Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
J Neurodev Disord ; 15(1): 39, 2023 11 13.
Article in English | MEDLINE | ID: mdl-37957569

ABSTRACT

BACKGROUND: ATRX is an ATP-dependent chromatin remodeling protein with essential roles in safeguarding genome integrity and modulating gene expression. Deficiencies in this protein cause ATR-X syndrome, a condition characterized by intellectual disability and an array of developmental abnormalities, including features of autism. Previous studies demonstrated that deleting ATRX in mouse forebrain excitatory neurons postnatally resulted in male-specific memory deficits, but no apparent autistic-like behaviours. METHODS: We generated mice with an earlier embryonic deletion of ATRX in forebrain excitatory neurons and characterized their behaviour using a series of memory and autistic-related paradigms. RESULTS: We found that mutant mice displayed a broader spectrum of impairments, including fear memory, decreased anxiety-like behaviour, hyperactivity, as well as self-injurious and repetitive grooming. Sex-specific alterations were also observed, including male-specific aggression, sensory gating impairments, and decreased social memory. CONCLUSIONS: Collectively, the findings indicate that early developmental abnormalities arising from ATRX deficiency in forebrain excitatory neurons contribute to the presentation of fear memory deficits as well as autistic-like behaviours.


Subject(s)
Autistic Disorder , Female , Mice , Male , Animals , Autistic Disorder/complications , Autistic Disorder/genetics , Neurons/physiology , Memory Disorders/etiology , Cognition
2.
Nat Commun ; 14(1): 7090, 2023 11 04.
Article in English | MEDLINE | ID: mdl-37925436

ABSTRACT

Myelin, an extension of the oligodendrocyte plasma membrane, wraps around axons to facilitate nerve conduction. Myelination is compromised in ATR-X intellectual disability syndrome patients, but the causes are unknown. We show that loss of ATRX leads to myelination deficits in male mice that are partially rectified upon systemic thyroxine administration. Targeted ATRX inactivation in either neurons or oligodendrocyte progenitor cells (OPCs) reveals OPC-intrinsic effects on myelination. OPCs lacking ATRX fail to differentiate along the oligodendrocyte lineage and acquire a more plastic state that favors astrocytic differentiation in vitro and in vivo. ATRX chromatin occupancy in OPCs greatly overlaps with that of the chromatin remodelers CHD7 and CHD8 as well as H3K27Ac, a mark of active enhancers. Overall, our data indicate that ATRX regulates the onset of myelination systemically via thyroxine, and by promoting OPC differentiation and suppressing astrogliogenesis. These functions of ATRX identified in mice could explain white matter pathogenesis observed in ATR-X syndrome patients.


Subject(s)
Myelin Sheath , Thyroxine , X-linked Nuclear Protein , Animals , Humans , Male , Mice , Cell Differentiation/physiology , Chromatin/metabolism , Myelin Sheath/metabolism , Neurogenesis , Oligodendroglia/metabolism , Thyroxine/metabolism , X-linked Nuclear Protein/genetics , X-linked Nuclear Protein/metabolism , Neuroglia
3.
Front Immunol ; 13: 943481, 2022.
Article in English | MEDLINE | ID: mdl-35983032

ABSTRACT

Increasing the number of CpG dinucleotides in RNA viral genomes, while preserving the original amino acid composition, leads to impaired infection which does not cause disease. Beneficially, impaired infection evokes antiviral host immune responses providing a cutting-edge vaccine approach. For example, we previously showed that CpG-enriched Zika virus variants cause attenuated infection phenotypes and protect against lethal challenge in mice. While CpG recoding is an emerging and promising vaccine approach, little is known about infection phenotypes caused by recoded viruses in vivo, particularly in non-rodent species. Here, we used well-established mouse and porcine models to study infection phenotypes of the CpG-enriched neurotropic and congenital virus-Zika virus, directly in the target tissues-the brain and placenta. Specifically, we used the uttermost challenge and directly injected mice intracerebrally to compare infection phenotypes caused by wild-type and two CpG-recoded Zika variants and model the scenario where vaccine strains breach the blood-brain barrier. Also, we directly injected porcine fetuses to compare in utero infection phenotypes and model the scenario where recoded vaccine strains breach the placental barrier. While overall infection kinetics were comparable between wild-type and recoded virus variants, we found convergent phenotypical differences characterized by reduced pathology in the mouse brain and reduced replication of CpG-enriched variants in fetal lymph nodes. Next, using next-generation sequencing for the whole virus genome, we compared the stability of de novo introduced CpG dinucleotides during prolonged virus infection in the brain and placenta. Most de novo introduced CpG dinucleotides were preserved in sequences of recoded Zika viruses showing the stability of vaccine variants. Altogether, our study emphasized further directions to fine-tune the CpG recoding vaccine approach for better safety and can inform future immunization strategies.


