1
|
Samiec M, Trzcińska M. From genome to epigenome: Who is a predominant player in the molecular hallmarks determining epigenetic mechanisms underlying ontogenesis? Reprod Biol 2024; 24:100965. [PMID: 39467448 DOI: 10.1016/j.repbio.2024.100965] [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: 07/26/2024] [Revised: 09/12/2024] [Accepted: 10/17/2024] [Indexed: 10/30/2024]
Abstract
Genetic factors are one of the basic determinants affecting ontogenesis in mammals. Nevertheless, on the one hand, epigenetic factors have been found to exert the preponderant and insightful impact on the intracellular mechanistic networks related to not only initiation and suppression, but also up- and downregulation of gene expression in all the phases of ontogenetic development in a variety of mammalian species. On the other hand, impairments in the epigenetic mechanisms underlying reprogramming of transcriptional activity of genes (termed epimutations) not only give rise to a broad spectrum of acute and chronic developmental abnormalities in mammalian embryos, foetuses and neonates, but also contribute to premature/expedited senescence or neoplastic transformation of cells and even neurodegenerative and mental disorders. The current article is focused on the unveiling the present knowledge aimed at the identification, classification and characterization of epigenetic agents as well as multifaceted interpretation of current and coming trends targeted at recognizing the epigenetic background of proper ontogenesis in mammals. Moreover, the next objective of this paper is to unravel the mechanistic insights into a wide array of disturbances leading to molecular imbalance taking place during epigenetic reprogramming of genomic DNA. The above-indicated imbalance seems to play a predominant role in the initiation and progression of anatomo-, histo-, and physiopathological processes throughout ontogenetic development. Conclusively, different modalities of epigenetically assisted therapeutic procedures that have been exemplified in the current article, might be the powerful and promiseful tools reliable and feasible in the medical treatments of several diseases triggered by dysfunctions in the epigenetic landscapes, e.g., myelodysplastic syndromes or epilepsy.
Collapse
Affiliation(s)
- Marcin Samiec
- Department of Reproductive Biotechnology and Cryoconservation, National Research Institute of Animal Production, Krakowska 1 Street, 32-083 Balice near Kraków, Poland.
| | - Monika Trzcińska
- Department of Reproductive Biotechnology and Cryoconservation, National Research Institute of Animal Production, Krakowska 1 Street, 32-083 Balice near Kraków, Poland.
| |
Collapse
|
2
|
Mohammadzadeh R, Fathi M, Pourseif MM, Omidi Y, Farhang S, Barzegar Jalali M, Valizadeh H, Nakhlband A, Adibkia K. Curcumin and nano-curcumin applications in psychiatric disorders. Phytother Res 2024. [PMID: 38965868 DOI: 10.1002/ptr.8265] [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: 11/08/2023] [Revised: 05/13/2024] [Accepted: 05/25/2024] [Indexed: 07/06/2024]
Abstract
Psychiatric disorders cause long-lasting disabilities across different age groups. While various medications are available for mental disorders, some patients do not fully benefit from them or experience treatment resistance. The pathogenesis of psychiatric disorders involves multiple mechanisms, including an increase in the inflammatory response. Targeting inflammatory mechanisms has shown promise as a therapeutic approach for these disorders. Curcumin, known for its anti-inflammatory properties and potential neuroprotective effects, has been the subject of studies investigating its potential as a treatment option for psychiatric disorders. This review comprehensively examines the potential therapeutic role of curcumin and its nanoformulations in psychiatric conditions, including major depressive disorder (MDD), bipolar disorder, schizophrenia, and anxiety disorders. There is lack of robust clinical trials across all the studied psychiatric disorders, particularly bipolar disorder and schizophrenia. More studies have focused on MDD. Studies on depression indicate that curcumin may be effective as an antidepressant agent, either alone or as an adjunct therapy. However, inconsistencies exist among study findings, highlighting the need for further research with improved blinding, optimized dosages, and treatment durations. Limited evidence supports the use of curcumin for bipolar disorder, making its therapeutic application challenging. Well-designed clinical trials are warranted to explore its potential therapeutic benefits. Exploring various formulations and delivery strategies, such as utilizing liposomes and nanoparticles, presents intriguing avenues for future research. More extensive clinical trials are needed to assess the efficacy of curcumin as a standalone or adjunctive treatment for psychiatric disorders, focusing on optimal dosages, formulations, and treatment durations.
Collapse
Affiliation(s)
- R Mohammadzadeh
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - M Fathi
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - M M Pourseif
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Y Omidi
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida, USA
| | - S Farhang
- Research Center of Psychiatry and Behavioral Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - M Barzegar Jalali
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - H Valizadeh
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - A Nakhlband
- Research Center of Psychiatry and Behavioral Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - K Adibkia
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
3
|
DelaCuesta-Barrutia J, Martínez-Peula O, Rivero G, Santas-Martín JA, Munarriz-Cuezva E, Brocos-Mosquera I, Miranda-Azpiazu P, Diez-Alarcia R, Morentin B, Honer WG, Callado LF, Erdozain AM, Ramos-Miguel A. Effect of antipsychotic drugs on group II metabotropic glutamate receptor expression and epigenetic control in postmortem brains of schizophrenia subjects. Transl Psychiatry 2024; 14:113. [PMID: 38396013 PMCID: PMC10891050 DOI: 10.1038/s41398-024-02832-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Antipsychotic-induced low availability of group II metabotropic glutamate receptors (including mGlu2R and mGlu3R) in brains of schizophrenia patients may explain the limited efficacy of mGlu2/3R ligands in clinical trials. Studies evaluating mGlu2/3R levels in well-designed, large postmortem brain cohorts are needed to address this issue. Postmortem samples from the dorsolateral prefrontal cortex of 96 schizophrenia subjects and matched controls were collected. Toxicological analyses identified cases who were (AP+) or were not (AP-) receiving antipsychotic treatment near the time of death. Protein and mRNA levels of mGlu2R and mGlu3R, as well as GRM2 and GRM3 promoter-attached histone posttranslational modifications, were quantified. Experimental animal models were used to compare with data obtained in human tissues. Compared to matched controls, schizophrenia cortical samples had lower mGlu2R protein amounts, regardless of antipsychotic medication. Downregulation of mGlu3R was observed in AP- schizophrenia subjects only. Greater predicted occupancy values of dopamine D2 and serotonin 5HT2A receptors correlated with higher density of mGlu3R, but not mGlu2R. Clozapine treatment and maternal immune activation in rodents mimicked the mGlu2R, but not mGlu3R regulation observed in schizophrenia brains. mGlu2R and mGlu3R mRNA levels, and the epigenetic control mechanisms did not parallel the alterations at the protein level, and in some groups correlated inversely. Insufficient cortical availability of mGlu2R and mGlu3R may be associated with schizophrenia. Antipsychotic treatment may normalize mGlu3R, but not mGlu2R protein levels. A model in which epigenetic feedback mechanisms controlling mGlu3R expression are activated to counterbalance mGluR loss of function is described.
Collapse
Affiliation(s)
| | | | - Guadalupe Rivero
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Leioa, Spain
| | - Jon A Santas-Martín
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
| | - Eva Munarriz-Cuezva
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Leioa, Spain
| | - Iria Brocos-Mosquera
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
| | | | - Rebeca Diez-Alarcia
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Leioa, Spain
| | - Benito Morentin
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Basque Institute of Legal Medicine, Bilbao, Spain
| | - William G Honer
- Department Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Luis F Callado
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Leioa, Spain
| | - Amaia M Erdozain
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Leioa, Spain
| | - Alfredo Ramos-Miguel
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain.
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain.
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Leioa, Spain.
| |
Collapse
|
4
|
Martínez-Peula O, Morentin B, Callado LF, Meana JJ, Rivero G, Ramos-Miguel A. Permissive epigenetic regulatory mechanisms at the histone level are enhanced in postmortem dorsolateral prefrontal cortex of individuals with schizophrenia. J Psychiatry Neurosci 2024; 49:E35-E44. [PMID: 38302137 PMCID: PMC10843339 DOI: 10.1503/jpn.230054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 09/28/2023] [Accepted: 11/03/2023] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Susceptibility to schizophrenia is determined by interactions between genes and environment, possibly via epigenetic mechanisms. Schizophrenia has been associated with a restrictive epigenome, and histone deacetylase (HDAC) inhibitors have been postulated as coadjuvant agents to potentiate the efficacy of current antipsychotic drugs. We aimed to evaluate global histone posttranslational modifications (HPTMs) and HDAC expression and activity in the dorsolateral prefrontal cortex (DLPFC) of individuals with schizophrenia. METHODS We used postmortem DLPFC samples of individuals with schizophrenia and controls matched for sex, age, and postmortem interval. Schizophrenia samples were classified into antipsychotic-treated or antipsychotic-free subgroups according to blood toxicology. Expression of HPTMs and HDAC was quantified by Western blot. HDAC activity was measured with a fluorometric assay. RESULTS H3K9ac, H3K27ac, and H3K4me3 were globally enhanced in the DLPFC of individuals with schizophrenia (+24%-42%, p < 0.05). HDAC activity (-17%, p < 0.01) and HDAC4 protein expression (-20%, p < 0.05) were downregulated in individuals with schizophrenia. Analyses of antipsychotic-free and antipsychotic-treated subgroups revealed enhanced H3K4me3 and H3K27ac (+24%-49%, p < 0.05) and reduced HDAC activity in the antipsychotic-treated, but not in the antipsychotic-free subgroup. LIMITATIONS Special care was taken to control the effect of confounding factors: age, sex, postmortem interval, and storage time. However, replication studies in bigger cohorts might strengthen the association between permissive HPTMs and schizophrenia. CONCLUSION We found global HPTM alterations consistent with an aberrantly permissive epigenome in schizophrenia. Further studies to elucidate the significance of enhanced permissive HPTMs in schizophrenia and its association with the mechanism of action of antipsychotic drugs are encouraged.
Collapse
Affiliation(s)
- Oihane Martínez-Peula
- From the Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain (Martínez-Peula, Callado, Meana, Rivero, Ramos-Miguel); Section of Forensic Pathology, Basque Institute of Legal Medicine, Bilbao, Spain (Morentin); the Biobizkaia Health Research Institute, Barakaldo, Spain ( Morentin, Callado, Meana, Rivero, Ramos-Miguel); and the Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, ISCIII, Spain (Callado, Meana, Rivero, Ramos-Miguel)
| | - Benito Morentin
- From the Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain (Martínez-Peula, Callado, Meana, Rivero, Ramos-Miguel); Section of Forensic Pathology, Basque Institute of Legal Medicine, Bilbao, Spain (Morentin); the Biobizkaia Health Research Institute, Barakaldo, Spain ( Morentin, Callado, Meana, Rivero, Ramos-Miguel); and the Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, ISCIII, Spain (Callado, Meana, Rivero, Ramos-Miguel)
| | - Luis F Callado
- From the Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain (Martínez-Peula, Callado, Meana, Rivero, Ramos-Miguel); Section of Forensic Pathology, Basque Institute of Legal Medicine, Bilbao, Spain (Morentin); the Biobizkaia Health Research Institute, Barakaldo, Spain ( Morentin, Callado, Meana, Rivero, Ramos-Miguel); and the Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, ISCIII, Spain (Callado, Meana, Rivero, Ramos-Miguel)
| | - J Javier Meana
- From the Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain (Martínez-Peula, Callado, Meana, Rivero, Ramos-Miguel); Section of Forensic Pathology, Basque Institute of Legal Medicine, Bilbao, Spain (Morentin); the Biobizkaia Health Research Institute, Barakaldo, Spain ( Morentin, Callado, Meana, Rivero, Ramos-Miguel); and the Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, ISCIII, Spain (Callado, Meana, Rivero, Ramos-Miguel)
| | - Guadalupe Rivero
- From the Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain (Martínez-Peula, Callado, Meana, Rivero, Ramos-Miguel); Section of Forensic Pathology, Basque Institute of Legal Medicine, Bilbao, Spain (Morentin); the Biobizkaia Health Research Institute, Barakaldo, Spain ( Morentin, Callado, Meana, Rivero, Ramos-Miguel); and the Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, ISCIII, Spain (Callado, Meana, Rivero, Ramos-Miguel)
| | - Alfredo Ramos-Miguel
- From the Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain (Martínez-Peula, Callado, Meana, Rivero, Ramos-Miguel); Section of Forensic Pathology, Basque Institute of Legal Medicine, Bilbao, Spain (Morentin); the Biobizkaia Health Research Institute, Barakaldo, Spain ( Morentin, Callado, Meana, Rivero, Ramos-Miguel); and the Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, ISCIII, Spain (Callado, Meana, Rivero, Ramos-Miguel)
| |
Collapse
|
5
|
Pordel S, Khorrami M, Saadatpour F, Rezaee D, Cho WC, Jahani S, Aghaei-Zarch SM, Hashemi E, Najafi S. The role of microRNA-185 in the pathogenesis of human diseases: A focus on cancer. Pathol Res Pract 2023; 249:154729. [PMID: 37639952 DOI: 10.1016/j.prp.2023.154729] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/29/2023] [Indexed: 08/31/2023]
Abstract
MicroRNAs (miRNAs) are a widely-studied class of non-coding RNAs characterized by their short length (18-25 nucleotides). The precise functions of miRNAs are not well-elucidated; however, an increasing number of studies suggest their involvement in various physiologic processes and deregulation in pathologic conditions. miRNA-185 (miR-185) is among the mostly-studied miRNAs in human diseases, which is found to play putative roles in conditions like metabolic disorders, asthma, frailty, schizophrenia, and hepatitis. Notably, many cancer studies report the downregulation of miR-185 in cell lines, tumor tissues, and plasma specimens of patients, while it demonstrates a suppressing role on the malignant properties of cancer cells in vitro and in vivo. Accordingly, miR-185 can be considered a tumor suppressor miRNA in human malignancies, while a few studies also report inconsistent findings. Being suggested as a prognostic/diagnostic biomarker, mi-185 is also found to offer clinical potentials, particularly for early diagnosis and prediction of the prognosis of cancer patients. In this review, we have outlined the studies that have evaluated the functions and clinical significance of miR-185 in different human diseases with a particular focus on cancer.
