1
|
Miyazawa Y, Furugen A, Aoyagi R, Kosugi H, Nishimura A, Umazume T, Narumi K, Kobayashi M. Alteration in folate carrier expression via histone deacetylase inhibition in BeWo human placental choriocarcinoma cells. Toxicol In Vitro 2024; 101:105934. [PMID: 39237058 DOI: 10.1016/j.tiv.2024.105934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 08/14/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024]
Abstract
Folates are essential nutrients for fetal development during pregnancy. Valproic acid (VPA), an inhibitor of histone deacetylases (HDACs), alters the expression of folate carriers in placental cells; however, the underlying mechanisms remain unclear. Here, we aimed to determine the profiles of folate carriers (folate receptor alpha [FOLR1], solute carrier [SLC]-19A1, and SLC46A1) after inhibition of HDACs, especially class I and IIa HDACs, using different inhibitors and gene knockdown tests. Quantitative polymerase chain reaction revealed that BeWo cells (a trophoblast model) expressed HDACs and folate carriers, similar to human placental villi. FOLR1 expression was upregulated by VPA, apicidin, and trichostatin A, but downregulated by MS-275 after 24 h treatment. VPA and apicidin upregulated the expression of SLC46A1. These inhibitors downregulated SLC19A1 expression. TMP269 (a class IIa inhibitor) did not affect folate carrier levels. HDAC1/2 knockdown upregulated FOLR1 and SLC46A1 levels, whereas HDAC1/3 knockdown downregulated FOLR1 levels. Our findings suggest that the pharmacological inhibition of class I HDACs alters the expression of folate carriers in BeWo cells. By contrast, HDAC inhibitors exert different regulatory effects on folate carriers. Moreover, HDAC1/2 inhibition may be a potential mechanism involved in altering FOLR1 and SLC46A1 levels.
Collapse
Affiliation(s)
- Yuki Miyazawa
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Ayako Furugen
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan.
| | - Ryoichi Aoyagi
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Haruna Kosugi
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Ayako Nishimura
- Department of Pharmacy, Hokkaido University Hospital, Kita-14-jo, Nishi-5-chome, Kita-ku, Sapporo 060-8648, Japan
| | - Takeshi Umazume
- Department of Obstetrics, Hokkaido University Hospital, Kita-14-jo, Nishi-5-chome, Kita-ku, Sapporo 060-8648, Japan
| | - Katsuya Narumi
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Masaki Kobayashi
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
2
|
Grand-Guillaume J, Mansi R, Gaonkar RH, Zanger S, Fani M, Eugster PJ, Beck Popovic M, Grouzmann E, Abid K. CUDC-907, a dual PI3K/histone deacetylase inhibitor, increases meta-iodobenzylguanidine uptake ( 123/131I-mIBG) in vitro and in vivo: a promising candidate for advancing theranostics in neuroendocrine tumors. J Transl Med 2023; 21:604. [PMID: 37679770 PMCID: PMC10485979 DOI: 10.1186/s12967-023-04466-z] [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: 04/24/2023] [Accepted: 08/22/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Neuroblastoma (NB) and pheochromocytoma/paraganglioma (PHEO/PGL) are neuroendocrine tumors. Imaging of these neoplasms is performed by scintigraphy after injection of radiolabeled meta-iodobenzylguanidine (mIBG), a norepinephrine analog taken up by tumoral cells through monoamine transporters. The pharmacological induction of these transporters is a promising approach to improve the imaging and therapy (theranostics) of these tumors. METHODS Transporters involved in mIBG internalization were identified by using transfected Human Embryonic Kidney (HEK) cells. Histone deacetylase inhibitors (HDACi) and inhibitors of the PI3K/AKT/mTOR pathway were tested in cell lines to study their effect on mIBG internalization. Studies in xenografted mice were performed to assess the effect of the most promising HDACi on 123I-mIBG uptake. RESULTS Transfected HEK cells demonstrated that the norepinephrine and dopamine transporter (NET and DAT) avidly internalizes mIBG. Sodium-4-phenylbutyrate (an HDACi), CUDC-907 (a dual HDACi and PI3K inhibitor), BGT226 (a PI3K inhibitor) and VS-5584 and rapamycin (two inhibitors of mTOR) increased mIBG internalization in a neuroblastoma cell line (IGR-NB8) by 2.9-, 2.1-, 2.5-, 1.5- and 1.3-fold, respectively, compared with untreated cells. CUDC-907 also increased mIBG internalization in two other NB cell lines and in one PHEO cell line. We demonstrated that mIBG internalization occurs primarily through the NET. In xenografted mice with IGR-NB8 cells, oral treatment with 5 mg/kg of CUDC-907 increased the tumor uptake of 123I-mIBG by 2.3- and 1.9-fold at 4 and 24 h post-injection, respectively, compared to the untreated group. CONCLUSIONS Upregulation of the NET by CUDC-907 lead to a better internalization of mIBG in vitro and in vivo.
Collapse
Affiliation(s)
- Joana Grand-Guillaume
- Catecholamine and Peptides Laboratory, Service of Clinical Pharmacology and Toxicology, Lausanne University Hospital and University of Lausanne, 1011, Lausanne, Switzerland
| | - Rosalba Mansi
- Division of Radiopharmaceutical Chemistry, Clinic of Radiology and Nuclear Medicine, University Hospital Basel, 4031, Basel, Switzerland
| | - Raghuvir H Gaonkar
- Division of Radiopharmaceutical Chemistry, Clinic of Radiology and Nuclear Medicine, University Hospital Basel, 4031, Basel, Switzerland
| | - Sandra Zanger
- Division of Radiopharmaceutical Chemistry, Clinic of Radiology and Nuclear Medicine, University Hospital Basel, 4031, Basel, Switzerland
| | - Melpomeni Fani
- Division of Radiopharmaceutical Chemistry, Clinic of Radiology and Nuclear Medicine, University Hospital Basel, 4031, Basel, Switzerland
| | - Philippe J Eugster
- Catecholamine and Peptides Laboratory, Service of Clinical Pharmacology and Toxicology, Lausanne University Hospital and University of Lausanne, 1011, Lausanne, Switzerland
| | - Maja Beck Popovic
- Pediatric Hematology-Oncology Unit, Woman-Mother-Child Department, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Eric Grouzmann
- Catecholamine and Peptides Laboratory, Service of Clinical Pharmacology and Toxicology, Lausanne University Hospital and University of Lausanne, 1011, Lausanne, Switzerland
| | - Karim Abid
- Catecholamine and Peptides Laboratory, Service of Clinical Pharmacology and Toxicology, Lausanne University Hospital and University of Lausanne, 1011, Lausanne, Switzerland.
