Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 52
Filtrer
1.
Brain ; 147(9): 3099-3112, 2024 Sep 03.
Article de Anglais | MEDLINE | ID: mdl-39028640

RÉSUMÉ

Huntington's disease and juvenile-onset schizophrenia have long been regarded as distinct disorders. However, both manifest cell-intrinsic abnormalities in glial differentiation, with resultant astrocytic dysfunction and hypomyelination. To assess whether a common mechanism might underlie the similar glial pathology of these otherwise disparate conditions, we used comparative correlation network approaches to analyse RNA-sequencing data from human glial progenitor cells (hGPCs) produced from disease-derived pluripotent stem cells. We identified gene sets preserved between Huntington's disease and schizophrenia hGPCs yet distinct from normal controls that included 174 highly connected genes in the shared disease-associated network, focusing on genes involved in synaptic signalling. These synaptic genes were largely suppressed in both schizophrenia and Huntington's disease hGPCs, and gene regulatory network analysis identified a core set of upstream regulators of this network, of which OLIG2 and TCF7L2 were prominent. Among their downstream targets, ADGRL3, a modulator of glutamatergic synapses, was notably suppressed in both schizophrenia and Huntington's disease hGPCs. Chromatin immunoprecipitation sequencing confirmed that OLIG2 and TCF7L2 each bound to the regulatory region of ADGRL3, whose expression was then rescued by lentiviral overexpression of these transcription factors. These data suggest that the disease-associated suppression of OLIG2 and TCF7L2-dependent transcription of glutamate signalling regulators may impair glial receptivity to neuronal glutamate. The consequent loss of activity-dependent mobilization of hGPCs may yield deficient oligodendrocyte production, and hence the hypomyelination noted in these disorders, as well as the disrupted astrocytic differentiation and attendant synaptic dysfunction associated with each. Together, these data highlight the importance of convergent glial molecular pathology in both the pathogenesis and phenotypic similarities of two otherwise unrelated disorders, Huntington's disease and schizophrenia.


Sujet(s)
Maladie de Huntington , Névroglie , Schizophrénie , Maladie de Huntington/génétique , Maladie de Huntington/métabolisme , Maladie de Huntington/anatomopathologie , Humains , Schizophrénie/génétique , Schizophrénie/métabolisme , Névroglie/métabolisme , Facteur de transcription-2 des oligodendrocytes/métabolisme , Facteur de transcription-2 des oligodendrocytes/génétique , Réseaux de régulation génique , Cellules souches pluripotentes/métabolisme
2.
Cell ; 187(10): 2465-2484.e22, 2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38701782

RÉSUMÉ

Remyelination failure in diseases like multiple sclerosis (MS) was thought to involve suppressed maturation of oligodendrocyte precursors; however, oligodendrocytes are present in MS lesions yet lack myelin production. We found that oligodendrocytes in the lesions are epigenetically silenced. Developing a transgenic reporter labeling differentiated oligodendrocytes for phenotypic screening, we identified a small-molecule epigenetic-silencing-inhibitor (ESI1) that enhances myelin production and ensheathment. ESI1 promotes remyelination in animal models of demyelination and enables de novo myelinogenesis on regenerated CNS axons. ESI1 treatment lengthened myelin sheaths in human iPSC-derived organoids and augmented (re)myelination in aged mice while reversing age-related cognitive decline. Multi-omics revealed that ESI1 induces an active chromatin landscape that activates myelinogenic pathways and reprograms metabolism. Notably, ESI1 triggered nuclear condensate formation of master lipid-metabolic regulators SREBP1/2, concentrating transcriptional co-activators to drive lipid/cholesterol biosynthesis. Our study highlights the potential of targeting epigenetic silencing to enable CNS myelin regeneration in demyelinating diseases and aging.


Sujet(s)
Épigenèse génétique , Gaine de myéline , Oligodendroglie , Remyélinisation , Animaux , Gaine de myéline/métabolisme , Humains , Souris , Remyélinisation/effets des médicaments et des substances chimiques , Oligodendroglie/métabolisme , Système nerveux central/métabolisme , Souris de lignée C57BL , Rajeunissement , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/effets des médicaments et des substances chimiques , Protéine-1 de liaison à l'élément de régulation des stérols/métabolisme , Organoïdes/métabolisme , Organoïdes/effets des médicaments et des substances chimiques , Maladies démyélinisantes/métabolisme , Maladies démyélinisantes/génétique , Différenciation cellulaire/effets des médicaments et des substances chimiques , Bibliothèques de petites molécules/pharmacologie , Mâle , Régénération/effets des médicaments et des substances chimiques , Sclérose en plaques/métabolisme , Sclérose en plaques/génétique , Sclérose en plaques/traitement médicamenteux , Sclérose en plaques/anatomopathologie
3.
Sci Rep ; 13(1): 12433, 2023 08 01.
Article de Anglais | MEDLINE | ID: mdl-37528157