Collapse
Affiliation(s)
- Safoora Pordel
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Immunology and Allergy, The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Motahare Khorrami
- Immunology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Saadatpour
- Pharmaceutical Biotechnology Lab, Department of Microbiology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran, Iran
| | - Delsuz Rezaee
- School of Allied Medical Sciences, Ilam University of Medical Sciences, Ilam, Iran
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, 30 Gascoigne Road, Hong Kong, China
| | | | - Seyed Mohsen Aghaei-Zarch
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Elham Hashemi
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Saha S, González-Maeso J. The crosstalk between 5-HT 2AR and mGluR2 in schizophrenia. Neuropharmacology 2023; 230:109489. [PMID: 36889432 PMCID: PMC10103009 DOI: 10.1016/j.neuropharm.2023.109489] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/26/2023] [Accepted: 03/05/2023] [Indexed: 03/08/2023]
Abstract
Schizophrenia is a severe brain disorder that usually produces a lifetime of disability. First generation or typical antipsychotics such as haloperidol and second generation or atypical antipsychotics such as clozapine and risperidone remain the current standard for schizophrenia treatment. In some patients with schizophrenia, antipsychotics produce complete remission of positive symptoms, such as hallucinations and delusions. However, antipsychotic drugs are ineffective against cognitive deficits and indeed treated schizophrenia patients have small improvements or even deterioration in several cognitive domains. This underlines the need for novel and more efficient therapeutic targets for schizophrenia treatment. Serotonin and glutamate have been identified as key parts of two neurotransmitter systems involved in fundamental brain processes. Serotonin (or 5-hydroxytryptamine) 5-HT2A receptor (5-HT2AR) and metabotropic glutamate 2 receptor (mGluR2) are G protein-coupled receptors (GPCRs) that interact at epigenetic and functional levels. These two receptors can form GPCR heteromeric complexes through which their pharmacology, function and trafficking becomes affected. Here we review past and current research on the 5-HT2AR-mGluR2 heterocomplex and its potential implication in schizophrenia and antipsychotic drug action. This article is part of the Special Issue on "The receptor-receptor interaction as a new target for therapy".
Collapse
Affiliation(s)
- Somdatta Saha
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | - Javier González-Maeso
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA.
| |
Collapse
|
7
|
Tovo PA, Marozio L, Abbona G, Calvi C, Frezet F, Gambarino S, Dini M, Benedetto C, Galliano I, Bergallo M. Pregnancy Is Associated with Impaired Transcription of Human Endogenous Retroviruses and of TRIM28 and SETDB1, Particularly in Mothers Affected by Multiple Sclerosis. Viruses 2023; 15:v15030710. [PMID: 36992419 PMCID: PMC10051116 DOI: 10.3390/v15030710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/01/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Accumulating evidence highlights the pathogenetic role of human endogenous retroviruses (HERVs) in eliciting and maintaining multiple sclerosis (MS). Epigenetic mechanisms, such as those regulated by TRIM 28 and SETDB1, are implicated in HERV activation and in neuroinflammatory disorders, including MS. Pregnancy markedly improves the course of MS, but no study explored the expressions of HERVs and of TRIM28 and SETDB1 during gestation. Using a polymerase chain reaction real-time Taqman amplification assay, we assessed and compared the transcriptional levels of pol genes of HERV-H, HERV-K, HERV-W; of env genes of Syncytin (SYN)1, SYN2, and multiple sclerosis associated retrovirus (MSRV); and of TRIM28 and SETDB1 in peripheral blood and placenta from 20 mothers affected by MS; from 27 healthy mothers, in cord blood from their neonates; and in blood from healthy women of child-bearing age. The HERV mRNA levels were significantly lower in pregnant than in nonpregnant women. Expressions of all HERVs were downregulated in the chorion and in the decidua basalis of MS mothers compared to healthy mothers. The former also showed lower mRNA levels of HERV-K-pol and of SYN1, SYN2, and MSRV in peripheral blood. Significantly lower expressions of TRIM28 and SETDB1 also emerged in pregnant vs. nonpregnant women and in blood, chorion, and decidua of mothers with MS vs. healthy mothers. In contrast, HERV and TRIM28/SETDB1 expressions were comparable between their neonates. These results show that gestation is characterized by impaired expressions of HERVs and TRIM28/SETDB1, particularly in mothers with MS. Given the beneficial effects of pregnancy on MS and the wealth of data suggesting the putative contribution of HERVs and epigenetic processes in the pathogenesis of the disease, our findings may further support innovative therapeutic interventions to block HERV activation and to control aberrant epigenetic pathways in MS-affected patients.
Collapse
Affiliation(s)
- Pier-Angelo Tovo
- Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy
- Correspondence: (P.-A.T.); (M.B.)
| | - Luca Marozio
- Department of Surgical Sciences, Obstetrics and Gynecology 1, University of Turin, 10126 Turin, Italy
| | - Giancarlo Abbona
- Pathology Unit, Department Laboratory Medicine, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Cristina Calvi
- Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy
- Pediatric Laboratory, Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy
| | - Federica Frezet
- Department of Surgical Sciences, Obstetrics and Gynecology 1, University of Turin, 10126 Turin, Italy
| | - Stefano Gambarino
- Pediatric Laboratory, Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy
| | - Maddalena Dini
- Pediatric Laboratory, Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy
| | - Chiara Benedetto
- Department of Surgical Sciences, Obstetrics and Gynecology 1, University of Turin, 10126 Turin, Italy
| | - Ilaria Galliano
- Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy
- Pediatric Laboratory, Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy
| | - Massimiliano Bergallo
- Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy
- Pediatric Laboratory, Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy
- Correspondence: (P.-A.T.); (M.B.)
| |
Collapse
|
8
|
Lavratti C, Iraci L, Ferreira A, Dorneles G, Pochmann D, da Rosa Boeira M, Peres A, Elsner V. Time course of epigenetic modulation in response to concurrent exercise training in patients with schizophrenia. COMPARATIVE EXERCISE PHYSIOLOGY 2022. [DOI: 10.3920/cep210013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
This study aimed to investigate the short and long-term effects of concurrent exercise training on anthropometric variables, HDCA2 activity and cortisol levels of individuals with schizophrenia (SZ). Therefore, 10 patients were submitted to the program (endurance and strength exercises in the same session, 60 min of duration, three times a week) and blood samples were collected before, 30 days and 180 days after the intervention started. Exercise training reduced the body mass index and body mass after 180 days of the intervention. A significant decrease on HDAC2 activity was found 180 days after intervention compared to before the intervention. The cortisol levels remained unchanged in any evaluated time-points. The concurrent exercise training was able to modulate HDAC2 activity in mononuclear cells and improve anthropometric variables in a time-dependent manner in patients with SZ.
Collapse
Affiliation(s)
- C. Lavratti
- Programa de Pós Graduação em Ciências da Reabilitação, Universidade Federal de Ciências da Saúde de Porto Alegre, Rua Coronel Joaquim Pedro Salgado 80, Rio Branco, CEP 90420-060 Porto Alegre, RS, Brazil
| | - L. Iraci
- Curso de Fisioterapia do Centro Universitário Metodista-IPA, Porto Alegre, RS, Brazil
| | - A. Ferreira
- Curso de Fisioterapia do Centro Universitário Metodista-IPA, Porto Alegre, RS, Brazil
| | - G. Dorneles
- Cellular and Molecular Immunology Lab., Department of Health Basic Sciences. Federal University of Health Sciences of Porto Alegre, Porto Alegre, RS, Brazil
| | - D. Pochmann
- Programa de Pós Graduação em Biociências e Reabilitação doCentro Universitário Metodista-IPA, Porto Alegre, RS, Brazil
| | - M.C. da Rosa Boeira
- Programa de Pós Graduação em Ciências da Reabilitação, Universidade Federal de Ciências da Saúde de Porto Alegre, Rua Coronel Joaquim Pedro Salgado 80, Rio Branco, CEP 90420-060 Porto Alegre, RS, Brazil
| | - A. Peres
- Cellular and Molecular Immunology Lab., Department of Health Basic Sciences. Federal University of Health Sciences of Porto Alegre, Porto Alegre, RS, Brazil
| | - V.R. Elsner
- Cellular and Molecular Immunology Lab., Department of Health Basic Sciences. Federal University of Health Sciences of Porto Alegre, Porto Alegre, RS, Brazil
- Programa de Pós Graduação em Biociências e Reabilitação doCentro Universitário Metodista-IPA, Porto Alegre, RS, Brazil
- Programa de Pós Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul, Brazil
| |
Collapse
|
9
|
Enhanced Expression of Human Endogenous Retroviruses, TRIM28 and SETDB1 in Autism Spectrum Disorder. Int J Mol Sci 2022; 23:ijms23115964. [PMID: 35682642 PMCID: PMC9180946 DOI: 10.3390/ijms23115964] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/12/2022] [Accepted: 05/23/2022] [Indexed: 02/01/2023] Open
Abstract
Human endogenous retroviruses (HERVs) are relics of ancestral infections and represent 8% of the human genome. They are no longer infectious, but their activation has been associated with several disorders, including neuropsychiatric conditions. Enhanced expression of HERV-K and HERV-H envelope genes has been found in the blood of autism spectrum disorder (ASD) patients, but no information is available on syncytin 1 (SYN1), SYN2, and multiple sclerosis-associated retrovirus (MSRV), which are thought to be implicated in brain development and immune responses. HERV activation is regulated by TRIM28 and SETDB1, which are part of the epigenetic mechanisms that organize the chromatin architecture in response to external stimuli and are involved in neural cell differentiation and brain inflammation. We assessed, through a PCR realtime Taqman amplification assay, the transcription levels of pol genes of HERV-H, -K, and -W families, of env genes of SYN1, SYN2, and MSRV, as well as of TRIM28 and SETDB1 in the blood of 33 ASD children (28 males, median 3.8 years, 25–75% interquartile range 3.0–6.0 y) and healthy controls (HC). Significantly higher expressions of TRIM28 and SETDB1, as well as of all the HERV genes tested, except for HERV-W-pol, were found in ASD, as compared with HC. Positive correlations were observed between the mRNA levels of TRIM28 or SETDB1 and every HERV gene in ASD patients, but not in HC. Overexpression of TRIM28/SETDB1 and several HERVs in children with ASD and the positive correlations between their transcriptional levels suggest that these may be main players in pathogenetic mechanisms leading to ASD.
