus on the role of epigenetic mechanisms in mental health has led to several studies examining the association of epigenetic processes with psychiatric conditions and neurodevelopmental traits. Some studies suggest that epigenetic changes might be causal in the development of the psychiatric condition under investigation. However, other scenarios are possible, e.g., statistical confounding or reverse causation, making it particularly challenging to derive conclusions on causality. In the present review, we examine the evidence from human population studies for a possible role of epigenetic mechanisms in neurodevelopment and mental health and discuss methodological approaches on how to strengthen causal inference, including the need for replication, (quasi-)experimental approaches and Mendelian randomization. We signpost openly accessible resources (e.g., "MR-Base" "EWAS catalog" as well as tissue-specific methylation and gene expression databases) to aid the application of these approaches.
Collapse
Affiliation(s)
- Esther Walton
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, BS8 2BN Bristol, UK.
- Population Health Sciences, Bristol Medical School, University of Bristol, BS8 2BN Bristol, UK.
- Department of Psychology, University of Bath, BA2 7AY Bath, UK.
| | - Caroline L Relton
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, BS8 2BN Bristol, UK.
- Population Health Sciences, Bristol Medical School, University of Bristol, BS8 2BN Bristol, UK.
| | - Doretta Caramaschi
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, BS8 2BN Bristol, UK.
- Population Health Sciences, Bristol Medical School, University of Bristol, BS8 2BN Bristol, UK.
| |
Collapse
|
40
|
Rey R, Chauvet-Gelinier JC, Suaud-Chagny MF, Ragot S, Bonin B, d'Amato T, Teyssier JR. Distinct Expression Pattern of Epigenetic Machinery Genes in Blood Leucocytes and Brain Cortex of Depressive Patients. Mol Neurobiol 2018; 56:4697-4707. [PMID: 30377985 PMCID: PMC6647377 DOI: 10.1007/s12035-018-1406-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 10/24/2018] [Indexed: 12/25/2022]
Abstract
In major depressive disorder (MDD), altered gene expression in brain cortex and blood leucocytes may be due to aberrant expression of epigenetic machinery coding genes. Here, we explore the expression of these genes both at the central and peripheral levels. Using real-time quantitative PCR technique, we first measured expression levels of genes encoding DNA and histone modifying enzymes in the dorsolateral prefrontal cortex (DLPFC) and cingulate cortex (CC) of MDD patients (n = 24) and healthy controls (n = 12). For each brain structure, transcripts levels were compared between subject groups. In an exploratory analysis, we then compared the candidate gene expressions between a subgroup of MDD patients with psychotic characteristics (n = 13) and the group of healthy subjects (n = 12). Finally, we compared transcript levels of the candidate genes in blood leucocytes between separate samples of MDD patients (n = 17) and healthy controls (n = 16). In brain and blood leucocytes of MDD patients, we identified an overexpression of genes encoding enzymes which transfer repressive transcriptional marks: HDAC4-5-6-8 and DNMT3B in the DLPFC, HDAC2 in the CC and blood leucocytes. In the DLPFC of patients with psychotic characteristics, two genes (KAT2A and UBE2A) were additionally overexpressed suggesting a shift to a more transcriptionally permissive conformation of chromatin. Aberrant activation of epigenetic repressive systems may be involved in MDD pathogenesis both in brain tissue and blood leucocytes.
Collapse
Affiliation(s)
- Romain Rey
- INSERM, U1028; CNRS, UMR5292; Lyon Neuroscience Research Center, Psychiatric Disorders: from Resistance to Response Team, F-69000, Lyon, France. .,University Lyon 1, F-69000, Villeurbanne, France. .,Schizophrenia Expert Centre, Le Vinatier Hospital, Bron, France. .,INSERM U1028; CNRS UMR5292; Université Claude Bernard Lyon 1; Centre de Recherche en Neurosciences de Lyon, Equipe PSYR2; Centre Hospitalier Le Vinatier, Pole Est, Centre Expert Schizophrénie, 95 boulevard Pinel BP 30039, 69678, Bron Cedex, France.