| |
Collapse
|
3
|
Santiago NA, He B, Howard SL, Beaudin S, Strupp BJ, Smith DR. Developmental Manganese Exposure Causes Lasting Attention Deficits Accompanied by Dysregulation of mTOR Signaling and Catecholaminergic Gene Expression in Brain Prefrontal Cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.16.549215. [PMID: 37503220 PMCID: PMC10370122 DOI: 10.1101/2023.07.16.549215] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Elevated manganese (Mn) exposure is associated with attentional deficits in children, and is an environmental risk factor for attention deficit hyperactivity disorder (ADHD). We have shown that developmental Mn exposure causes lasting attention and sensorimotor deficits in a rat model of early childhood Mn exposure, and that these deficits are associated with a hypofunctioning catecholaminergic system in the prefrontal cortex (PFC), though the mechanistic basis for these deficits is not well understood. To address this, male Long-Evans rats were exposed orally to Mn (50 mg/kg/d) over PND 1-21 and attentional function was assessed in adulthood using the 5-Choice Serial Reaction Time Task. Targeted catecholaminergic system and epigenetic gene expression, followed by unbiased differential DNA methylation and gene regulation expression transcriptomics in the PFC, were performed in young adult littermates. Results show that developmental Mn exposure causes lasting focused attention deficits that are associated with reduced gene expression of tyrosine hydroxylase, dopamine transporter, and DNA methyltransferase 3a. Further, developmental Mn exposure causes broader lasting methylation and gene expression dysregulation associated with epigenetic regulation, inflammation, cell development, and hypofunctioning catecholaminergic neuronal systems. Pathway enrichment analyses uncovered mTOR and Wnt signaling pathway genes as significant transcriptomic regulators of the Mn altered transcriptome, and Western blot of total, C1 and C2 phospho-mTOR confirmed mTOR pathway dysregulation. Our findings deepen our understanding of the mechanistic basis of how developmental Mn exposure leads to lasting catecholaminergic dysfunction and attention deficits, which may aid future therapeutic interventions of environmental exposure associated disorders. Significance Statement Attention deficit hyperactivity disorder (ADHD) is associated with environmental risk factors, including exposure to neurotoxic agents. Here we used a rodent model of developmental manganese (Mn) exposure producing lasting attention deficits to show broad epigenetic and gene expression changes in the prefrontal cortex, and to identify disrupted mTOR and Wnt signaling pathways as a novel mechanism for how developmental Mn exposure may induce lasting attention and catecholaminergic system impairments. Importantly, our findings establish early development as a critical period of susceptibility to lasting deficits in attentional function caused by elevated environmental toxicant exposure. Given that environmental health threats disproportionately impact communities of color and low socioeconomic status, our findings can aid future studies to assess therapeutic interventions for vulnerable populations.
Collapse
|
4
|
Yatham LN, Liddle PF, Gonzalez M, Saraf G, Vafai N, Lam RW, Sossi V. A Positron Emission Tomography Study of Dopamine Transporter Density in Patients With Bipolar Disorder With Current Mania and Those With Recently Remitted Mania. JAMA Psychiatry 2022; 79:1217-1224. [PMID: 36322065 PMCID: PMC9631223 DOI: 10.1001/jamapsychiatry.2022.3541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 09/07/2022] [Indexed: 12/12/2022]
Abstract
Importance Although dopamine is implicated in the pathophysiology of bipolar disorder (BD), the precise alterations in the dopaminergic system remain unknown. Objective To assess dopamine transporter (DAT) density in the striatum in patients with BD with current and recently remitted mania in comparison to healthy control individuals and its correlation with severity of manic symptoms. Design, Setting, and Participants This cross-sectional study conducted in a tertiary care referral center for mood disorders in Vancouver, British Columbia, Canada, recruited 26 patients with BD (9 with current mania; 17 with recently remitted mania) and 21 matched healthy control individuals. DAT density was measured using positron emission tomography with [11C]d-threo-methylphenidate (MP). The differences between the groups in nondisplaceable binding potential (BPND) for DAT was assessed using statistical parametric mapping. The study was conducted from November 2001 to February 2007 and the data were analyzed from November 2020 to December 2021. Main Outcomes and Measures DAT density as indexed by BPND for MP across groups; manic symptom severity as measured with the Young Mania Rating Scale (YMRS) and correlated with BPND values in patients with BD. Results Of 47 total participants (mean [SD] age, 37.8 [14.4] years), 27 (57.4%) were female; 26 individuals had BD (9 with current mania and 17 with recently remitted mania) and there were 21 healthy control individuals. MP BPND was significantly lower in patients with BD in the right putamen and nucleus accumbens (mean reduction [MR] = 22%; cluster level familywise error [FWE]-corrected P < .001) as well as left putamen and caudate (MR = 24%; cluster level FWE-corrected P < .001). The reduction in BPND was more extensive and pronounced in patients with current mania, while patients with recently remitted mania had lower BPND in the left striatum but not the right. There was a significant negative correlation between YMRS scores and MP BPND in the right striatum in patients with current mania (ρ = -0.93; 95% CI, -0.99 to -0.69; P < .001) and those with recently remitted mania (ρ = 0.64; 95% CI, -0.86 to -0.23; P = .005) but not in the left striatum in either group. Conclusions and Relevance These findings indicate that mania was associated with reduced DAT density and remitted mania was associated with DAT levels that approximated those present in individuals without BD. These results have potential implications for drug development for mania.