RÉSUMÉ

Low-grade and secondary high-grade gliomas frequently contain mutations in the IDH1 or IDH2 metabolic enzymes that are hypothesized to drive tumorigenesis by inhibiting many of the chromatin-regulating enzymes that regulate DNA structure. Histone deacetylase inhibitors are promising anti-cancer agents and have already been used in clinical trials. However, a clear understanding of their mechanism or gene targets is lacking. In this study, the authors genetically dissect patient-derived IDH1 mutant cultures to determine which HDAC enzymes drive growth in IDH1 mutant gliomas. A panel of patient-derived gliomasphere cell lines (2 IDH1 mutant lines, 3 IDH1 wildtype lines) were subjected to a drug-screen of epigenetic modifying drugs from different epigenetic classes. The effect of LBH (panobinostat) on gene expression and chromatin structure was tested on patient-derived IDH1 mutant lines. The role of each of the highly expressed HDAC enzymes was molecularly dissected using lentiviral RNA interference knock-down vectors and a patient-derived IDH1 mutant in vitro model of glioblastoma (HK252). These results were then confirmed in an in vivo xenotransplant model (BT-142). The IDH1 mutation leads to gene down-regulation, DNA hypermethylation, increased DNA accessibility and H3K27 hypo-acetylation in two distinct IDH1 mutant over-expression models. The drug screen identified histone deacetylase inhibitors (HDACi) and panobinostat (LBH) more specifically as the most selective compounds to inhibit growth in IDH1 mutant glioma lines. Of the eleven annotated HDAC enzymes (HDAC1-11) only six are expressed in IDH1 mutant glioma tissue samples and patient-derived gliomasphere lines (HDAC1-4, HDAC6, and HDAC9). Lentiviral knock-down experiments revealed that HDAC1 and HDAC6 are the most consistently essential for growth both in vitro and in vivo and target very different gene modules. Knock-down of HDAC1 or HDAC6 in vivo led to a more circumscribed less invasive tumor. The gene dysregulation induced by the IDH1 mutation is wide-spread and only partially reversible by direct IDH1 inhibition. This study identifies HDAC1 and HDAC6 as important and drug-targetable enzymes that are necessary for growth and invasiveness in IDH1 mutant gliomas.


Sujet(s)
Antinéoplasiques , Tumeurs du cerveau , Gliome , Humains , Panobinostat/pharmacologie , Panobinostat/usage thérapeutique , Inhibiteurs de désacétylase d'histone/pharmacologie , Inhibiteurs de désacétylase d'histone/usage thérapeutique , Gliome/métabolisme , Antinéoplasiques/usage thérapeutique , Chromatine , Isocitrate dehydrogenases/génétique , Isocitrate dehydrogenases/métabolisme , Mutation , Tumeurs du cerveau/anatomopathologie , Histone Deacetylase 1/génétique , Histone deacetylase 6/génétique
4.
Nat Cell Biol ; 25(2): 323-336, 2023 02.
Article de Anglais | MEDLINE | ID: mdl-36732631

RÉSUMÉ

Nuclear localization of HIPPO-YAP fusion proteins has been implicated in supratentorial ependymoma development. Here, unexpectedly, we find that liquid-liquid phase separation, rather than nuclear localization, of recurrent patient-derived YAP fusions, YAP-MAMLD1 and C11ORF95-YAP, underlies ependymoma tumourigenesis from neural progenitor cells. Mutagenesis and chimaera assays demonstrate that an intrinsically disordered region promotes oligomerization of the YAP fusions into nuclear, puncta-like, membrane-less condensates. Oligomerization and nuclear condensates induced by YAP fusion with a coiled-coil domain of transcriptional activator GCN4 also promote ependymoma formation. YAP-MAMLD1 concentrates transcription factors and co-activators, including BRD4, MED1 and TEAD, in condensates while excluding transcriptional repressive PRC2, and induces long-range enhancer-promoter interactions that promote transcription and oncogenic programmes. Blocking condensate-mediated transcriptional co-activator activity inhibits tumourigenesis, indicating a critical role of liquid phase separation for YAP fusion oncogenic activity in ependymoma. YAP fusions containing the intrinsically disordered region features are common in human tumours, suggesting that nuclear condensates could be targeted to treat YAP-fusion-induced cancers.


Sujet(s)
Épendymome , Facteurs de transcription , Humains , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Carcinogenèse/génétique , Protéines du cycle cellulaire/métabolisme , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/métabolisme , Épendymome/génétique , Épendymome/métabolisme , Épendymome/anatomopathologie , Protéines nucléaires/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Protéines de signalisation YAP , Noyau de la cellule/métabolisme , Transcription génétique
5.
Nat Commun ; 14(1): 762, 2023 02 10.
Article de Anglais | MEDLINE | ID: mdl-36765089

