1
|
Harter AM, Kim C, Yamazaki A, Lee L, Ji MT, Nemesh M, Redei EE. Stress enhances aggression in male rats with genetic stress hyper-reactivity. GENES, BRAIN, AND BEHAVIOR 2024; 23:e70005. [PMID: 39422001 PMCID: PMC11487273 DOI: 10.1111/gbb.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024]
Abstract
The current study investigated stress-induced aggressive behavior in the resident-intruder test in males of the genetically stress hyper-reactive Wistar Kyoto More Immobile (WMI), and the nearly isogenic, control Wistar Kyoto Less Immobile (WLI) strains. Tests were carried out against same-age intruders during adolescence, and same-age and juvenile intruders in adulthood. In adolescence and adulthood, prior acute restraint stress decreased social interactions and decreased aggressive behaviors of adolescents and adult WLIs. However, prior stress precipitated aggression in the adult WMI males toward both same-age, and juvenile intruders compared with control WMIs and WLIs. Trunk blood levels of testosterone and androstenedione increased in stressed WLIs, but not in WMIs, suggesting no direct role of androgens in the increased aggression of WMIs. Expressions of aggression-relevant genes showed patterns commensurate with being causative in aggressive behavior. The methyl-CpG binding protein 2 was lower in the frontal cortex of control WMIs, and in the amygdala of stressed WMIs compared with their respective WLIs. Frontal cortex expression of vasopressin receptor 1a and serotonin transporter increased, solely in WMI males after stress. As behaviors were the same toward same-age and non-threatening juvenile intruders, the stress-induced increase in confrontational behavior of the adult WMI male was not because of enhanced fear or anxiety. These results suggest that genetic stress hyper-reactivity is a risk factor for stress-induced increases in aggression in males. Additionally, as known aggression-related genes showed expression patterns paralleling aggressive behavior, this model system could identify novel molecular pathways leading to stress-enhanced aggression.
Collapse
Affiliation(s)
- Aspen M. Harter
- Department of Psychiatry and Behavioral Sciences, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Chris Kim
- Department of Psychiatry and Behavioral Sciences, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Anna Yamazaki
- Department of Psychiatry and Behavioral Sciences, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Luca Lee
- Department of Psychiatry and Behavioral Sciences, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Michelle T. Ji
- Department of Psychiatry and Behavioral Sciences, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Mariya Nemesh
- Department of Psychiatry and Behavioral Sciences, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Eva E. Redei
- Department of Psychiatry and Behavioral Sciences, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| |
Collapse
|
2
|
Medeiros D, Ayala-Baylon K, Egido-Betancourt H, Miller E, Chapleau C, Robinson H, Phillips ML, Yang T, Longo FM, Li W, Pozzo-Miller L. A small-molecule TrkB ligand improves dendritic spine phenotypes and atypical behaviors in female Rett syndrome mice. Dis Model Mech 2024; 17:dmm050612. [PMID: 38785269 PMCID: PMC11139040 DOI: 10.1242/dmm.050612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 03/06/2024] [Indexed: 05/25/2024] Open
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder caused by mutations in MECP2, which encodes methyl-CpG-binding protein 2, a transcriptional regulator of many genes, including brain-derived neurotrophic factor (BDNF). BDNF levels are lower in multiple brain regions of Mecp2-deficient mice, and experimentally increasing BDNF levels improve atypical phenotypes in Mecp2 mutant mice. Due to the low blood-brain barrier permeability of BDNF itself, we tested the effects of LM22A-4, a brain-penetrant, small-molecule ligand of the BDNF receptor TrkB (encoded by Ntrk2), on dendritic spine density and form in hippocampal pyramidal neurons and on behavioral phenotypes in female Mecp2 heterozygous (HET) mice. A 4-week systemic treatment of Mecp2 HET mice with LM22A-4 restored spine volume in MeCP2-expressing neurons to wild-type (WT) levels, whereas spine volume in MeCP2-lacking neurons remained comparable to that in neurons from female WT mice. Female Mecp2 HET mice engaged in aggressive behaviors more than WT mice, the levels of which were reduced to WT levels by the 4-week LM22A-4 treatment. These data provide additional support to the potential usefulness of novel therapies not only for RTT but also to other BDNF-related disorders.
Collapse
Affiliation(s)
- Destynie Medeiros
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Karen Ayala-Baylon
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Hailey Egido-Betancourt
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Eric Miller
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Christopher Chapleau
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Holly Robinson
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Mary L. Phillips
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Tao Yang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Frank M. Longo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Wei Li
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lucas Pozzo-Miller
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
3
|
Medeiros D, Ayala-Baylon K, Egido-Betancourt H, Miller E, Chapleau CA, Robinson HA, Phillips ML, Yang T, Longo F, Li W, Pozzo-Miller L. A small-molecule TrkB ligand improves dendritic spine phenotypes and atypical behaviors in female Rett syndrome mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.09.566435. [PMID: 37986936 PMCID: PMC10659425 DOI: 10.1101/2023.11.09.566435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder caused by mutations in methyl-CpG-binding protein-2 (MECP2), encoding a transcriptional regulator of many genes, including brain-derived neurotrophic factor (Bdnf). BDNF mRNA and protein levels are lower in RTT autopsy brains and in multiple brain regions of Mecp2-deficient mice, and experimentally increasing BDNF levels improve atypical phenotypes in Mecp2 mutant mice. Due to the low blood-brain barrier permeability of BDNF itself, we tested the effects of a brain penetrant, small molecule ligand of its TrkB receptors. Applied in vitro, LM22A-4 increased dendritic spine density in pyramidal neurons in cultured hippocampal slices from postnatal day (P) 7 male Mecp2 knockout (KO) mice as much as recombinant BDNF, and both effects were prevented by the TrkB receptor inhibitors K-252a and ANA-12. Consistent with its partial agonist activity, LM22A-4 did not affect spine density in CA1 pyramidal neurons in slice cultures from male wildtype (WT) mice, where typical BDNF levels outcompete its binding to TrkB. To identify neurons of known genotypes in the "mosaic" brain of female Mecp2 heterozygous (HET) mice, we treated 4-6-month-old female MeCP2-GFP WT and HET mice with peripheral injections of LM22A-4 for 4 weeks. Surprisingly, mutant neurons lacking MeCP2-GFP showed dendritic spine volumes comparable to that in WT controls, while MeCP2-GFP-expressing neurons showed larger spines, similar to the phenotype we described in symptomatic male Mecp2 KO mice where all neurons lack MeCP2. Consistent with this non-cell-autonomous mechanism, a 4-week systemic treatment with LM22A-4 had an effect only in MeCP2-GFP-expressing neurons in female Mecp2 HET mice, bringing dendritic spine volumes down to WT control levels, and without affecting spines of MeCP2-GFP-lacking neurons. At the behavioral level, we found that female Mecp2 HET mice engaged in aggressive behaviors significantly more than WT controls, which were reduced to WT levels by a 4-week systemic treatment with LM22A-4. Altogether, these data revealed differences in dendritic spine size and altered behaviors in Mecp2 HET mice, while providing support to the potential usefulness of BDNF-related therapeutic approaches such as the partial TrkB agonist LM22A-4.