Collapse
Affiliation(s)
- Lakshmi N. Yatham
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Peter F. Liddle
- Institute of Mental Health, University of Nottingham, Nottingham, United Kingdom
| | - Marjorie Gonzalez
- Department of Nuclear Medicine, Interior Health Authority, Kelowna, British Columbia, Canada
| | - Gayatri Saraf
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Psychiatry, University of Ottawa, Ottawa, Ontario, Canada
| | - Nasim Vafai
- Positron Emission Tomography and Magnetic Resonance Imaging, David Mowafaghian Centre for Brain Health, Vancouver, British Columbia, Canada
| | - Raymond W. Lam
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Vesna Sossi
- Positron Emission Tomography and Magnetic Resonance Imaging, David Mowafaghian Centre for Brain Health, Vancouver, British Columbia, Canada
- Department of Physics and Astronomy, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
5
|
Su Y, Lian J, Chen S, Zhang W, Deng C. Epigenetic histone acetylation modulating prenatal Poly I:C induced neuroinflammation in the prefrontal cortex of rats: a study in a maternal immune activation model. Front Cell Neurosci 2022; 16:1037105. [DOI: 10.3389/fncel.2022.1037105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 11/08/2022] [Indexed: 11/29/2022] Open
Abstract
Introduction: Neuroinflammation in the central nervous system, particularly the prefrontal cortex (PFC), plays a role in the pathogenesis of schizophrenia, which has been found to be associated with maternal immune activation (MIA). Recent evidence suggests that epigenetic regulation involves in the MIA-induced neurodevelopmental disturbance. However, it is not well-understood how epigenetic modulation is involved in the neuroinflammation and pathogenesis of schizophrenia.Methods: This study explored the modulation of histone acetylation in both neuroinflammation and neurotransmission using an MIA rat model induced by prenatal polyriboinosinic-polyribocytidylic acid (Poly I:C) exposure, specifically examining those genes that were previously observed to be impacted by the exposure, including a subunit of nuclear factor kappa-B (Rela), Nod-Like-Receptor family Pyrin domain containing 3 (Nlrp3), NMDA receptor subunit 2A (Grin2a), 5-HT2A (Htr2a), and GABAA subunit β3 (Gabrb3).Results: Our results revealed global changes of histone acetylation on H3 (H3ace) and H4 (H4ace) in the PFC of offspring rats with prenatal Poly I:C exposure. In addition, it revealed enhancement of both H3ace and H4ace binding on the promoter region of Rela, as well as positive correlations between Rela and genes encoding histone acetyltransferases (HATs) including CREB-binding protein (CBP) and E1A-associated protein p300 (EP300). Although there was no change in H3ace or H4ace enrichment on the promoter region of Nlrp3, a significant enhancement of histone deacetylase 6 (HDAC6) binding on the promoter region of Nlrp3 and a positive correlation between Nlrp3 and Hdac6 were also observed. However, prenatal Poly I:C treatment did not lead to any specific changes of H3ace and H4ace on the promoter region of the target genes encoding neurotransmitter receptors in this study.Discussion: These findings demonstrated that epigenetic modulation contributes to NF-κB/NLRP3 mediated neuroinflammation induced by prenatal Poly I:C exposure via enhancement of histone acetylation of H3ace and H4ace on Rela and HDAC6-mediated NLRP3 transcriptional activation. This may further lead to deficits in neurotransmissions and schizophrenia-like behaviors observed in offspring.
Collapse
|
6
|
Yang KC, Chen YY, Liu MN, Yang BH, Chou YH. Interactions between dopamine transporter and N-methyl-d-aspartate receptor-related amino acids on cognitive impairments in schizophrenia. Schizophr Res 2022; 248:263-270. [PMID: 36115191 DOI: 10.1016/j.schres.2022.09.022] [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: 04/25/2022] [Revised: 07/21/2022] [Accepted: 09/07/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Cognitive impairments, the main determinants of functional outcomes in schizophrenia, had limited treatment responses and need a better understanding of the mechanisms. Dysfunctions of the dopamine system and N-methyl-d-aspartate receptor (NMDAR), the primary pathophysiologies of schizophrenia, may impair cognition. This study explored the effects and interactions of striatal dopamine transporter (DAT) and plasma NMDAR-related amino acids on cognitive impairments in schizophrenia. METHODS We recruited 36 schizophrenia patients and 36 age- and sex-matched healthy controls (HC). All participants underwent cognitive assessments of attention, memory, and executive function. Single-photon emission computed tomography with 99mTc-TRODAT and ultra-performance liquid chromatography were applied to determine DAT availability and plasma concentrations of eight amino acids, respectively. RESULTS Compared with HC, schizophrenia patients had lower cognitive performance, higher methionine concentrations, decreased concentrations of glutamic acid, cysteine, aspartic acid, arginine, the ratio of glutamic acid to gamma-aminobutyric acid (Glu/GABA), and DAT availability in the left caudate nucleus (CN) and putamen. Regarding memory scores, Glu/GABA and the DAT availability in left CN and putamen exhibited positive relationships, while methionine concentrations showed negative associations in all participants. The DAT availability in left CN mediated the methionine-memory relationship. An exploratory backward stepwise regression analysis for the four biological markers associated with memory indicated that DAT availability in left CN and Glu/GABA remained in the final model. CONCLUSIONS This study demonstrated the interactions of striatal DAT and NMDAR-related amino acids on cognitive impairments in schizophrenia. Future studies to comprehensively evaluate their complex interactions and treatment implications are warranted.
Collapse
Affiliation(s)
- Kai-Chun Yang
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yen-Yu Chen
- Department of Education and Research, Taipei City Hospital, Taipei, Taiwan
| | - Mu-N Liu
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Bang-Hung Yang
- Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yuan-Hwa Chou
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Center for Quality Management, Taipei Veterans General Hospital, Taipei, Taiwan.