Collapse
|
10
|
Ghafouri-Fard S, Eghtedarian R, Taheri M, Beatrix Brühl A, Sadeghi-Bahmani D, Brand S. A Review on the Expression Pattern of Non-coding RNAs in Patients With Schizophrenia: With a Special Focus on Peripheral Blood as a Source of Expression Analysis. Front Psychiatry 2021; 12:640463. [PMID: 34220567 PMCID: PMC8249727 DOI: 10.3389/fpsyt.2021.640463] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 05/06/2021] [Indexed: 12/14/2022] Open
Abstract
Schizophrenia is a destructive neuropsychiatric disease with a median prevalence of 4.0 per 1,000 during the whole life. Genome-wide association studies have shown the role of copy number variants (generally deletions) and certain alleles of common single nucleotide polymorphisms in the pathogenesis of schizophrenia. This disorder predominantly follows the polygenic inheritance model. Schizophrenia has also been linked with various alterations in the transcript and protein content of the brain tissue. Recent studies indicate that alterations in non-coding RNAs (ncRNAs) signature underlie a proportion of this dysregulation. High throughput microarray investigations have demonstrated momentous alterations in the expression of long non-coding RNAs (lncRNA) and microRNAs (miRNAs) in the circulation or post-mortem brain tissues of patients with schizophrenia compared with control samples. While Gomafu, PINT, GAS5, TCONS_l2_00021339, IFNG-AS1, FAS-AS1, PVT1, and TUG1 are among down-regulated lncRNAs in schizophrenia, MEG3, THRIL, HOXA-AS2, Linc-ROR, SPRY4-IT1, UCA1, and MALAT1 have been up-regulated in these patients. Moreover, several miRNAs, such as miR-30e, miR-130b, hsa-miR-130b, miR-193a-3p, hsa-miR-193a-3p, hsa-miR-181b, hsa-miR-34a, hsa-miR-346, and hsa-miR-7 have been shown to be dysregulated in blood or brain samples of patients with schizophrenia. Dysregulation of these transcripts in schizophrenia not only provides insight into the pathogenic processes of this disorder, it also suggests these transcripts could serve as diagnostic markers for schizophrenia. In the present paper, we explore the changes in the expression of miRNAs and lncRNAs in patients with schizophrenia.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reyhane Eghtedarian
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Annette Beatrix Brühl
- Psychiatric Clinics, Center for Affective, Stress and Sleep Disorders, University of Basel, Basel, Switzerland
| | - Dena Sadeghi-Bahmani
- Psychiatric Clinics, Center for Affective, Stress and Sleep Disorders, University of Basel, Basel, Switzerland
- Exercise Neuroscience Research Laboratory, The University of Alabama at Birmingham, Birmingham, AL, United States
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Serge Brand
- Psychiatric Clinics, Center for Affective, Stress and Sleep Disorders, University of Basel, Basel, Switzerland
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Substance Abuse Prevention Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Division of Sport Science and Psychosocial Health, Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
- Department of Psychiatry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Liu YR, Wang JQ, Huang ZG, Chen RN, Cao X, Zhu DC, Yu HX, Wang XR, Zhou HY, Xia Q, Li J. Histone deacetylase‑2: A potential regulator and therapeutic target in liver disease (Review). Int J Mol Med 2021; 48:131. [PMID: 34013366 PMCID: PMC8136123 DOI: 10.3892/ijmm.2021.4964] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 04/12/2021] [Indexed: 12/12/2022] Open
Abstract
Histone acetyltransferases are responsible for histone acetylation, while histone deacetylases (HDACs) counteract histone acetylation. An unbalanced dynamic between histone acetylation and deacetylation may lead to aberrant chromatin landscape and chromosomal function. HDAC2, a member of class I HDAC family, serves a crucial role in the modulation of cell signaling, immune response and gene expression. HDAC2 has emerged as a promising therapeutic target for liver disease by regulating gene transcription, chromatin remodeling, signal transduction and nuclear reprogramming, thus receiving attention from researchers and clinicians. The present review introduces biological information of HDAC2 and its physiological and biochemical functions. Secondly, the functional roles of HDAC2 in liver disease are discussed in terms of hepatocyte apoptosis and proliferation, liver regeneration, hepatocellular carcinoma, liver fibrosis and non-alcoholic steatohepatitis. Moreover, abnormal expression of HDAC2 may be involved in the pathogenesis of liver disease, and its expression levels and pharmacological activity may represent potential biomarkers of liver disease. Finally, research on selective HDAC2 inhibitors and non-coding RNAs relevant to HDAC2 expression in liver disease is also reviewed. The aim of the present review was to improve understanding of the multifunctional role and potential regulatory mechanism of HDAC2 in liver disease.
Collapse
Affiliation(s)
- Ya-Ru Liu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Jie-Quan Wang
- Department of Pharmacy, Affiliated Psychological Hospital of Anhui Medical University, Hefei, Anhui 230000, P.R. China
| | - Zhao-Gang Huang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Ruo-Nan Chen
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Xi Cao
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Dong-Chun Zhu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Hai-Xia Yu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Xiu-Rong Wang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Hai-Yun Zhou
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Quan Xia
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Jun Li
- The Key Laboratory of Anti‑inflammatory Immune Medicines, School of Pharmacy, Anhui Medical University, Ministry of Education, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
12
|
Nie FY, Zhang MR, Shang SS, Zhang QX, Zhang R, Chen P, Ma J. Methylome-wide association study of first-episode schizophrenia reveals a hypermethylated CpG site in the promoter region of the TNIK susceptibility gene. Prog Neuropsychopharmacol Biol Psychiatry 2021; 106:110081. [PMID: 32853717 DOI: 10.1016/j.pnpbp.2020.110081] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 08/16/2020] [Accepted: 08/18/2020] [Indexed: 12/11/2022]
Abstract
Accumulating evidence suggests that epigenetics plays an important role in the etiology of schizophrenia. Here, we performed a methylome-wide association study (MWAS) of first-onset schizophrenia patients and controls from the Han Chinese population using microarray technology. The DNA methylation profiles revealed 4494 differentially methylated CpG sites. Gene ontology (GO) analysis showed that the functions of differentially methylated genes were primarily involved in enzymatic activity, cytoskeleton organization and cell adhesion, and the TNIK (encoding TRAF2- and NCK-interacting kinase) gene was enriched in most of these terms. By combining the MWAS results with those of previous genome-wide association studies (GWASs), we identified 72 candidate genes located in 49 human genome loci. Among the overlapping genes, the most significantly methylated CpG sites were in the transcriptional start site (TSS) 200 region (cg21413905, Punadjusted = 3.20 × 10-5) of TNIK. TNIK was listed in the top 50 differentially methylated loci. The results of pyrosequencing and TNIK mRNA expression were consistent with those of the microarray study. Bioinformatics analyses, dual-luciferase reporter assays and chromatin immunoprecipitation (ChIP) studies showed that TNIK interacted with genes associated with schizophrenia and NRF1 was identified as a novel transcription factor (TF) that binds to TNIK in its TSS200 region. Thus, the regulatory function of NRF1 may be influenced by the status of the methylated CpG site in this region. In summary, our study provides new insights into the epigenetic mechanisms that regulate schizophrenia. Studies of the functions of TNIK methylation should be performed in vitro and in vivo to provide a better understanding of the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Fa-Yi Nie
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Medical Research Center, Xi'an No.3 Hospital, Xi'an, Shaanxi 710018, China
| | - Miao-Ran Zhang
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Shan-Shan Shang
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Qiao-Xia Zhang
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Rui Zhang
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China
| | - Peng Chen
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China.
| | - Jie Ma
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Medical Research Center, Xi'an No.3 Hospital, Xi'an, Shaanxi 710018, China.
| |
Collapse
|
13
|
Yin P, Zhao C, Li Y, Liu X, Chen L, Hong N. Changes in Brain Structure, Function, and Network Properties in Patients With First-Episode Schizophrenia Treated With Antipsychotics. Front Psychiatry 2021; 12:735623. [PMID: 34916969 PMCID: PMC8668948 DOI: 10.3389/fpsyt.2021.735623] [Citation(s) in RCA: 6] [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: 07/13/2021] [Accepted: 11/09/2021] [Indexed: 01/10/2023] Open
Abstract
Purpose: Comprehensive and longitudinal brain analysis is of great significance for understanding the pathological changes of antipsychotic drug treatment in patients with schizophrenia. This study aimed to investigate the changes of structure, function, and network properties in patients with first-episode schizophrenia (FES) after antipsychotic therapy and their relationship with clinical symptoms. Materials and Methods: A total of 30 patients diagnosed with FES and 30 healthy subjects matched for sex and age were enrolled in our study. Patients at baseline were labeled as antipsychotic-naive first-episode schizophrenia (AN-FES), and patients after antipsychotic treatment were labeled as antipsychotic treatment first-episode schizophrenia (AT-FES). The severity of illness was measured by using the PANSS and CGI score. Structural and functional MRI data were also performed. Differences in GMV, ALFF, and ReHo between the FES group and healthy control group were tested using a voxel-wise two-sample t-test, and the comparison of AN-FES group and AT-FES group was evaluated by paired-sample t-test. Results: After the 1-year follow-up, the FES patients showed increased GMV in the right cerebellum, right inferior temporal gyrus, left middle frontal gyrus, parahippocampal gyrus, bilateral inferior parietal lobule, and reduced GMV in the left occipital lobe, gyrus rectus, right orbital frontal cortex. The patients also showed increased ALFF in the medial superior frontal gyrus and right precentral gyrus. For network properties, the patients showed reduced characteristic path length and increased global efficiency. The GMV of the right inferior parietal lobule was negatively correlated with the clinical symptoms. Conclusions: Our study showed that the antipsychotic treatment contributed to the structural alteration and functional improvement, and the GMV alteration may be associated with the improvement of clinical symptoms.
Collapse
Affiliation(s)
- Ping Yin
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Chao Zhao
- Department of Interventional Radiology, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yang Li
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Xiaoyi Liu
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Lei Chen
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Nan Hong
- Department of Radiology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
14
|
Chen R, Chen J, Gao C, Wu C, Pan D, Zhang J, Zhou J, Wang K, Zhang Q, Yang Q, Jian X, Zhao Y, Wen Y, Wang Z, Shi Y, Li Z. Association analysis of potentially functional variants within 8p12 with schizophrenia in the Han Chinese population. World J Biol Psychiatry 2021; 22:27-33. [PMID: 32129128 DOI: 10.1080/15622975.2020.1738550] [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] [Indexed: 10/24/2022]
Abstract
OBJECTIVES Chromosome 8p12 was first identified as a schizophrenia (SCZ) risk locus in Chinese populations and replicated in European populations. However, the underlying functional variants still need to be further explored. In this study, we sought to identify plausible causal variants within this locus. METHODS A total of 386 potentially functional variants from 29 genes within the 8p12 locus were analysed in 2403 SCZ cases and 2594 control subjects in the Han Chinese population using Affymetrix customised genotyping assays. SHEsisplus was used for association analysis. A multiple testing corrected p value (false discovery rate (FDR)) < .05 was considered significant, and an unadjusted p value < .05 was considered nominal evidence of an association. RESULTS We did not find significant associations between the tested variants and SCZ. However, nominal associations were found for rs201292574 (unadjusted p = .033, FDR p = .571; 95% confidence interval (CI): 0.265-0.945; TACC1, NP_006274.2:p.Ala211Thr) and rs45563241 (unadjusted p = .039, FDR p = .571; 95% CI: 1.023-1.866; a synonymous mutation in ADRB3). CONCLUSIONS Our results provide limited evidence for the associations between variants from protein coding regions in 8p12 and SCZ in the Chinese population. Analyses of both coding and regulatory variants in larger sample sizes are required to further clarify the causal variants for SCZ with this risk locus.
Collapse
Affiliation(s)
- Ruirui Chen
- School of Basic Medicine, Qingdao University, Qingdao, China.,Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China
| | - Jianhua Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Chengwen Gao
- Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China
| | - Chuanhong Wu
- Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China
| | - Dun Pan
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Jinmai Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Juan Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Ke Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Qian Zhang
- Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China
| | - Qiangzhen Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Xuemin Jian
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Yalin Zhao
- School of Basic Medicine, Qingdao University, Qingdao, China.,Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China
| | - Yanqin Wen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Zhuo Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Yongyong Shi
- School of Basic Medicine, Qingdao University, Qingdao, China.,Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China.,Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China.,Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiqiang Li
- School of Basic Medicine, Qingdao University, Qingdao, China.,Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China.,Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China.,Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
15
|
Transient receptor potential vanilloid 1 antagonism in neuroinflammation, neuroprotection and epigenetic regulation: potential therapeutic implications for severe psychiatric disorders treatment. Psychiatr Genet 2020; 30:39-48. [PMID: 32097233 DOI: 10.1097/ypg.0000000000000249] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
16
|
Abstract
The brain-derived neurotrophic factor (BDNF) is a secretory growth factor that promotes neuronal proliferation and survival, synaptic plasticity and long-term potentiation in the central nervous system. Brain-derived neurotrophic factor biosynthesis and secretion are chrono-topically regulated processes at the cellular level, accounting for specific localizations and functions. Given its role in regulating brain development and activity, BDNF represents a potentially relevant gene for schizophrenia, and indeed BDNF and its non-synonymous functional variant, rs6265 (C → T, Val → Met) have been widely studied in psychiatric genetics. Human and animal studies have indicated that brain-derived neurotrophic factor is relevant for schizophrenia-related phenotypes, and that: (1) fine-tuned regulation of brain-derived neurotrophic factor secretion and activity is necessary to guarantee brain optimal development and functioning; (2) the Val → Met substitution is associated with impaired activity-dependent secretion of brain-derived neurotrophic factor; (3) disruption of brain-derived neurotrophic factor signaling is associated with altered synaptic plasticity and neurodevelopment. However, genome-wide association studies failed to associate the BDNF locus with schizophrenia, even though a sub-threshold association exists. Here, we will review studies focused on the relationship between the genetic variation of BDNF and schizophrenia, trying to fill the gap between genetic risk per se and insights from molecular biology. A deeper understanding of brain-derived neurotrophic factor biology and of the epigenetic regulation of brain-derived neurotrophic factor and its interactome during development may help clarifying the potential role of this gene in schizophrenia, thus informing development of brain-derived neurotrophic factor-based strategies of prevention and treatment of this disorder.
