Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 4879, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849354

ABSTRACT

The mammalian neocortex comprises an enormous diversity regarding cell types, morphology, and connectivity. In this work, we discover a post-transcriptional mechanism of gene expression regulation, protein translation, as a determinant of cortical neuron identity. We find specific upregulation of protein synthesis in the progenitors of later-born neurons and show that translation rates and concomitantly protein half-lives are inherent features of cortical neuron subtypes. In a small molecule screening, we identify Ire1α as a regulator of Satb2 expression and neuronal polarity. In the developing brain, Ire1α regulates global translation rates, coordinates ribosome traffic, and the expression of eIF4A1. Furthermore, we demonstrate that the Satb2 mRNA translation requires eIF4A1 helicase activity towards its 5'-untranslated region. Altogether, we show that cortical neuron diversity is generated by mechanisms operating beyond gene transcription, with Ire1α-safeguarded proteostasis serving as an essential regulator of brain development.


Subject(s)
Matrix Attachment Region Binding Proteins , Neocortex , Neurons , Protein Biosynthesis , Protein Serine-Threonine Kinases , Animals , Neocortex/metabolism , Neocortex/cytology , Neocortex/embryology , Neurons/metabolism , Neurons/cytology , Mice , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Matrix Attachment Region Binding Proteins/metabolism , Matrix Attachment Region Binding Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Gene Expression Regulation, Developmental , Proteostasis , Neurogenesis/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , 5' Untranslated Regions/genetics , Ribosomes/metabolism , Ribosomes/genetics , Humans , Endoribonucleases/metabolism , Endoribonucleases/genetics , Cell Differentiation/genetics
2.
Int J Mol Sci ; 24(23)2023 Dec 04.
Article in English | MEDLINE | ID: mdl-38069426

ABSTRACT

Epilepsy is one of the common neurological diseases that affects not only adults but also infants and children. Because epilepsy has been studied for a long time, there are several pharmacologically effective anticonvulsants, which, however, are not suitable as therapy for all patients. The genesis of epilepsy has been extensively investigated in terms of its occurrence after injury and as a concomitant disease with various brain diseases, such as tumors, ischemic events, etc. However, in the last decades, there are multiple reports that both genetic and epigenetic factors play an important role in epileptogenesis. Therefore, there is a need for further identification of genes and loci that can be associated with higher susceptibility to epileptic seizures. Use of mouse knockout models of epileptogenesis is very informative, but it has its limitations. One of them is due to the fact that complete deletion of a gene is not, in many cases, similar to human epilepsy-associated syndromes. Another approach to generating mouse models of epilepsy is N-Ethyl-N-nitrosourea (ENU)-directed mutagenesis. Recently, using this approach, we generated a novel mouse strain, soc (socrates, formerly s8-3), with epileptiform activity. Using molecular biology methods, calcium neuroimaging, and immunocytochemistry, we were able to characterize the strain. Neurons isolated from soc mutant brains retain the ability to differentiate in vitro and form a network. However, soc mutant neurons are characterized by increased spontaneous excitation activity. They also demonstrate a high degree of Ca2+ activity compared to WT neurons. Additionally, they show increased expression of NMDA receptors, decreased expression of the Ca2+-conducting GluA2 subunit of AMPA receptors, suppressed expression of phosphoinositol 3-kinase, and BK channels of the cytoplasmic membrane involved in protection against epileptogenesis. During embryonic and postnatal development, the expression of several genes encoding ion channels is downregulated in vivo, as well. Our data indicate that soc mutation causes a disruption of the excitation-inhibition balance in the brain, and it can serve as a mouse model of epilepsy.


Subject(s)
Epilepsy, Reflex , Child , Animals , Humans , Mice , Epilepsy, Reflex/genetics , Epilepsy, Reflex/metabolism , Ethylnitrosourea/toxicity , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Anticonvulsants/pharmacology , Brain/metabolism , Disease Models, Animal
3.
Nucleic Acids Res ; 51(19): 10218-10237, 2023 10 27.
Article in English | MEDLINE | ID: mdl-37697438