| |
Collapse
|
7
|
Eid A, Mhatre-Winters I, Sammoura FM, Edler MK, von Stein R, Hossain MM, Han Y, Lisci M, Carney K, Konsolaki M, Hart RP, Bennett JW, Richardson JR. Effects of DDT on Amyloid Precursor Protein Levels and Amyloid Beta Pathology: Mechanistic Links to Alzheimer's Disease Risk. ENVIRONMENTAL HEALTH PERSPECTIVES 2022; 130:87005. [PMID: 35946953 PMCID: PMC9364816 DOI: 10.1289/ehp10576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
BACKGROUND The interaction of aging-related, genetic, and environmental factors is thought to contribute to the etiology of late-onset, sporadic Alzheimer's disease (AD). We previously reported that serum levels of p,p'-dichlorodiphenyldichloroethylene (DDE), a long-lasting metabolite of the organochlorine pesticide dichlorodiphenyltrichloroethane (DDT), were significantly elevated in patients with AD and associated with the risk of AD diagnosis. However, the mechanism by which DDT may contribute to AD pathogenesis is unknown. OBJECTIVES This study sought to assess effects of DDT exposure on the amyloid pathway in multiple in vitro and in vivo models. METHODS Cultured cells (SH-SY5Y and primary neurons), transgenic flies overexpressing amyloid beta (Aβ), and C57BL/6J and 3xTG-AD mice were treated with DDT to assess impacts on the amyloid pathway. Real time quantitative polymerase chain reaction, multiplex assay, western immunoblotting and immunohistochemical methods were used to assess the effects of DDT on amyloid precursor protein (APP) and other contributors to amyloid processing and deposition. RESULTS Exposure to DDT revealed significantly higher APP mRNA and protein levels in immortalized and primary neurons, as well as in wild-type and AD-models. This was accompanied by higher levels of secreted Aβ in SH-SY5Y cells, an effect abolished by the sodium channel antagonist tetrodotoxin. Transgenic flies and 3xTG-AD mice had more Aβ pathology following DDT exposure. Furthermore, loss of the synaptic markers synaptophysin and PSD95 were observed in the cortex of the brains of 3xTG-AD mice. DISCUSSION Sporadic Alzheimer's disease risk involves contributions from genetic and environmental factors. Here, we used multiple model systems, including primary neurons, transgenic flies, and mice to demonstrate the effects of DDT on APP and its pathological product Aβ. These data, combined with our previous epidemiological findings, provide a mechanistic framework by which DDT exposure may contribute to increased risk of AD by impacting the amyloid pathway. https://doi.org/10.1289/EHP10576.
Collapse
Affiliation(s)
- Aseel Eid
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA
| | - Isha Mhatre-Winters
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA
- School of Biomedical Sciences, Kent State University, Kent, Ohio, USA
| | - Ferass M. Sammoura
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA
| | - Melissa K. Edler
- School of Biomedical Sciences, Kent State University, Kent, Ohio, USA
- Department of Anthropology, Kent State University, Kent, Ohio, USA
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
| | - Richard von Stein
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, New Jersey, USA
| | - Muhammad M. Hossain
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, New Jersey, USA
| | - Yoonhee Han
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA
| | - Miriam Lisci
- Department of Genetics, Rutgers University, Piscataway, New Jersey, USA
| | - Kristina Carney
- Department of Genetics, Rutgers University, Piscataway, New Jersey, USA
| | - Mary Konsolaki
- Department of Genetics, Rutgers University, Piscataway, New Jersey, USA
- Federated Department of Biological Sciences, New Jersey Institute of Technology, Newark, New Jersey, USA
| | - Ronald P. Hart
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey, USA
| | - Joan W. Bennett
- Department of Plant Sciences, Rutgers University, New Brunswick, New Jersey, USA
| | - Jason R. Richardson
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, New Jersey, USA
- Center for Neurodegenerative Disease and Aging, Northeast Ohio Medical University, Rootstown, Ohio, USA
| |
Collapse
|
8
|
Chen G, Shi F, Yin W, Guo Y, Liu A, Shuai J, Sun J. Gut microbiota dysbiosis: The potential mechanisms by which alcohol disrupts gut and brain functions. Front Microbiol 2022; 13:916765. [PMID: 35966709 PMCID: PMC9372561 DOI: 10.3389/fmicb.2022.916765] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/29/2022] [Indexed: 11/24/2022] Open
Abstract
Alcohol use disorder (AUD) is a high-risk psychiatric disorder and a key cause of death and disability in individuals. In the development of AUD, there is a connection known as the microbiota-gut-brain axis, where alcohol use disrupts the gut barrier, resulting in changes in intestinal permeability as well as the gut microbiota composition, which in turn impairs brain function and worsens the patient’s mental status and gut activity. Potential mechanisms are explored by which alcohol alters gut and brain function through the effects of the gut microbiota and their metabolites on immune and inflammatory pathways. Alcohol and microbiota dysregulation regulating neurotransmitter release, including DA, 5-HT, and GABA, are also discussed. Thus, based on the above discussion, it is possible to speculate on the gut microbiota as an underlying target for the treatment of diseases associated with alcohol addiction. This review will focus more on how alcohol and gut microbiota affect the structure and function of the gut and brain, specific changes in the composition of the gut microbiota, and some measures to mitigate the changes caused by alcohol exposure. This leads to a potential intervention for alcohol addiction through fecal microbiota transplantation, which could normalize the disruption of gut microbiota after AUD.
Collapse
Affiliation(s)
- Ganggang Chen
- Department of Anatomy and Neurobiology, School of Basic Medicine, Shandong University, Jinan, China
| | - Fenglei Shi
- Department of Othopaedics, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Yin
- Department of Anatomy and Neurobiology, School of Basic Medicine, Shandong University, Jinan, China
| | - Yao Guo
- Shandong Provincial Mental Health Center, Jinan, China
| | - Anru Liu
- Department of Anatomy and Neurobiology, School of Basic Medicine, Shandong University, Jinan, China
| | - Jiacheng Shuai
- Department of Anatomy and Neurobiology, School of Basic Medicine, Shandong University, Jinan, China
| | - Jinhao Sun
- Department of Anatomy and Neurobiology, School of Basic Medicine, Shandong University, Jinan, China
- *Correspondence: Jinhao Sun,
| |
Collapse
|
9
|
Monayo SM, Liu X. The Prospective Application of Melatonin in Treating Epigenetic Dysfunctional Diseases. Front Pharmacol 2022; 13:867500. [PMID: 35668933 PMCID: PMC9163742 DOI: 10.3389/fphar.2022.867500] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/14/2022] [Indexed: 01/09/2023] Open
Abstract
In the past, different human disorders were described by scientists from the perspective of either environmental factors or just by genetically related mechanisms. The rise in epigenetic studies and its modifications, i.e., heritable alterations in gene expression without changes in DNA sequences, have now been confirmed in diseases. Modifications namely, DNA methylation, posttranslational histone modifications, and non-coding RNAs have led to a better understanding of the coaction between epigenetic alterations and human pathologies. Melatonin is a widely-produced indoleamine regulator molecule that influences numerous biological functions within many cell types. Concerning its broad spectrum of actions, melatonin should be investigated much more for its contribution to the upstream and downstream mechanistic regulation of epigenetic modifications in diseases. It is, therefore, necessary to fill the existing gaps concerning corresponding processes associated with melatonin with the physiological abnormalities brought by epigenetic modifications. This review outlines the findings on melatonin’s action on epigenetic regulation in human diseases including neurodegenerative diseases, diabetes, cancer, and cardiovascular diseases. It summarizes the ability of melatonin to act on molecules such as proteins and RNAs which affect the development and progression of diseases.