Collapse
|
17
|
Endogenous Retroviruses Activity as a Molecular Signature of Neurodevelopmental Disorders. Int J Mol Sci 2019; 20:ijms20236050. [PMID: 31801288 PMCID: PMC6928979 DOI: 10.3390/ijms20236050] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 11/26/2019] [Accepted: 11/28/2019] [Indexed: 12/20/2022] Open
Abstract
Human endogenous retroviruses (HERVs) are genetic elements resulting from relics of ancestral infection of germline cells, now recognized as cofactors in the etiology of several complex diseases. Here we present a review of findings supporting the role of the abnormal HERVs activity in neurodevelopmental disorders. The derailment of brain development underlies numerous neuropsychiatric conditions, likely starting during prenatal life and carrying on during subsequent maturation of the brain. Autism spectrum disorders, attention deficit hyperactivity disorders, and schizophrenia are neurodevelopmental disorders that arise clinically during early childhood or adolescence, currently attributed to the interplay among genetic vulnerability, environmental risk factors, and maternal immune activation. The role of HERVs in human embryogenesis, their intrinsic responsiveness to external stimuli, and the interaction with the immune system support the involvement of HERVs in the derailed neurodevelopmental process. Although definitive proofs that HERVs are involved in neurobehavioral alterations are still lacking, both preclinical models and human studies indicate that the abnormal expression of ERVs could represent a neurodevelopmental disorders-associated biological trait in affected individuals and their parents.
Collapse
|
18
|
Post-translational histone modifications and their interaction with sex influence normal brain development and elaboration of neuropsychiatric disorders. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1968-1981. [DOI: 10.1016/j.bbadis.2018.10.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/05/2018] [Accepted: 10/08/2018] [Indexed: 02/06/2023]
|
19
|
Sidoli S, Kori Y, Lopes M, Yuan ZF, Kim HJ, Kulej K, Janssen KA, Agosto LM, Cunha JPCD, Andrews AJ, Garcia BA. One minute analysis of 200 histone posttranslational modifications by direct injection mass spectrometry. Genome Res 2019; 29:978-987. [PMID: 31123082 PMCID: PMC6581051 DOI: 10.1101/gr.247353.118] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 05/13/2019] [Indexed: 01/11/2023]
Abstract
DNA and histone proteins define the structure and composition of chromatin. Histone posttranslational modifications (PTMs) are covalent chemical groups capable of modeling chromatin accessibility, mostly due to their ability in recruiting enzymes responsible for DNA readout and remodeling. Mass spectrometry (MS)-based proteomics is the methodology of choice for large-scale identification and quantification of protein PTMs, including histones. High sensitivity proteomics requires online MS coupling with relatively low throughput and poorly robust nano-liquid chromatography (nanoLC) and, for histone proteins, a 2-d sample preparation that includes histone purification, derivatization, and digestion. We present a new protocol that achieves quantitative data on about 200 histone PTMs from tissue or cell lines in 7 h from start to finish. This protocol includes 4 h of histone extraction, 3 h of derivatization and digestion, and only 1 min of MS analysis via direct injection (DI-MS). We demonstrate that this sample preparation can be parallelized for 384 samples by using multichannel pipettes and 96-well plates. We also engineered the sequence of a synthetic "histone-like" peptide to spike into the sample, of which derivatization and digestion benchmarks the quality of the sample preparation. We ensure that DI-MS does not introduce biases in histone peptide ionization as compared to nanoLC-MS/MS by producing and analyzing a library of synthetically modified histone peptides mixed in equal molarity. Finally, we introduce EpiProfileLite for comprehensive analysis of this new data type. Altogether, our workflow is suitable for high-throughput screening of >1000 samples per day using a single mass spectrometer.
Collapse
Affiliation(s)
- Simone Sidoli
- Epigenetics Institute, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Yekaterina Kori
- Epigenetics Institute, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Mariana Lopes
- Epigenetics Institute, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Laboratório Especial de Ciclo Celular, Center of Toxins, Immune Response and Cell Signaling - CeTICS, Instituto Butantan, São Paulo, 05503-900, Brazil
| | - Zuo-Fei Yuan
- Epigenetics Institute, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Hee Jong Kim
- Epigenetics Institute, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Katarzyna Kulej
- Epigenetics Institute, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Kevin A Janssen
- Epigenetics Institute, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Laura M Agosto
- Epigenetics Institute, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Julia Pinheiro Chagas da Cunha
- Laboratório Especial de Ciclo Celular, Center of Toxins, Immune Response and Cell Signaling - CeTICS, Instituto Butantan, São Paulo, 05503-900, Brazil
| | - Andrew J Andrews
- Cancer Epigenetics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111, USA
| | - Benjamin A Garcia
- Epigenetics Institute, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
20
|
Xiang B, Wang Q, Lei W, Li M, Li Y, Zhao L, Ma X, Wang Y, Yu H, Li X, Meng Y, Guo W, Deng W, Ren H, Li T. Genes in immune pathways associated with abnormal white matter integrity in first-episode and treatment-naïve patients with schizophrenia. Br J Psychiatry 2019; 214:281-287. [PMID: 30722794 DOI: 10.1192/bjp.2018.297] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Previous studies have inferred a strong genetic component in schizophrenia. However, the genetic variants involved in the susceptibility to schizophrenia remain unclear.AimsTo detect potential gene pathways and networks associated with schizophrenia, and to explore the relationship between common and rare variants in these pathways and abnormal white matter integrity in schizophrenia. METHOD The analysis included 100 first-episode treatment-naïve patients with schizophrenia and 140 healthy controls. A network-based analysis was carried out on the data collected from the Psychiatric Genomics Consortium Phase I (PGC-I). Based on our genome-wide association study and whole-exome sequencing data-sets, we performed a gene-set analysis to detect associations between the combining effects of common and rare genetic variants and abnormal white matter integrity in schizophrenia. RESULTS Patients had significantly reduced functional anisotropy in the left and right anterior cingulate cortex, left and right precuneus and extra-nuclear (t = 4.61-5.10, PFDR < 0.01), compared with controls. Generated from co-expression network analysis of the PGC-1 summary statistics of schizophrenia, a subnetwork of 207 genes associated with schizophrenia was identified (P < 0.01), and 176 genes were co-expressed in four gene modules. Functional enrichment analysis for genes in each module revealed that the yellow module was enriched with highly co-expressed, innate immune response genes. Furthermore, rare variants of enriched genes in the yellow module were associated with reduced functional anisotropy in the left anterior cingulate cortex (P = 0.006; Padjusted = 0.024) in patients only. CONCLUSIONS The pathogenesis of schizophrenia may be substantially influenced by genes involved in the immune system, via both pathway and network.Declaration of interestsNone.
Collapse
Affiliation(s)
- Bo Xiang
- Assistant Professor,Mental Health Center and Psychiatric Laboratory,State Key Laboratory of Biotherapy,West China Brain Research Center,West China Hospital of Sichuan University; andDepartment of Psychiatry,Affiliated Hospital of Southwest Medical University,China
| | - Qiang Wang
- Professor,Mental Health Center and Psychiatric Laboratory,State Key Laboratory of Biotherapy,West China Brain Research Center,West China Hospital of Sichuan University,China
| | - Wei Lei
- Assistant Professor,Mental Health Center and Psychiatric Laboratory,State Key Laboratory of Biotherapy,West China Brain Research Center,West China Hospital of Sichuan University; andDepartment of Psychiatry,Affiliated Hospital of Southwest Medical University,China
| | - Mingli Li
- Associate Professor,Mental Health Center and Psychiatric Laboratory,State Key Laboratory of Biotherapy,West China Brain Research Center,West China Hospital of Sichuan University,China
| | - Yinfei Li
- Attending Doctor,Mental Health Center and Psychiatric Laboratory,State Key Laboratory of Biotherapy,West China Brain Research Center,West China Hospital of Sichuan University,China
| | - Liansheng Zhao
- Assistant Professor,Mental Health Center and Psychiatric Laboratory,State Key Laboratory of Biotherapy,West China Brain Research Center,West China Hospital of Sichuan University,China
| | - Xiaohong Ma
- Professor,Mental Health Center and Psychiatric Laboratory,State Key Laboratory of Biotherapy,West China Brain Research Center,West China Hospital of Sichuan University,China
| | - Yingcheng Wang
- Assistant Professor,Mental Health Center and Psychiatric Laboratory,State Key Laboratory of Biotherapy,West China Brain Research Center,West China Hospital of Sichuan University,China
| | - Hua Yu
- Attending Doctor,Mental Health Center and Psychiatric Laboratory,State Key Laboratory of Biotherapy,West China Brain Research Center,West China Hospital of Sichuan University,China
| | - Xiaojing Li
- Attending Doctor,Mental Health Center and Psychiatric Laboratory,State Key Laboratory of Biotherapy,West China Brain Research Center,West China Hospital of Sichuan University,China
| | - Yajing Meng
- Attending Doctor,Mental Health Center and Psychiatric Laboratory,State Key Laboratory of Biotherapy,West China Brain Research Center,West China Hospital of Sichuan University,China
| | - Wanjun Guo
- Associate Professor,Mental Health Center and Psychiatric Laboratory,State Key Laboratory of Biotherapy,West China Brain Research Center,West China Hospital of Sichuan University,China
| | - Wei Deng
- Associate Professor,Mental Health Center and Psychiatric Laboratory,State Key Laboratory of Biotherapy,West China Brain Research Center,West China Hospital of Sichuan University,China
| | - Hongyan Ren
- Attending Doctor,Mental Health Center and Psychiatric Laboratory,State Key Laboratory of Biotherapy,West China Brain Research Center,West China Hospital of Sichuan University,China
| | - Tao Li
- Professor,Mental Health Center and Psychiatric Laboratory,State Key Laboratory of Biotherapy,West China Brain Research Center,West China Hospital of Sichuan University,China
| |
Collapse
|
21
|
Hu TM, Chen SJ, Hsu SH, Cheng MC. Functional analyses and effect of DNA methylation on the EGR1 gene in patients with schizophrenia. Psychiatry Res 2019; 275:276-282. [PMID: 30952071 DOI: 10.1016/j.psychres.2019.03.044] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 03/14/2019] [Accepted: 03/26/2019] [Indexed: 11/29/2022]
Abstract
EGR1, involved in the regulation of synaptic plasticity, learning, and memory, is considered a candidate gene for schizophrenia. We resequenced the exonic regions of EGR1 in 516 patients with schizophrenia and conducted a reporter gene assay. We found two mutations including a rare mutation (c.-8C>T, rs561524195) and one common SNP (c.308-42C>T, rs11743810). The reporter gene assay showed c.-8C>T mutant did not affect promoter activity. Gene expression analyses showed that the average EGR1 mRNA and protein levels in lymphoblastoid cell lines of schizophrenia in male, but not female, were significantly higher than those in controls. We conducted in vitro DNA methylation reaction, luciferase activity assay, and pyrosequencing to assess DNA methylation of EGR1 expression underlying the pathophysiology of schizophrenia. DNA methylation of the EGR1 promoter region attenuated reporter activity, suggesting that DNA methylation regulates EGR1 expression. There were no statistically significant differences in DNA methylation levels of 17 CpG sites at the EGR1 promoter region between 64 patients with schizophrenia compared with 64 controls. These results suggest that the exonic mutations in EGR1 and DNA methylation regulating EGR1 expression might not be associated with schizophrenia. However, the gender-specific association of elevated EGR1 expression might be involved in the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Tsung-Ming Hu
- Department of Psychiatry, Yuli Branch, Taipei Veterans General Hospital, Hualien County, Taiwan; Department of Long-Term Care, University of Kang Ning, Taipei City, Taiwan
| | - Shaw-Ji Chen
- Department of Psychiatry, Mackay Medical College, New Taipei City, Taiwan; Department of Psychiatry, Mackay Memorial Hospital, Taitung Branch, Taitung County, Taiwan
| | - Shih-Hsin Hsu
- Department of Psychiatry, Yuli Branch, Taipei Veterans General Hospital, Hualien County, Taiwan
| | - Min-Chih Cheng
- Department of Psychiatry, Yuli Branch, Taipei Veterans General Hospital, Hualien County, Taiwan.
| |
Collapse
|
22
|
First-episode schizophrenia is associated with a reduction of HERV-K methylation in peripheral blood. Psychiatry Res 2019; 271:459-463. [PMID: 30537669 DOI: 10.1016/j.psychres.2018.12.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/02/2018] [Accepted: 12/03/2018] [Indexed: 12/20/2022]
Abstract
Human endogenous retroviruses (HERV) have been widely associated with schizophrenia etiology. Aberrant epigenetic processes may play a role in the etiology of schizophrenia. In this study, we tested whether schizophrenia patients at different stages of illness might present alterations in the levels of HERV-K methylation. We recruited 49 first-episode schizophrenia (FES) patients with 47 age- and sex-matched healthy controls (HCs), and 100 multi-episode schizophrenia (MES) patients with 50 age- and sex-matched HCs. Based on the Schedule for Deficit Schizophrenia, patients with MES were also divided into two subgroups: deficit (D-SCZ) and non-deficit schizophrenia (ND-SCZ). DNA methylation levels of HERV-K sequences were examined in peripheral blood leukocytes. We found significantly lower levels of HERV-K methylation in FES patients compared to HCs. Patients with MES and matched HCs had similar levels of HERV-K methylation. There was a significant positive correlation between chlorpromazine equivalent dosage and HERV-K methylation levels in MES patients, but not in FES individuals. No significant differences in HERV-K methylation levels between D-SCZ and ND-SCZ as well as HCs were found. Our results indicate lower HERV-K methylation levels at early stages of schizophrenia. This difference might normalize with subsequent exacerbations of schizophrenia, likely due to the effects of antipsychotics.