RÉSUMÉ

MYC-driven medulloblastomas are highly aggressive childhood brain tumors, however, the molecular and genetic events triggering MYC amplification and malignant transformation remain elusive. Here we report that mutations in CTDNEP1, a CTD nuclear-envelope-phosphatase, are the most significantly enriched recurrent alterations in MYC-driven medulloblastomas, and define high-risk subsets with poorer prognosis. Ctdnep1 ablation promotes the transformation of murine cerebellar progenitors into Myc-amplified medulloblastomas, resembling their human counterparts. CTDNEP1 deficiency stabilizes and activates MYC activity by elevating MYC serine-62 phosphorylation, and triggers chromosomal instability to induce p53 loss and Myc amplifications. Further, phosphoproteomics reveals that CTDNEP1 post-translationally modulates the activities of key regulators for chromosome segregation and mitotic checkpoint regulators including topoisomerase TOP2A and checkpoint kinase CHEK1. Co-targeting MYC and CHEK1 activities synergistically inhibits CTDNEP1-deficient MYC-amplified tumor growth and prolongs animal survival. Together, our studies demonstrate that CTDNEP1 is a tumor suppressor in highly aggressive MYC-driven medulloblastomas by controlling MYC activity and mitotic fidelity, pointing to a CTDNEP1-dependent targetable therapeutic vulnerability.


Sujet(s)
Tumeurs du cerveau , Tumeurs du cervelet , Médulloblastome , Humains , Souris , Animaux , Enfant , Médulloblastome/anatomopathologie , Phosphoric monoester hydrolases/génétique , Tumeurs du cervelet/anatomopathologie , Transformation cellulaire néoplasique/génétique , Instabilité du génome , Protéines proto-oncogènes c-myc/génétique , Phosphoprotein Phosphatases/génétique
6.
Nature ; 612(7941): 787-794, 2022 12.
Article de Anglais | MEDLINE | ID: mdl-36450980

RÉSUMÉ

Medulloblastoma (MB) is the most common malignant childhood brain tumour1,2, yet the origin of the most aggressive subgroup-3 form remains elusive, impeding development of effective targeted treatments. Previous analyses of mouse cerebella3-5 have not fully defined the compositional heterogeneity of MBs. Here we undertook single-cell profiling of freshly isolated human fetal cerebella to establish a reference map delineating hierarchical cellular states in MBs. We identified a unique transitional cerebellar progenitor connecting neural stem cells to neuronal lineages in developing fetal cerebella. Intersectional analysis revealed that the transitional progenitors were enriched in aggressive MB subgroups, including group 3 and metastatic tumours. Single-cell multi-omics revealed underlying regulatory networks in the transitional progenitor populations, including transcriptional determinants HNRNPH1 and SOX11, which are correlated with clinical prognosis in group 3 MBs. Genomic and Hi-C profiling identified de novo long-range chromatin loops juxtaposing HNRNPH1/SOX11-targeted super-enhancers to cis-regulatory elements of MYC, an oncogenic driver for group 3 MBs. Targeting the transitional progenitor regulators inhibited MYC expression and MYC-driven group 3 MB growth. Our integrated single-cell atlases of human fetal cerebella and MBs show potential cell populations predisposed to transformation and regulatory circuitries underlying tumour cell states and oncogenesis, highlighting hitherto unrecognized transitional progenitor intermediates predictive of disease prognosis and potential therapeutic vulnerabilities.


Sujet(s)
Tumeurs du cerveau , Transformation cellulaire néoplasique , Foetus , Médulloblastome , Humains , Tumeurs du cerveau/anatomopathologie , Transformation cellulaire néoplasique/anatomopathologie , Tumeurs du cervelet/anatomopathologie , Cervelet/cytologie , Cervelet/anatomopathologie , Foetus/cytologie , Foetus/anatomopathologie , Médulloblastome/anatomopathologie , Cellules souches neurales/cytologie , Cellules souches neurales/anatomopathologie , Pronostic
7.
J Neurosci ; 42(45): 8542-8555, 2022 11 09.
Article de Anglais | MEDLINE | ID: mdl-36198499

RÉSUMÉ

The oligodendrocyte (OL) lineage transcription factor Olig2 is expressed throughout oligodendroglial development and is essential for oligodendroglial progenitor specification and differentiation. It was previously reported that deletion of Olig2 enhanced the maturation and myelination of immature OLs and accelerated the remyelination process. However, by analyzing multiple Olig2 conditional KO mouse lines (male and female), we conclude that Olig2 has the opposite effect and is required for OL maturation and remyelination. We found that deletion of Olig2 in immature OLs driven by an immature OL-expressing Plp1 promoter resulted in defects in OL maturation and myelination, and did not enhance remyelination after demyelination. Similarly, Olig2 deletion during premyelinating stages in immature OLs using Mobp or Mog promoter-driven Cre lines also did not enhance OL maturation in the CNS. Further, we found that Olig2 was not required for myelin maintenance in mature OLs but was critical for remyelination after lysolecithin-induced demyelinating injury. Analysis of genomic occupancy in immature and mature OLs revealed that Olig2 targets the enhancers of key myelination-related genes for OL maturation from immature OLs. Together, by leveraging multiple immature OL-expressing Cre lines, these studies indicate that Olig2 is essential for differentiation and myelination of immature OLs and myelin repair. Our findings raise fundamental questions about the previously proposed role of Olig2 in opposing OL myelination and highlight the importance of using Cre-dependent reporter(s) for lineage tracing in studying cell state progression.SIGNIFICANCE STATEMENT Identification of the regulators that promote oligodendrocyte (OL) myelination and remyelination is important for promoting myelin repair in devastating demyelinating diseases. Olig2 is expressed throughout OL lineage development. Ablation of Olig2 was reported to induce maturation, myelination, and remyelination from immature OLs. However, lineage-mapping analysis of Olig2-ablated cells was not conducted. Here, by leveraging multiple immature OL-expressing Cre lines, we observed no evidence that Olig2 ablation promotes maturation or remyelination of immature OLs. Instead, we find that Olig2 is required for immature OL maturation, myelination, and myelin repair. These data raise fundamental questions about the proposed inhibitory role of Olig2 against OL maturation and remyelination. Our findings highlight the importance of validating genetic manipulation with cell lineage tracing in studying myelination.