Collapse
|
4
|
He XB, Guo F, Li K, Yan J, Lee SH. Timing of MeCP2 Expression Determines Midbrain Dopamine Neuron Phenotype Specification. Stem Cells 2022; 40:1043-1055. [PMID: 36041430 DOI: 10.1093/stmcls/sxac061] [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: 05/08/2022] [Accepted: 08/22/2022] [Indexed: 11/14/2022]
Abstract
Midbrain dopamine (DA) neurons are associated with locomotor and psychiatric disorders. DA phenotype is specified in ancestral neural precursor cells (NPCs) and maintained throughout neuronal differentiation. Here we show that endogenous expression of MeCP2 coincides with DA phenotype specification in mouse mesencephalon, and premature expression of MeCP2 prevents in vitro cultured NPCs from acquiring DA phenotype through interfering NURR1 transactivation of DA phenotype genes. By contrast, ectopic MeCP2 expression does not disturb DA phenotype in the DA neurons. By analyzing the dynamic change of DNA methylation along DA neuronal differentiation at the promoter of DA phenotype gene tyrosine hydroxylase (Th), we show that Th expression is determined by TET1-mediated de-methylation of NURR1 binding sites within Th promoter. Chromatin immunoprecipitation assays demonstrate that premature MeCP2 dominates the DNA binding of the corresponding sites thereby blocking TET1 function in DA NPCs, whereas TET1-mediated de-methylation prevents excessive MeCP2 binding in DA neurons. The significance of temporal DNA methylation status is further confirmed by targeted methylation/demethylation experiments showing that targeted de-methylation in DA NPCs protects DA phenotype specification from ectopic MeCP2 expression, whereas targeted methylation disturbs phenotype maintenance in MeCP2-overexpressed DA neurons. These findings suggest the appropriate timing of MeCP2 expression as a novel determining factor for guiding NPCs into DA lineage.
Collapse
Affiliation(s)
- Xi-Biao He
- Laboratory of Stem Cell Biology and Epigenetics, College of Basic Medical Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, People's Republic of China
| | - Fang Guo
- Laboratory of Stem Cell Biology and Epigenetics, College of Basic Medical Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, People's Republic of China
| | - Kexuan Li
- Laboratory of Stem Cell Biology and Epigenetics, College of Basic Medical Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, People's Republic of China
| | - Jiaqing Yan
- Laboratory of Stem Cell Biology and Epigenetics, College of Basic Medical Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, People's Republic of China
| | - Sang-Hun Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
5
|
Tayel SI, Muharram NM, Fotoh DS, Elbarbary HS, Abd-Elhafiz HI, El-Masry EA, Taha AE, Soliman SE. Prognostic Impact of Genetic Variants of MECP2 and TIRAP on Clinical Outcomes of Systemic Lupus Erythematosus with and without Nephritis. Biomolecules 2021; 11:1378. [PMID: 34572591 PMCID: PMC8466489 DOI: 10.3390/biom11091378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/11/2021] [Accepted: 09/12/2021] [Indexed: 12/16/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune illness with a growing prevalence in many populations. Few studies have examined genetic predisposition to SLE, so we aimed to examine the clinical impact of the genetic polymorphisms MECP2 rs2734647and TIRAP rs8177374 on the outcomes and therapeutic precision of SLE with and without nephritis. This study included 110 SLE patients-divided into 63 with lupus nephritis (LN), and 47 without nephritis-and 100 controls. Laboratory measurements including CRP, ESR, ACR, CBC, anti-ds-DNA, vitamin A, C3, and C4 were carried out, along with genotyping of MECP2 rs2734647and TIRAP rs8177374 by real-time PCR and sequencing. Treg %, vitamin A, C3, and C4 were lower, whereas Th17 % was higher, in patients vs. controls (p < 0.001). The T allele of MECP2 rs2734647 was higher in LN than in non-nephritis and control subjects. Moreover, the T allele of TIRAP rs8177374 was higher in LN than in non-nephritis and control subjects. The MECP2 and TIRAP genes could play a role in predisposition to SLE, and can also predict disease progress to nephritis, helping to personalize medicine.
Collapse
Affiliation(s)
- Safaa I. Tayel
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Menoufia University, Shebin el Kom 32511, Egypt; (N.M.M.); (S.E.S.)
- Medical Biochemistry Unit, College of Medicine, Al Baha University, Al Baha 65779, Saudi Arabia
| | - Nashwa M. Muharram
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Menoufia University, Shebin el Kom 32511, Egypt; (N.M.M.); (S.E.S.)
- Medical Biochemistry Unit, College of Medicine, Al Baha University, Al Baha 65779, Saudi Arabia
| | - Dina S. Fotoh
- Physical Medicine, Rheumatology and Rehabilitation Department, Faculty of Medicine, Menoufia University, Shebin el Kom 32511, Egypt;
| | - Hany S. Elbarbary
- Renal Unit, Department of Internal Medicine, Faculty of Medicine, Menoufia University, Shebin el Kom 32511, Egypt;
- Renal Unit, Department of Internal Medicine, College of Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Huda I. Abd-Elhafiz
- Clinical Pharmacology Department, Faculty of Medicine, Menoufia University, Shebin el Kom 32511, Egypt;
| | - Eman A. El-Masry
- Microbiology and Immunology Unit, Department of Pathology, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia; (E.A.E.-M.); (A.E.T.)
- Medical Microbiology and Immunology Department, Faculty of Medicine, Menoufia University, Shebin el Kom 32511, Egypt
| | - Ahmed E. Taha
- Microbiology and Immunology Unit, Department of Pathology, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia; (E.A.E.-M.); (A.E.T.)
- Medical Microbiology and Immunology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Shimaa E. Soliman
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Menoufia University, Shebin el Kom 32511, Egypt; (N.M.M.); (S.E.S.)
- Medical Biochemistry Unit, Department of Pathology, College of Medicine, Qassim University, Buraydah 51452, Saudi Arabia
| |
Collapse
|
6
|
Rodrigues DC, Mufteev M, Ellis J. Regulation, diversity and function of MECP2 exon and 3'UTR isoforms. Hum Mol Genet 2021; 29:R89-R99. [PMID: 32681172 PMCID: PMC7530521 DOI: 10.1093/hmg/ddaa154] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/12/2020] [Accepted: 07/14/2020] [Indexed: 12/11/2022] Open
Abstract
The methyl-CpG-binding protein 2 (MECP2) is a critical global regulator of gene expression. Mutations in MECP2 cause neurodevelopmental disorders including Rett syndrome (RTT). MECP2 exon 2 is spliced into two alternative messenger ribonucleic acid (mRNA) isoforms encoding MECP2-E1 or MECP2-E2 protein isoforms that differ in their N-termini. MECP2-E2, isolated first, was used to define the general roles of MECP2 in methyl-deoxyribonucleic acid (DNA) binding, targeting of transcriptional regulatory complexes, and its disease-causing impact in RTT. It was later found that MECP2-E1 is the most abundant isoform in the brain and its exon 1 is also mutated in RTT. MECP2 transcripts undergo alternative polyadenylation generating mRNAs with four possible 3'untranslated region (UTR) lengths ranging from 130 to 8600 nt. Together, the exon and 3'UTR isoforms display remarkable abundance disparity across cell types and tissues during development. These findings indicate discrete means of regulation and suggest that protein isoforms perform non-overlapping roles. Multiple regulatory programs have been explored to explain these disparities. DNA methylation patterns of the MECP2 promoter and first intron impact MECP2-E1 and E2 isoform levels. Networks of microRNAs and RNA-binding proteins also post-transcriptionally regulate the stability and translation efficiency of MECP2 3'UTR isoforms. Finally, distinctions in biophysical properties in the N-termini between MECP2-E1 and E2 lead to variable protein stabilities and DNA binding dynamics. This review describes the steps taken from the discovery of MECP2, the description of its key functions, and its association with RTT, to the emergence of evidence revealing how MECP2 isoforms are differentially regulated at the transcriptional, post-transcriptional and post-translational levels.