Collapse
|
23
|
de la Fuente Revenga M, Ibi D, Cuddy T, Toneatti R, Kurita M, Ijaz MK, Miles MF, Wolstenholme JT, González-Maeso J. Chronic clozapine treatment restrains via HDAC2 the performance of mGlu2 receptor agonism in a rodent model of antipsychotic activity. Neuropsychopharmacology 2019; 44:443-454. [PMID: 30038413 PMCID: PMC6300555 DOI: 10.1038/s41386-018-0143-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 06/06/2018] [Accepted: 06/25/2018] [Indexed: 01/25/2023]
Abstract
Preclinical findings in rodent models pointed toward activation of metabotropic glutamate 2/3 (mGlu2/3) receptors as a new pharmacological approach to treat psychosis. However, more recent studies failed to show clinical efficacy of mGlu2/3 receptor agonism in schizophrenia patients. We previously proposed that long-term antipsychotic medication restricted the therapeutic effects of these glutamatergic agents. However, little is known about the molecular mechanism underlying the potential repercussion of previous antipsychotic exposure on the therapeutic performance of mGlu2/3 receptor agonists. Here we show that this maladaptive effect of antipsychotic treatment is mediated mostly via histone deacetylase 2 (HDAC2). Chronic treatment with the antipsychotic clozapine led to a decrease in mouse frontal cortex mGlu2 mRNA, an effect that required expression of both HDAC2 and the serotonin 5-HT2A receptor. This transcriptional alteration occurred in association with HDAC2-dependent repressive histone modifications at the mGlu2 promoter. We found that chronic clozapine treatment decreased via HDAC2 the capabilities of the mGlu2/3 receptor agonist LY379268 to activate G-proteins in the frontal cortex of mice. Chronic clozapine treatment blunted the antipsychotic-related behavioral effects of LY379268, an effect that was not observed in HDAC2 knockout mice. More importantly, co-administration of the class I and II HDAC inhibitor SAHA (vorinostat) preserved the antipsychotic profile of LY379268 and frontal cortex mGlu2/3 receptor density in wild-type mice. These findings raise concerns on the design of previous clinical studies with mGlu2/3 agonists, providing the rationale for the development of HDAC2 inhibitors as a new epigenetic-based approach to improve the currently limited response to treatment with glutamatergic antipsychotics.
Collapse
Affiliation(s)
- Mario de la Fuente Revenga
- 0000 0004 0458 8737grid.224260.0Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298 USA
| | - Daisuke Ibi
- 0000 0004 0458 8737grid.224260.0Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298 USA ,0000 0001 0670 2351grid.59734.3cDepartment Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA ,grid.259879.8Department of Chemical Pharmacology, Meijo University, Nagoya, 468-8503 Japan
| | - Travis Cuddy
- 0000 0004 0458 8737grid.224260.0Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298 USA
| | - Rudy Toneatti
- 0000 0004 0458 8737grid.224260.0Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298 USA
| | - Mitsumasa Kurita
- 0000 0001 0670 2351grid.59734.3cDepartment Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA ,0000 0004 1797 168Xgrid.417741.0Present Address: Dainippon Sumitomo Pharma Co., Ltd., Osaka, 564-0053 Japan
| | - Maryum K. Ijaz
- 0000 0004 0458 8737grid.224260.0Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298 USA
| | - Michael F. Miles
- 0000 0004 0458 8737grid.224260.0Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298 USA ,0000 0004 0458 8737grid.224260.0VCU Alcohol Research Center, Virginia Commonwealth University School of Medicine, Richmond, VA 23298 USA
| | - Jennifer T. Wolstenholme
- 0000 0004 0458 8737grid.224260.0Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298 USA ,0000 0004 0458 8737grid.224260.0VCU Alcohol Research Center, Virginia Commonwealth University School of Medicine, Richmond, VA 23298 USA
| | - Javier González-Maeso
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA. .,Department Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA. .,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA. .,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
24
|
Gao J, Yi H, Tang X, Feng X, Yu M, Sha W, Wang X, Zhang X, Zhang X. DNA Methylation and Gene Expression of Matrix Metalloproteinase 9 Gene in Deficit and Non-deficit Schizophrenia. Front Genet 2018; 9:646. [PMID: 30619470 PMCID: PMC6297256 DOI: 10.3389/fgene.2018.00646] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Accepted: 11/29/2018] [Indexed: 12/03/2022] Open
Abstract
The biological pathology of deficit schizophrenia (DS) remains unclear. Matrix metalloproteinase 9 (MMP9) might be associated with neural plasticity and glutamate regulation, involved in schizophrenia pathogenesis. This study explores gene expression and DNA methylation of MMP9 in peripheral blood mononuclear cells (PBMCs) and their relationship with clinical symptoms in DS and non-deficit schizophrenia (NDS). Pyrosequencing was used to determine DNA methylation at CpG sites in exon 4 and exon 5 of MMP9 in 51 DS patients, 53 NDS patients and 50 healthy subjects (HC). RT-qPCR was used to detect MMP9 expression. Clinical symptoms were assessed by BPRS, SANS and SAPS scales. MMP9 expression in PBMCs was significantly higher in DS than NDS and HC subjects. Compared to NDS patients, DS patients had significantly lower DNA methylation at individual CpG sites in exon 4 and exon 5 of MMP9. Correlation analysis showed that DNA methylation in exon 4 was negatively correlated with gene expression in DS group. Positive correlation was found between MMP9 expression and negative symptoms in total schizophrenic patients. The social amotivation factor of SANS and negative syndrome of BPRS was negatively correlated with DNA methylation of CpG5-1 in DS patients but not in NDS patients. DS patients showed a specific abnormality of peripheral MMP9 expression and DNA methylation, indicating a pathological mechanism underlying DS as a specific subgroup of schizophrenia.
Collapse
Affiliation(s)
- Ju Gao
- Department of Geriatric Psychiatry, Nanjing Brain Hospital, Affiliated to Nanjing Medical University, Nanjing, China
- Centers of Disease Prevention and Control for Mental Disorders, Shanghai Changning Mental Health Center, Shanghai, China
| | - Hongwei Yi
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, China
| | - Xiaowei Tang
- Department of Psychiatry, Affiliated WuTaiShan Hospital of Medical College, Yangzhou University, Yangzhou, China
| | - Xiaotang Feng
- Department of Psychiatry, Nanjing Qing Long Mountain Psychiatric Hospital, Nanjing, China
| | - Miao Yu
- Department of Geriatric Psychiatry, Nanjing Brain Hospital, Affiliated to Nanjing Medical University, Nanjing, China
| | - Weiwei Sha
- Department of Psychiatry, Affiliated WuTaiShan Hospital of Medical College, Yangzhou University, Yangzhou, China
| | - Xiang Wang
- Medical Psychological Institute of the Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiaobin Zhang
- Department of Psychiatry, Affiliated WuTaiShan Hospital of Medical College, Yangzhou University, Yangzhou, China
| | - Xiangrong Zhang
- Department of Geriatric Psychiatry, Nanjing Brain Hospital, Affiliated to Nanjing Medical University, Nanjing, China
| |
Collapse
|
25
|
Association between family history of mental disorders and outcome in psychotic disorders. Psychiatry Res 2018; 270:616-621. [PMID: 30384280 DOI: 10.1016/j.psychres.2018.10.047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 10/17/2018] [Accepted: 10/19/2018] [Indexed: 01/20/2023]
Abstract
We investigated the association of family history of mental disorders, especially psychosis, with occupational and clinical outcome in psychotic disorders in a longitudinal population-based cohort. The Northern Finland Birth Cohort 1986 (n = 9432) was used to gather the data. In total 189 individuals with psychosis were identified by age of 28. The outcome was assessed by using register information regarding occupational activity, disability pension and hospital treatments due to psychiatric cause. Parental psychosis and any psychiatric disorder were used as predictors of outcome. The results showed that presence of any parental psychiatric disorder was associated with higher number of days spent at hospital and higher number of hospitalizations in psychotic disorders, but was not associated with occupational outcome or disability pension. The presence of parental psychosis was not associated with outcome. These findings suggest that the presence of any psychiatric disorder among parents may increase the risk of poorer outcome in psychoses in terms of need of hospitalisations. Based on this study the presence of parental psychosis is not associated with outcome, but the result should be interpreted with caution due to the small sample size and conflict with the results of earlier studies.
Collapse
|
26
|
Qvist P, Eskildsen SF, Hansen B, Baragji M, Ringgaard S, Roovers J, Paternoster V, Molgaard S, Corydon TJ, Stødkilde-Jørgensen H, Glerup S, Mors O, Wegener G, Nyengaard JR, Børglum AD, Christensen JH. Brain volumetric alterations accompanied with loss of striatal medium-sized spiny neurons and cortical parvalbumin expressing interneurons in Brd1 +/- mice. Sci Rep 2018; 8:16486. [PMID: 30405140 PMCID: PMC6220279 DOI: 10.1038/s41598-018-34729-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/22/2018] [Indexed: 12/17/2022] Open
Abstract
Schizophrenia is a common and severe mental disorder arising from complex gene-environment interactions affecting brain development and functioning. While a consensus on the neuroanatomical correlates of schizophrenia is emerging, much of its fundamental pathobiology remains unknown. In this study, we explore brain morphometry in mice with genetic susceptibility and phenotypic relevance to schizophrenia (Brd1+/− mice) using postmortem 3D MR imaging coupled with histology, immunostaining and regional mRNA marker analysis. In agreement with recent large-scale schizophrenia neuroimaging studies, Brd1+/− mice displayed subcortical abnormalities, including volumetric reductions of amygdala and striatum. Interestingly, we demonstrate that structural alteration in striatum correlates with a general loss of striatal neurons, differentially impacting subpopulations of medium-sized spiny neurons and thus potentially striatal output. Akin to parvalbumin interneuron dysfunction in patients, a decline in parvalbumin expression was noted in the developing cortex of Brd1+/− mice, mainly driven by neuronal loss within or near cortical layer V, which is rich in corticostriatal projection neurons. Collectively, our study highlights the translational value of the Brd1+/− mouse as a pre-clinical tool for schizophrenia research and provides novel insight into its developmental, structural, and cellular pathology.