Sujet(s)
Maladies démyélinisantes , Remyélinisation , Animaux , Femelle , Mâle , Souris , Différenciation cellulaire , Maladies démyélinisantes/métabolisme , Gaine de myéline/métabolisme , Facteur de transcription-2 des oligodendrocytes/génétique , Facteur de transcription-2 des oligodendrocytes/métabolisme , Oligodendroglie/métabolisme , Souris knockout
8.
J Neurosci ; 42(44): 8373-8392, 2022 11 02.
Article de Anglais | MEDLINE | ID: mdl-36127134

RÉSUMÉ

The chromatin remodeler CHD8 represents a high-confidence risk factor in autism, a multistage progressive neurologic disorder, however the underlying stage-specific functions remain elusive. In this study, by analyzing Chd8 conditional knock-out mice (male and female), we find that CHD8 controls cortical neural stem/progenitor cell (NSC) proliferation and survival in a stage-dependent manner. Strikingly, inducible genetic deletion reveals that CHD8 is required for the production and fitness of transit-amplifying intermediate progenitors (IPCs) essential for upper-layer neuron expansion in the embryonic cortex. p53 loss of function partially rescues apoptosis and neurogenesis defects in the Chd8-deficient brain. Further, transcriptomic and epigenomic profiling indicates that CHD8 regulates the chromatin accessibility landscape to activate neurogenesis-promoting factors including TBR2, a key regulator of IPC neurogenesis, while repressing DNA damage- and p53-induced apoptotic programs. In the adult brain, CHD8 depletion impairs forebrain neurogenesis by impeding IPC differentiation from NSCs in both subventricular and subgranular zones; however, unlike in embryos, it does not affect NSC proliferation and survival. Treatment with an antidepressant approved by the Federal Drug Administration (FDA), fluoxetine, partially restores adult hippocampal neurogenesis in Chd8-ablated mice. Together, our multistage functional studies identify temporally specific roles for CHD8 in developmental and adult neurogenesis, pointing to a potential strategy to enhance neurogenesis in the CHD8-deficient brain.SIGNIFICANCE STATEMENT The role of the high-confidence autism gene CHD8 in neurogenesis remains incompletely understood. Here, we identify a stage-specific function of CHD8 in development of NSCs in developing and adult brains by conserved, yet spatiotemporally distinct, mechanisms. In embryonic cortex, CHD8 is critical for the proliferation, survival, and differentiation of both NSC and IPCs during cortical neurogenesis. In adult brain, CHD8 is required for IPC generation but not the proliferation and survival of adult NSCs. Treatment with FDA-approved antidepressant fluoxetine partially rescues the adult neurogenesis defects in CHD8 mutants. Thus, our findings help resolve CHD8 functions throughout life during embryonic and adult neurogenesis and point to a potential avenue to promote neurogenesis in CHD8 deficiency.


Sujet(s)
Trouble autistique , Chromatine , Protéines de liaison à l'ADN , Neurogenèse , Animaux , Femelle , Mâle , Souris , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Fluoxétine , Hippocampe/métabolisme , Souris knockout , Neurogenèse/physiologie , Protéine p53 suppresseur de tumeur , Prosencéphale
9.
Sci Rep ; 12(1): 8221, 2022 05 17.
Article de Anglais | MEDLINE | ID: mdl-35581287