Collapse
Affiliation(s)
- Deivid Carvalho Rodrigues
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto ON M5G 0A4, Canada
| | - Marat Mufteev
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto ON M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Toronto ON M5S 1A8, Canada
| | - James Ellis
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto ON M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Toronto ON M5S 1A8, Canada
| |
Collapse
|
7
|
D'Mello SR. MECP2 and the Biology of MECP2 Duplication Syndrome. J Neurochem 2021; 159:29-60. [PMID: 33638179 DOI: 10.1111/jnc.15331] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/21/2021] [Accepted: 02/18/2021] [Indexed: 11/27/2022]
Abstract
MECP2 duplication syndrome (MDS), a rare X-linked genomic disorder affecting predominantly males, is caused by duplication of the chromosomal region containing the methyl CpG binding protein-2 (MECP2) gene, which encodes methyl-CpG-binding protein 2 (MECP2), a multi-functional protein required for proper brain development and maintenance of brain function during adulthood. Disease symptoms include severe motor and cognitive impairment, delayed or absent speech development, autistic features, seizures, ataxia, recurrent respiratory infections and shortened lifespan. The cellular and molecular mechanisms by which a relatively modest increase in MECP2 protein causes such severe disease symptoms are poorly understood and consequently there are no treatments available for this fatal disorder. This review summarizes what is known to date about the structure and complex regulation of MECP2 and its many functions in the developing and adult brain. Additionally, recent experimental findings on the cellular and molecular underpinnings of MDS based on cell culture and mouse models of the disorder are reviewed. The emerging picture from these studies is that MDS is a neurodegenerative disorder in which neurons die in specific parts of the central nervous system, including the cortex, hippocampus, cerebellum and spinal cord. Neuronal death likely results from astrocytic dysfunction, including a breakdown of glutamate homeostatic mechanisms. The role of elevations in the expression of glial acidic fibrillary protein (GFAP) in astrocytes and the microtubule-associated protein, Tau, in neurons to the pathogenesis of MDS is discussed. Lastly, potential therapeutic strategies to potentially treat MDS are discussed.
Collapse
|
8
|
Martínez-Rodríguez E, Martín-Sánchez A, Kul E, Bose A, Martínez-Martínez FJ, Stork O, Martínez-García F, Lanuza E, Santos M, Agustín-Pavón C. Male-specific features are reduced in Mecp2-null mice: analyses of vasopressinergic innervation, pheromone production and social behaviour. Brain Struct Funct 2020; 225:2219-2238. [PMID: 32749543 DOI: 10.1007/s00429-020-02122-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 07/23/2020] [Indexed: 11/28/2022]
Abstract
Deficits in arginine vasopressin (AVP) and oxytocin (OT), two neuropeptides closely implicated in the modulation of social behaviours, have been reported in some early developmental disorders and autism spectrum disorders. Mutations in the X-linked methyl-CpG-binding protein 2 (MECP2) gene are associated to Rett syndrome and other neuropsychiatric conditions. Thus, we first analysed AVP and OT expression in the brain of Mecp2-mutant mice by immunohistochemistry. Our results revealed no significant differences in these systems in young adult Mecp2-heterozygous females, as compared to WT littermates. By contrast, we found a significant reduction in the sexually dimorphic, testosterone-dependent, vasopressinergic innervation in several nuclei of the social brain network and oxytocinergic innervation in the lateral habenula of Mecp2-null males, as compared to WT littermates. Analysis of urinary production of pheromones shows that Mecp2-null males lack the testosterone-dependent pheromone darcin, strongly suggesting low levels of androgens in these males. In addition, resident-intruder tests revealed lack of aggressive behaviour in Mecp2-null males and decreased chemoinvestigation of the intruder. By contrast, Mecp2-null males exhibited enhanced social approach, as compared to WT animals, in a 3-chamber social interaction test. In summary, Mecp2-null males, which display internal testicles, display a significant reduction of some male-specific features, such as vasopressinergic innervation within the social brain network, male pheromone production and aggressive behaviour. Thus, atypical social behaviours in Mecp2-null males may be caused, at least in part, by the effect of lack of MeCP2 over sexual differentiation.
Collapse
Affiliation(s)
- Elena Martínez-Rodríguez
- Unitat Mixta d'Investigació Neuroanatomia Funcional, Departament de Biologia Cel·lular, Funcional i Antropologia Física, Universitat de València, Av. Vicent Andrés Estellés, s/n, 46100, Burjassot, Spain
| | - Ana Martín-Sánchez
- Unitat Mixta d'Investigació Neuroanatomia Funcional, Unitat Predepartamental de Medicina, Universitat Jaume I de Castelló, Castelló, Spain.,Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,Neuroscience Research Program, IMIM-Hospital del Mar Research Institute, Barcelona, Spain
| | - Emre Kul
- Department of Genetics and Molecular Neurobiology, Institute of Biology, and Center for Behavioral Sciences, Otto-von-Guericke University, Magdeburg, Germany
| | - Aparajita Bose
- Department of Genetics and Molecular Neurobiology, Institute of Biology, and Center for Behavioral Sciences, Otto-von-Guericke University, Magdeburg, Germany.,Neurologie, Ammerland-Klinik GmbH, Westerstede, Germany
| | - Francisco José Martínez-Martínez
- Unitat Mixta d'Investigació Neuroanatomia Funcional, Departament de Biologia Cel·lular, Funcional i Antropologia Física, Universitat de València, Av. Vicent Andrés Estellés, s/n, 46100, Burjassot, Spain
| | - Oliver Stork
- Department of Genetics and Molecular Neurobiology, Institute of Biology, and Center for Behavioral Sciences, Otto-von-Guericke University, Magdeburg, Germany
| | - Fernando Martínez-García
- Unitat Mixta d'Investigació Neuroanatomia Funcional, Unitat Predepartamental de Medicina, Universitat Jaume I de Castelló, Castelló, Spain
| | - Enrique Lanuza
- Unitat Mixta d'Investigació Neuroanatomia Funcional, Departament de Biologia Cel·lular, Funcional i Antropologia Física, Universitat de València, Av. Vicent Andrés Estellés, s/n, 46100, Burjassot, Spain
| | - Mónica Santos
- Department of Genetics and Molecular Neurobiology, Institute of Biology, and Center for Behavioral Sciences, Otto-von-Guericke University, Magdeburg, Germany. .,CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Faculdade de Medicina, pólo I, 2º andar, 3004-504, Coimbra, Portugal.