Collapse
|
10
|
Zhang H, Lu P, Tang HL, Yan HJ, Jiang W, Shi H, Chen SY, Gao MM, Zeng XD, Long YS. Valproate-Induced Epigenetic Upregulation of Hypothalamic Fto Expression Potentially Linked with Weight Gain. Cell Mol Neurobiol 2021; 41:1257-1269. [PMID: 32500354 DOI: 10.1007/s10571-020-00895-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 06/01/2020] [Indexed: 12/17/2022]
Abstract
Valproate (VPA), a widely-used antiepileptic drug, is a selective inhibitor of histone deacetylase (HDAC) that play important roles in epigenetic regulation. The patient with different diseases receiving this drug tend to exhibit weight gain and abnormal metabolic phenotypes, but the underlying mechanisms remain largely unknown. Here we show that VPA increases the Fto mRNA and protein expression in mouse hypothalamic GT1-7 cells. Interestingly, VPA promotes histone H3/H4 acetylation and the FTO expression which could be reversed by C646, an inhibitor for histone acetyltransferase. Furthermore, VPA weakens the FTO's binding and enhances the binding of transcription factor TAF1 to the Fto promoter, and C646 leads to reverse effect of the VPA, suggesting an involvement of the dynamic of histone H3/H4 acetylation in the regulation of FTO expression. In addition, the mice exhibit an increase in the food intake and body weight at the beginning of 2-week treatment with VPA. Simultaneously, in the hypothalamus of the VPA-treated mice, the FTO expression is upregulated and the H3/H4 acetylation is increased; further the FTO's binding to the Fto promoter is decreased and the TAF1's binding to the promoter is enhanced, suggesting that VPA promotes the assembly of the basal transcriptional machinery of the Fto gene. Finally, the inhibitor C646 could restore the effects of VPA on FTO expression, H3/H4 acetylation, body weight, and food intake; and loss of FTO could reverse the VPA-induced increase of body weight and food intake. Taken together, this study suggests an involvement of VPA in the epigenetic upregulation of hypothalamic FTO expression that is potentially associated with the VPA-induced weight gain.
Collapse
Affiliation(s)
- Huan Zhang
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, 250 Changang East Road, Guangzhou, 510260, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, China
| | - Ping Lu
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, 250 Changang East Road, Guangzhou, 510260, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, China
| | - Hui-Ling Tang
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, 250 Changang East Road, Guangzhou, 510260, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, China
| | - Hua-Juan Yan
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, 250 Changang East Road, Guangzhou, 510260, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, China
| | - Wei Jiang
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, 250 Changang East Road, Guangzhou, 510260, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, China
| | - Hang Shi
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, 250 Changang East Road, Guangzhou, 510260, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, China
| | - Si-Yu Chen
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, 250 Changang East Road, Guangzhou, 510260, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, China
| | - Mei-Mei Gao
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, 250 Changang East Road, Guangzhou, 510260, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, China
| | - Xiang-Da Zeng
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, 250 Changang East Road, Guangzhou, 510260, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, China
| | - Yue-Sheng Long
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, 250 Changang East Road, Guangzhou, 510260, China.
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, China.
| |
Collapse
|
11
|
Cope ZA, Kenton JA, Minassian A, Martin MV, Perry W, Bundgaard C, Arnt J, van Enkhuizen J, Geyer MA, Young JW. Chronic antipsychotic treatment exerts limited effects on the mania-like behavior of dopamine transporter knockdown mice. Behav Brain Res 2021; 405:113167. [PMID: 33577882 PMCID: PMC10729608 DOI: 10.1016/j.bbr.2021.113167] [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: 08/27/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Bipolar disorder is a life-threatening disorder linked to dopamine transporter (DAT) polymorphisms, with reduced DAT levels seen in positron emission tomography and postmortem brains. AIMS The purpose of this study was to examine the effects of approved antipsychotics on DAT dysfunction-mediated mania behavior in mice. METHODS DAT knockdown mice received either D2-family receptor antagonist risperidone or asenapine and mania-related behaviors were assessed in the clinically-relevant behavioral pattern monitor to assess spontaneous exploration. RESULTS Chronic risperidone did not reverse mania-like behavior in DAT knockdown mice. Chronic asenapine reduced mania behavior but this effect was more pronounced in wild-type littermates than in DAT knockdown mice. CONCLUSION Taken together, these findings suggest that while acute antipsychotic treatment may be beneficial in management of bipolar mania, more targeted therapeutics may be necessary for long-term treatment. Specific investigation into DAT-targeting drugs could improve future treatment of bipolar mania.
Collapse
Affiliation(s)
- Zackary A Cope
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, United States
| | - Johnny A Kenton
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, United States
| | - Arpi Minassian
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, United States; Center of Excellence for Stress and Mental Health and Research Service, VA San Diego Healthcare System, United States
| | - Maureen V Martin
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, United States
| | - William Perry
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, United States
| | - Christoffer Bundgaard
- H. Lundbeck A/S. Neuroscience Research, Ottiliavej 9, DK-2500, Copenhagen, Valby, Denmark
| | - Jørn Arnt
- Sunred Pharma Consulting, Solrød Strand, Denmark
| | - Jordy van Enkhuizen
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, United States
| | - Mark A Geyer
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, United States; Research Service, VA San Diego Healthcare System, San Diego, CA, United States
| | - Jared W Young
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, United States; Research Service, VA San Diego Healthcare System, San Diego, CA, United States.