RÉSUMÉ

The pathological changes in epigenetics and gene regulation that accompany the progression of low-grade to high-grade gliomas are under-studied. The authors use a large set of paired atac-seq and RNA-seq data from surgically resected glioma specimens to infer gene regulatory relationships in glioma. Thirty-eight glioma patient samples underwent atac-seq sequencing and 16 samples underwent additional RNA-seq analysis. Using an atac-seq/RNA-seq correlation matrix, atac-seq peaks were paired with genes based on high correlation values (|r2| > 0.6). Samples clustered by IDH1 status but not by grade. Surprisingly there was a trend for IDH1 mutant samples to have more peaks. The majority of peaks are positively correlated with survival and positively correlated with gene expression. Constructing a model of the top six atac-seq peaks created a highly accurate survival prediction model (r2 = 0.68). Four of these peaks were still significant after controlling for age, grade, pathology, IDH1 status and gender. Grade II, III, and IV (primary) samples have similar transcription factors and gene modules. However, grade IV (recurrent) samples have strikingly few peaks. Patient-derived glioma cultures showed decreased peak counts following radiation indicating that this may be radiation-induced. This study supports the notion that IDH1 mutant and IDH1 wildtype gliomas have different epigenetic landscapes and that accessible chromatin sites mapped by atac-seq peaks tend to be positively correlated with expression. The data in this study leads to a new model of treatment response wherein glioma cells respond to radiation therapy by closing open regions of DNA.


Sujet(s)
Gliome , Chromatine/génétique , Séquençage après immunoprécipitation de la chromatine , ADN/génétique , Gliome/génétique , Gliome/anatomopathologie , Humains , Facteurs de transcription/génétique
10.
Blood ; 138(3): 221-233, 2021 07 22.
Article de Anglais | MEDLINE | ID: mdl-34292326

RÉSUMÉ

The Chd8 gene encodes a member of the chromodomain helicase DNA-binding (CHD) family of SNF2H-like adenosine triphosphate (ATP)-dependent chromatin remodeler, the mutations of which define a subtype of autism spectrum disorders. Increasing evidence from recent studies indicates that ATP-dependent chromatin-remodeling genes are involved in the control of crucial gene-expression programs in hematopoietic stem/progenitor cell (HSPC) regulation. In this study, we identified CHD8 as a specific and essential regulator of normal hematopoiesis. Loss of Chd8 leads to severe anemia, pancytopenia, bone marrow failure, and engraftment failure related to a drastic depletion of HSPCs. CHD8 forms a complex with ATM and its deficiency increases chromatin accessibility and drives genomic instability in HSPCs causing an activation of ATM kinase that further stabilizes P53 protein by phosphorylation and leads to increased HSPC apoptosis. Deletion of P53 rescues the apoptotic defects of HSPCs and restores overall hematopoiesis in Chd8-/- mice. Our findings demonstrate that chromatin organization by CHD8 is uniquely necessary for the maintenance of hematopoiesis by integrating the ATM-P53-mediated survival of HSPCs.


Sujet(s)
Protéines de liaison à l'ADN/métabolisme , Hématopoïèse , Cellules souches hématopoïétiques/cytologie , Protéine p53 suppresseur de tumeur/métabolisme , Animaux , Apoptose , Protéines mutées dans l'ataxie-télangiectasie/métabolisme , Survie cellulaire , Cellules cultivées , Protéines de liaison à l'ADN/génétique , Délétion de gène , Cellules souches hématopoïétiques/métabolisme , Souris , Pancytopénie/génétique , Pancytopénie/métabolisme , Stabilité protéique
11.
Transl Psychiatry ; 11(1): 225, 2021 04 17.
Article de Anglais | MEDLINE | ID: mdl-33866329

RÉSUMÉ

Previous genome-wide association studies (GWAS) have identified potential genetic variants associated with the risk of major depressive disorder (MDD), but the underlying biological interpretation remains largely unknown. We aimed to prioritize genes that were pleiotropically or potentially causally associated with MDD. We applied the summary data-based Mendelian randomization (SMR) method integrating GWAS and gene expression quantitative trait loci (eQTL) data in 13 brain regions to identify genes that were pleiotropically associated with MDD. In addition, we repeated the analysis by using the meta-analyzed version of the eQTL summary data in the brain (brain-eMeta). We identified multiple significant genes across different brain regions that may be involved in the pathogenesis of MDD. The prime-specific gene BTN3A2 (corresponding probe: ENSG00000186470.9) was the top hit showing pleiotropic association with MDD in 9 of the 13 brain regions and in brain-eMeta, after correction for multiple testing. Many of the identified genes are located in the human major histocompatibility complex (MHC) region on chromosome 6 and are mainly involved in the immune response. Our SMR analysis indicated that multiple genes showed pleiotropic association with MDD across the brain regions. These findings provided important leads to a better understanding of the mechanism of MDD and revealed potential therapeutic targets for the prevention and effective treatment of MDD.