Subject(s)
Viruses , Zika Virus Infection , Zika Virus , Animals , Brain , Female , Fetus , Genome, Viral , Lymph Nodes , Mice , Phenotype , Placenta , Pregnancy , Swine , Viruses/genetics
4.
Front Vet Sci ; 9: 931232, 2022.
Article in English | MEDLINE | ID: mdl-35990278

ABSTRACT

An effective single-dose vaccine that protects the dam and her suckling offspring against infectious disease would be widely beneficial to livestock animals. We assessed whether a single-dose intramuscular (i.m.) porcine epidemic diarrhea virus (PEDV) vaccine administered to the gilt 30 days post-breeding could generate mucosal and systemic immunity and sufficient colostral and mature milk antibodies to protect suckling piglets against infectious challenge. The vaccine was comprised of polymeric poly-(lactide-co-glycolide) (PGLA)-nanoparticle (NP) encapsulating recombinant PEDV spike protein 1 (PEDVS1) associated with ARC4 and ARC7 adjuvants, a muramyl dipeptide analog and a monophosphoryl lipid A (MPLA) analog, respectively (NP-PEDVS1). To establish whether prior mucosal exposure could augment the i.m. immune response and/or contribute to mucosal tolerance, gilts were immunized with the NP-PEDVS1 vaccine via the intrauterine route at breeding, followed by the i.m. vaccine 30 days later. Archived colostrum from gilts that were challenged with low-dose PEDV plus alum was used as positive reference samples for neutralizing antibodies and passive protection. On day 100 of gestation (70 days post i.m. immunization), both vaccinated groups showed significant PEDVS1-specific IgG and IgA in the serum, as well as in uterine tissue collected on the day of euthanasia. Anti-PEDVS1 colostral IgG antibody titers collected at farrowing were significantly higher relative to the negative control gilts indicating that the NP vaccine was effective in contributing to the colostral antibodies. The PEDVS1-specific colostral IgA and anti-PEDVS1 IgG and IgA antibodies in the mature milk collected 6 days after farrowing were low for both vaccinated groups. No statistical differences between the vaccinated groups were observed, suggesting that the i.u. priming vaccine did not induce mucosal tolerance. Piglets born to either group of vaccinated gilts did not receive sufficient neutralizing antibodies to protect them against infectious PEDV at 3 days of age. In summary, a single i.m. NP vaccine administered 30 days after breeding and a joint i.u./i.m. vaccine administered at breeding and 30 days post-breeding induced significant anti-PEDVS1 immunity in systemic and mucosal sites but did not provide passive protection in suckling offspring.

5.
J Neurosci Methods ; 369: 109480, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-35026308

ABSTRACT

BACKGROUND: Isolation of cell types of interest from the brain for molecular applications presents several challenges, including cellular damage during tissue dissociation or enrichment procedures, and low cell number in the tissue in some cases. Techniques have been developed to enrich distinct cell populations using immunopanning or fluorescence activated cell/nuclei sorting. However, these techniques often involve fixation, immunolabeling and DNA staining steps, which could potentially influence downstream omics applications. NEW METHOD: Taking advantage of readily available genetically modified mice with fluorescent-tagged nuclei, we describe a technique for the purification of cell-type specific brain nuclei, optimized to decrease sample preparation time and to limit potential artefacts for downstream omics applications. We demonstrate the applicability of this approach for the purification of glial cell nuclei and show that the resulting cell-type specific nuclei obtained can be used effectively for omics applications, including ATAC-seq and RNA-seq. RESULTS: We demonstrate excellent enrichment of fluorescently-tagged glial nuclei, yielding high quality RNA and chromatin. We identify several critical steps during nuclei isolation that help limit nuclei rupture and clumping, including quick homogenization, dilution before filtration and loosening of the pellet before resuspension, thus improving yield. Sorting of fluorescent nuclei can be achieved without fixation, antibody labelling, or DAPI staining, reducing potential artifactual results in RNA-seq and ATAC-seq analyses. We show that reproducible glial cell type-specific profiles can be obtained in transcriptomic and chromatin accessibility assays using this rapid protocol. COMPARISON WITH EXISTING METHODS: Our method allows for rapid enrichment of glial nuclei populations from the mouse brain with minimal processing steps, while still providing high quality RNA and chromatin required for reliable omics analyses. CONCLUSIONS: We provide a reproducible method to obtain nucleic material from glial cells in the mouse brain with a quick and limited sample preparation.