Collapse
Affiliation(s)
- Per Qvist
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark. .,Department of Biomedicine, Aarhus University, Aarhus, Denmark. .,iSEQ, Centre for Integrative Sequencing, Aarhus University, Aarhus, Denmark.
| | - Simon F Eskildsen
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Brian Hansen
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Steffen Ringgaard
- The MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jolien Roovers
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Veerle Paternoster
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark.,iSEQ, Centre for Integrative Sequencing, Aarhus University, Aarhus, Denmark
| | - Simon Molgaard
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Thomas Juhl Corydon
- Department of Biomedicine, Aarhus University, Aarhus, Denmark. .,Department of Ophthalmology, Aarhus University Hospital, Aarhus, Denmark.
| | | | - Simon Glerup
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Ole Mors
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark.,iSEQ, Centre for Integrative Sequencing, Aarhus University, Aarhus, Denmark.,Psychosis Research Unit, Aarhus University Hospital, Risskov, Denmark
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Aarhus University Hospital, Aarhus, Denmark
| | - Jens R Nyengaard
- Core Center for Molecular Morphology, Section for Stereology and Microscopy, Centre for Stochastic Geometry and Advanced Bioimaging, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Anders D Børglum
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark.,iSEQ, Centre for Integrative Sequencing, Aarhus University, Aarhus, Denmark.,Psychosis Research Unit, Aarhus University Hospital, Risskov, Denmark
| | - Jane H Christensen
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark.,iSEQ, Centre for Integrative Sequencing, Aarhus University, Aarhus, Denmark
| |
Collapse
|
27
|
Zhou C, Chen J, Tang X, Feng X, Yu M, Sha W, Wang X, Zhang X, Yi H, Zhang X. DNA methylation and gene expression of the chemokine (C-X-C motif) ligand 1 in patients with deficit and non-deficit schizophrenia. Psychiatry Res 2018; 268:82-86. [PMID: 30015110 DOI: 10.1016/j.psychres.2018.06.067] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 06/12/2018] [Accepted: 06/30/2018] [Indexed: 12/27/2022]
Abstract
This study detected the differences in gene expression and DNA methylation of CpG sites in CXCL1 gene and further investigated their associations with clinical symptoms in deficit schizophrenia (DS) and non-deficit schizophrenia (NDS). Pyrosequencing and RT-qPCR were separately used to determine DNA methylation and mRNA expression of CXCL1 gene. Both DNA methylation and expression were significantly different among DS, NDS and healthy control (HC) groups. Correlation analysis revealed that CXCL1 gene expression was associated with the negative syndrome in NDS patients, while no association in DS patients was observed. All together, these results suggest that DS may be a specific subgroup of schizophrenia with the characteristic abnormality of peripheral CXCL1 DNA methylation and gene expression.
Collapse
Affiliation(s)
- Chao Zhou
- Department of Geriatric Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Jiu Chen
- Institute of Neuropsychiatry, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Institute of Brain Functional Imaging, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Xiaowei Tang
- Department of Geriatric Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China; Department of Psychiatry, Affiliated WuTaiShan Hospital of Medical College of Yangzhou University, Yangzhou, Jiangsu 225003, China
| | - Xiaotang Feng
- Department of Psychiatry, Nanjing Qing Long Mountain Psychiatric Hospital, Nanjing, China
| | - Miao Yu
- Department of Neurology, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Weiwei Sha
- Department of Psychiatry, Affiliated WuTaiShan Hospital of Medical College of Yangzhou University, Yangzhou, Jiangsu 225003, China
| | - Xiang Wang
- Medical Psychological Institute of the Second Xiangya Hospital, Central South University, 139 Renmin (M) Road, Changsha, Hunan 410011, China
| | - Xiaobin Zhang
- Department of Psychiatry, Affiliated WuTaiShan Hospital of Medical College of Yangzhou University, Yangzhou, Jiangsu 225003, China
| | - Hongwei Yi
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China.
| | - Xiangrong Zhang
- Department of Geriatric Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China.
| |
Collapse
|
28
|
Zebrafish models of epigenetic regulation of CNS functions. Brain Res Bull 2018; 142:344-351. [DOI: 10.1016/j.brainresbull.2018.08.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 08/22/2018] [Accepted: 08/30/2018] [Indexed: 12/12/2022]
|
29
|
Impact of antipsychotic treatment on methylation status of Interleukin-6 [IL-6] gene in Schizophrenia. J Psychiatr Res 2018; 104:88-95. [PMID: 30005373 DOI: 10.1016/j.jpsychires.2018.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 06/03/2018] [Accepted: 07/06/2018] [Indexed: 12/14/2022]
Abstract
Immunopathogenesis of schizophrenia has emerged as one of the predominant research paradigms in recent times. Based on the altered serum levels as well as gene expression, IL-6 has been considered as a peripheral biomarker of schizophrenia. However, the precise mechanism underlying the altered expression of IL6 in schizophrenia is inadequately known. Given the profound influence of environmental factors on schizophrenia risk, it is important to understand the effect of epigenetic changes on schizophrenia risk. Further, it is not known whether epigenetic changes modulate the expression of IL6 and its subsequent effects on the risk and progression of schizophrenia. In this study, we analysed and compared the methylation status of IL6 promoter sequence from -1200bp to +27bp in antipsychotic-naïve/free schizophrenia patients (N = 47) and matched healthy controls (N = 47) using bisulfite sequencing method. In addition, we also examined the methylation status in these patients at least after 3-months of treatment with antipsychotics (N = 40). At baseline, a state of hypomethylation was observed in the IL6 promoter of schizophrenia subjects in comparison to healthy controls. This state of hypomethylation was shown to be reversed by the administration of antipsychotics. In summary, our observations emphasize a significant role for IL-6 promoter methylation in schizophrenia pathogenesis as well as treatment with antipsychotic medications.
Collapse
|
30
|
de la Fuente Revenga M, Ibi D, Saunders JM, Cuddy T, Ijaz MK, Toneatti R, Kurita M, Holloway T, Shen L, Seto J, Dozmorov MG, González-Maeso J. HDAC2-dependent Antipsychotic-like Effects of Chronic Treatment with the HDAC Inhibitor SAHA in Mice. Neuroscience 2018; 388:102-117. [PMID: 30025863 DOI: 10.1016/j.neuroscience.2018.07.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/08/2018] [Accepted: 07/05/2018] [Indexed: 01/20/2023]
Abstract
Antipsychotic drugs, including both typical such as haloperidol and atypical such as clozapine, remain the current standard for schizophrenia treatment. These agents are relatively effective in treating hallucinations and delusions. However, cognitive deficits are at present essentially either persistent or exacerbated following chronic antipsychotic drug exposure. This underlines the need of new therapeutic approaches to improve cognition in treated schizophrenia patients. Our previous findings suggested that upregulation of histone deacetylase 2 (HDAC2) expression upon chronic antipsychotic treatment may lead to negative effects on cognition and cortical synaptic structure. Here we tested different phenotypes of psychosis, synaptic plasticity, cognition and antipsychotic drug action in HDAC2 conditional knockout (HDAC2-cKO) mice and controls. Conditional depletion of HDAC2 function in glutamatergic pyramidal neurons led to a protective phenotype against behavior models induced by psychedelic and dissociative drugs, such as DOI and MK801, respectively. Immunoreactivity toward synaptophysin, which labels presynaptic terminals of functional synapses, was decreased in the frontal cortex of control mice chronically treated with clozapine - an opposite effect occurred in HDAC2-cKO mice. Chronic treatment with the class I and class II HDAC inhibitor SAHA prevented via HDAC2 the disruptive effects of MK801 on recognition memory. Additionally, chronic SAHA treatment affected transcription of numerous plasticity-related genes in the frontal cortex of control mice, an effect that was not observed in HDAC2-cKO animals. Together, these findings suggest that HDAC2 may represent a novel target to improve synaptic plasticity and cognition in treated schizophrenia patients.
Collapse
Affiliation(s)
- Mario de la Fuente Revenga
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, United States
| | - Daisuke Ibi
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, United States; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Department of Chemical Pharmacology, Meijo University, Nagoya 468-8503, Japan
| | - Justin M Saunders
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, United States
| | - Travis Cuddy
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, United States
| | - Maryum K Ijaz
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, United States
| | - Rudy Toneatti
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, United States
| | - Mitsumasa Kurita
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Terrell Holloway
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Li Shen
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Jeremy Seto
- Department of Biological Sciences, New York City College of Technology, Brooklyn, NY 11201, United States
| | - Mikhail G Dozmorov
- Department of Biostatistics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, United States
| | - Javier González-Maeso
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, United States; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States.
| |
Collapse
|
31
|
Gibbons A, Udawela M, Dean B. Non-Coding RNA as Novel Players in the Pathophysiology of Schizophrenia. Noncoding RNA 2018; 4:E11. [PMID: 29657307 PMCID: PMC6027250 DOI: 10.3390/ncrna4020011] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 03/29/2018] [Accepted: 04/06/2018] [Indexed: 12/22/2022] Open
Abstract
Schizophrenia is associated with diverse changes in the brain's transcriptome and proteome. Underlying these changes is the complex dysregulation of gene expression and protein production that varies both spatially across brain regions and temporally with the progression of the illness. The growing body of literature showing changes in non-coding RNA in individuals with schizophrenia offers new insights into the mechanisms causing this dysregulation. A large number of studies have reported that the expression of microRNA (miRNA) is altered in the brains of individuals with schizophrenia. This evidence is complemented by findings that single nucleotide polymorphisms (SNPs) in miRNA host gene sequences can confer an increased risk of developing the disorder. Additionally, recent evidence suggests the expression of other non-coding RNAs, such as small nucleolar RNA and long non-coding RNA, may also be affected in schizophrenia. Understanding how these changes in non-coding RNAs contribute to the development and progression of schizophrenia offers potential avenues for the better treatment and diagnosis of the disorder. This review will focus on the evidence supporting the involvement of non-coding RNA in schizophrenia and its therapeutic potential.
Collapse
Affiliation(s)
- Andrew Gibbons
- The Florey Institute for Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC 3052, Australia.
- The Department of Psychiatry, the University of Melbourne, Parkville, Victoria, Australia.
| | - Madhara Udawela
- The Florey Institute for Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC 3052, Australia.
| | - Brian Dean
- The Florey Institute for Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC 3052, Australia.
- The Centre for Mental Health, Swinburne University of Technology, Hawthorn, Victoria, Australia.
| |
Collapse
|
32
|
Goldsmith DR, Crooks CL, Walker EF, Cotes RO. An Update on Promising Biomarkers in Schizophrenia. FOCUS: JOURNAL OF LIFE LONG LEARNING IN PSYCHIATRY 2018; 16:153-163. [PMID: 31975910 DOI: 10.1176/appi.focus.20170046] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Given the heterogeneity of symptoms in patients with schizophrenia and current treatment limitations, biomarkers may play an important role in diagnosis, subtype stratification, and the assessment of treatment response. Though many potential biomarkers have been studied, we have chosen to focus on some of the most promising and potentially clinically relevant biomarkers to review herein. These include markers of inflammation, neuroimaging biomarkers, brain-derived neurotrophic factor, genetic/epigenetic markers, and speech analysis. This will provide a broad overview of putative biomarkers that could become clinically relevant in the future, though none currently appear ready to assist the clinician in identifying cases of schizophrenia, subtypes of the disorder, treatment choice, or response. Nonetheless, some biomarkers, such as C-reactive protein (CRP), may be useful at identifying individuals who may be more highly inflamed, which could drive treatment choice. Though checking CRP is not a standard of practice, this is one example of how biomarkers may drive treatment decisions in the future, supporting precision medicine. Similarly, technological advances may one day allow clinicians to detect changes in speech patterns, which could represent a noninvasive, clinically useful tool in the future. We conclude the review by highlighting two important potential clinical uses for biomarkers in schizophrenia: the identification of individuals who may convert from clinical high risk and the stratification of patients via different biomarkers that may supersede clinical diagnosis. Given the enormous burden of illness of schizophrenia, the search for clinically relevant biomarkers is of great importance to improve the lives of patients with the disorder.
Collapse
Affiliation(s)
- David R Goldsmith
- Dr. Goldsmith, Dr. Crooks, and Dr. Cotes are with the Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia. Dr. Crooks is also with the Electronic Systems Laboratory, Georgia Tech Research Institute, Atlanta. Dr. Walker is with the Department of Psychology, Emory University
| | - Courtney L Crooks
- Dr. Goldsmith, Dr. Crooks, and Dr. Cotes are with the Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia. Dr. Crooks is also with the Electronic Systems Laboratory, Georgia Tech Research Institute, Atlanta. Dr. Walker is with the Department of Psychology, Emory University
| | - Elaine F Walker
- Dr. Goldsmith, Dr. Crooks, and Dr. Cotes are with the Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia. Dr. Crooks is also with the Electronic Systems Laboratory, Georgia Tech Research Institute, Atlanta. Dr. Walker is with the Department of Psychology, Emory University
| | - Robert O Cotes
- Dr. Goldsmith, Dr. Crooks, and Dr. Cotes are with the Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia. Dr. Crooks is also with the Electronic Systems Laboratory, Georgia Tech Research Institute, Atlanta. Dr. Walker is with the Department of Psychology, Emory University
| |
Collapse
|
33
|
Kular L, Kular S. Epigenetics applied to psychiatry: Clinical opportunities and future challenges. Psychiatry Clin Neurosci 2018; 72:195-211. [PMID: 29292553 DOI: 10.1111/pcn.12634] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/12/2017] [Accepted: 12/26/2017] [Indexed: 12/11/2022]
Abstract
Psychiatric disorders are clinically heterogeneous and debilitating chronic diseases resulting from a complex interplay between gene variants and environmental factors. Epigenetic processes, such as DNA methylation and histone posttranslational modifications, instruct the cell/tissue to correctly interpret external signals and adjust its functions accordingly. Given that epigenetic modifications are sensitive to environment, stable, and reversible, epigenetic studies in psychiatry could represent a promising approach to better understanding and treating disease. In the present review, we aim to discuss the clinical opportunities and challenges arising from the epigenetic research in psychiatry. Using selected examples, we first recapitulate key findings supporting the role of adverse life events, alone or in combination with genetic risk, in epigenetic programming of neuropsychiatric systems. Epigenetic studies further report encouraging findings about the use of methylation changes as diagnostic markers of disease phenotype and predictive tools of progression and response to treatment. Then we discuss the potential of using targeted epigenetic pharmacotherapy, combined with psychosocial interventions, for future personalized medicine for patients. Finally, we review the methodological limitations that could hinder interpretation of epigenetic data in psychiatry. They mainly arise from heterogeneity at the individual and tissue level and require future strategies in order to reinforce the biological relevance of epigenetic data and its translational use in psychiatry. Overall, we suggest that epigenetics could provide new insights into a more comprehensive interpretation of mental illness and might eventually improve the nosology, treatment, and prevention of psychiatric disorders.