| |
Collapse
|
12
|
Hsueh YS, Lin CY, Chiu NT, Yang YK, Chen PS, Chang HH. Changes in striatal dopamine transporters in bipolar disorder and valproate treatment. Eur Psychiatry 2021; 64:e9. [PMID: 33413711 PMCID: PMC8057387 DOI: 10.1192/j.eurpsy.2021.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Previous studies suggested that a disturbance of the dopamine system underlies the pathophysiology of bipolar disorder (BD). In addition, the therapeutic action of medications for treating BD, such as valproate (VPA), might modulate dopamine system activity, but it remains unclear. Here, we aimed to investigate the role of the striatal dopamine transporter (DAT) in BD patients and in social defeat (SD) mice treated with VPA. Methods We enrolled community-dwelling controls (N = 18) and BD patients (N = 23) who were treated with VPA in a euthymic stage. The striatal DAT availabilities were approached by TRODAT-1 single photon emission computed tomography. We also established a chronic SD mouse model and treated mice with 350 mg/kg VPA for 3 weeks. Behavioral tests were administered, and striatal DAT expression levels were determined. Results In humans, the level of striatal DAT availability was significantly higher in euthymic BD patients (1.52 ± 0.17 and 1.37 ± 0.23, p = 0.015). Moreover, the level of striatal DAT availability was also negatively correlated with the VPA concentration in BD patients (r = −0.653, p = 0.003). In SD mice, the expression of striatal DAT significantly increased (p < 0.001), and the SD effect on DAT expression was rescued by VPA treatment. Conclusions The striatal DAT might play a role in the pathophysiology of BD and in the therapeutic mechanism of VPA. The homeostasis of DAT might represent a new therapeutic strategy for BD patients.
Collapse
Affiliation(s)
- Yuan-Shuo Hsueh
- Department of Medical Science Industries, College of Health Sciences, Chang Jung Christian University, Tainan, Taiwan.,Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, Taiwan
| | - Chih-Ying Lin
- Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Nan-Tsing Chiu
- Department of Nuclear Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yen Kuang Yang
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Psychiatry, National Cheng Kung University Hospital Dou-Liou Branch, Dou-Liou, Yunlin, Taiwan
| | - Po See Chen
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Behavioral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hui Hua Chang
- Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Pharmacy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Pharmacy, National Cheng Kung University Hospital, Dou-Liou Branch, Yunlin, Taiwan
| |
Collapse
|
13
|
Kong FC, Ma CL, Zhong MK. Epigenetic Effects Mediated by Antiepileptic Drugs and their Potential Application. Curr Neuropharmacol 2020; 18:153-166. [PMID: 31660836 PMCID: PMC7324883 DOI: 10.2174/1570159x17666191010094849] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 08/01/2019] [Accepted: 10/03/2019] [Indexed: 12/20/2022] Open
Abstract
An epigenetic effect mainly refers to a heritable modulation in gene expression in the short term but does not involve alterations in the DNA itself. Epigenetic molecular mechanisms include DNA methylation, histone modification, and untranslated RNA regulation. Antiepileptic drugs have drawn attention to biological and translational medicine because their impact on epigenetic mechanisms will lead to the identification of novel biomarkers and possible therapeutic strategies for the prevention and treatment of various diseases ranging from neuropsychological disorders to cancers and other chronic conditions. However, these transcriptional and posttranscriptional alterations can also result in adverse reactions and toxicity in vitro and in vivo. Hence, in this review, we focus on recent findings showing epigenetic processes mediated by antiepileptic drugs to elucidate their application in medical experiments and shed light on epigenetic research for medicinal purposes.
Collapse
Affiliation(s)
- Fan-Cheng Kong
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Chun-Lai Ma
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Ming-Kang Zhong
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
14
|
Regulation of Social Stress and Neural Degeneration by Activity-Regulated Genes and Epigenetic Mechanisms in Dopaminergic Neurons. Mol Neurobiol 2020; 57:4500-4510. [PMID: 32748368 PMCID: PMC7515954 DOI: 10.1007/s12035-020-02037-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 07/22/2020] [Indexed: 02/08/2023]
Abstract
Transcriptional and epigenetic regulation of both dopaminergic neurons and their accompanying glial cells is of great interest in the search for therapies for neurodegenerative disorders such as Parkinson’s disease (PD). In this review, we collate transcriptional and epigenetic changes identified in adult Drosophila melanogaster dopaminergic neurons in response to either prolonged social deprivation or social enrichment, and compare them with changes identified in mammalian dopaminergic neurons during normal development, stress, injury, and neurodegeneration. Surprisingly, a small set of activity-regulated genes (ARG) encoding transcription factors, and a specific pattern of epigenetic marks on gene promoters, are conserved in dopaminergic neurons over the long evolutionary period between mammals and insects. In addition to their classical function as immediate early genes to mark acute neuronal activity, these ARG transcription factors are repurposed in both insects and mammals to respond to chronic perturbations such as social enrichment, social stress, nerve injury, and neurodegeneration. We suggest that these ARG transcription factors and epigenetic marks may represent important targets for future therapeutic intervention strategies in various neurodegenerative disorders including PD.
Collapse
|
15
|
The early overgrowth theory of autism spectrum disorder: Insight into convergent mechanisms from valproic acid exposure and translational models. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020. [PMID: 32711813 DOI: 10.1016/bs.pmbts.2020.04.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
The development of new approaches for the clinical management of autism spectrum disorder (ASD) can only be realized through a better understanding of the neurobiological changes associated with ASD. One strategy for gaining deeper insight into the neurobiological mechanisms associated with ASD is to identify converging pathogenic processes associated with human idiopathic clinicopathology that are conserved in translational models of ASD. In this chapter, we first present the early overgrowth theory of ASD. Second, we introduce valproic acid (VPA), one of the most robust and well-known environmental risk factors associated with ASD, and we summarize the rapidly growing body of animal research literature using VPA as an ASD translational model. Lastly, we will detail the mechanisms of action of VPA and its impact on functional neural systems, as well as discuss future research directions that could have a lasting impact on the field.