| | - Carmen Agustín-Pavón
- Unitat Mixta d'Investigació Neuroanatomia Funcional, Departament de Biologia Cel·lular, Funcional i Antropologia Física, Universitat de València, Av. Vicent Andrés Estellés, s/n, 46100, Burjassot, Spain.
| |
Collapse
|
9
|
Zhang X, Li Y, Ma L, Zhang G, Liu M, Wang C, Zheng Y, Li R. A new sex-specific underlying mechanism for female schizophrenia: accelerated skewed X chromosome inactivation. Biol Sex Differ 2020; 11:39. [PMID: 32680558 PMCID: PMC7368719 DOI: 10.1186/s13293-020-00315-6] [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: 01/15/2020] [Accepted: 07/02/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND X chromosome inactivation (XCI) is the mechanism by which the X-linked gene dosage is adjusted between the sexes. Evidence shows that many sex-specific diseases have their basis in X chromosome biology. While female schizophrenia patients often have a delayed age of disease onset and clinical phenotypes that are different from those of males, it is unknown whether the sex differences in schizophrenia are associated with X-linked gene dosage and the choice of X chromosome silencing in female cells. Previous studies demonstrated that sex chromosome aneuploidies may be related to the pathogeneses of some psychiatric diseases. Here, we examined the changes in skewed XCI in patients with schizophrenia. METHODS A total of 109 female schizophrenia (SCZ) patients and 80 age- and sex-matched healthy controls (CNTLs) were included in this study. We evaluated clinical features including disease onset age, disease duration, clinical symptoms by the Positive and Negative Syndrome Scale (PANSS) and antipsychotic treatment dosages. The XCI skewing patterns were analyzed by the methylation profile of the HUMARA gene found in DNA isolated from SCZ patient and CNTL leukocytes in the three age groups. RESULTS First, we found that the frequency of skewed XCI in SCZ patients was 4 times more than that in the age- and sex-matched CNTLs (p < 0.01). Second, we found an earlier onset of severe XCI skewing in the SCZ patients than in CNTLs. Third, we demonstrated a close relationship between the severity of skewed XCI and schizophrenic symptoms (PANSS score ≥ 90) as well as the age of disease onset. Fourth, we demonstrated that the skewed XCI in SCZ patients was not transmitted from the patients' mothers. LIMITATIONS The XCI skewing pattern might differ depending on tissues or organs. Although this is the first study to explore skewed XCI in SCZ, in the future, samples from different tissues or cells in SCZ patients might be important for understanding the impact of skewed XCI in this disease. CONCLUSION Our study, for the first time, investigated skewed XCI in female SCZ patients and presented a potential mechanism for the sex differences in SCZ. Our data also suggested that XCI might be a potential target for the development of female-specific interventions for SCZ.
Collapse
Affiliation(s)
- Xinzhu Zhang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Yuhong Li
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Lei Ma
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Guofu Zhang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Min Liu
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Chuanyue Wang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Yi Zheng
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Rena Li
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China. .,The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
10
|
Manchia M, Comai S, Pinna M, Pinna F, Fanos V, Denovan-Wright E, Carpiniello B. Biomarkers in aggression. Adv Clin Chem 2019; 93:169-237. [PMID: 31655730 DOI: 10.1016/bs.acc.2019.07.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Aggressive behavior exerts an enormous impact on society remaining among the main causes of worldwide premature death. Effective primary interventions, relying on predictive models of aggression that show adequate sensitivity and specificity are currently lacking. One strategy to increase the accuracy and precision of prediction would be to include biological data in the predictive models. Clearly, to be included in such models, biological markers should be reliably associated with the specific trait under study (i.e., diagnostic biomarkers). Aggression, however, is phenotypically highly heterogeneous, an element that has hindered the identification of reliable biomarkers. However, current research is trying to overcome these challenges by focusing on more homogenous aggression subtypes and/or by studying large sample size of aggressive individuals. Further advance is coming by bioinformatics approaches that are allowing the integration of inter-species biological data as well as the development of predictive algorithms able to discriminate subjects on the basis of the propensity toward aggressive behavior. In this review we first present a brief summary of the available evidence on neuroimaging of aggression. We will then treat extensively the data on genetic determinants, including those from hypothesis-free genome-wide association studies (GWAS) and candidate gene studies. Transcriptomic and neurochemical biomarkers will then be reviewed, and we will dedicate a section on the role of metabolomics in aggression. Finally, we will discuss how biomarkers can inform the development of new pharmacological tools as well as increase the efficacy of preventive strategies.
Collapse
Affiliation(s)
- Mirko Manchia
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy; Department of Pharmacology, Dalhousie University, Halifax, NS, Canada.
| | - Stefano Comai
- San Raffaele Scientific Institute and Vita Salute University, Milano, Italy; Department of Psychiatry, McGill University, Montreal, QC, Canada.
| | - Martina Pinna
- Forensic Psychiatry Unit, Sardinia Health Agency, Cagliari, Italy
| | - Federica Pinna
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Vassilios Fanos
- Department of Surgical Sciences, University of Cagliari, Cagliari, Italy; Puericulture Institute and Neonatal Section, University Hospital Agency of Cagliari, Cagliari, Italy
| | | | - Bernardo Carpiniello
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| |
Collapse
|
11
|
Lee LC, Su MT, Cho YC, Lee-Chen GJ, Yeh TK, Chang CY. Multiple epigenetic biomarkers for evaluation of students' academic performance. GENES BRAIN AND BEHAVIOR 2019; 18:e12559. [PMID: 30806012 DOI: 10.1111/gbb.12559] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 01/18/2019] [Accepted: 02/07/2019] [Indexed: 11/28/2022]
Abstract
Several reports have shown that methyl CpG-binding protein 2 (MeCP2), brain-derived neurotrophic factor (BDNF), phospho-cAMP response element-binding protein (p-CREB) and microRNAs may be important in regulating academic performance because of their roles in neuropsychiatry and cognitive diseases. The first goal of this study was to explore the associations among MeCP2, BDNF, CREB and academic performance. This study also examined the pathway responsible for the effects of MeCP2, BDNF, p-CREB and microRNAs on academic performance. Scores from the basic competency test, an annual national competitive entrance examination, were used to evaluate academic performance. Subjects' plasma RNA was extracted and analyzed. This study determined that participants in the higher academic performance group had a significant difference in MECP2 mRNA expression compared with the lower academic performance group. We then used neuronal human derived neuroblastoma cell line (SH-SY5Y) cells with inducible MeCP2 expression from a second copy of the gene as a gain-of-function model and found that MeCP2 overexpression positively affected p-CREB and BDNF expression initially. After negative feedback, the p-CREB and BDNF levels subsequently decreased. In the neuronal phenotype examination, we found a significant reduction in total outgrowth and branches in MeCP2-induced cells compared with noninduced cells. This work describes pathways that may be responsible for the effects of MeCP2, BDNF, p-CREB and microRNAs on academic performance. These results may shed light on the development of promising clinical treatment strategies in the area of neuropsychological adjustment.