Subject(s)
Cell Nucleus , Chromatin , Animals , Brain , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromatin/metabolism , Mice , RNA-Seq
6.
Viruses ; 13(9)2021 09 14.
Article in English | MEDLINE | ID: mdl-34578408

ABSTRACT

The host's immune status may affect virus evolution. Little is known about how developing fetal and placental immune milieus affect virus heterogeneity. This knowledge will help us better understand intra-host virus evolution and how new virus variants emerge. The goal of our study was to find out whether the isolated in utero environment-an environment with specialized placental immunity and developing fetal immunity-supports the emergence of RNA and DNA virus variants. We used well-established porcine models for isolated Zika virus (RNA virus) and porcine circovirus 2 (DNA virus) fetal infections. We found that the isolated in utero environment was conducive to the emergence of RNA and DNA virus variants. Next-generation sequencing of nearly whole virus genomes and validated bioinformatics pipelines identified both unique and convergent single nucleotide variations in virus genomes isolated from different fetuses. Zika virus and PCV2 in utero evolution also resulted in single nucleotide variations previously reported in the human and porcine field samples. These findings should encourage further studies on virus evolution in placenta and fetuses, to better understand how virus variants emerge and how in utero viral evolution affects congenital virus transmission and pathogenicity.


Subject(s)
Circovirus/genetics , Directed Molecular Evolution , Genetic Variation , Placenta/virology , Uterus/virology , Zika Virus/genetics , Animals , Cellular Microenvironment , Chlorocebus aethiops , Circovirus/physiology , Female , Fetus/virology , Genetic Heterogeneity , High-Throughput Nucleotide Sequencing , Placenta/immunology , Pregnancy , Swine , Uterus/immunology , Vero Cells , Viral Load , Zika Virus/physiology
7.
Cell Rep ; 31(13): 107838, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32610139

ABSTRACT

ATRX gene mutations have been identified in syndromic and non-syndromic intellectual disabilities in humans. ATRX is known to maintain genomic stability in neuroprogenitor cells, but its function in differentiated neurons and memory processes remains largely unresolved. Here, we show that the deletion of neuronal Atrx in mice leads to distinct hippocampal structural defects, fewer presynaptic vesicles, and an enlarged postsynaptic area at CA1 apical dendrite-axon junctions. We identify male-specific impairments in long-term contextual memory and in synaptic gene expression, linked to altered miR-137 levels. We show that ATRX directly binds to the miR-137 locus and that the enrichment of the suppressive histone mark H3K27me3 is significantly reduced upon the loss of ATRX. We conclude that the ablation of ATRX in excitatory forebrain neurons leads to sexually dimorphic effects on miR-137 expression and on spatial memory, identifying a potential therapeutic target for neurological defects caused by ATRX dysfunction.


Subject(s)
Gene Deletion , Gene Expression Regulation , Memory Disorders/genetics , Memory Disorders/physiopathology , MicroRNAs/genetics , Sex Characteristics , Spatial Learning , X-linked Nuclear Protein/deficiency , Animals , Base Sequence , CA1 Region, Hippocampal/pathology , CA1 Region, Hippocampal/ultrastructure , Conditioning, Operant , Dendrites/metabolism , Dendrites/ultrastructure , Female , Genotype , Histones/metabolism , Lysine/metabolism , Magnetic Resonance Imaging , Male , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/metabolism , Neurons , Organ Specificity , RNA, Messenger/genetics , RNA, Messenger/metabolism , Synapses/metabolism , Synapses/ultrastructure , X-linked Nuclear Protein/metabolism
8.
J Neurodev Disord ; 12(1): 17, 2020 06 24.
Article in English | MEDLINE | ID: mdl-32580781

ABSTRACT

BACKGROUND: Alpha-thalassemia/mental retardation, X-linked, or ATRX, is an autism susceptibility gene that encodes a chromatin remodeler. Mutations of ATRX result in the ATR-X intellectual disability syndrome and have been identified in autism spectrum disorder (ASD) patients. The mechanisms by which ATRX mutations lead to autism and autistic-like behaviours are not yet known. To address this question, we generated mice with postnatal Atrx inactivation in excitatory neurons of the forebrain and performed a battery of behavioural assays that assess autistic-like behaviours. METHODS: Male and female mice with a postnatal conditional ablation of ATRX were generated using the Cre/lox system under the control of the αCaMKII gene promoter. These mice were tested in a battery of behavioural tests that assess autistic-like features. We utilized paradigms that measure social behaviour, repetitive, and stereotyped behaviours, as well as sensory gating. Statistics were calculated by two-way repeated measures ANOVA with Sidak's multiple comparison test or unpaired Student's t tests as indicated. RESULTS: The behaviour tests revealed no significant differences between Atrx-cKO and control mice. We identified sexually dimorphic changes in odor habituation and discrimination; however, these changes did not correlate with social deficits. CONCLUSION: The postnatal knockout of Atrx in forebrain excitatory neurons does not lead to autism-related behaviours in male or female mice.