Collapse
Affiliation(s)
- Lara Kular
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Sonia Kular
- Adult Psychiatry Unit of Laval Secteur Est, Laval, France
| |
Collapse
|
34
|
Deng JV, Orsini CA, Shimp KG, Setlow B. MeCP2 Expression in a Rat Model of Risky Decision Making. Neuroscience 2017; 369:212-221. [PMID: 29155278 DOI: 10.1016/j.neuroscience.2017.11.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 10/23/2017] [Accepted: 11/10/2017] [Indexed: 01/13/2023]
Abstract
Many neuropsychiatric disorders are associated with abnormal decision making involving risk of punishment, but the underlying molecular basis remains poorly understood. Methyl CpG-binding protein 2 (MeCP2) is an epigenetic factor that regulates transcription by directly binding to methylated DNA. Here, we evaluated MeCP2 expression in the context of risk-taking behaviors using the Risky Decision-making Task (RDT), in which rats make discrete choices between a small "safe" food reward and a large "risky" food reward accompanied by varying probabilities of punishment. In Experiment 1, expression of MeCP2 as assessed by immunoblotting in the medial prefrontal cortex (mPFC), but not the striatum, was inversely correlated with the degree of preference for the large, risky reward (risk taking) seven days after the last RDT test. In Experiment 2, MeCP2 expression 90 min after RDT testing, assessed using immunohistochemistry, was suppressed in both the dorsal mPFC (dmPFC) and nucleus accumbens compared to home cage controls, indicating that MeCP2 expression is modulated by RDT performance. Additional experiments revealed that RDT performance increased expression of MeCP2 phosphorylated at Ser421 (associated with neuronal activity and activation of gene expression) in dmPFC principal neurons. Finally, as in Experiment 1, lower expression of MeCP2 in the ventral mPFC was associated with greater risk taking under baseline conditions. Together, these findings indicate a complex regulatory role of MeCP2 in risky decision making, and suggest that epigenetic factors may be an important component of the molecular mechanisms underlying such decision-making processes.
Collapse
Affiliation(s)
- Jay V Deng
- Department of Psychiatry, University of Florida College of Medicine, United States; Center for Addiction Research and Education, University of Florida, United States.
| | - Caitlin A Orsini
- Department of Psychiatry, University of Florida College of Medicine, United States
| | - Kristy G Shimp
- Department of Neuroscience, University of Florida College of Medicine, United States
| | - Barry Setlow
- Department of Psychiatry, University of Florida College of Medicine, United States; Department of Neuroscience, University of Florida College of Medicine, United States; Department of Psychology, University of Florida, United States; Center for Addiction Research and Education, University of Florida, United States.
| |
Collapse
|
35
|
Paparelli A, Iwata K, Wakuda T, Iyegbe C, Murray RM, Takei N. Perinatal Asphyxia in Rat Alters Expression of Novel Schizophrenia Risk Genes. Front Mol Neurosci 2017; 10:341. [PMID: 29163023 PMCID: PMC5663725 DOI: 10.3389/fnmol.2017.00341] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 10/06/2017] [Indexed: 11/17/2022] Open
Abstract
Epidemiological studies suggest that obstetric complications, particularly those related to hypoxia during labor and delivery, are a risk factor for development of schizophrenia. The impact of perinatal asphyxia on postnatal life has been studied in a rodent model of global hypoxia, which is accompanied by cesarean section birth. This asphyxia model shows several behavioral, pharmacological, neurochemical, and neuroanatomical abnormalities in adulthood that have relevance to schizophrenia. Further, it is suggested that schizophrenia has a strong genetic component, and indeed novel candidate genes were recently identified by a genome-wide association study. Here, we examined alteration in the novel schizophrenia risk genes, CNNM2, CSMD1, and MMP16 in the brains of rats undergoing cesarean section with or without global hypoxia. The brain regions studied were the prefrontal cortex, striatum, and hippocampus, which are all relevant to schizophrenia. Risk gene expression was measured at three time periods: neonatal, adolescence, and adulthood. We also performed an in vitro analysis to determine involvement of these genes in CNS maturation during differentiation of human neuronal and glial cell lines. Cnnm2 expression was altered in the brains of asphyxia model rats. However, Csmd1 and Mmp16 showed altered expression by exposure to cesarean section only. These findings suggest that altered expression of these risk genes via asphyxia and cesarean section may be associated, albeit through distinct pathways, with the pathobiology of schizophrenia.
Collapse
Affiliation(s)
- Alessandra Paparelli
- Department of Psychosis Studies, Institute of Psychiatry, King's College London, London, United Kingdom
| | - Keiko Iwata
- Research Center for Child Mental Development, University of Fukui, Fukui, Japan.,Department of Biology, University of Padova, Padova, Italy
| | - Tomoyasu Wakuda
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Conrad Iyegbe
- Department of Psychosis Studies, Institute of Psychiatry, King's College London, London, United Kingdom
| | - Robin M Murray
- Department of Psychosis Studies, Institute of Psychiatry, King's College London, London, United Kingdom
| | - Nori Takei
- Department of Psychosis Studies, Institute of Psychiatry, King's College London, London, United Kingdom.,Division of Neuropsychological Development and Health Sciences, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Hamamatsu, Japan.,Research Center for Child Mental Development, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
36
|
Saia-Cereda VM, Santana AG, Schmitt A, Falkai P, Martins-de-Souza D. The Nuclear Proteome of White and Gray Matter from Schizophrenia Postmortem Brains. MOLECULAR NEUROPSYCHIATRY 2017; 3:37-52. [PMID: 28879200 PMCID: PMC5582429 DOI: 10.1159/000477299] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 05/03/2017] [Indexed: 12/14/2022]
Abstract
Schizophrenia (SCZ) is a serious neuropsychiatric disorder that manifests through several symptoms from early adulthood. Numerous studies over the last decades have led to significant advances in increasing our understanding of the factors involved in SCZ. For example, mass spectrometry-based proteomic analysis has provided important insights by uncovering protein dysfunctions inherent to SCZ. Here, we present a comprehensive analysis of the nuclear proteome of postmortem brain tissues from corpus callosum (CC) and anterior temporal lobe (ATL). We show an overview of the role of deregulated nuclear proteins in these two main regions of the brain: the first, mostly composed of glial cells and axons of neurons, and the second, represented mainly by neuronal cell bodies. These samples were collected from SCZ patients in an attempt to characterize the role of the nucleus in the disease process. With the ATL nucleus enrichment, we found 224 proteins present at different levels, and 76 of these were nuclear proteins. In the CC analysis, we identified 119 present at different levels, and 24 of these were nuclear proteins. The differentially expressed nuclear proteins of ATL are mainly associated with the spliceosome, whereas those of the CC region are associated with calcium/calmodulin signaling.
Collapse
Affiliation(s)
- Verônica M. Saia-Cereda
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Aline G. Santana
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Andrea Schmitt
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University (LMU), Munich, Germany
- Laboratory of Neurosciences (LIM-27), Institute of Psychiatry, University of São Paulo, São Paulo, Brazil
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University (LMU), Munich, Germany
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
- UNICAMP's Neurobiology Center, Campinas, Brazil
| |
Collapse
|
37
|
Differences in 5-HT2A and mGlu2 Receptor Expression Levels and Repressive Epigenetic Modifications at the 5-HT2A Promoter Region in the Roman Low- (RLA-I) and High- (RHA-I) Avoidance Rat Strains. Mol Neurobiol 2017; 55:1998-2012. [PMID: 28265857 DOI: 10.1007/s12035-017-0457-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 02/13/2017] [Indexed: 01/12/2023]
Abstract
The serotonin 2A (5-HT2A) and metabotropic glutamate 2 (mGlu2) receptors regulate each other and are associated with schizophrenia. The Roman high- (RHA-I) and the Roman low- (RLA-I) avoidance rat strains present well-differentiated behavioral profiles, with the RHA-I strain emerging as a putative genetic rat model of schizophrenia-related features. The RHA-I strain shows increased 5-HT2A and decreased mGlu2 receptor binding levels in prefrontal cortex (PFC). Here, we looked for differences in gene expression and transcriptional regulation of these receptors. The striatum (STR) was included in the analysis. 5-HT2A, 5-HT1A, and mGlu2 mRNA and [3H]ketanserin binding levels were measured in brain homogenates. As expected, 5-HT2A binding was significantly increased in PFC in the RHA-I rats, while no difference in binding was observed in STR. Surprisingly, 5-HT2A gene expression was unchanged in PFC but significantly decreased in STR. mGlu2 receptor gene expression was significantly decreased in both PFC and STR. No differences were observed for the 5-HT1A receptor. Chromatin immunoprecipitation assay revealed increased trimethylation of histone 3 at lysine 27 (H3K27me3) at the promoter region of the HTR2A gene in the STR. We further looked at the Akt/GSK3 signaling pathway, a downstream point of convergence of the serotonin and glutamate system, and found increased phosphorylation levels of GSK3β at tyrosine 216 and increased β-catenin levels in the PFC of the RHA-I rats. These results reveal region-specific regulation of the 5-HT2A receptor in the RHA-I rats probably due to absence of mGlu2 receptor that may result in differential regulation of downstream pathways.
Collapse
|
38
|
Käkelä J, Marttila R, Keskinen E, Veijola J, Isohanni M, Koivumaa-Honkanen H, Haapea M, Jääskeläinen E, Miettunen J. Association between family history of psychiatric disorders and long-term outcome in schizophrenia - The Northern Finland Birth Cohort 1966 study. Psychiatry Res 2017; 249:16-22. [PMID: 28063393 DOI: 10.1016/j.psychres.2016.12.040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 10/05/2016] [Accepted: 12/26/2016] [Indexed: 01/13/2023]
Abstract
Family history of psychiatric disorders has been associated with impaired outcome in schizophrenia, but very few studies have investigated its long-term social and occupational outcome. We investigated the association of family history of psychiatric disorders, especially psychosis, with long-term social, occupational, clinical and global outcome in schizophrenia. The study sample comprises of the Northern Finland Birth Cohort 1966. Cohort members with psychosis were detected by Finnish national registers. Altogether 69 individuals with schizophrenia spectrum diagnosis participated, mean age 43, after on average 17 years since onset of illness. The information regarding family history of psychiatric disorders were gathered from registers and interviews. A Strauss-Carpenter Outcome Scale, PANSS and SOFAS were conducted to assess the outcome. Results showed that the family history of any psychiatric disorder was associated with more severe positive and emotional symptoms in PANSS. The family history of psychosis was not associated with outcomes. These findings suggest that family history of psychiatric disorders has a small association with outcome in schizophrenia. Despite family history of psychosis being a strong risk factor for schizophrenia, after years of illness it does not seem to affect outcome.