ABSTRACT

The seat of higher-order cognitive abilities in mammals, the neocortex, is a complex structure, organized in several layers. The different subtypes of principal neurons are distributed in precise ratios and at specific positions in these layers and are generated by the same neural progenitor cells (NPCs), steered by a spatially and temporally specified combination of molecular cues that are incompletely understood. Recently, we discovered that an alternatively spliced isoform of the TrkC receptor lacking the kinase domain, TrkC-T1, is a determinant of the corticofugal projection neuron (CFuPN) fate. Here, we show that the finely tuned balance between TrkC-T1 and the better known, kinase domain-containing isoform, TrkC-TK+, is cell type-specific in the developing cortex and established through the antagonistic actions of two RNA-binding proteins, Srsf1 and Elavl1. Moreover, our data show that Srsf1 promotes the CFuPN fate and Elavl1 promotes the callosal projection neuron (CPN) fate in vivo via regulating the distinct ratios of TrkC-T1 to TrkC-TK+. Taken together, we connect spatio-temporal expression of Srsf1 and Elavl1 in the developing neocortex with the regulation of TrkC alternative splicing and transcript stability and neuronal fate choice, thus adding to the mechanistic and functional understanding of alternative splicing in vivo.


Subject(s)
Neocortex , Receptor, trkC , Animals , Alternative Splicing , Mammals/metabolism , Neocortex/metabolism , Neurons/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptor, trkC/chemistry , Receptor, trkC/genetics , Receptor, trkC/metabolism , Mice , Cell Line, Tumor
4.
5.
Front Immunol ; 12: 738958, 2021.
Article in English | MEDLINE | ID: mdl-34721405

ABSTRACT

Immune checkpoint blockade (ICB) relieves CD8+ T-cell exhaustion in most mutated tumors, and TCF-1 is implicated in converting progenitor exhausted cells to functional effector cells. However, identifying mechanisms that can prevent functional senescence and potentiate CD8+ T-cell persistence for ICB non-responsive and resistant tumors remains elusive. We demonstrate that targeting Cbx3/HP1γ in CD8+ T cells augments transcription initiation and chromatin remodeling leading to increased transcriptional activity at Lef1 and Il21r. LEF-1 and IL-21R are necessary for Cbx3/HP1γ-deficient CD8+ effector T cells to persist and control ovarian cancer, melanoma, and neuroblastoma in preclinical models. The enhanced persistence of Cbx3/HP1γ-deficient CD8+ T cells facilitates remodeling of the tumor chemokine/receptor landscape ensuring their optimal invasion at the expense of CD4+ Tregs. Thus, CD8+ T cells heightened effector function consequent to Cbx3/HP1γ deficiency may be distinct from functional reactivation by ICB, implicating Cbx3/HP1γ as a viable cancer T-cell-based therapy target for ICB resistant, non-responsive solid tumors.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Chromobox Protein Homolog 5/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphoid Enhancer-Binding Factor 1/metabolism , Melanoma, Experimental/metabolism , Neuroblastoma/metabolism , Ovarian Neoplasms/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Cell Differentiation , Cell Line, Tumor , Chromobox Protein Homolog 5/genetics , Chromosomal Proteins, Non-Histone/genetics , Coculture Techniques , Female , Gene Expression Regulation, Neoplastic , Immunotherapy, Adoptive , Interleukin-21 Receptor alpha Subunit/genetics , Interleukin-21 Receptor alpha Subunit/metabolism , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Lymphoid Enhancer-Binding Factor 1/genetics , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neuroblastoma/genetics , Neuroblastoma/immunology , Neuroblastoma/therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Tumor Burden , Tumor Microenvironment
6.
JCI Insight ; 5(7)2020 04 09.
Article in English | MEDLINE | ID: mdl-32271161

ABSTRACT

Although human endogenous retroviruses (HERVs) represent a substantial proportion of the human genome and some HERVs, such as HERV-K(HML-2), are reported to be involved in neurological disorders, little is known about their biological function. We report that RNA from an HERV-K(HML-2) envelope gene region binds to and activates human Toll-like receptor (TLR) 8, as well as murine Tlr7, expressed in neurons and microglia, thereby causing neurodegeneration. HERV-K(HML-2) RNA introduced into the cerebrospinal fluid (CSF) of either C57BL/6 wild-type mice or APPPS1 mice, a mouse model for Alzheimer's disease (AD), resulted in neurodegeneration and microglia accumulation. Tlr7-deficient mice were protected against neurodegenerative effects but were resensitized toward HERV-K(HML-2) RNA when neurons ectopically expressed murine Tlr7 or human TLR8. Transcriptome data sets of human AD brain samples revealed a distinct correlation of upregulated HERV-K(HML-2) and TLR8 RNA expression. HERV-K(HML-2) RNA was detectable more frequently in CSF from individuals with AD compared with controls. Our data establish HERV-K(HML-2) RNA as an endogenous ligand for species-specific TLRs 7/8 and imply a functional contribution of human endogenous retroviral transcripts to neurodegenerative processes, such as AD.