Subject(s)
Autistic Disorder/genetics , Mental Retardation, X-Linked/genetics , alpha-Thalassemia/genetics , Animals , Chromatin Assembly and Disassembly , Female , Male , Mice , Mice, Knockout , Mutation , Neurons/metabolism , Postpartum Period , X-linked Nuclear Protein
9.
Viruses ; 12(5)2020 05 25.
Article in English | MEDLINE | ID: mdl-32466170

ABSTRACT

We studied whether cytosine phosphate-guanine (CpG) recoding in a viral genome may provide oncolytic candidates with reduced infection kinetics in nonmalignant brain cells, but with high virulence in glioblastoma stem cells (GSCs). As a model, we used well-characterized CpG-recoded Zika virus vaccine candidates that previously showed genetic stability and safety in animal models. In vitro, one of the CpG-recoded Zika virus variants had reduced infection kinetics in nonmalignant brain cells but high infectivity and oncolytic activity in GSCs as represented by reduced cell proliferation. The recoded virus also efficiently replicated in GSC-derived tumors in ovo with a significant reduction of tumor growth. We also showed that some GSCs may be resistant to Zika virus oncolytic activity, emphasizing the need for personalized oncolytic therapy or a strategy to overcome resistance in GSCs. Collectively, we demonstrated the potential of the CpG recoding approach for oncolytic virus development that encourages further research towards a better understanding of host-tumor-CpG-recoded virus interactions.


Subject(s)
CpG Islands/physiology , Glioblastoma/metabolism , Stem Cells/metabolism , Zika Virus/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , CpG Islands/genetics , Glioblastoma/virology , Humans , Oncolytic Viruses/genetics , Phosphates , Virus Replication , Zika Virus Infection
10.
Cell Death Dis ; 11(5): 311, 2020 05 04.
Article in English | MEDLINE | ID: mdl-32366868

ABSTRACT

Cell fusion occurs when several cells combine to form a multinuclear aggregate (syncytium). In human placenta, a syncytialized trophoblast (syncytiotrophoblast) layer forms the primary interface between maternal and fetal tissue, facilitates nutrient and gas exchange, and produces hormones vital for pregnancy. Syncytiotrophoblast development occurs by differentiation of underlying progenitor cells called cytotrophoblasts, which then fuse into the syncytiotrophoblast layer. Differentiation is associated with chromatin remodeling and specific changes in gene expression mediated, at least in part, by histone acetylation. However, the epigenetic regulation of human cytotrophoblast differentiation and fusion is poorly understood. In this study, we found that human syncytiotrophoblast development was associated with deacetylation of multiple core histone residues. Chromatin immunoprecipitation sequencing revealed chromosomal regions that exhibit dynamic alterations in histone H3 acetylation during differentiation. These include regions containing genes classically associated with cytotrophoblast differentiation (TEAD4, TP63, OVOL1, CGB), as well as near genes with novel regulatory roles in trophoblast development and function, such as LHX4 and SYDE1. Prevention of histone deacetylation using both pharmacological and genetic approaches inhibited trophoblast fusion, supporting a critical role of this process for trophoblast differentiation. Finally, we identified the histone deacetylases (HDACs) HDAC1 and HDAC2 as the critical mediators driving cytotrophoblast differentiation. Collectively, these findings provide novel insights into the epigenetic mechanisms underlying trophoblast fusion during human placental development.


Subject(s)
Cell Differentiation , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Placenta/cytology , Stem Cells/cytology , Trophoblasts/cytology , Trophoblasts/enzymology , Acetylation/drug effects , Cell Differentiation/drug effects , Cell Fusion , Cells, Cultured , Female , Gene Knockdown Techniques , Genome, Human , Giant Cells/drug effects , Giant Cells/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Humans , Pregnancy , Stem Cells/drug effects , Stem Cells/metabolism , Trophoblasts/drug effects
11.
Hippocampus ; 30(6): 565-581, 2020 06.
Article in English | MEDLINE | ID: mdl-31713968

ABSTRACT

α-Thalassemia X-linked intellectual disability (ATR-X) syndrome is a neurodevelopmental disorder caused by mutations in the ATRX gene that encodes a SNF2-type chromatin-remodeling protein. The ATRX protein regulates chromatin structure and gene expression in the developing mouse brain and early inactivation leads to DNA replication stress, extensive cell death, and microcephaly. However, the outcome of Atrx loss of function postnatally in neurons is less well understood. We recently reported that conditional inactivation of Atrx in postnatal forebrain excitatory neurons (ATRX-cKO) causes deficits in long-term hippocampus-dependent spatial memory. Thus, we hypothesized that ATRX-cKO mice will display impaired hippocampal synaptic transmission and plasticity. In the present study, evoked field potentials and current source density analysis were recorded from a multichannel electrode in male, urethane-anesthetized mice. Three major excitatory synapses, the Schaffer collaterals to basal dendrites and proximal apical dendrites, and the temporoammonic path to distal apical dendrites on hippocampal CA1 pyramidal cells were assessed by their baseline synaptic transmission, including paired-pulse facilitation (PPF) at 50-ms interpulse interval, and by their long-term potentiation (LTP) induced by theta-frequency burst stimulation. Baseline single-pulse excitatory response at each synapse did not differ between ATRX-cKO and control mice, but baseline PPF was reduced at the CA1 basal dendritic synapse in ATRX-cKO mice. While basal dendritic LTP of the first-pulse excitatory response was not affected in ATRX-cKO mice, proximal and distal apical dendritic LTP were marginally and significantly reduced, respectively. These results suggest that ATRX is required in excitatory neurons of the forebrain to achieve normal hippocampal LTP and PPF at the CA1 apical and basal dendritic synapses, respectively. Such alterations in hippocampal synaptic transmission and plasticity could explain the long-term spatial memory deficits in ATRX-cKO mice and provide insight into the physiological mechanisms underlying intellectual disability in ATR-X syndrome patients.