Collapse
Affiliation(s)
- Li-Ching Lee
- Science Education Center and Graduate Institute of Science Education, National Taiwan Normal University, Taipei, Taiwan
| | - Ming-Tsan Su
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Ying-Chun Cho
- Science Education Center and Graduate Institute of Science Education, National Taiwan Normal University, Taipei, Taiwan
| | - Guey-Jen Lee-Chen
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Ting-Kuang Yeh
- Science Education Center and Graduate Institute of Science Education, National Taiwan Normal University, Taipei, Taiwan.,Institute of Marine Environment Science and Technology, National Taiwan Normal University, Taipei, Taiwan.,Department of Earth Science, National Taiwan Normal University, Taipei, Taiwan
| | - Chun-Yen Chang
- Science Education Center and Graduate Institute of Science Education, National Taiwan Normal University, Taipei, Taiwan.,Department of Earth Science, National Taiwan Normal University, Taipei, Taiwan
| |
Collapse
|
12
|
Kurtenbach S, Gießl A, Strömberg S, Kremers J, Atorf J, Rasche S, Neuhaus EM, Hervé D, Brandstätter JH, Asan E, Hatt H, Kilimann MW. The BEACH Protein LRBA Promotes the Localization of the Heterotrimeric G-protein G olf to Olfactory Cilia. Sci Rep 2017; 7:8409. [PMID: 28814779 PMCID: PMC5559528 DOI: 10.1038/s41598-017-08543-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 07/10/2017] [Indexed: 02/07/2023] Open
Abstract
BEACH domain proteins are involved in membrane protein traffic and human diseases, but their molecular mechanisms are not understood. The BEACH protein LRBA has been implicated in immune response and cell proliferation, and human LRBA mutations cause severe immune deficiency. Here, we report a first functional and molecular phenotype outside the immune system of LRBA-knockout mice: compromised olfaction, manifesting in reduced electro-olfactogram response amplitude, impaired food-finding efficiency, and smaller olfactory bulbs. LRBA is prominently expressed in olfactory and vomeronasal chemosensory neurons of wild-type mice. Olfactory impairment in the LRBA-KO is explained by markedly reduced concentrations (20–40% of wild-type levels) of all three subunits αolf, β1 and γ13 of the olfactory heterotrimeric G-protein, Golf, in the sensory cilia of olfactory neurons. In contrast, cilia morphology and the concentrations of many other proteins of olfactory cilia are not or only slightly affected. LRBA is also highly expressed in photoreceptor cells, another cell type with a specialized sensory cilium and heterotrimeric G-protein-based signalling; however, visual function appeared unimpaired by the LRBA-KO. To our knowledge, this is the first observation that a BEACH protein is required for the efficient subcellular localization of a lipid-anchored protein, and of a ciliary protein.
Collapse
Affiliation(s)
- Stefan Kurtenbach
- Department of Cell Physiology, Ruhr University Bochum, D-44780, Bochum, Germany
| | - Andreas Gießl
- Department of Biology, Animal Physiology, University of Erlangen-Nürnberg, D-91058, Erlangen, Germany
| | - Siv Strömberg
- Department of Neuroscience, Uppsala University, S-75124, Uppsala, Sweden
| | - Jan Kremers
- Department of Ophthalmology, University Hospital Erlangen, D-91054, Erlangen, Germany.,Department of Anatomy II, Friedrich-Alexander University Erlangen-Nürnberg, D-91054, Erlangen, Germany
| | - Jenny Atorf
- Department of Ophthalmology, University Hospital Erlangen, D-91054, Erlangen, Germany
| | - Sebastian Rasche
- Department of Cell Physiology, Ruhr University Bochum, D-44780, Bochum, Germany
| | - Eva M Neuhaus
- Department of Pharmacology and Toxikology, University Hospital Jena, D-07747, Jena, Germany
| | - Denis Hervé
- Inserm UMR-S839, Institut du Fer a Moulin, Universite Pierre et Marie Curie, F-75005, Paris, France
| | | | - Esther Asan
- Institute of Anatomy and Cell Biology, University of Würzburg, D-97070, Würzburg, Germany
| | - Hanns Hatt
- Department of Cell Physiology, Ruhr University Bochum, D-44780, Bochum, Germany
| | - Manfred W Kilimann
- Department of Neuroscience, Uppsala University, S-75124, Uppsala, Sweden. .,Department of Molecular Neurobiology, Max Planck Institute for Experimental Medicine, D-37075, Göttingen, Germany.
| |
Collapse
|
13
|
Gupta T, Morgan HR, Andrews JC, Brewer ER, Certel SJ. Methyl-CpG binding domain proteins inhibit interspecies courtship and promote aggression in Drosophila. Sci Rep 2017; 7:5420. [PMID: 28710457 PMCID: PMC5511146 DOI: 10.1038/s41598-017-05844-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 06/05/2017] [Indexed: 11/10/2022] Open
Abstract
Reproductive isolation and speciation are driven by the convergence of environmental and genetic variation. The integration of these variation sources is thought to occur through epigenetic marks including DNA methylation. Proteins containing a methyl-CpG-binding domain (MBD) bind methylated DNA and interpret epigenetic marks, providing a dynamic yet evolutionarily adapted cellular output. Here, we report the Drosophila MBD-containing proteins, dMBD-R2 and dMBD2/3, contribute to reproductive isolation and survival behavioral strategies. Drosophila melanogaster males with a reduction in dMBD-R2 specifically in octopamine (OA) neurons exhibit courtship toward divergent interspecies D. virilis and D. yakuba females and a decrease in conspecific mating success. Conspecific male-male courtship is increased between dMBD-R2-deficient males while aggression is reduced. These changes in adaptive behavior are separable as males with a hypermethylated OA neuronal genome exhibited a decrease in aggression without altering male-male courtship. These results suggest Drosophila MBD-containing proteins are required within the OA neural circuitry to inhibit interspecies and conspecific male-male courtship and indicate that the genetically hard-wired neural mechanisms enforcing behavioral reproductive isolation include the interpretation of the epigenome.