Subject(s)
Alzheimer Disease , Endogenous Retroviruses , Membrane Glycoproteins , RNA, Viral , Toll-Like Receptor 7 , Toll-Like Receptor 8 , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Disease Models, Animal , Endogenous Retroviruses/genetics , Endogenous Retroviruses/metabolism , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , RNA, Viral/genetics , RNA, Viral/metabolism , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/genetics , Toll-Like Receptor 8/metabolism
7.
R Soc Open Sci ; 7(2): 191976, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32257349

ABSTRACT

The relationship between compartmentalization of the genome and epigenetics is long and hoary. In 1928, Heitz defined heterochromatin as the largest differentiated chromatin compartment in eukaryotic nuclei. Müller's discovery of position-effect variegation in 1930 went on to show that heterochromatin is a cytologically visible state of heritable (epigenetic) gene repression. Current insights into compartmentalization have come from a high-throughput top-down approach where contact frequency (Hi-C) maps revealed the presence of compartmental domains that segregate the genome into heterochromatin and euchromatin. It has been argued that the compartmentalization seen in Hi-C maps is owing to the physiochemical process of phase separation. Oddly, the insights provided by these experimental and conceptual advances have remained largely silent on how Hi-C maps and phase separation relate to epigenetics. Addressing this issue directly in mammals, we have made use of a bottom-up approach starting with the hallmarks of constitutive heterochromatin, heterochromatin protein 1 (HP1) and its binding partner the H3K9me2/3 determinant of the histone code. They are key epigenetic regulators in eukaryotes. Both hallmarks are also found outside mammalian constitutive heterochromatin as constituents of larger (0.1-5 Mb) heterochromatin-like domains and smaller (less than 100 kb) complexes. The well-documented ability of HP1 proteins to function as bridges between H3K9me2/3-marked nucleosomes contributes to polymer-polymer phase separation that packages epigenetically heritable chromatin states during interphase. Contacts mediated by HP1 'bridging' are likely to have been detected in Hi-C maps, as evidenced by the B4 heterochromatic subcompartment that emerges from contacts between large KRAB-ZNF heterochromatin-like domains. Further, mutational analyses have revealed a finer, innate, compartmentalization in Hi-C experiments that probably reflect contacts involving smaller domains/complexes. Proteins that bridge (modified) DNA and histones in nucleosomal fibres-where the HP1-H3K9me2/3 interaction represents the most evolutionarily conserved paradigm-could drive and generate the fundamental compartmentalization of the interphase nucleus. This has implications for the mechanism(s) that maintains cellular identity, be it a terminally differentiated fibroblast or a pluripotent embryonic stem cell.

8.
Cell Rep ; 29(11): 3460-3471.e7, 2019 Dec 10.
Article in English | MEDLINE | ID: mdl-31825829

ABSTRACT

Microglia express Toll-like receptors (TLRs) that sense pathogen- and host-derived factors, including single-stranded RNA. In the brain, let-7 microRNA (miRNA) family members are abundantly expressed, and some have recently been shown to serve as TLR7 ligands. We investigated whether let-7 miRNA family members differentially control microglia biology in health and disease. We found that a subset of let-7 miRNA family members function as signaling molecules to induce microglial release of inflammatory cytokines, modulate antigen presentation, and attenuate cell migration in a TLR7-dependent manner. The capability of the let-7 miRNAs to control microglial function is sequence specific, mapping to a let-7 UUGU motif. In human and murine glioblastoma/glioma, let-7 miRNAs are differentially expressed and reduce murine GL261 glioma growth in the same sequence-specific fashion through microglial TLR7. Taken together, these data establish let-7 miRNAs as key TLR7 signaling activators that serve to regulate the diverse functions of microglia in health and glioma.