Subject(s)
Hippocampus/metabolism , Neuronal Plasticity/physiology , Prosencephalon/metabolism , Synapses/metabolism , X-linked Nuclear Protein/deficiency , Animals , Excitatory Postsynaptic Potentials/physiology , Hippocampus/cytology , Male , Mice , Mice, Knockout , Mice, Transgenic , Prosencephalon/cytology , X-linked Nuclear Protein/genetics
12.
PLoS Pathog ; 15(11): e1008038, 2019 11.
Article in English | MEDLINE | ID: mdl-31725819

ABSTRACT

Zika virus (ZIKV) infection during human pregnancy may lead to severe fetal pathology and debilitating impairments in offspring. However, the majority of infections are subclinical and not associated with evident birth defects. Potentially detrimental life-long health outcomes in asymptomatic offspring evoke high concerns. Thus, animal models addressing sequelae in offspring may provide valuable information. To induce subclinical infection, we inoculated selected porcine fetuses at the mid-stage of development. Inoculation resulted in trans-fetal virus spread and persistent infection in the placenta and fetal membranes for two months. Offspring did not show congenital Zika syndrome (e.g., microcephaly, brain calcifications, congenital clubfoot, arthrogryposis, seizures) or other visible birth defects. However, a month after birth, a portion of offspring exhibited excessive interferon alpha (IFN-α) levels in blood plasma in a regular environment. Most affected offspring also showed dramatic IFN-α shutdown during social stress providing the first evidence for the cumulative impact of prenatal ZIKV exposure and postnatal environmental insult. Other eleven cytokines tested before and after stress were not altered suggesting the specific IFN-α pathology. While brains from offspring did not have histopathology, lesions, and ZIKV, the whole genome expression analysis of the prefrontal cortex revealed profound sex-specific transcriptional changes that most probably was the result of subclinical in utero infection. RNA-seq analysis in the placenta persistently infected with ZIKV provided independent support for the sex-specific pattern of in utero-acquired transcriptional responses. Collectively, our results provide strong evidence that two hallmarks of fetal ZIKV infection, altered type I IFN response and molecular brain pathology can persist after birth in offspring in the absence of congenital Zika syndrome.


Subject(s)
Brain/pathology , Fetal Diseases/epidemiology , Fetus/virology , Interferon-alpha/metabolism , Pregnancy Complications, Infectious/epidemiology , Uterus/virology , Zika Virus Infection/virology , Animals , Antiviral Agents/metabolism , Brain/metabolism , Brain/virology , Communicable Diseases/transmission , Communicable Diseases/virology , Female , Fetal Diseases/metabolism , Fetal Diseases/virology , Fetus/metabolism , Fetus/pathology , Male , Pregnancy , Pregnancy Complications, Infectious/metabolism , Pregnancy Complications, Infectious/virology , Sex Factors , Swine , Uterus/metabolism , Uterus/pathology , Zika Virus/pathogenicity , Zika Virus Infection/pathology , Zika Virus Infection/transmission , Zika Virus Infection/veterinary
13.
Emerg Microbes Infect ; 8(1): 1098-1107, 2019.
Article in English | MEDLINE | ID: mdl-31340725

ABSTRACT

Studies in mice showed that African Zika virus (ZIKV) strains cause more damage in embryos. These studies, however, were limited to the mouse-adapted African MR766 strain or infection at early gestation. Here, we compared infection of Asian and African strains in the fetal pig model at midgestation. Both strains caused fetal infection. ZIKV was detected in placenta, amniotic membrane, amniotic fluid, fetal blood, and brain. The African strain produced more vigorous in utero infection as represented by more efficient virus transmission between siblings, and higher viral loads in fetal organs and membranes. Infection with both strains was associated with reduced fetal brain weight and increased number of placental CD163-positive cells, as well as elevated in utero interferon alpha and cortisol levels. This is the first large animal model study which demonstrated that African strain of ZIKV, with no passage history in experimental animals, can cause persistent infection in fetuses and fetal membranes at midgestation. Our studies also suggest that similar to Asian strains, ZIKV of African lineage might cause silent pathology which is difficult to identify in deceptively healthy fetuses. The findings emphasize the need for further studies to highlight the impact of ZIKV heterogeneity on infection outcomes during pregnancy.