| | - Jean-Christophe Chauvet-Gelinier
- Psychiatry Unit, Neurosciences Department, Le Bocage University Hospital, Marion Building, Dijon, France.,Laboratory of Psychopathology and Medical Psychology (IFR 100), Bourgogne University, Dijon, France
| | - Marie-Françoise Suaud-Chagny
- INSERM, U1028; CNRS, UMR5292; Lyon Neuroscience Research Center, Psychiatric Disorders: from Resistance to Response Team, F-69000, Lyon, France.,University Lyon 1, F-69000, Villeurbanne, France.,Schizophrenia Expert Centre, Le Vinatier Hospital, Bron, France
| | - Sylviane Ragot
- Department of Genetics and Laboratory of Molecular Genetics, University Hospital, Dijon, France
| | - Bernard Bonin
- Psychiatry Unit, Neurosciences Department, Le Bocage University Hospital, Marion Building, Dijon, France.,Laboratory of Psychopathology and Medical Psychology (IFR 100), Bourgogne University, Dijon, France
| | - Thierry d'Amato
- INSERM, U1028; CNRS, UMR5292; Lyon Neuroscience Research Center, Psychiatric Disorders: from Resistance to Response Team, F-69000, Lyon, France.,University Lyon 1, F-69000, Villeurbanne, France.,Schizophrenia Expert Centre, Le Vinatier Hospital, Bron, France
| | - Jean-Raymond Teyssier
- Department of Genetics and Laboratory of Molecular Genetics, University Hospital, Dijon, France
| |
Collapse
|
41
|
Larsen K, Kristensen KK, Callesen H. DNA methyltransferases and tRNA methyltransferase DNMT2 in developing pig brain - expression and promoter methylation. GENE REPORTS 2018. [DOI: 10.1016/j.genrep.2018.02.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
42
|
Gong Z, Zhou Q. Dnmt3a in the dorsal dentate gyrus is a key regulator of fear renewal. Sci Rep 2018; 8:5093. [PMID: 29572461 PMCID: PMC5865109 DOI: 10.1038/s41598-018-23533-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 03/14/2018] [Indexed: 12/13/2022] Open
Abstract
Renewal of extinguished fear memory in an altered context is widely believed to be a major limiting issue for exposure therapy in treating various psychiatric diseases. Effective prevention of fear renewal will significantly improve the efficacy of exposure therapy. DNA methyltransferase (DNMTs) mediated epigenetic processes play critical roles in long term memory, but little is known about their functions in fear memory extinction or renewal. Here we investigated whether DNMTs regulate fear renewal after extinction. We found that elevated Dnmt3a level in the dorsal dentate gyrus (dDG) of hippocampus was associated with the absence of fear renewal in an altered context after extinction training. Overexpression and knockdown of Dnmt3a in the dDG regulated the occurrence of fear renewal in a bi-directional manner. In addition, Dnmt3a overexpression was associated with elevated expression of c-Fos in the dDG during extinction training. Furthermore, we found that renewal of remote fear memory can be prevented, and the absence of renewal was concurrent with an elevated Dnmt3a level. Our results indicate that Dnmt3a in the dDG is a key regulator of fear renewal after extinction, and Dnmt3a may play a critical role in controlling fear memory return and thus has therapeutic values.
Collapse
Affiliation(s)
- Zhiting Gong
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Qiang Zhou
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China.
| |
Collapse
|
43
|
Antidepressant administration modulates stress-induced DNA methylation and DNA methyltransferase expression in rat prefrontal cortex and hippocampus. Behav Brain Res 2018; 343:8-15. [PMID: 29378290 DOI: 10.1016/j.bbr.2018.01.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 01/18/2018] [Accepted: 01/19/2018] [Indexed: 12/23/2022]
Abstract
Stress and antidepressant treatment can modulate DNA methylation in promoter region of genes related to neuroplasticity and mood regulation, thus implicating this epigenetic mechanism in depression neurobiology and treatment. Accordingly, systemic administration of DNA methyltransferase (DNMT) inhibitors induces antidepressant-like effects in rodents. DNA methylation is conveyed by DNMT 1, 3a and 3b isoforms, which are differentially expressed in the brain. In order to investigate if the behavioral effects of antidepressants could be associated with changes in DNA methylation and DNMT expression, we investigated the effects induced by acute and repeated antidepressant treatment on DNA methylation and DNMT expression (1, 3a and 3b isoforms) in different brain regions of rats exposed to a stress model of depression, the learned helplessness (LH). Therefore, rats were exposed to pretest and treated with one or seven injections of vehicle or imipramine (15 mg kg-1), with test session performed one hour after the last injection. Chronic, but not acute, imipramine administration attenuated escape failures during the test, a well described antidepressant-like effect in this model. DNA methylation and DNMT (1, 3a and 3b) levels were measured in the dorsal and ventral hippocampus (dHPC, vHPC) and in the prefrontal cortex (PFC) of rats exposed to stress and treatment. Stress increased DNA methylation, DNMT3a and DNMT3b expression in the dHPC and PFC. Chronic, but not acute, imipramine administration attenuated stress effects only in the PFC. These results suggest the regulation of DNA methylation in the PFC may be an important mechanism for antidepressant-like effects in the LH model.