Collapse
Affiliation(s)
- Tarun Gupta
- Neuroscience Graduate Program, The University of Montana, Missoula, MT, United States
| | - Hannah R Morgan
- Division of Biological Sciences, The University of Montana, Missoula, MT, United States
| | - Jonathan C Andrews
- Division of Biological Sciences, The University of Montana, Missoula, MT, United States
| | - Edmond R Brewer
- Division of Biological Sciences, The University of Montana, Missoula, MT, United States
| | - Sarah J Certel
- Neuroscience Graduate Program, The University of Montana, Missoula, MT, United States. .,Division of Biological Sciences, The University of Montana, Missoula, MT, United States.
| |
Collapse
|
14
|
Long-term consumption of sugar-sweetened beverage during the growth period promotes social aggression in adult mice with proinflammatory responses in the brain. Sci Rep 2017; 7:45693. [PMID: 28393871 PMCID: PMC5385878 DOI: 10.1038/srep45693] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 03/06/2017] [Indexed: 12/29/2022] Open
Abstract
Overconsumption of sugar-sweetened beverages (SSBs) is known to be a key contributor to the obesity epidemic; however, its effects on behavioral changes are yet to be fully studied. In the present study, we examined the long-term effects of SSB on social aggression in mice. Three-week-old weaned mice started to drink either a 30 w/v% sucrose solution (S30), plain water (CT), or an aspartame solution with sweetness equivalent to the sucrose solution (A30) and continued to drink until they were 11-week-old adults. Aggressive behaviors were assessed by the resident-intruder test. We found that SSB significantly promoted social aggression, accompanied by heightened serum corticosterone and reduced body weight. To understand the underlying mechanism, we performed transcriptome analyses of brain. The profiles of mice on S30 were dramatically different from those on CT or A30. Transcriptional networks related to immunological function were significantly dysregulated by SSB. FACS analysis of mice on S30 revealed increased numbers of inflammatory cells in peripheral blood. Interestingly, the artificial sweetener failed to mimic the effects of sugar on social aggression and inflammatory responses. These results demonstrate that SSB promotes aggressive behaviors and provide evidence that sugar reduction strategies may be useful in efforts to prevent social aggression.
Collapse
|
15
|
Tomasoni R, Morini R, Lopez-Atalaya JP, Corradini I, Canzi A, Rasile M, Mantovani C, Pozzi D, Garlanda C, Mantovani A, Menna E, Barco A, Matteoli M. Lack of IL-1R8 in neurons causes hyperactivation of IL-1 receptor pathway and induces MECP2-dependent synaptic defects. eLife 2017; 6. [PMID: 28347403 PMCID: PMC5370184 DOI: 10.7554/elife.21735] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 02/26/2017] [Indexed: 12/12/2022] Open
Abstract
Inflammation modifies risk and/or severity of a variety of brain diseases through still elusive molecular mechanisms. Here we show that hyperactivation of the interleukin 1 pathway, through either ablation of the interleukin 1 receptor 8 (IL-1R8, also known as SIGIRR or Tir8) or activation of IL-1R, leads to up-regulation of the mTOR pathway and increased levels of the epigenetic regulator MeCP2, bringing to disruption of dendritic spine morphology, synaptic plasticity and plasticity-related gene expression. Genetic correction of MeCP2 levels in IL-1R8 KO neurons rescues the synaptic defects. Pharmacological inhibition of IL-1R activation by Anakinra corrects transcriptional changes, restores MeCP2 levels and spine plasticity and ameliorates cognitive defects in IL-1R8 KO mice. By linking for the first time neuronal MeCP2, a key player in brain development, to immune activation and demonstrating that synaptic defects can be pharmacologically reversed, these data open the possibility for novel treatments of neurological diseases through the immune system modulation. DOI:http://dx.doi.org/10.7554/eLife.21735.001 Errors that occur while the brain is developing can lead to conditions such as autism and schizophrenia. They can also lead to rare disorders like Rett syndrome and MeCP2 duplication syndromes, which are characterized by severe cognitive and physical disabilities. Many people with these neurodevelopmental disorders have mutations in genes that encode proteins found at synapses, which are the junctions between neurons where the cells exchange information with one another. However, not everyone with these mutations develops a neurodevelopmental disorder, which indicates that other, non-genetic factors also play a part. One of the main non-genetic factors that can influence the risk and severity of neurodevelopmental disorders is inflammation of the brain. Inflammation is a normal part of the body’s immune response to threats such as invading microorganisms or tissue damage. However, abnormal activation of the immune system in early life can trigger excessive inflammation. This increases the risk of a neurodevelopmental disorder, but it is not clear exactly how it does so. Tomasoni et al. set out to test whether the missing link between inflammation and neurodevelopmental disorders might be damage to synapses. The experiments revealed that genetically modified mice with inflammation of the brain have abnormal synapses and are unable to learn properly. These mutant mice also have excessive levels of a protein that influences how synapses function called MeCP2, which is missing in the brains of people with Rett syndrome and abnormally increased in brains of patients affected by MeCP2 Duplication Syndrome. This is thus the first evidence that directly links inflammation of the brain to a synapse protein implicated in a disorder of brain development. Tomasoni et al. also found that a drug called anakinra – which is used to treat an inflammatory disease called rheumatoid arthritis – reduced levels of MeCP2 in the mutant mice and improved their performance in cognitive tasks. Together, these results raise the possibility that anti-inflammatory medications may be beneficial in the treatment of neurodevelopment disorders. DOI:http://dx.doi.org/10.7554/eLife.21735.002
Collapse
Affiliation(s)
- Romana Tomasoni
- IRCCS Humanitas, Rozzano, Italy.,Instituto de Neurociencias (Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas), Alicante, Spain
| | | | - Jose P Lopez-Atalaya
- Instituto de Neurociencias (Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas), Alicante, Spain
| | | | - Alice Canzi
- IRCCS Humanitas, Rozzano, Italy.,Hunimed University, Rozzano, Italy
| | - Marco Rasile
- IRCCS Humanitas, Rozzano, Italy.,Hunimed University, Rozzano, Italy
| | | | - Davide Pozzi
- IRCCS Humanitas, Rozzano, Italy.,Hunimed University, Rozzano, Italy
| | | | | | | | - Angel Barco
- Instituto de Neurociencias (Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas), Alicante, Spain
| | | |
Collapse
|
16
|
Hypersocial behavior and biological redundancy in mice with reduced expression of PSD95 or PSD93. Behav Brain Res 2017; 352:35-45. [PMID: 28189758 DOI: 10.1016/j.bbr.2017.02.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 02/07/2017] [Accepted: 02/08/2017] [Indexed: 12/27/2022]
Abstract
The postsynaptic density proteins 95 (PSD95) and 93 (PSD93) belong to a family of scaffolding proteins, the membrane-associated guanylate kinases (MAGUKs), which are highly enriched in synapses and responsible for organizing the numerous protein complexes required for synaptic development and plasticity. Genetic studies have associated MAGUKs with diseases like autism and schizophrenia, but knockout mice show severe, complex defects with difficult-to-interpret behavioral abnormalities due to major motor dysfunction which is atypical for psychiatric phenotypes. Therefore, rather than studying loss-of-function mutants, we comprehensively investigated the behavioral consequences of reduced PSD95 expression, using heterozygous PSD95 knockout mice (PSD95+/-). Specifically, we asked whether heterozygous PSD95 deficient mice would exhibit alterations in the processing of social stimuli and social behavior. Additionally, we investigated whether PSD95 and PSD93 would reveal any indication of functional or biological redundancy. Therefore, homozygous and heterozygous PSD93 deficient mice were examined in a similar behavioral battery as PSD95 mutants. We found robust hypersocial behavior in the dyadic interaction test in both PSD95+/- males and females. Additionally, male PSD95+/- mice exhibited higher levels of aggression and territoriality, while female PSD95+/- mice showed increased vocalization upon exposure to an anesthetized female mouse. Both male and female PSD95+/- mice revealed mild hypoactivity in the open field but no obvious motor deficit. Regarding PSD93 mutants, homozygous (but not heterozygous) knockout mice displayed prominent hypersocial behavior comparable to that observed in PSD95+/- mice, despite a more severe motor phenotype, which precluded several behavioral tests or their interpretation. Considering that PSD95 and PSD93 reduction provoke strikingly similar behavioral consequences, we explored a potential substitution effect and found increased PSD93 protein expression in hippocampal synaptic enrichment preparations of PSD95+/- mice. These data suggest that both PSD95 and PSD93 are involved in processing of social stimuli and control of social behavior. This important role may be partly assured by functional/behavioral and biological/biochemical redundancy.