Subject(s)
Fetal Diseases/virology , Pregnancy Complications, Infectious/virology , Zika Virus Infection/virology , Zika Virus/physiology , Animals , Brain/virology , Disease Models, Animal , Female , Humans , Placenta/virology , Pregnancy , Swine , Uterus/virology , Zika Virus/classification , Zika Virus/genetics , Zika Virus Infection/transmission
14.
Front Immunol ; 10: 3077, 2019.
Article in English | MEDLINE | ID: mdl-32038625

ABSTRACT

Experimental increase of CpG dinucleotides in an RNA virus genome impairs infection providing a promising approach for vaccine development. While CpG recoding is an emerging and promising vaccine approach, little is known about infection phenotypes caused by recoded viruses in vivo. For example, infection phenotypes, immunogenicity, and protective efficacy induced by CpG-recoded viruses in different age groups were not studied yet. This is important, because attenuation of infection phenotypes caused by recoded viruses may depend on the population-based expression of cellular components targeting viral CpG dinucleotides. In the present study, we generated several Zika virus (ZIKV) variants with the increasing CpG content and compared infection in neonatal and adult mice. Increasing the CpG content caused host-age-dependent attenuation of infection with considerable attenuation in neonates and high attenuation in adults. Expression of the zinc-finger antiviral protein (ZAP)-the host protein targeting viral CpG dinucleotides-was also age-dependent. Similar to the wild-type virus, ZIKV variants with the increased CpG content evoked robust cellular and humoral immune responses and protection against lethal challenge. Collectively, the host age should be accounted for in future studies on mechanisms targeting viral CpG dinucleotides, development of safe dinucleotide recoding strategies, and applications of CpG-recoded vaccines.


Subject(s)
CpG Islands/genetics , Genome, Viral/genetics , Viral Vaccines/genetics , Zika Virus Infection/immunology , Zika Virus/physiology , Age Factors , Animals , Animals, Newborn , Cell Line , Disease Resistance , Humans , Immunity, Cellular , Immunity, Heterologous , Immunity, Humoral , Mice , Mice, Inbred C57BL , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
15.
J Neurosci ; 39(1): 177-192, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30377227

ABSTRACT

The CCCTC-binding factor (CTCF) is a central regulator of chromatin topology recently linked to neurodevelopmental disorders such as intellectual disability, autism, and schizophrenia. The aim of this study was to identify novel roles of CTCF in the developing mouse brain. We provide evidence that CTCF is required for the expression of the LIM homeodomain factor LHX6 involved in fate determination of cortical interneurons (CINs) that originate in the medial ganglionic eminence (MGE). Conditional Ctcf ablation in the MGE of mice of either sex leads to delayed tangential migration, abnormal distribution of CIN in the neocortex, a marked reduction of CINs expressing parvalbumin and somatostatin (Sst), and an increased number of MGE-derived cells expressing Lhx8 and other markers of basal forebrain projection neurons. Likewise, Ctcf-null MGE cells transplanted into the cortex of wild-type hosts generate fewer Sst-expressing CINs and exhibit lamination defects that are efficiently rescued upon reexpression of LHX6. Collectively, these data indicate that CTCF regulates the dichotomy between Lhx6 and Lhx8 to achieve correct specification and migration of MGE-derived CINs.SIGNIFICANCE STATEMENT This work provides evidence that CCCTC-binding factor (CTCF) controls an early fate decision point in the generation of cortical interneurons mediated at least in part by Lhx6. Importantly, the abnormalities described could reflect early molecular and cellular events that contribute to human neurological disorders previously linked to CTCF, including schizophrenia, autism, and intellectual disability.


Subject(s)
CCCTC-Binding Factor/physiology , Cerebral Cortex/physiology , Interneurons/physiology , Median Eminence/physiology , Animals , CCCTC-Binding Factor/genetics , Cell Count , Cell Movement/genetics , Cell Movement/physiology , Cerebral Cortex/cytology , Female , LIM-Homeodomain Proteins/biosynthesis , LIM-Homeodomain Proteins/genetics , Male , Median Eminence/cytology , Mice , Mice, Inbred C57BL , Neocortex/cytology , Neocortex/physiology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Parvalbumins/metabolism , Somatostatin/metabolism , Telencephalon/cytology , Telencephalon/growth & development , Transcription Factors/biosynthesis , Transcription Factors/genetics , gamma-Aminobutyric Acid/physiology
16.
Aging (Albany NY) ; 10(6): 1223-1238, 2018 Jun 07.
Article in English | MEDLINE | ID: mdl-29883366