Subject(s)
Brain Neoplasms/genetics , Glioma/genetics , MicroRNAs/metabolism , Microglia/metabolism , Toll-Like Receptor 7/genetics , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cells, Cultured , Female , Gene Expression Regulation, Neoplastic , Glioma/metabolism , Glioma/pathology , Humans , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Signal Transduction , Toll-Like Receptor 7/metabolism
9.
J Biosci ; 44(4)2019 Sep.
Article in English | MEDLINE | ID: mdl-31502583

ABSTRACT

It has been proposed that age reprogramming enables old cells to be rejuvenated without passage through an embryonic stage (Singh and Zacouto in J. Biosci. 35 315-319, 2010). As such, age reprogramming stands apart from the induced pluripotent stem (iPS) and nuclear transfer-embryonic stem (NT-ES) cell therapies where histo-compatible cells are produced only after passage through an embryonic stage. It avoids many of the disadvantages associated with iPS and NT-ES cell therapies. Experimental evidence in support of age reprogramming is burgeoning. Here, we discuss possible new approaches to enhance age reprogramming, which will have considerable benefits for regenerative therapies.


Subject(s)
Aging/genetics , Cellular Reprogramming/genetics , Epigenesis, Genetic/genetics , Embryonic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Regenerative Medicine
10.
Brain Res ; 1705: 24-31, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30266271

ABSTRACT

Zeb2 (Sip1, Zfhx1b) is a transcription factor that plays essential role in neuronal development. Sip1 mutation in humans was shown to cause Mowat-Wilson syndrome, a syndromic form of Hirschprung's disease. Affected individuals exhibit multiple severe neurodevelopmental defects. Zeb2 can act as both transcriptional repressor and activator. It controls expression of a wide number of genes that regulate various aspects of neuronal development. This review addresses the molecular pathways acting downstream of Zeb2 that cause brain development disorders.


Subject(s)
Brain/embryology , Zinc Finger E-box Binding Homeobox 2/genetics , Zinc Finger E-box Binding Homeobox 2/metabolism , Brain/metabolism , Cell Differentiation/genetics , Cell Movement/genetics , Cerebral Cortex/metabolism , Embryonic Development , Facies , Gene Expression Regulation, Developmental/genetics , Hippocampus/metabolism , Hirschsprung Disease , Homeodomain Proteins/genetics , Humans , Intellectual Disability , Microcephaly , Neurogenesis/physiology , Neuroglia/metabolism , Neurons/metabolism , Repressor Proteins/metabolism , Stem Cells/physiology , Transcription Factors/metabolism
11.
Epigenetics Chromatin ; 11(1): 73, 2018 12 20.
Article in English | MEDLINE | ID: mdl-30572909

ABSTRACT

Age reprogramming represents a novel method for generating patient-specific tissues for transplantation. It bypasses the de-differentiation/redifferentiation cycle that is characteristic of the induced pluripotent stem (iPS) and nuclear transfer-embryonic stem (NT-ES) cell technologies that drive current interest in regenerative medicine. Despite the obvious potential of iPS and NT-ES cell-based therapies, there are several problems that must be overcome before these therapies are safe and routine. As an alternative, age reprogramming aims to rejuvenate the specialized functions of an old cell without de-differentiation; age reprogramming does not require developmental reprogramming through an embryonic stage, unlike the iPS and NT-ES cell-based therapies. Tests of age reprogramming have largely focused on one aspect, the epigenome. Epigenetic rejuvenation has been achieved in vitro in the absence of de-differentiation using iPS cell reprogramming factors. Studies on the dynamics of epigenetic age (eAge) reprogramming have demonstrated that the separation of eAge from developmental reprogramming can be explained largely by their different kinetics. Age reprogramming has also been achieved in vivo and shown to increase lifespan in a premature ageing mouse model. We conclude that age and developmental reprogramming can be disentangled and regulated independently in vitro and in vivo.