Collapse
|
16
|
Magno LAV, Tenza-Ferrer H, Collodetti M, Nicolau EDS, Khlghatyan J, Del'Guidice T, Romano-Silva MA, Beaulieu JM. Contribution of neuronal calcium sensor 1 (Ncs-1) to anxiolytic-like and social behavior mediated by valproate and Gsk3 inhibition. Sci Rep 2020; 10:4566. [PMID: 32165725 PMCID: PMC7067888 DOI: 10.1038/s41598-020-61248-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 02/24/2020] [Indexed: 12/20/2022] Open
Abstract
Peripheral biomarker and post-mortem brains studies have shown alterations of neuronal calcium sensor 1 (Ncs-1) expression in people with bipolar disorder or schizophrenia. However, its engagement by psychiatric medications and potential contribution to behavioral regulation remains elusive. We investigated the effect on Ncs-1 expression of valproic acid (VPA), a mood stabilizer used for the management of bipolar disorder. Treatment with VPA induced Ncs-1 gene expression in cell line while chronic administration of this drug to mice increased both Ncs-1 protein and mRNA levels in the mouse frontal cortex. Inhibition of histone deacetylases (HDACs), a known biochemical effect of VPA, did not alter the expression of Ncs-1. In contrast, pharmacological inhibition or genetic downregulation of glycogen synthase kinase 3β (Gsk3β) increased Ncs-1 expression, whereas overexpression of a constitutively active Gsk3β had the opposite effect. Moreover, adeno-associated virus-mediated Ncs-1 overexpression in mouse frontal cortex caused responses similar to those elicited by VPA or lithium in tests evaluating social and mood-related behaviors. These findings indicate that VPA increases frontal cortex Ncs-1 gene expression as a result of Gsk3 inhibition. Furthermore, behavioral changes induced by Ncs-1 overexpression support a contribution of this mechanism in the regulation of behavior by VPA and potentially other psychoactive medications inhibiting Gsk3 activity.
Collapse
Affiliation(s)
- Luiz Alexandre Viana Magno
- Centro de Tecnologia em Medicina Molecular, Belo Horizonte, Brazil.,Departamento de Saúde Mental, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, CEP, 30130-100, Brazil.,Department of Psychiatry and Neuroscience, Laval University, Québec, Canada
| | - Helia Tenza-Ferrer
- Centro de Tecnologia em Medicina Molecular, Belo Horizonte, Brazil.,Departamento de Saúde Mental, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, CEP, 30130-100, Brazil
| | - Mélcar Collodetti
- Centro de Tecnologia em Medicina Molecular, Belo Horizonte, Brazil.,Departamento de Saúde Mental, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, CEP, 30130-100, Brazil
| | - Eduardo de Souza Nicolau
- Centro de Tecnologia em Medicina Molecular, Belo Horizonte, Brazil.,Departamento de Saúde Mental, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, CEP, 30130-100, Brazil
| | - Jivan Khlghatyan
- Department of Psychiatry and Neuroscience, Laval University, Québec, Canada.,Department of Pharmacology & Toxicology, University of Toronto, Toronto, Canada
| | - Thomas Del'Guidice
- Department of Psychiatry and Neuroscience, Laval University, Québec, Canada.,Feldan Therapeutics, Québec City, Canada
| | - Marco Aurélio Romano-Silva
- Centro de Tecnologia em Medicina Molecular, Belo Horizonte, Brazil. .,Departamento de Saúde Mental, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, CEP, 30130-100, Brazil.
| | - Jean Martin Beaulieu
- Department of Psychiatry and Neuroscience, Laval University, Québec, Canada. .,Department of Pharmacology & Toxicology, University of Toronto, Toronto, Canada.
| |
Collapse
|
17
|
Carbone C, Brancato A, Adinolfi A, Lo Russo SLM, Alleva E, Cannizzaro C, Adriani W. Motor Transitions' Peculiarity of Heterozygous DAT Rats When Offspring of an Unconventional KOxWT Mating. Neuroscience 2020; 433:108-120. [PMID: 32171819 DOI: 10.1016/j.neuroscience.2020.03.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 12/23/2022]
Abstract
Causal factors of psychiatric diseases are unclear, due to gene × environment interactions. Evaluation of consequences, after a dopamine-transporter (DAT) gene knock-out (DAT-KO), has enhanced our understanding into the pathological dynamics of several brain disorders, such as Attention-Deficit/Hyperactivity and Bipolar-Affective disorders. Recently, our attention has shifted to DAT hypo-functional (heterozygous, HET) rodents: HET dams display less maternal care and HET females display marked hypo-locomotion if cared by HET dams (Mariano et al., 2019). We assessed phenotypes of male DAT-heterozygous rats as a function of their parents: we compared "maternal" origin (MAT-HET, obtained by breeding KO-male rats with WT-female dams) to "mixed" origin (MIX-HET, obtained by classical breeding, both heterozygous parents) of the allele. MAT-HET subjects had significantly longer rhythms of daily locomotor activity than MIX-HET and WT-control subjects. Furthermore, acute methylphenidate (MPH: 0, 1, 2 mg/kg) revealed elevated threshold for locomotor stimulation in MAT-HETs, with no response to the lower dose. Finally, by Porsolt-Test, MAT-HETs showed enhanced escape-seeking (diving) with more transitions towards behavioral despair (floating). When comparing both MAT- and MIX-HET to WT-control rats, decreased levels of DAT and HDAC4 were evident in the ventral-striatum; moreover, with respect to MIX-HET subjects, MAT-HET ones displayed increased DAT density in dorsal-striatum. MAT-HET rats displayed region-specific changes in DAT expression, compared to "classical" MIX-HET subjects: greater DAT availability may elevate threshold for dopamine action. Further behavioral and epigenetic characterizations of MAT-HETs, together with deeper characterization of maternal roles, could help to explore parent-of-origin mechanisms for such a peculiar phenotype.