ABSTRACT

ATRX is an ATP-dependent chromatin remodeler required for the maintenance of genomic integrity. We previously reported that conditional Atrx ablation in the mouse embryonic forebrain and anterior pituitary using the Foxg1cre driver causes reduced health and lifespan. In these mice, premature aging-like phenotypes were accompanied by low circulating levels of insulin-like growth factor 1 (IGF-1) and thyroxine (T4), hormones that maintain stem cell pools and normal metabolic profiles, respectively. Based on emerging evidence that T4 stimulates expression of IGF-1 in pre-pubertal mice, we tested whether T4 supplementation in Atrx Foxg1cre mice could restore IGF-1 levels and ameliorate premature aging-like phenotypes. Despite restoration of normal serum T4 levels, we did not observe improvements in circulating IGF-1. In the liver, thyroid hormone target genes were differentially affected upon T4 treatment, with Igf1 and several other thyroid hormone responsive genes failing to recover normal expression levels. These findings hinted at Cre-mediated Atrx inactivation in the liver of Atrx Foxg1cre mice, which we confirmed. We conclude that the phenotypes observed in the Atrx Foxg1cre mice can be explained in part by a role of ATRX in the liver to promote T4-mediated Igf1 expression, thus explaining the inefficacy of T4 therapy observed in this study.


Subject(s)
Aging/physiology , Forkhead Transcription Factors/metabolism , Liver/metabolism , Nerve Tissue Proteins/metabolism , Thyroxine/pharmacology , X-linked Nuclear Protein/metabolism , Animals , Blood Glucose/drug effects , Forkhead Transcription Factors/genetics , Gene Expression Regulation/drug effects , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Subcutaneous Fat/physiology , X-linked Nuclear Protein/genetics
17.
J Neurosci ; 37(45): 10773-10782, 2017 11 08.
Article in English | MEDLINE | ID: mdl-29118205

ABSTRACT

Intellectual disability (ID) is a prevailing neurodevelopmental condition associated with impaired cognitive and adaptive behaviors. Many chromatin-modifying enzymes and other epigenetic regulators have been genetically associated with ID disorders (IDDs). Here we review how alterations in the function of histone modifiers, chromatin remodelers, and methyl-DNA binding proteins contribute to neurodevelopmental defects and altered brain plasticity. We also discuss how progress in human genetics has led to the generation of mouse models that unveil the molecular etiology of ID, and outline the direction in which this field is moving to identify therapeutic strategies for IDDs. Importantly, because the chromatin regulators linked to IDDs often target common downstream genes and cellular processes, the impact of research in individual syndromes goes well beyond each syndrome and can also contribute to the understanding and therapy of other IDDs. Furthermore, the investigation of these disorders helps us to understand the role of chromatin regulators in brain development, plasticity, and gene expression, thereby answering fundamental questions in neurobiology.


Subject(s)
Epigenesis, Genetic/genetics , Intellectual Disability/etiology , Intellectual Disability/genetics , Epigenomics , Humans
18.
Epigenetics Chromatin ; 10: 10, 2017.
Article in English | MEDLINE | ID: mdl-28293299

ABSTRACT

BACKGROUND: Alpha thalassemia/mental retardation X-linked syndrome (ATR-X) is caused by a mutation at the chromatin regulator gene ATRX. The mechanisms involved in the ATR-X pathology are not completely understood, but may involve epigenetic modifications. ATRX has been linked to the regulation of histone H3 and DNA methylation, while mutations in the ATRX gene may lead to the downstream epigenetic and transcriptional effects. Elucidating the underlying epigenetic mechanisms altered in ATR-X will provide a better understanding about the pathobiology of this disease, as well as provide novel diagnostic biomarkers. RESULTS: We performed genome-wide DNA methylation assessment of the peripheral blood samples from 18 patients with ATR-X and compared it to 210 controls. We demonstrated the evidence of a unique and highly specific DNA methylation "epi-signature" in the peripheral blood of ATRX patients, which was corroborated by targeted bisulfite sequencing experiments. Although genomically represented, differentially methylated regions showed evidence of preferential clustering in pericentromeric and telometric chromosomal regions, areas where ATRX has multiple functions related to maintenance of heterochromatin and genomic integrity. CONCLUSION: Most significant methylation changes in the 14 genomic loci provide a unique epigenetic signature for this syndrome that may be used as a highly sensitive and specific diagnostic biomarker to support the diagnosis of ATR-X, particularly in patients with phenotypic complexity and in patients with ATRX gene sequence variants of unknown significance.


Subject(s)
Epigenesis, Genetic , Mental Retardation, X-Linked/genetics , alpha-Thalassemia/genetics , Adolescent , Adult , Child , Child, Preschool , CpG Islands , DNA/chemistry , DNA/isolation & purification , DNA/metabolism , DNA Methylation , Genotype , Humans , Infant , Male , Mental Retardation, X-Linked/pathology , Middle Aged , Promoter Regions, Genetic , Sequence Analysis, DNA , X-linked Nuclear Protein/genetics , X-linked Nuclear Protein/metabolism , Young Adult , alpha-Thalassemia/pathology
19.
Dis Model Mech ; 10(2): 119-126, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28093507

ABSTRACT

The rapid modulation of chromatin organization is thought to play a crucial role in cognitive processes such as memory consolidation. This is supported in part by the dysregulation of many chromatin-remodelling proteins in neurodevelopmental and psychiatric disorders. A key example is ATRX, an X-linked gene commonly mutated in individuals with syndromic and nonsyndromic intellectual disability. The consequences of Atrx inactivation for learning and memory have been difficult to evaluate because of the early lethality of hemizygous-null animals. In this study, we evaluated the outcome of brain-specific Atrx deletion in heterozygous female mice. These mice exhibit a mosaic pattern of ATRX protein expression in the central nervous system attributable to the location of the gene on the X chromosome. Although the hemizygous male mice die soon after birth, heterozygous females survive to adulthood. Body growth is stunted in these animals, and they have low circulating concentrations of insulin growth factor 1. In addition, they are impaired in spatial, contextual fear and novel object recognition memory. Our findings demonstrate that mosaic loss of ATRX expression in the central nervous system leads to endocrine defects and decreased body size and has a negative impact on learning and memory.


Subject(s)
Central Nervous System/pathology , Memory Disorders/physiopathology , Mosaicism , X-linked Nuclear Protein/metabolism , Animals , Body Weight , Brain/metabolism , Brain/pathology , Brain/physiopathology , Central Nervous System/metabolism , Central Nervous System/physiopathology , Fear , Female , Gene Deletion , Gene Expression Regulation , Growth and Development , Hand Strength , Heterozygote , Hindlimb/pathology , Insulin-Like Growth Factor I/metabolism , Maze Learning , Memory Disorders/blood , Memory Disorders/genetics , Memory, Short-Term , Mice , Motor Activity , Phenotype , Spatial Memory , Survival Analysis , X-linked Nuclear Protein/genetics
20.
Virol J ; 13: 57, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27036203

ABSTRACT

BACKGROUND: Porcine epidemic diarrhea virus (PEDV) is a highly contagious virus infecting pigs of all ages with high morbidity and mortality among newborn piglets. Currently, there is no effective vaccine available to protect the pigs from PEDV. The N-terminal subunit of spike protein (S1) is responsible for virus binding to the cellular receptor and contains a number of neutralizing antibody epitopes. Thus, we expressed and produced recombinant S1 protein to protect newborn piglets by immunization of sows. METHODS: Affinity tagged PEDV S1 protein was expressed in a secretory form in yeast, insect and mammalian cells to identify the most suitable production system. Purified recombinant protein was analysed by SDS-PAGE, Western blot and deglycosylation assay. A pregnant sow was intramuscularly immunized three times with adjuvanted recombinant protein prior to farrowing. PEDV-specific immune responses in sera and colostrum of the sow and piglets were assayed by ELISA and virus neutralization assays. Piglets were challenged orally with PEDV, and clinical parameters were monitored for 6 days post-challenge. RESULTS AND CONCLUSION: Of three eukaryotic expression systems tested (yeast, insect-cell, and mammalian), expression by HEK-293 T cells gave the highest yield of protein that was N-glycosylated and was the most appropriate candidate for vaccination. Administration of the subunit vaccine in a sow resulted in induction of S1-specific IgG and IgA that were passively transferred to the suckling piglets. Also, high virus neutralization titres were observed in the serum of the vaccinated sow and its piglets. After PEDV challenge, piglets born to the vaccinated sow exhibited less severe signs of disease and significantly lower mortality compared to the piglets of a control sow. However, there were no significant differences in diarrhea, body weight and virus shedding. Thus, vaccination with S1 subunit vaccine failed to provide complete protection to suckling piglets after challenge exposure, and further improvements are needed for the development of a subunit vaccine that fully protects against PEDV infection.


Subject(s)
Antigens, Viral/immunology , Coronavirus Infections/veterinary , Porcine epidemic diarrhea virus/immunology , Spike Glycoprotein, Coronavirus/immunology , Viral Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Neutralizing/analysis , Antibodies, Viral/analysis , Antigens, Viral/genetics , Colostrum/immunology , Coronavirus Infections/pathology , Coronavirus Infections/prevention & control , Enzyme-Linked Immunosorbent Assay , Female , Injections, Intramuscular , Neutralization Tests , Porcine epidemic diarrhea virus/genetics , Pregnancy , Serum/immunology , Spike Glycoprotein, Coronavirus/genetics , Swine , Treatment Outcome , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...