Sujet(s)
Trouble dépressif majeur , Étude d'association pangénomique , Trouble dépressif majeur/génétique , Prédisposition génétique à une maladie , Humains , Analyse de randomisation mendélienne , Polymorphisme de nucléotide simple , Locus de caractère quantitatif
12.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Article de Anglais | MEDLINE | ID: mdl-33443202

RÉSUMÉ

The mechanistic target of rapamycin (mTOR) is a central regulator of cell growth and an attractive anticancer target that integrates diverse signals to control cell proliferation. Previous studies using mTOR inhibitors have shown that mTOR targeting suppresses gene expression and cell proliferation. To date, however, mTOR-targeted therapies in cancer have seen limited efficacy, and one key issue is related to the development of evasive resistance. In this manuscript, through the use of a gene targeting mouse model, we have found that inducible deletion of mTOR in hematopoietic stem cells (HSCs) results in a loss of quiescence and increased proliferation. Adaptive to the mTOR loss, mTOR-/- HSCs increase chromatin accessibility and activate global gene expression, contrary to the effects of short-term inhibition by mTOR inhibitors. Mechanistically, such genomic changes are due to a rewiring and adaptive activation of the ERK/MNK/eIF4E signaling pathway that enhances the protein translation of RNA polymerase II, which in turn leads to increased c-Myc gene expression, allowing the HSCs to thrive despite the loss of a functional mTOR pathway. This adaptive mechanism can also be utilized by leukemia cells undergoing long-term mTOR inhibitor treatment to confer resistance to mTOR drug targeting. The resistance can be counteracted by MNK, CDK9, or c-Myc inhibition. These results provide insights into the physiological role of mTOR in mammalian stem cell regulation and implicate a mechanism of evasive resistance in the context of mTOR targeting.


Sujet(s)
Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules souches hématopoïétiques/métabolisme , Sirolimus/pharmacologie , Sérine-thréonine kinases TOR/antagonistes et inhibiteurs , Sérine-thréonine kinases TOR/génétique , Animaux , Cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , Chromatine/métabolisme , Séquençage après immunoprécipitation de la chromatine , Kinase-9 cycline-dépendante/métabolisme , Facteur-4E d'initiation eucaryote/métabolisme , Extracellular Signal-Regulated MAP Kinases/métabolisme , Ciblage de gène , Gènes myc/génétique , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/génétique , Souris , Souris knockout , Phosphorylation/effets des médicaments et des substances chimiques , Inhibiteurs de protéines kinases/pharmacologie , RNA polymerase II/métabolisme , Sérine-thréonine kinases TOR/métabolisme
13.
J Infect ; 82(1): 126-132, 2021 01.
Article de Anglais | MEDLINE | ID: mdl-33259846

RÉSUMÉ

OBJECTIVES: COVID-19 has caused a large global pandemic. Patients with COVID-19 exhibited considerable variation in disease behavior. Pervious genome-wide association studies have identified potential genetic variants involved in the risk and prognosis of COVID-19, but the underlying biological interpretation remains largely unclear. METHODS: We applied the summary data-based Mendelian randomization (SMR) method to identify genes that were pleiotropically associated with the risk and various outcomes of COVID-19, including severe respiratory confirmed COVID-19 and hospitalized COVID-19. RESULTS: In blood, we identified 2 probes, ILMN_1765146 and ILMN_1791057 tagging IFNAR2, that showed pleiotropic association with hospitalized COVID-19 (ß [SE]=0.42 [0.09], P = 4.75 × 10-06 and ß [SE]=-0.48 [0.11], P = 6.76 × 10-06, respectively). Although no other probes were significant after correction for multiple testing in both blood and lung, multiple genes as tagged by the top 5 probes were involved in inflammation or antiviral immunity, and several other tagged genes, such as PON2 and HPS5, were involved in blood coagulation. CONCLUSIONS: We identified IFNAR2 and other potential genes that could be involved in the susceptibility or prognosis of COVID-19. These findings provide important leads to a better understanding of the mechanisms of cytokine storm and venous thromboembolism in COVID-19 and potential therapeutic targets for the effective treatment of COVID-19.


Sujet(s)
COVID-19/épidémiologie , Variation génétique/génétique , Étude d'association pangénomique/méthodes , Analyse de randomisation mendélienne , SARS-CoV-2/génétique , Aryldialkylphosphatase/génétique , Coagulation sanguine/génétique , COVID-19/mortalité , Protéines de transport/génétique , Syndrome de libération de cytokines/génétique , Syndrome de libération de cytokines/anatomopathologie , Prédisposition génétique à une maladie/génétique , Humains , Pronostic , Récepteur à l'interféron alpha-bêta/génétique , Risque , Thromboembolisme veineux/génétique , Thromboembolisme veineux/anatomopathologie
14.
medRxiv ; 2020 Sep 04.
Article de Anglais | MEDLINE | ID: mdl-32909000

RÉSUMÉ

OBJECTIVES: COVID-19 has caused a large global pandemic. Patients with COVID-19 exhibited considerable variation in disease behavior. Pervious genome-wide association studies have identified potential genetic variants involved in the risk and prognosis of COVID-19, but the underlying biological interpretation remains largely unclear. METHODS: We applied the summary data-based Mendelian randomization (SMR) method to identify genes that were pleiotropically associated with the risk and various outcomes of COVID-19, including severe respiratory confirmed COVID-19 and hospitalized COVID-19. RESULTS: In blood, we identified 2 probes, ILMN_1765146 and ILMN_1791057 tagging IFNAR2, that showed pleiotropic association with hospitalized COVID-19 (Beta; [SE]=0.42 [0.09], P=4.75E-06 and Beta; [SE]=-0.48 [0.11], P=6.76E-06, respectively). Although no other probes were significant after correction for multiple testing in both blood and lung, multiple genes as tagged by the top 5 probes were involved in inflammation or antiviral immunity, and several other tagged genes, such as PON2 and HPS5, were involved in blood coagulation. CONCLUSIONS: We identified IFNAR2 and other potential genes that could be involved in the susceptibility or prognosis of COVID-19. These findings provide important leads to a better understanding of the mechanisms of cytokine storm and venous thromboembolism in COVID-19 and potential therapeutic targets for the effective treatment of COVID-19.