Collapse
|
17
|
Krueger-Burg D, Winkler D, Mitkovski M, Daher F, Ronnenberg A, Schlüter OM, Dere E, Ehrenreich H. The SocioBox: A Novel Paradigm to Assess Complex Social Recognition in Male Mice. Front Behav Neurosci 2016; 10:151. [PMID: 27563287 PMCID: PMC4980394 DOI: 10.3389/fnbeh.2016.00151] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 07/19/2016] [Indexed: 12/27/2022] Open
Abstract
Impairments in social skills are central to mental disease, and developing tools for their assessment in mouse models is essential. Here we present the SocioBox, a new behavioral paradigm to measure social recognition. Using this paradigm, we show that male wildtype mice of different strains can readily identify an unfamiliar mouse among 5 newly acquainted animals. In contrast, female mice exhibit lower locomotor activity during social exploration in the SocioBox compared to males and do not seem to discriminate between acquainted and unfamiliar mice, likely reflecting inherent differences in gender-specific territorial tasks. In addition to a simple quantification of social interaction time of mice grounded on predefined spatial zones (zone-based method), we developed a set of unbiased, data-driven analysis tools based on heat map representations and characterized by greater sensitivity. First proof-of-principle that the SocioBox allows diagnosis of social recognition deficits is provided using male PSD-95 heterozygous knockout mice, a mouse model related to psychiatric pathophysiology.
Collapse
Affiliation(s)
| | - Daniela Winkler
- Max Planck Institute of Experimental MedicineGöttingen, Germany; DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the BrainGöttingen, Germany
| | - Mišo Mitkovski
- Max Planck Institute of Experimental Medicine Göttingen, Germany
| | - Fernanda Daher
- Max Planck Institute of Experimental Medicine Göttingen, Germany
| | - Anja Ronnenberg
- Max Planck Institute of Experimental Medicine Göttingen, Germany
| | - Oliver M Schlüter
- DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the BrainGöttingen, Germany; European Neuroscience InstituteGöttingen, Germany
| | - Ekrem Dere
- Max Planck Institute of Experimental MedicineGöttingen, Germany; DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the BrainGöttingen, Germany
| | - Hannelore Ehrenreich
- Max Planck Institute of Experimental MedicineGöttingen, Germany; DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the BrainGöttingen, Germany
| |
Collapse
|
18
|
Pearson BL, Defensor EB, Blanchard DC, Blanchard RJ. Applying the ethoexperimental approach to neurodevelopmental syndrome research reveals exaggerated defensive behavior in Mecp2 mutant mice. Physiol Behav 2016; 146:98-104. [PMID: 26066729 DOI: 10.1016/j.physbeh.2015.03.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 03/25/2015] [Accepted: 03/26/2015] [Indexed: 02/07/2023]
Abstract
Rett syndrome is a Pervasive Developmental Disorder (PDD) associated with de novo mutations of the methyl CpG-binding protein 2 (MECP2) gene. Mecp2 functions as a transcription factor that regulates the expression of hundreds of genes. Identification of the role of Mecp2 in specific neurodevelopmental symptoms remains an important research aim. We previously demonstrated that male mice possessing a truncation mutation in Mecp2 are hyper-social. We predicted that reduced fear or anxiety might underlie this enhanced affiliation. In order to probe risk assessment and anxiety-like behavior, we compared Mecp2 truncation mutants to their wild-type littermates in the elevated plus maze and elevated zero maze. Additionally, subjects were administered the mouse defense test battery to evaluate unconditioned fear- and panic-like behavior to a graded set of threat scenarios and a predator stimulus. Mutant mice showed no significant changes in anxiety-like behavior. Yet, they displayed hyper-reactive escape and defensive behaviors to an animate predatory threat stimulus. Notably, mutant mice engaged in exaggerated active defense responding to threat stimuli at nearly all phases of the fear battery. These results reveal abnormalities in emotion regulation in Mecp2 mutants particularly in response to ecologically relevant threats. This hyper-responsivity suggests that transcriptional targets of Mecp2 are critical to emotion regulation. Moreover, we suggest that detailed analysis of defensive behavior and aggression with ethologically relevant tasks provides an avenue to interrogate gene-behavior mechanisms of neurodevelopmental and other psychiatric conditions.
Collapse
|
19
|
McGowan H, Pang ZP. Regulatory functions and pathological relevance of the MECP2 3'UTR in the central nervous system. CELL REGENERATION 2015; 4:9. [PMID: 26516454 PMCID: PMC4625459 DOI: 10.1186/s13619-015-0023-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 09/18/2015] [Indexed: 11/10/2022]
Abstract
Methyl-CpG-binding protein 2 (MeCP2), encoded by the gene MECP2, is a transcriptional regulator and chromatin-remodeling protein, which is ubiquitously expressed and plays an essential role in the development and maintenance of the central nervous system (CNS). Highly enriched in post-migratory neurons, MeCP2 is needed for neuronal maturation, including dendritic arborization and the development of synapses. Loss-of-function mutations in MECP2 cause Rett syndrome (RTT), a debilitating neurodevelopmental disorder characterized by a phase of normal development, followed by the progressive loss of milestones and cognitive disability. While a great deal has been discovered about the structure, function, and regulation of MeCP2 in the time since its discovery as the genetic cause of RTT, including its involvement in a number of RTT-related syndromes that have come to be known as MeCP2-spectrum disorders, much about this multifunctional protein remains enigmatic. One unequivocal fact that has become apparent is the importance of maintaining MeCP2 protein levels within a narrow range, the limits of which may depend upon the cell type and developmental time point. As such, MeCP2 is amenable to complex, multifactorial regulation. Here, we summarize the role of the MECP2 3' untranslated region (UTR) in the regulation of MeCP2 protein levels and how mutations in this region contribute to autism and other non-RTT neuropsychiatric disorders.