Collapse
Affiliation(s)
- Juha Käkelä
- Research Unit of Clinical Neuroscience, Department of Psychiatry, University of Oulu, Oulu, Finland; Center for Life Course Health Research, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.
| | - Riikka Marttila
- Center for Life Course Health Research, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Emmi Keskinen
- Research Unit of Clinical Neuroscience, Department of Psychiatry, University of Oulu, Oulu, Finland; Center for Life Course Health Research, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Juha Veijola
- Research Unit of Clinical Neuroscience, Department of Psychiatry, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland; Department of Psychiatry, Oulu University Hospital, Oulu, Finland
| | - Matti Isohanni
- Research Unit of Clinical Neuroscience, Department of Psychiatry, University of Oulu, Oulu, Finland; Department of Psychiatry, Oulu University Hospital, Oulu, Finland
| | - Heli Koivumaa-Honkanen
- Institute of Clinical Medicine, Psychiatry, University of Eastern Finland, Kuopio, Finland; Department of Psychiatry, Kuopio University Hospital, Kuopio, Finland; South-Savonia Hospital District, Mikkeli, Finland; North Karelia Central Hospital, Joensuu, Finland; SOSTERI, Savonlinna, Finland; SOTE, Iisalmi, Finland; Lapland Hospital District, Rovaniemi, Finland
| | - Marianne Haapea
- Center for Life Course Health Research, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Erika Jääskeläinen
- Research Unit of Clinical Neuroscience, Department of Psychiatry, University of Oulu, Oulu, Finland; Center for Life Course Health Research, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland; Department of Psychiatry, Oulu University Hospital, Oulu, Finland; Oulu Occupational Health, Oulu, Finland
| | - Jouko Miettunen
- Research Unit of Clinical Neuroscience, Department of Psychiatry, University of Oulu, Oulu, Finland; Center for Life Course Health Research, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland; Department of Psychiatry, Oulu University Hospital, Oulu, Finland
| |
Collapse
|
39
|
Alfimova MV, Kondratiev NV, Golimbet VE. Results and promises of genetics of cognitive impairment in schizophrenia: epigenetic approaches. Zh Nevrol Psikhiatr Im S S Korsakova 2017. [DOI: 10.17116/jnevro201711721130-135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
40
|
Lavratti C, Dorneles G, Pochmann D, Peres A, Bard A, de Lima Schipper L, Dal Lago P, Wagner LC, Elsner VR. Exercise-induced modulation of histone H4 acetylation status and cytokines levels in patients with schizophrenia. Physiol Behav 2016; 168:84-90. [PMID: 27810494 DOI: 10.1016/j.physbeh.2016.10.021] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 10/07/2016] [Accepted: 10/27/2016] [Indexed: 12/17/2022]
Abstract
The present study aimed to investigate the short and long-term effects of a concurrent exercise protocol on global histone H4 acetylation levels and inflammatory markers (interleukin-4 (IL-4), interleukin-6 (IL-6), interferon gamma (IFN-γ) and cortisol) in phytohemagglutinin-stimulated peripheral blood mononuclear cells (PBMC) and in peripheral blood of patients with schizophrenia (SZ), as well the intervention impact on anthropometric characteristics. Seventeen individuals were submitted to the intervention three times a week and blood samples were collected pre, 30, 60 and 90days after the intervention started. A remarkable reduction on body mass index and body mass were observed following intervention. The protocol also induced a histone H4 hypoacetylation status in PBMC all times evaluated when compared to the pre intervention period. Although the IL-4 and cortisol levels were not altered in response to the intervention, a reduction in IL-6 production during the 60 and 90days compared to the pre intervention period was observed. Finally, diminished IFN-γ production was found in the 90days period compared to the pre intervention and 30days after periods. In addition, systemic IL-6 levels were lower at 60 and 90days compared to the pre intervention. The concurrent exercise protocol was able to improve anthropometric characteristics in patients with SZ, engaging the modulation of cytokine and histone H4 acetylation levels.
Collapse
Affiliation(s)
- Caroline Lavratti
- Programa de Pós Graduação em Biociências e Reabilitação do Centro Universitário Metodista-IPA, Porto Alegre, RS, Brazil
| | - Gilson Dorneles
- Cellular and Molecular Immunology Lab., Department of Health Basic Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
| | - Daniela Pochmann
- Programa de Pós Graduação em Biociências e Reabilitação do Centro Universitário Metodista-IPA, Porto Alegre, RS, Brazil
| | - Alessandra Peres
- Programa de Pós Graduação em Biociências e Reabilitação do Centro Universitário Metodista-IPA, Porto Alegre, RS, Brazil; Cellular and Molecular Immunology Lab., Department of Health Basic Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
| | - Andréia Bard
- Curso de Fisioterapia do Centro Universitário Metodista-IPA, Porto Alegre, RS, Brazil
| | | | - Pedro Dal Lago
- Programa de Pós Graduação em Ciências da Reabilitação, Universidade Federal de Ciências da Saúde de Porto Alegre, RS, Brazil
| | - Luciane Carniel Wagner
- Programa de Pós Graduação em Biociências e Reabilitação do Centro Universitário Metodista-IPA, Porto Alegre, RS, Brazil
| | - Viviane Rostirola Elsner
- Programa de Pós Graduação em Biociências e Reabilitação do Centro Universitário Metodista-IPA, Porto Alegre, RS, Brazil.
| |
Collapse
|
41
|
Bakulski KM, Halladay A, Hu VW, Mill J, Fallin MD. Epigenetic Research in Neuropsychiatric Disorders: the "Tissue Issue". Curr Behav Neurosci Rep 2016; 3:264-274. [PMID: 28093577 PMCID: PMC5235359 DOI: 10.1007/s40473-016-0083-4] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
PURPOSE OF REVIEW Evidence has linked neuropsychiatric disorders with epigenetic marks as either a biomarker of disease, biomarker of exposure, or mechanism of disease processes. Neuropsychiatric epidemiologic studies using either target brain tissue or surrogate blood tissue each have methodological challenges and distinct advantages. RECENT FINDINGS Brain tissue studies are challenged by small sample sizes of cases and controls, incomplete phenotyping, post-mortem timing, and cellular heterogeneity, but the use of a primary disease relevant tissue is critical. Blood-based studies have access to much larger sample sizes and more replication opportunities, as well as the potential for longitudinal measurements, both prior to onset and during the course of treatments. Yet, blood studies also are challenged by cell-type heterogeneity, and many question the validity of using peripheral tissues as a brain biomarker. Emerging evidence suggests that these limitations to blood-based epigenetic studies are surmountable, but confirmation in target tissue remains important. SUMMARY Epigenetic mechanisms have the potential to help elucidate biology connecting experiential risk factors with neuropsychiatric disease manifestation. Cross-tissue studies as well as advanced epidemiologic methods should be employed to more effectively conduct neuropsychiatric epigenetic research.
Collapse
Affiliation(s)
- Kelly M Bakulski
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Alycia Halladay
- Autism Science Foundation, New York City, New York, USA; Department of Pharmacology and Toxicology, Rutgers University, New Brunswick, New Jersey, USA
| | - Valerie W Hu
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC, USA
| | - Jonathan Mill
- University of Exeter Medical School, University of Exeter, Exeter, UK; Institute for Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - M Daniele Fallin
- Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA; Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
42
|
Cariaga-Martinez A, Saiz-Ruiz J, Alelú-Paz R. From Linkage Studies to Epigenetics: What We Know and What We Need to Know in the Neurobiology of Schizophrenia. Front Neurosci 2016; 10:202. [PMID: 27242407 PMCID: PMC4862989 DOI: 10.3389/fnins.2016.00202] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 04/25/2016] [Indexed: 01/15/2023] Open
Abstract
Schizophrenia is a complex psychiatric disorder characterized by the presence of positive, negative, and cognitive symptoms that lacks a unifying neuropathology. In the present paper, we will review the current understanding of molecular dysregulation in schizophrenia, including genetic and epigenetic studies. In relation to the latter, basic research suggests that normal cognition is regulated by epigenetic mechanisms and its dysfunction occurs upon epigenetic misregulation, providing new insights into missing heritability of complex psychiatric diseases, referring to the discrepancy between epidemiological heritability and the proportion of phenotypic variation explained by DNA sequence difference. In schizophrenia the absence of consistently replicated genetic effects together with evidence for lasting changes in gene expression after environmental exposures suggest a role of epigenetic mechanisms. In this review we will focus on epigenetic modifications as a key mechanism through which environmental factors interact with individual's genetic constitution to affect risk of psychotic conditions throughout life.
Collapse
Affiliation(s)
- Ariel Cariaga-Martinez
- Laboratory for Neuroscience of Mental Disorders Elena Pessino, Department of Medicine and Medical Specialties, School of Medicine, Alcalá University Madrid, Spain
| | - Jerónimo Saiz-Ruiz
- Department of Psychiatry, Ramón y Cajal Hospital, IRYCISMadrid, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM)Madrid, Spain
| | - Raúl Alelú-Paz
- Laboratory for Neuroscience of Mental Disorders Elena Pessino, Department of Medicine and Medical Specialties, School of Medicine, Alcalá UniversityMadrid, Spain; Department of Psychiatry, Ramón y Cajal Hospital, IRYCISMadrid, Spain
| |
Collapse
|
43
|
Flores G, Morales-Medina JC, Diaz A. Neuronal and brain morphological changes in animal models of schizophrenia. Behav Brain Res 2016; 301:190-203. [DOI: 10.1016/j.bbr.2015.12.034] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 12/15/2015] [Accepted: 12/18/2015] [Indexed: 12/14/2022]
|
44
|
Abstract
Over the past 60 years, a large number of selective neurotoxins were discovered and developed, making it possible to animal-model a broad range of human neuropsychiatric and neurodevelopmental disorders. In this paper, we highlight those neurotoxins that are most commonly used as neuroteratologic agents, to either produce lifelong destruction of neurons of a particular phenotype, or a group of neurons linked by a specific class of transporter proteins (i.e., dopamine transporter) or body of receptors for a specific neurotransmitter (i.e., NMDA class of glutamate receptors). Actions of a range of neurotoxins are described: 6-hydroxydopamine (6-OHDA), 6-hydroxydopa, DSP-4, MPTP, methamphetamine, IgG-saporin, domoate, NMDA receptor antagonists, and valproate. Their neuroteratologic features are outlined, as well as those of nerve growth factor, epidermal growth factor, and that of stress. The value of each of these neurotoxins in animal modeling of human neurologic, neurodegenerative, and neuropsychiatric disorders is discussed in terms of the respective value as well as limitations of the derived animal model. Neuroteratologic agents have proven to be of immense importance for understanding how associated neural systems in human neural disorders may be better targeted by new therapeutic agents.
Collapse
Affiliation(s)
- Trevor Archer
- Department of Psychology, University of Gothenburg, Box 500, 430 50, Gothenburg, Sweden.
| | - Richard M Kostrzewa
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, PO Box 70577, Johnson City, TN, 37614, USA
| |
Collapse
|
45
|
Hu X, Fan Q, Hou H, Yan R. Neurological dysfunctions associated with altered BACE1-dependent Neuregulin-1 signaling. J Neurochem 2016; 136:234-49. [PMID: 26465092 PMCID: PMC4833723 DOI: 10.1111/jnc.13395] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 09/23/2015] [Accepted: 09/25/2015] [Indexed: 01/09/2023]
Abstract
Inhibition of BACE1 is being pursued as a therapeutic target to treat patients suffering from Alzheimer's disease because BACE1 is the sole β-secretase that generates β-amyloid peptide. Knowledge regarding other cellular functions of BACE1 is therefore critical for the safe use of BACE1 inhibitors in human patients. Neuregulin-1 (Nrg1) is a BACE1 substrate and BACE1 cleavage of Nrg1 is critical for signaling functions in myelination, remyelination, synaptic plasticity, normal psychiatric behaviors, and maintenance of muscle spindles. This review summarizes the most recent discoveries associated with BACE1-dependent Nrg1 signaling in these areas. This body of knowledge will help to provide guidance for preventing unwanted Nrg1-based side effects following BACE1 inhibition in humans. To initiate its signaling cascade, membrane anchored Neuregulin (Nrg), mainly type I and III β1 Nrg1 isoforms and Nrg3, requires ectodomain shedding. BACE1 is one of such indispensable sheddases to release the functional Nrg signaling fragment. The dependence of Nrg on the cleavage by BACE1 is best manifested by disrupting the critical role of Nrg in the control of axonal myelination, schizophrenic behaviors as well as the formation and maintenance of muscle spindles.
Collapse
Affiliation(s)
- Xiangyou Hu
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Qingyuan Fan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Hailong Hou
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Riqiang Yan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| |
Collapse
|
46
|
Ayhan Y, McFarland R, Pletnikov MV. Animal models of gene-environment interaction in schizophrenia: A dimensional perspective. Prog Neurobiol 2015; 136:1-27. [PMID: 26510407 DOI: 10.1016/j.pneurobio.2015.10.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 09/07/2015] [Accepted: 10/22/2015] [Indexed: 12/12/2022]
Abstract
Schizophrenia has long been considered as a disorder with multifactorial origins. Recent discoveries have advanced our understanding of the genetic architecture of the disease. However, even with the increase of identified risk variants, heritability estimates suggest an important contribution of non-genetic factors. Various environmental risk factors have been proposed to play a role in the etiopathogenesis of schizophrenia. These include season of birth, maternal infections, obstetric complications, adverse events at early childhood, and drug abuse. Despite the progress in identification of genetic and environmental risk factors, we still have a limited understanding of the mechanisms whereby gene-environment interactions (G × E) operate in schizophrenia and psychoses at large. In this review we provide a critical analysis of current animal models of G × E relevant to psychotic disorders and propose that dimensional perspective will advance our understanding of the complex mechanisms of these disorders.
Collapse
Affiliation(s)
- Yavuz Ayhan
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, USA; Hacettepe University Faculty of Medicine, Department of Psychiatry, Turkey
| | - Ross McFarland
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, USA; Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, USA
| | - Mikhail V Pletnikov
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, USA; Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, USA; Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, USA; Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, USA.
| |
Collapse
|