Collapse
Affiliation(s)
- Cristiana Carbone
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Anna Brancato
- Dept Sciences of Health Promotion & Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Italy
| | - Annalisa Adinolfi
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | | | - Enrico Alleva
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Carla Cannizzaro
- Dept Sciences of Health Promotion & Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Italy
| | - Walter Adriani
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
18
|
Green AL, Eid A, Zhan L, Zarbl H, Guo GL, Richardson JR. Epigenetic Regulation of the Ontogenic Expression of the Dopamine Transporter. Front Genet 2019; 10:1099. [PMID: 31749842 PMCID: PMC6844290 DOI: 10.3389/fgene.2019.01099] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 10/11/2019] [Indexed: 01/19/2023] Open
Abstract
The dopamine transporter (DAT) is a plasma membrane transport protein responsible for regulating the duration and intensity of dopaminergic signaling. Altered expression of DAT is linked to neurodevelopmental disorders, including attention deficit hyperactivity disorder and autism spectrum disorder, and is shown to contribute to the response of psychotropic drugs and neurotoxicants. Although the postnatal levels of DAT have been characterized, there are few data regarding the mechanisms that regulate postnatal DAT expression. Here, we examine the ontogeny of DAT mRNA from postnatal days 0 to 182 in the rat brain and define a role for epigenetic mechanisms regulating DAT expression. DAT mRNA and protein significantly increased between PND 0 and 6 months in rat midbrain and striatum, respectively. The epigenetic modifiers Dnmt1, Dnmt3a, Dnmt3b, and Hdac2 demonstrated age associated decreases in mRNA expression whereas Hdac5 and Hdac8 showed increased mRNA expression with age. Chromatin immunoprecipitation studies revealed increased protein enrichment of acetylated histone 3 at lysines 9 and 14 and the dopaminergic transcription factors Nurr1 and Pitx3 within the DAT promoter in an age-related manner. Together these studies provide evidence for the role of epigenetic modifications in the regulation of DAT during development. The identification of these mechanisms may contribute to potential therapeutic interventions aimed at neurodevelopmental disorders of the dopaminergic system.
Collapse
Affiliation(s)
- Ashley L. Green
- Environmental and Occupational Health Sciences Institute and Department of Environmental and Occupational Medicine, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - Aseel Eid
- Department of Environmental Health Sciences, Robert Stempel School of Public Health and Social Work, Florida International University, Miami, FL, United States
| | - Le Zhan
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, United States
| | - Helmut Zarbl
- Environmental and Occupational Health Sciences Institute and Department of Environmental and Occupational Medicine, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - Grace L. Guo
- Environmental and Occupational Health Sciences Institute and Department of Environmental and Occupational Medicine, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, United States,Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, United States
| | - Jason R. Richardson
- Environmental and Occupational Health Sciences Institute and Department of Environmental and Occupational Medicine, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, United States,Department of Environmental Health Sciences, Robert Stempel School of Public Health and Social Work, Florida International University, Miami, FL, United States,*Correspondence: Jason R. Richardson,
| |
Collapse
|
19
|
Gottschalk CG, Roy A, Jana M, Kundu M, Pahan K. Activation of Peroxisome Proliferator-Activated Receptor-α Increases the Expression of Nuclear Receptor Related 1 Protein (Nurr1) in Dopaminergic Neurons. Mol Neurobiol 2019; 56:7872-7887. [PMID: 31127527 DOI: 10.1007/s12035-019-01649-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 05/13/2019] [Indexed: 12/31/2022]
Abstract
Nuclear receptor related 1 protein (Nurr1) is an important transcription factor required for differentiation and maintenance of midbrain dopaminergic (DA) neurons. Since decrease in Nurr1 function either due to diminished expression or rare mutation is associated with Parkinson's disease (PD), upregulation of Nurr1 may be beneficial for PD. However, such mechanisms are poorly understood. This study underlines the importance of peroxisome proliferator-activated receptor (PPAR)α in controlling the transcription of Nurr1. Our mRNA analyses followed by different immunoassays clearly indicated that PPARα agonist gemfibrozil strongly upregulated the expression of Nurr1 in wild-type, but not PPARα-/-, DA neurons. Moreover, identification of conserved PPRE in the promoter of Nurr1 gene followed by chromatin immunoprecipitation analysis, PPRE luciferase assay, and manipulation of Nurr1 gene by viral transduction of different PPARα plasmids confirmed that PPARα was indeed involved in the expression of Nurr1. Finally, oral administration of gemfibrozil increased Nurr1 expression in vivo in nigra of wild-type, but not PPARα-/-, mice identifying PPARα as a novel regulator of Nurr1 expression and associated protection of DA neurons.
Collapse
Affiliation(s)
- Carl G Gottschalk
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St, Suite Cohn 310, Chicago, IL, 60612, USA
| | - Avik Roy
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St, Suite Cohn 310, Chicago, IL, 60612, USA.,Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| | - Malabendu Jana
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St, Suite Cohn 310, Chicago, IL, 60612, USA.,Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| | - Madhuchhanda Kundu
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St, Suite Cohn 310, Chicago, IL, 60612, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St, Suite Cohn 310, Chicago, IL, 60612, USA. .,Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA.
| |
Collapse
|
20
|
Montagud-Romero S, Blanco-Gandía MC, Reguilón MD, Ferrer-Pérez C, Ballestín R, Miñarro J, Rodríguez-Arias M. Social defeat stress: Mechanisms underlying the increase in rewarding effects of drugs of abuse. Eur J Neurosci 2018; 48:2948-2970. [PMID: 30144331 DOI: 10.1111/ejn.14127] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/11/2018] [Accepted: 08/14/2018] [Indexed: 12/27/2022]
Abstract
Social interaction is known to be the main source of stress in human beings, which explains the translational importance of this research in animals. Evidence reported over the last decade has revealed that, when exposed to social defeat experiences (brief episodes of social confrontations during adolescence and adulthood), the rodent brain undergoes remodeling and functional modifications, which in turn lead to an increase in the rewarding and reinstating effects of different drugs of abuse. The mechanisms by which social stress cause changes in the brain and behavior are unknown, and so the objective of this review is to contemplate how social defeat stress induces long-lasting consequences that modify the reward system. First of all, we will describe the most characteristic results of the short- and long-term consequences of social defeat stress on the rewarding effects of drugs of abuse such as psychostimulants and alcohol. Secondly, and throughout the review, we will carefully assess the neurobiological mechanisms underlying these effects, including changes in the dopaminergic system, corticotrophin releasing factor signaling, epigenetic modifications and the neuroinflammatory response. To conclude, we will consider the advantages and disadvantages and the translational value of the social defeat stress model, and will discuss challenges and future directions.
Collapse
Affiliation(s)
- Sandra Montagud-Romero
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain
| | | | - Marina D Reguilón
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain
| | - Carmen Ferrer-Pérez
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain
| | - Raul Ballestín
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain
| | - Jose Miñarro
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain
| | - Marta Rodríguez-Arias
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain
| |
Collapse
|