Collapse
Affiliation(s)
- Heather McGowan
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers University Robert Wood Johnson Medical School, 89 French Street, Room 3277, New Brunswick, NJ 08901 USA
| | - Zhiping P Pang
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers University Robert Wood Johnson Medical School, 89 French Street, Room 3277, New Brunswick, NJ 08901 USA
| |
Collapse
|
20
|
Codina-Solà M, Rodríguez-Santiago B, Homs A, Santoyo J, Rigau M, Aznar-Laín G, Del Campo M, Gener B, Gabau E, Botella MP, Gutiérrez-Arumí A, Antiñolo G, Pérez-Jurado LA, Cuscó I. Integrated analysis of whole-exome sequencing and transcriptome profiling in males with autism spectrum disorders. Mol Autism 2015; 6:21. [PMID: 25969726 PMCID: PMC4427998 DOI: 10.1186/s13229-015-0017-0] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 03/19/2015] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Autism spectrum disorders (ASD) are a group of neurodevelopmental disorders with high heritability. Recent findings support a highly heterogeneous and complex genetic etiology including rare de novo and inherited mutations or chromosomal rearrangements as well as double or multiple hits. METHODS We performed whole-exome sequencing (WES) and blood cell transcriptome by RNAseq in a subset of male patients with idiopathic ASD (n = 36) in order to identify causative genes, transcriptomic alterations, and susceptibility variants. RESULTS We detected likely monogenic causes in seven cases: five de novo (SCN2A, MED13L, KCNV1, CUL3, and PTEN) and two inherited X-linked variants (MAOA and CDKL5). Transcriptomic analyses allowed the identification of intronic causative mutations missed by the usual filtering of WES and revealed functional consequences of some rare mutations. These included aberrant transcripts (PTEN, POLR3C), deregulated expression in 1.7% of mutated genes (that is, SEMA6B, MECP2, ANK3, CREBBP), allele-specific expression (FUS, MTOR, TAF1C), and non-sense-mediated decay (RIT1, ALG9). The analysis of rare inherited variants showed enrichment in relevant pathways such as the PI3K-Akt signaling and the axon guidance. CONCLUSIONS Integrative analysis of WES and blood RNAseq data has proven to be an efficient strategy to identify likely monogenic forms of ASD (19% in our cohort), as well as additional rare inherited mutations that can contribute to ASD risk in a multifactorial manner. Blood transcriptomic data, besides validating 88% of expressed variants, allowed the identification of missed intronic mutations and revealed functional correlations of genetic variants, including changes in splicing, expression levels, and allelic expression.
Collapse
Affiliation(s)
- Marta Codina-Solà
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, C/Doctor Aiguader 88, 422, Barcelona, 08003 Spain ; Hospital del Mar Research Institute (IMIM), C/Doctor Aiguader 88, Barcelona, 08003 Spain ; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), C/ Monforte de Lemos 3-5, Madrid, 28029 Spain
| | | | - Aïda Homs
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, C/Doctor Aiguader 88, 422, Barcelona, 08003 Spain ; Hospital del Mar Research Institute (IMIM), C/Doctor Aiguader 88, Barcelona, 08003 Spain ; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), C/ Monforte de Lemos 3-5, Madrid, 28029 Spain
| | - Javier Santoyo
- Medical Genome Project, Genomics and Bioinformatics Platform of Andalusia (GBPA), C/Albert Einstein, Cartuja Scientific and Technology Park, INSUR Builiding, Sevilla, 41092 Spain
| | - Maria Rigau
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, C/Doctor Aiguader 88, 422, Barcelona, 08003 Spain
| | - Gemma Aznar-Laín
- Pediatric Neurology, Hospital del Mar, Passeig Marítim 25-29, Barcelona, 08003 Spain
| | - Miguel Del Campo
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, C/Doctor Aiguader 88, 422, Barcelona, 08003 Spain ; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), C/ Monforte de Lemos 3-5, Madrid, 28029 Spain ; Servicio de Genética, Hospital Vall d'Hebron, Passeig Vall d'Hebron, 119-129, Barcelona, 08015 Spain
| | - Blanca Gener
- Genetics Service, BioCruces Health Research Institute, Hospital Universitario Cruces, Plaza de Cruces 12, Barakaldo, Bizkaia 48093 Spain
| | - Elisabeth Gabau
- Pediatrics Service, Corporació Sanitària Parc Taulí, Parc Taulí 1, Sabadell, 08208 Spain
| | - María Pilar Botella
- Pediatric Neurology, Hospital de Txagorritxu, C/José de Atxotegui s/n, Victoria-Gasteiz, 01009 Spain
| | - Armand Gutiérrez-Arumí
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, C/Doctor Aiguader 88, 422, Barcelona, 08003 Spain ; Hospital del Mar Research Institute (IMIM), C/Doctor Aiguader 88, Barcelona, 08003 Spain ; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), C/ Monforte de Lemos 3-5, Madrid, 28029 Spain
| | - Guillermo Antiñolo
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), C/ Monforte de Lemos 3-5, Madrid, 28029 Spain ; Medical Genome Project, Genomics and Bioinformatics Platform of Andalusia (GBPA), C/Albert Einstein, Cartuja Scientific and Technology Park, INSUR Builiding, Sevilla, 41092 Spain ; Department of Genetics, Reproduction and Fetal Medicine, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, Avda Manuel Siurot s/n, Sevilla, 41013 Spain
| | - Luis Alberto Pérez-Jurado
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, C/Doctor Aiguader 88, 422, Barcelona, 08003 Spain ; Hospital del Mar Research Institute (IMIM), C/Doctor Aiguader 88, Barcelona, 08003 Spain ; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), C/ Monforte de Lemos 3-5, Madrid, 28029 Spain
| | - Ivon Cuscó
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, C/Doctor Aiguader 88, 422, Barcelona, 08003 Spain ; Hospital del Mar Research Institute (IMIM), C/Doctor Aiguader 88, Barcelona, 08003 Spain ; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), C/ Monforte de Lemos 3-5, Madrid, 28029 Spain
| |
Collapse
|
21
|
|
22
|
Serguera C, Bemelmans AP. Gene therapy of the central nervous system: general considerations on viral vectors for gene transfer into the brain. Rev Neurol (Paris) 2014; 170:727-38. [PMID: 25459120 DOI: 10.1016/j.neurol.2014.09.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 08/13/2014] [Accepted: 09/10/2014] [Indexed: 02/04/2023]
Abstract
The last decade has nourished strong doubts on the beneficial prospects of gene therapy for curing fatal diseases. However, this climate of reservation is currently being transcended by the publication of several successful clinical protocols, restoring confidence in the appropriateness of therapeutic gene transfer. A strong sign of this present enthusiasm for gene therapy by clinicians and industrials is the market approval of the therapeutic viral vector Glybera, the first commercial product in Europe of this class of drug. This new field of medicine is particularly attractive when considering therapies for a number of neurological disorders, most of which are desperately waiting for a satisfactory treatment. The central nervous system is indeed a very compliant organ where gene transfer can be stable and successful if provided through an appropriate strategy. The purpose of this review is to present the characteristics of the most efficient virus-derived vectors used by researchers and clinicians to genetically modify particular cell types or whole regions of the brain. In addition, we discuss major issues regarding side effects, such as genotoxicity and immune response associated to the use of these vectors.
Collapse
Affiliation(s)
- C Serguera
- CEA, DSV, I(2)BM, Molecular Imaging Research Center (MIRCen) and CNRS, CEA URA 2210, 18, route du Panorama, 92265 Fontenay-aux-Roses, France
| | - A-P Bemelmans
- CEA, DSV, I(2)BM, Molecular Imaging Research Center (MIRCen) and CNRS, CEA URA 2210, 18, route du Panorama, 92265 Fontenay-aux-Roses, France.
| |
Collapse
|