Collapse
|
44
|
Wright EC, Johnson SA, Hao R, Kowalczyk AS, Greenberg GD, Sanchez EO, Laman-Maharg A, Trainor BC, Rosenfeld CS. Exposure to extrinsic stressors, social defeat or bisphenol A, eliminates sex differences in DNA methyltransferase expression in the amygdala. J Neuroendocrinol 2017; 29:10.1111/jne.12475. [PMID: 28406523 PMCID: PMC5501704 DOI: 10.1111/jne.12475] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 03/22/2017] [Accepted: 03/30/2017] [Indexed: 12/24/2022]
Abstract
Chemical and psychological stressors can exert long lasting changes in brain function and behaviour. Changes in DNA methylation have been shown to be an important mechanism mediating long lasting changes in neural function and behaviour, especially for anxiety-like or stress responses. In the present study, we examined the effects of either a social or chemical stressor on DNA methyltransferase (DNMT) gene expression in the amygdala, an important brain region modulating stress responses and anxiety. In adult California mice (Peromyscus californicus) that were naïve to social defeat, females had higher levels of Dnmt1 expression in punch samples of the central amygdala (CeA) than males. In addition, mice that underwent social defeat stress showed reduced Dnmt1 and Dnmt3a expression in the CeA of females but not males. A second study using more anatomically specific punch samples replicated these effects for Dnmt1. Perinatal exposure (spanning from periconception through lactation) to bisphenol A or ethinyl oestradiol (oestrogens in birth control pills) also abolished sex differences in Dnmt1 expression in the CeA but not the basolateral amygdala. These findings identify a robust sex difference in Dnmt1 expression in the CeA that is sensitive to both psychological and chemical stressors. Future studies should aim to examine the impact of psychological and chemical stressors on DNA methylation in the CeA and also investigate whether Dnmt1 may have an underappreciated role in plasticity in behaviour.
Collapse
Affiliation(s)
- Emily C. Wright
- Department of Psychology, University of California, Davis, CA, USA
| | - Sarah A. Johnson
- Bond Life Science Center, Department of Biomedical Sciences, Department of Animal Science, University of Missouri, Columbia, MO, USA
| | - Rebecca Hao
- Department of Psychology, University of California, Davis, CA, USA
| | | | - Gian D. Greenberg
- Neuroscience Graduate Group, University of California, Davis, CA, USA
| | | | | | - Brian C. Trainor
- Department of Psychology, University of California, Davis, CA, USA
- Neuroscience Graduate Group, University of California, Davis, CA, USA
| | - Cheryl S. Rosenfeld
- Bond Life Science Center, Department of Biomedical Sciences, Department of Animal Science, University of Missouri, Columbia, MO, USA
- Genetics Area Program and Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, Columbia, MO, USA
| |
Collapse
|
45
|
Campos AC, Fogaça MV, Scarante FF, Joca SRL, Sales AJ, Gomes FV, Sonego AB, Rodrigues NS, Galve-Roperh I, Guimarães FS. Plastic and Neuroprotective Mechanisms Involved in the Therapeutic Effects of Cannabidiol in Psychiatric Disorders. Front Pharmacol 2017; 8:269. [PMID: 28588483 PMCID: PMC5441138 DOI: 10.3389/fphar.2017.00269] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 05/01/2017] [Indexed: 12/25/2022] Open
Abstract
Beneficial effects of cannabidiol (CBD) have been described for a wide range of psychiatric disorders, including anxiety, psychosis, and depression. The mechanisms responsible for these effects, however, are still poorly understood. Similar to clinical antidepressant or atypical antipsychotic drugs, recent findings clearly indicate that CBD, either acutely or repeatedly administered, induces plastic changes. For example, CBD attenuates the decrease in hippocampal neurogenesis and dendrite spines density induced by chronic stress and prevents microglia activation and the decrease in the number of parvalbumin-positive GABA neurons in a pharmacological model of schizophrenia. More recently, it was found that CBD modulates cell fate regulatory pathways such as autophagy and others critical pathways for neuronal survival in neurodegenerative experimental models, suggesting the potential benefit of CBD treatment for psychiatric/cognitive symptoms associated with neurodegeneration. These changes and their possible association with CBD beneficial effects in psychiatric disorders are reviewed here.