15.
Nat Commun ; 11(1): 4133, 2020 08 17.
Article de Anglais | MEDLINE | ID: mdl-32807777

RÉSUMÉ

Chromatin organization is critical for cell growth, differentiation, and disease development, however, its functions in peripheral myelination and myelin repair remain elusive. In this report, we demonstrate that the CCCTC-binding factor (CTCF), a crucial chromatin organizer, is essential for Schwann cell myelination and myelin regeneration after nerve injury. Inhibition of CTCF or its deletion blocks Schwann cell differentiation at the pro-myelinating stage, whereas overexpression of CTCF promotes the myelination program. We find that CTCF establishes chromatin interaction loops between enhancer and promoter regulatory elements and promotes expression of a key pro-myelinogenic factor EGR2. In addition, CTCF interacts with SUZ12, a component of polycomb-repressive-complex 2 (PRC2), to repress the transcriptional program associated with negative regulation of Schwann cell maturation. Together, our findings reveal a dual role of CTCF-dependent chromatin organization in promoting myelinogenic programs and recruiting chromatin-repressive complexes to block Schwann cell differentiation inhibitors to control peripheral myelination and repair.


Sujet(s)
Facteur de liaison à la séquence CCCTC/métabolisme , Chromatine/métabolisme , Facteur de transcription EGR-2/métabolisme , Gaine de myéline/métabolisme , Complexe répresseur Polycomb-2/métabolisme , Animaux , Facteur de liaison à la séquence CCCTC/génétique , Cellules cultivées , Immunoprécipitation de la chromatine , Facteur de transcription EGR-2/génétique , Souris , Gaine de myéline/génétique , Complexe répresseur Polycomb-2/génétique , Rats , Cellules de Schwann/métabolisme
16.
Sci Adv ; 6(33): eaaz6477, 2020 08.
Article de Anglais | MEDLINE | ID: mdl-32851157

RÉSUMÉ

Mutations in the polycomb repressive complex 2 (PRC2) can cause Weaver-like syndrome, wherein a patient cohort exhibits abnormal white matter; however, PRC2 functions in CNS myelination and regeneration remain elusive. We show here that H3K27me3, the PRC2 catalytic product, increases during oligodendrocyte maturation. Depletion of embryonic ectoderm development (EED), a core PRC2 subunit, reduces differentiation of oligodendrocyte progenitors (OPCs), and causes an OPC-to-astrocyte fate switch in a region-specific manner. Although dispensable for myelin maintenance, EED is critical for oligodendrocyte remyelination. Genomic occupancy and transcriptomic analyses indicate that EED establishes a chromatin landscape that selectively represses inhibitory WNT and bone morphogenetic protein (BMP) signaling, and senescence-associated programs. Blocking WNT or BMP pathways partially restores differentiation defects in EED-deficient OPCs. Thus, our findings reveal that EED/PRC2 is a crucial epigenetic programmer of CNS myelination and repair, while demonstrating a spatiotemporal-specific role of PRC2-mediated chromatin silencing in shaping oligodendrocyte identity and lineage plasticity.

17.
Glia ; 68(8): 1604-1618, 2020 08.
Article de Anglais | MEDLINE | ID: mdl-32460418

RÉSUMÉ

Oligodendrocytes, the myelinating cells in the vertebrate central nervous system, produce myelin sheaths to enable saltatory propagation of action potentials. The process of oligodendrocyte myelination entails a stepwise progression from precursor specification to differentiation, which is coordinated by a series of transcriptional and chromatin remodeling events. ATP-dependent chromatin remodeling enzymes, which utilize ATP as an energy source to control chromatin dynamics and regulate the accessibility of chromatin to transcriptional regulators, are critical for oligodendrocyte lineage development and regeneration. In this review, we focus on the latest insights into the spatial and temporal specificity of chromatin remodelers during oligodendrocyte development, myelinogenesis, and regeneration. We will also bring together various plausible mechanisms by which lineage specific transcriptional regulators coordinate with chromatin remodeling factors for programming genomic landscapes to specifically modulate these different processes during developmental myelination and remyelination upon injury.


Sujet(s)
Assemblage et désassemblage de la chromatine/physiologie , Gaine de myéline/physiologie , Oligodendroglie/cytologie , Remyélinisation/physiologie , Animaux , Différenciation cellulaire/physiologie , Système nerveux central/métabolisme , Humains
18.
Ann Palliat Med ; 9(3): 1351, 2020 05.
Article de Anglais | MEDLINE | ID: mdl-32389022

RÉSUMÉ

This corrects the article DOI: 10.21037/apm.2020.03.29.