Subject(s)
Aging/physiology , Cellular Reprogramming/physiology , Rejuvenation/physiology , Age Factors , Animals , Cell Differentiation , Embryonic Stem Cells/physiology , Epigenesis, Genetic/genetics , Epigenesis, Genetic/physiology , Epigenomics , Humans , Induced Pluripotent Stem Cells/physiology , Nuclear Transfer Techniques , Pluripotent Stem Cells/physiology , Regenerative Medicine/methods , Stem Cell Transplantation/methods
13.
Neurobiol Aging ; 35(5): 1012-23, 2014 May.
Article in English | MEDLINE | ID: mdl-24262201

ABSTRACT

A neuro-inflammatory response has been implicated in human patients and animal models of Alzheimer's disease (AD). Type-1 interferons are pleiotropic cytokines involved in the initiation and regulation of the pro-inflammatory response; however, their role in AD is unknown. This study investigated the contribution of type-1 IFN signaling in the neuro-inflammatory response to amyloid-beta (Aß) in vitro and in the APP/PS1 transgenic mouse model of AD. Enzyme-linked immunosorbent assay confirmed a 2-fold increase in IFNα in APP/PS1 brains compared with control brains. Quantitative polymerase chain reaction also identified increased IFNα and IFNß expression in human pre-frontal cortex from AD patients. In vitro studies in primary neurons demonstrated Aß-induced type-1 IFN expression preceded that of other classical pro-inflammatory cytokines, IL1-ß, and IL-6. Significantly, ablation of type-1 interferon-α receptor 1 expression in BE(2)M17 neuroblastoma cells and primary neurons afforded protection against Aß-induced toxicity. This study supports a role for type-1 interferons in the pro-inflammatory response and neuronal cell death in AD and suggests that blocking type-1 interferon-α receptor 1 maybe a therapeutic target to limit the disease progression.


Subject(s)
Alzheimer Disease/genetics , Inflammation/genetics , Interferon Type I/physiology , Signal Transduction/genetics , Aged , Alzheimer Disease/drug therapy , Alzheimer Disease/etiology , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Cell Death/genetics , Cell Line , Cytokines/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Mice , Mice, Transgenic , Molecular Targeted Therapy , Neuroblastoma/metabolism , Neurons/metabolism , Neurons/pathology , Polymerase Chain Reaction , Receptor, Interferon alpha-beta/antagonists & inhibitors
14.
Learn Mem ; 18(10): 678-87, 2011.
Article in English | MEDLINE | ID: mdl-21969490

ABSTRACT

The identity and distribution of neurons that are involved in any learning or memory event is not known. In previous studies, we identified a discrete population of neurons in the lateral amygdala that show learning-specific activation of a c-fos-regulated transgene following context fear conditioning. Here, we have extended these studies to look throughout the amygdala for learning-specific activation. We identified two further neuronal populations, in the amygdalo-striatal transition area and medial amygdala, that show learning-specific activation. We also identified a population of hypothalamic neurons that show strong learning-specific activation. In addition, we asked whether these neurons are activated following recall of fear-conditioning memory. None of the populations of neurons we identified showed significant memory-recall-related activation. These findings suggest that a series of discrete populations of neurons are involved in fear learning in amygdala and hypothalamus. The lack of reactivation during memory recall suggests that these neurons either do not undergo substantial change following recall, or that c-fos is not involved in any such activation and change.


Subject(s)
Amygdala/physiology , Association Learning/physiology , Fear/physiology , Hypothalamus/physiology , Neurons/physiology , Amygdala/metabolism , Animals , Conditioning, Psychological/physiology , Hypothalamus/metabolism , Mental Recall/physiology , Mice , Neurons/metabolism , Proto-Oncogene Proteins c-fos/metabolism
15.
Anesth Analg ; 103(1): 124-5, table of contents, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16790639

ABSTRACT

We describe two cases in which keyed filling devices for sevoflurane were inadvertently screwed onto isoflurane bottles. The mishaps were possible because the collars on sevoflurane and isoflurane bottles are mirror images of each other. The particular keyed filling device was designed with a flexible outer sleeve and could be screwed onto the wrong bottle while slightly gouging its soft plastic collar. The keyed filling adapters for sevoflurane and isoflurane could each be manipulated to fit the other's bottle. A manufacturer (Southmedic, Inc., Barrie, Canada) has modified their keyed filling adapters to prevent this unusual circumstance from recurring.


Subject(s)
Anesthetics, Inhalation , Drug Packaging , Isoflurane , Medication Errors , Methyl Ethers , Sevoflurane
SELECTION OF CITATIONS
SEARCH DETAIL