Collapse
Affiliation(s)
- Alline C Campos
- Department of Pharmacology, Centre for Interdisciplinary Research on Applied Neurosciences (NAPNA), School of Medicine of Ribeirão Preto, University of São PauloRibeirão Preto, Brazil
| | - Manoela V Fogaça
- Department of Pharmacology, Centre for Interdisciplinary Research on Applied Neurosciences (NAPNA), School of Medicine of Ribeirão Preto, University of São PauloRibeirão Preto, Brazil
| | - Franciele F Scarante
- Department of Pharmacology, Centre for Interdisciplinary Research on Applied Neurosciences (NAPNA), School of Medicine of Ribeirão Preto, University of São PauloRibeirão Preto, Brazil
| | - Sâmia R L Joca
- Department of Physical and Chemical, School of Pharmaceutical Science of Ribeirão Preto, University of São PauloRibeirão Preto, Brazil
| | - Amanda J Sales
- Department of Physical and Chemical, School of Pharmaceutical Science of Ribeirão Preto, University of São PauloRibeirão Preto, Brazil
| | - Felipe V Gomes
- Department of Neuroscience, University of PittsburghPittsburgh, PA, United States
| | - Andreza B Sonego
- Department of Pharmacology, Centre for Interdisciplinary Research on Applied Neurosciences (NAPNA), School of Medicine of Ribeirão Preto, University of São PauloRibeirão Preto, Brazil
| | - Naielly S Rodrigues
- Department of Pharmacology, Centre for Interdisciplinary Research on Applied Neurosciences (NAPNA), School of Medicine of Ribeirão Preto, University of São PauloRibeirão Preto, Brazil
| | - Ismael Galve-Roperh
- Department of Biochemistry and Molecular Biology I, School of Biology, Complutense UniversityMadrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Universitario de Investigación en Neuroquímica and Instituto Ramón y Cajal de Investigación SanitariaMadrid, Spain
| | - Francisco S Guimarães
- Department of Pharmacology, Centre for Interdisciplinary Research on Applied Neurosciences (NAPNA), School of Medicine of Ribeirão Preto, University of São PauloRibeirão Preto, Brazil
| |
Collapse
|
46
|
Bayraktar G, Kreutz MR. Neuronal DNA Methyltransferases: Epigenetic Mediators between Synaptic Activity and Gene Expression? Neuroscientist 2017; 24:171-185. [PMID: 28513272 PMCID: PMC5846851 DOI: 10.1177/1073858417707457] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
DNMT3A and 3B are the main de novo DNA methyltransferases (DNMTs) in the brain that introduce new methylation marks to non-methylated DNA in postmitotic neurons. DNA methylation is a key epigenetic mark that is known to regulate important cellular processes in neuronal development and brain plasticity. Accumulating evidence disclosed rapid and dynamic changes in DNA methylation of plasticity-relevant genes that are important for learning and memory formation. To understand how DNMTs contribute to brain function and how they are regulated by neuronal activity is a prerequisite for a deeper appreciation of activity-dependent gene expression in health and disease. This review discusses the functional role of de novo methyltransferases and in particular DNMT3A1 in the adult brain with special emphasis on synaptic plasticity, memory formation, and brain disorders.
Collapse
Affiliation(s)
- Gonca Bayraktar
- 1 RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Michael R Kreutz
- 1 RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany.,2 Leibniz Group "Dendritic Organelles and Synaptic Function", ZMNH, Magdeburg, Germany
| |
Collapse
|
47
|
The Crucial Role of DNA Methylation and MeCP2 in Neuronal Function. Genes (Basel) 2017; 8:genes8050141. [PMID: 28505093 PMCID: PMC5448015 DOI: 10.3390/genes8050141] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/25/2017] [Accepted: 05/05/2017] [Indexed: 12/16/2022] Open
Abstract
A neuron is unique in its ability to dynamically modify its transcriptional output in response to synaptic activity while maintaining a core gene expression program that preserves cellular identity throughout a lifetime that is longer than almost every other cell type in the body. A contributing factor to the immense adaptability of a neuron is its unique epigenetic landscape that elicits locus-specific alterations in chromatin architecture, which in turn influences gene expression. One such epigenetic modification that is sensitive to changes in synaptic activity, as well as essential for maintaining cellular identity, is DNA methylation. The focus of this article is on the importance of DNA methylation in neuronal function, summarizing recent studies on critical players in the establishment of (the “writing”), the modification or erasure of (the “editing”), and the mediation of (the “reading”) DNA methylation in neurodevelopment and neuroplasticity. One “reader” of DNA methylation in particular, methyl-CpG-binding protein 2 (MeCP2), is highlighted, given its undisputed importance in neuronal function.
Collapse
|