19.
Ann Palliat Med ; 9(2): 451-458, 2020 03.
Article de Anglais | MEDLINE | ID: mdl-32233643

RÉSUMÉ

BACKGROUND: Chronic osteomyelitis is a serious complication of orthopedic trauma. Residual bacteria after incomplete debridement and/or bacterial colonization, bacterial biofilm formation, and generation of antibiotic-resistant bacterial strains in the microtubule system of compact bones due to irrational use of antibiotics often make the condition more prolonged, recurrent, and refractory. The passive immunotherapy targeting the protein components of bacteria has become an area of intense research interest, for which identifying the bacterial isolates in different areas at different time points remains a key step. Few multicenter randomized controlled trials have investigated the epidemiological data of pathogens in different areas, and there is a lack of timely and dynamic data that can inform clinical treatment. METHODS: A total of 5,268 patients with limb fractures were treated in our center from January 1, 2012, to December 31, 2015, among whom 108 were diagnosed with post-traumatic osteomyelitis (PTO) based on clinical manifestations, imaging findings, and pathology. Bacteria cultures showed positive results in 84 patients. The clinical manifestations (including the infection site) were analyzed. The distribution and drug resistance of pathogens were analyzed and summarized based on the M-100-S22 protocol [Clinical and Laboratory Standards Institute® (CLSI) 2012, USA]. RESULTS: The incidence of PTO in limbs was 2.1% (n=108), and the bacterial cultures were positive in 84 patients (84/108, 77.8%). The infection sites included the tibia and fibula (n=40, 47.6%), femur (n=20, 23.8%), ulna and radium (n=11, 13.1%), humerus (n=5, 6%), patella (n=5, 6%), and calcaneus (n=3, 3.6%). In total, 104 of the following bacterial strains were identified: 56 strains of gram-positive bacteria (53.9%), among which Staphylococcus aureus (n=39, 37.5%) and Staphylococcus epidermis (n=6, 5.8%) were the most dominant bacteria, with both being sensitive to ampicillin, quinupristin, linazolamide, tigarycline, nitrofurantoin, and vancomycin; 48 strains of gram-negative bacteria (46.1%), among which Escherichia coli (n=16, 15.4%) and Enterobacter cloacae (n=11, 10.6%) were the most common bacteria, with both being sensitive to thiomycin; mixed infections were detected in 18 cases (21.4%). CONCLUSIONS: The incidence of PTO in the Zunyi area is similar to the national level. The most common site of infection is the lower extremity. Bacterial infections (mainly infection caused by a single bacterial type) were observed in 77.8% of the cases. Staphylococcus aureus is the most common pathogenic bacteria, followed by Escherichia coli and Enterobacter cloacae. The antibiotic-resistant bacteria have characteristic distributions in different regions.


Sujet(s)
Fractures osseuses/microbiologie , Infections bactériennes à Gram négatif/microbiologie , Infections bactériennes à Gram positif/microbiologie , Ostéomyélite/microbiologie , Adulte , Antibactériens/usage thérapeutique , Femelle , Fractures osseuses/complications , Infections bactériennes à Gram négatif/traitement médicamenteux , Infections bactériennes à Gram positif/traitement médicamenteux , Humains , Mâle , Adulte d'âge moyen , Ostéomyélite/traitement médicamenteux , Résultat thérapeutique
20.
Front Neurosci ; 13: 829, 2019.
Article de Anglais | MEDLINE | ID: mdl-31440130

RÉSUMÉ

Oligodendrocyte precursor cells (OPCs), also known as NG2 glia, arise from neural progenitor cells in the embryonic ganglionic eminences that also generate inhibitory neurons. They are ubiquitously distributed in the central nervous system, remain proliferative through life, and generate oligodendrocytes in both gray and white matter. OPCs exhibit some lineage plasticity, and attempts have been made to reprogram them into neurons, with varying degrees of success. However, little is known about how epigenetic mechanisms affect the ability of OPCs to undergo fate switch and whether OPCs have a unique chromatin environment around neuronal genes that might contribute to their lineage plasticity. Our bioinformatic analysis of histone posttranslational modifications at interneuron genes in OPCs revealed that OPCs had significantly fewer bivalent and repressive histone marks at interneuron genes compared to astrocytes or fibroblasts. Conversely, OPCs had a greater degree of deposition of active histone modifications at bivalently marked interneuron genes than other cell types, and this was correlated with higher expression levels of these genes in OPCs. Furthermore, a significantly higher proportion of interneuron genes in OPCs than in other cell types lacked the histone posttranslational modifications examined. These genes had a moderately high level of expression, suggesting that the "no mark" interneuron genes could be in a transcriptionally "poised" or "transitional" state. Thus, our findings suggest that OPCs have a unique histone code at their interneuron genes that may obviate the need for erasure of repressive marks during their fate switch to inhibitory neurons.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE