Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 12.112
Filter
1.
Sci Rep ; 14(1): 10573, 2024 05 08.
Article in English | MEDLINE | ID: mdl-38719983

ABSTRACT

Multiple sclerosis (MS) is a chronic neurological disease characterized by inflammatory demyelination that disrupts neuronal transmission resulting in neurodegeneration progressive disability. While current treatments focus on immunosuppression to limit inflammation and further myelin loss, no approved therapies effectively promote remyelination to mitigate the progressive disability associated with chronic demyelination. Lysophosphatidic acid (LPA) is a pro-inflammatory lipid that is upregulated in MS patient plasma and cerebrospinal fluid (CSF). LPA activates the LPA1 receptor, resulting in elevated CNS cytokine and chemokine levels, infiltration of immune cells, and microglial/astrocyte activation. This results in a neuroinflammatory response leading to demyelination and suppressed remyelination. A medicinal chemistry effort identified PIPE-791, an oral, brain-penetrant, LPA1 antagonist. PIPE-791 was characterized in vitro and in vivo and was found to be a potent, selective LPA1 antagonist with slow receptor off-rate kinetics. In vitro, PIPE-791 induced OPC differentiation and promoted remyelination following a demyelinating insult. PIPE-791 further mitigated the macrophage-mediated inhibition of OPC differentiation and inhibited microglial and fibroblast activation. In vivo, the compound readily crossed the blood-brain barrier and blocked LPA1 in the CNS after oral dosing. Direct dosing of PIPE-791 in vivo increased oligodendrocyte number, and in the mouse experimental autoimmune encephalomyelitis (EAE) model of MS, we observed that PIPE-791 promoted myelination, reduced neuroinflammation, and restored visual evoked potential latencies (VEP). These findings support targeting LPA1 for remyelination and encourage development of PIPE-791 for treating MS patients with advantages not seen with current immunosuppressive disease modifying therapies.


Subject(s)
Multiple Sclerosis , Receptors, Lysophosphatidic Acid , Remyelination , Animals , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Receptors, Lysophosphatidic Acid/metabolism , Remyelination/drug effects , Humans , Mice , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Oligodendroglia/metabolism , Oligodendroglia/drug effects , Brain/metabolism , Brain/drug effects , Brain/pathology , Cell Differentiation/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Mice, Inbred C57BL , Myelin Sheath/metabolism , Myelin Sheath/drug effects , Lysophospholipids/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects
2.
Acta Neuropathol ; 147(1): 82, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38722375

ABSTRACT

Aging affects all cell types in the CNS and plays an important role in CNS diseases. However, the underlying molecular mechanisms driving these age-associated changes and their contribution to diseases are only poorly understood. The white matter in the aging brain as well as in diseases, such as Multiple sclerosis is characterized by subtle abnormalities in myelin sheaths and paranodes, suggesting that oligodendrocytes, the myelin-maintaining cells of the CNS, lose the capacity to preserve a proper myelin structure and potentially function in age and certain diseases. Here, we made use of directly converted oligodendrocytes (dchiOL) from young, adult and old human donors to study age-associated changes. dchiOL from all three age groups differentiated in an comparable manner into O4 + immature oligodendrocytes, but the proportion of MBP + mature dchiOL decreased with increasing donor age. This was associated with an increased ROS production and upregulation of cellular senescence markers such as CDKN1A, CDKN2A in old dchiOL. Comparison of the transcriptomic profiles of dchiOL from adult and old donors revealed 1324 differentially regulated genes with limited overlap with transcriptomic profiles of the donors' fibroblasts or published data sets from directly converted human neurons or primary rodent oligodendroglial lineage cells. Methylome analyses of dchiOL and human white matter tissue samples demonstrate that chronological and epigenetic age correlate in CNS white matter as well as in dchiOL and resulted in the identification of an age-specific epigenetic signature. Furthermore, we observed an accelerated epigenetic aging of the myelinated, normal appearing white matter of multiple sclerosis (MS) patients compared to healthy individuals. Impaired differentiation and upregulation of cellular senescence markers could be induced in young dchiOL in vitro using supernatants from pro-inflammatory microglia. In summary, our data suggest that physiological aging as well as inflammation-induced cellular senescence contribute to oligodendroglial pathology in inflammatory demyelinating diseases such as MS.


Subject(s)
Aging , Cellular Senescence , Multiple Sclerosis , Oligodendroglia , Humans , Oligodendroglia/pathology , Oligodendroglia/metabolism , Cellular Senescence/physiology , Aging/pathology , Multiple Sclerosis/pathology , Multiple Sclerosis/metabolism , Adult , Aged , Middle Aged , Male , Female , Young Adult , Inflammation/pathology , Inflammation/metabolism , White Matter/pathology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p21
3.
J Neuroinflammation ; 21(1): 127, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38741181

ABSTRACT

HIV-associated neurocognitive disorders (HAND) are a spectrum of cognitive impairments that continue to affect approximately half of all HIV-positive individuals despite effective viral suppression through antiretroviral therapy (ART). White matter pathologies have persisted in the ART era, and the degree of white matter damage correlates with the degree of neurocognitive impairment in patients with HAND. The HIV protein Nef has been implicated in HAND pathogenesis, but its effect on white matter damage has not been well characterized. Here, utilizing in vivo, ex vivo, and in vitro methods, we demonstrate that Nef-containing extracellular vesicles (Nef EVs) disrupt myelin sheaths and inflict damage upon oligodendrocytes within the murine central nervous system. Intracranial injection of Nef EVs leads to reduced myelin basic protein (MBP) staining and a decreased number of CC1 + oligodendrocytes in the corpus callosum. Moreover, cerebellar slice cultures treated with Nef EVs exhibit diminished MBP expression and increased presence of unmyelinated axons. Primary mixed brain cultures and enriched oligodendrocyte precursor cell cultures exposed to Nef EVs display a decreased number of O4 + cells, indicative of oligodendrocyte impairment. These findings underscore the potential contribution of Nef EV-mediated damage to oligodendrocytes and myelin maintenance in the pathogenesis of HAND.


Subject(s)
Extracellular Vesicles , HIV-1 , Mice, Inbred C57BL , Oligodendroglia , nef Gene Products, Human Immunodeficiency Virus , Animals , Oligodendroglia/metabolism , Oligodendroglia/pathology , Oligodendroglia/virology , Mice , Extracellular Vesicles/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism , HIV-1/metabolism , Myelin Sheath/metabolism , Myelin Sheath/pathology , Central Nervous System/metabolism , Central Nervous System/pathology , Central Nervous System/virology , Cells, Cultured , Humans , Male
4.
Science ; 384(6698): eadh2602, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38781372

ABSTRACT

Genomic profiling in postmortem brain from autistic individuals has consistently revealed convergent molecular changes. What drives these changes and how they relate to genetic susceptibility in this complex condition are not well understood. We performed deep single-nucleus RNA sequencing (snRNA-seq) to examine cell composition and transcriptomics, identifying dysregulation of cell type-specific gene regulatory networks (GRNs) in autism spectrum disorder (ASD), which we corroborated using single-nucleus assay for transposase-accessible chromatin with sequencing (snATAC-seq) and spatial transcriptomics. Transcriptomic changes were primarily cell type specific, involving multiple cell types, most prominently interhemispheric and callosal-projecting neurons, interneurons within superficial laminae, and distinct glial reactive states involving oligodendrocytes, microglia, and astrocytes. Autism-associated GRN drivers and their targets were enriched in rare and common genetic risk variants, connecting autism genetic susceptibility and cellular and circuit alterations in the human brain.


Subject(s)
Autism Spectrum Disorder , Gene Regulatory Networks , Neurons , Single-Cell Analysis , Transcriptome , Humans , Autism Spectrum Disorder/genetics , Neurons/metabolism , Genetic Predisposition to Disease , Astrocytes/metabolism , Brain/metabolism , Genomics , Oligodendroglia/metabolism , Microglia/metabolism , RNA-Seq , Male , Interneurons/metabolism , Chromatin/metabolism , Female , Sequence Analysis, RNA
5.
Dis Model Mech ; 17(4)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38721692

ABSTRACT

Duchenne muscular dystrophy (DMD) is caused by mutations in the DMD gene, resulting in the loss of dystrophin, a large cytosolic protein that links the cytoskeleton to extracellular matrix receptors in skeletal muscle. Aside from progressive muscle damage, many patients with DMD also have neurological deficits of unknown etiology. To investigate potential mechanisms for DMD neurological deficits, we assessed postnatal oligodendrogenesis and myelination in the Dmdmdx mouse model. In the ventricular-subventricular zone (V-SVZ) stem cell niche, we found that oligodendrocyte progenitor cell (OPC) production was deficient, with reduced OPC densities and proliferation, despite a normal stem cell niche organization. In the Dmdmdx corpus callosum, a large white matter tract adjacent to the V-SVZ, we also observed reduced OPC proliferation and fewer oligodendrocytes. Transmission electron microscopy further revealed significantly thinner myelin, an increased number of abnormal myelin structures and delayed myelin compaction, with hypomyelination persisting into adulthood. Our findings reveal alterations in oligodendrocyte development and myelination that support the hypothesis that changes in diffusion tensor imaging seen in patients with DMD reflect developmental changes in myelin architecture.


Subject(s)
Mice, Inbred mdx , Muscular Dystrophy, Duchenne , Myelin Sheath , Oligodendroglia , Animals , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Oligodendroglia/pathology , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/genetics , Cell Proliferation , Dystrophin/metabolism , Dystrophin/deficiency , Dystrophin/genetics , Corpus Callosum/pathology , Corpus Callosum/metabolism , Mice, Inbred C57BL , Mice , Oligodendrocyte Precursor Cells/metabolism , Oligodendrocyte Precursor Cells/pathology , Lateral Ventricles/pathology , Lateral Ventricles/metabolism , Disease Models, Animal , Cell Differentiation , Male
6.
Int J Mol Sci ; 25(10)2024 May 17.
Article in English | MEDLINE | ID: mdl-38791505

ABSTRACT

In contrast to the hypothesis that aging results from cell-autonomous deterioration processes, the programmed longevity theory proposes that aging arises from a partial inactivation of a "longevity program" aimed at maintaining youthfulness in organisms. Supporting this hypothesis, age-related changes in organisms can be reversed by factors circulating in young blood. Concordantly, the endocrine secretion of exosomal microRNAs (miRNAs) by hypothalamic neural stem cells (htNSCs) regulates the aging rate by enhancing physiological fitness in young animals. However, the specific molecular mechanisms through which hypothalamic-derived miRNAs exert their anti-aging effects remain unexplored. Using experimentally validated miRNA-target gene interactions and single-cell transcriptomic data of brain cells during aging and heterochronic parabiosis, we identify the main pathways controlled by these miRNAs and the cell-type-specific gene networks that are altered due to age-related loss of htNSCs and the subsequent decline in specific miRNA levels in the cerebrospinal fluid (CSF). Our bioinformatics analysis suggests that these miRNAs modulate pathways associated with senescence and cellular stress response, targeting crucial genes such as Cdkn2a, Rps27, and Txnip. The oligodendrocyte lineage appears to be the most responsive to age-dependent loss of exosomal miRNA, leading to significant derepression of several miRNA target genes. Furthermore, heterochronic parabiosis can reverse age-related upregulation of specific miRNA-targeted genes, predominantly in brain endothelial cells, including senescence promoting genes such as Cdkn1a and Btg2. Our findings support the presence of an anti-senescence mechanism triggered by the endocrine secretion of htNSC-derived exosomal miRNAs, which is associated with a youthful transcriptional signature.


Subject(s)
Aging , Exosomes , Hypothalamus , MicroRNAs , Neural Stem Cells , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Exosomes/metabolism , Hypothalamus/metabolism , Aging/genetics , Aging/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Gene Regulatory Networks , Cellular Senescence/genetics , Brain/metabolism , Mice , Parabiosis , Oligodendroglia/metabolism , Transcriptome , Gene Expression Regulation , Gene Expression Profiling
7.
Mol Brain ; 17(1): 28, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38790036

ABSTRACT

The aggregated alpha-synuclein (αsyn) in oligodendrocytes (OLGs) is one of the pathological hallmarks in multiple system atrophy (MSA). We have previously reported that αsyn accumulates not only in neurons but also in OLGs long after the administration of αsyn preformed fibrils (PFFs) in mice. However, detailed spatial and temporal analysis of oligodendroglial αsyn aggregates was technically difficult due to the background neuronal αsyn aggregates. The aim of this study is to create a novel mouse that easily enables sensitive and specific detection of αsyn aggregates in OLGs and the comparable analysis of the cellular tropism of αsyn aggregates in MSA brains. To this end, we generated transgenic (Tg) mice expressing human αsyn-green fluorescent protein (GFP) fusion proteins in OLGs under the control of the 2', 3'-cyclic nucleotide 3'-phosphodiesterase (CNP) promoter (CNP-SNCAGFP Tg mice). Injection of αsyn PFFs in these mice induced distinct GFP-positive aggregates in the processes of OLGs as early as one month post-inoculation (mpi), and their number and size increased in a centripetal manner. Moreover, MSA-brain homogenates (BH) induced significantly more oligodendroglial αsyn aggregates than neuronal αsyn aggregates compared to DLB-BH in CNP-SNCAGFP Tg mice, suggestive of their potential tropism of αsyn seeds for OLGs. In conclusion, CNP-SNCAGFP Tg mice are useful for studying the development and tropism of αsyn aggregates in OLGs and could contribute to the development of therapeutics targeting αsyn aggregates in OLGs.


Subject(s)
Disease Models, Animal , Inclusion Bodies , Mice, Transgenic , Multiple System Atrophy , Oligodendroglia , Protein Aggregates , alpha-Synuclein , Animals , alpha-Synuclein/metabolism , Oligodendroglia/metabolism , Oligodendroglia/pathology , Multiple System Atrophy/pathology , Multiple System Atrophy/metabolism , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Humans , Green Fluorescent Proteins/metabolism , Cytoplasm/metabolism , Mice , Brain/pathology , Brain/metabolism , Protein Aggregation, Pathological/metabolism
8.
Cell Death Dis ; 15(5): 350, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773070

ABSTRACT

Seipin is one key mediator of lipid metabolism that is highly expressed in adipose tissues as well as in the brain. Lack of Seipin gene, Bscl2, leads to not only severe lipid metabolic disorders but also cognitive impairments and motor disabilities. Myelin, composed mainly of lipids, facilitates nerve transmission and is important for motor coordination and learning. Whether Seipin deficiency-leaded defects in learning and motor coordination is underlined by lipid dysregulation and its consequent myelin abnormalities remains to be elucidated. In the present study, we verified the expression of Seipin in oligodendrocytes (OLs) and their precursors, oligodendrocyte precursor cells (OPCs), and demonstrated that Seipin deficiency compromised OPC differentiation, which led to decreased OL numbers, myelin protein, myelinated fiber proportion and thickness of myelin. Deficiency of Seipin resulted in impaired spatial cognition and motor coordination in mice. Mechanistically, Seipin deficiency suppressed sphingolipid metabolism-related genes in OPCs and caused morphological abnormalities in lipid droplets (LDs), which markedly impeded OPC differentiation. Importantly, rosiglitazone, one agonist of PPAR-gamma, substantially restored phenotypes resulting from Seipin deficiency, such as aberrant LDs, reduced sphingolipids, obstructed OPC differentiation, and neurobehavioral defects. Collectively, the present study elucidated how Seipin deficiency-induced lipid dysregulation leads to neurobehavioral deficits via impairing myelination, which may pave the way for developing novel intervention strategy for treating metabolism-involved neurological disorders.


Subject(s)
Cell Differentiation , Cognitive Dysfunction , GTP-Binding Protein gamma Subunits , Myelin Sheath , Oligodendrocyte Precursor Cells , Animals , GTP-Binding Protein gamma Subunits/metabolism , GTP-Binding Protein gamma Subunits/genetics , Mice , Oligodendrocyte Precursor Cells/metabolism , Myelin Sheath/metabolism , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Cognitive Dysfunction/genetics , Lipid Metabolism , Oligodendroglia/metabolism , Oligodendroglia/pathology , Mice, Inbred C57BL , PPAR gamma/metabolism , PPAR gamma/genetics , Mice, Knockout , Male , Rosiglitazone/pharmacology
9.
Int J Biol Macromol ; 269(Pt 2): 131964, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38692525

ABSTRACT

This study aims to identify FDA-approved drugs that can target the kappa-opioid receptor (KOR) for the treatment of demyelinating diseases. Demyelinating diseases are characterized by myelin sheath destruction or formation that results in severe neurological dysfunction. Remission of this disease is largely dependent on the differentiation of oligodendrocyte precursor cells (OPCs) into mature oligodendrocytes (OLGs) in demyelinating lesions. KOR is an important regulatory protein and drug target for the treatment of demyelinating diseases. However, no drug targeting KOR has been developed due to the long clinical trials for drug discovery. Here, a structure-based virtual screening was applied to identify drugs targeting KOR among 1843 drugs of FDA-approved drug libraries, and famotidine was screen out by its high affinity cooperation with KOR as well as the clinical safety. We discovered that famotidine directly promoted OPC maturation and remyelination using the complementary in vitro and in vivo models. Administration of famotidine was not only effectively enhanced CNS myelinogenesis, but also promoted remyelination. Mechanically speaking, famotidine promoted myelinogenesis or remyelination through KOR/STAT3 signaling pathway. In general, our study provided evidence of new clinical applicability of famotidine for the treatment of demyelinating diseases for which there is currently no effective therapy.


Subject(s)
Cell Differentiation , Famotidine , Receptors, Opioid, kappa , Remyelination , STAT3 Transcription Factor , Signal Transduction , Famotidine/pharmacology , STAT3 Transcription Factor/metabolism , Animals , Signal Transduction/drug effects , Cell Differentiation/drug effects , Remyelination/drug effects , Receptors, Opioid, kappa/metabolism , Oligodendrocyte Precursor Cells/drug effects , Oligodendrocyte Precursor Cells/metabolism , Oligodendrocyte Precursor Cells/cytology , Central Nervous System/drug effects , Central Nervous System/metabolism , Mice , Rats , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Oligodendroglia/cytology , Myelin Sheath/metabolism , Myelin Sheath/drug effects , Demyelinating Diseases/drug therapy , Demyelinating Diseases/metabolism , Humans
10.
Cell ; 187(10): 2465-2484.e22, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38701782

ABSTRACT

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.


Subject(s)
Epigenesis, Genetic , Myelin Sheath , Oligodendroglia , Remyelination , Animals , Myelin Sheath/metabolism , Humans , Mice , Remyelination/drug effects , Oligodendroglia/metabolism , Central Nervous System/metabolism , Mice, Inbred C57BL , Rejuvenation , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/drug effects , Sterol Regulatory Element Binding Protein 1/metabolism , Organoids/metabolism , Organoids/drug effects , Demyelinating Diseases/metabolism , Demyelinating Diseases/genetics , Cell Differentiation/drug effects , Small Molecule Libraries/pharmacology , Male , Regeneration/drug effects , Multiple Sclerosis/metabolism , Multiple Sclerosis/genetics , Multiple Sclerosis/drug therapy , Multiple Sclerosis/pathology
11.
Elife ; 132024 May 09.
Article in English | MEDLINE | ID: mdl-38722021

ABSTRACT

Neural stem cells (NSCs) are multipotent and correct fate determination is crucial to guarantee brain formation and homeostasis. How NSCs are instructed to generate neuronal or glial progeny is not well understood. Here, we addressed how murine adult hippocampal NSC fate is regulated and described how scaffold attachment factor B (SAFB) blocks oligodendrocyte production to enable neuron generation. We found that SAFB prevents NSC expression of the transcription factor nuclear factor I/B (NFIB) by binding to sequences in the Nfib mRNA and enhancing Drosha-dependent cleavage of the transcripts. We show that increasing SAFB expression prevents oligodendrocyte production by multipotent adult NSCs, and conditional deletion of Safb increases NFIB expression and oligodendrocyte formation in the adult hippocampus. Our results provide novel insights into a mechanism that controls Drosha functions for selective regulation of NSC fate by modulating the post-transcriptional destabilization of Nfib mRNA in a lineage-specific manner.


Subject(s)
Hippocampus , NFI Transcription Factors , Neural Stem Cells , RNA, Messenger , Ribonuclease III , Animals , NFI Transcription Factors/metabolism , NFI Transcription Factors/genetics , Hippocampus/metabolism , Hippocampus/cytology , Ribonuclease III/metabolism , Ribonuclease III/genetics , Mice , Neural Stem Cells/metabolism , RNA, Messenger/metabolism , RNA, Messenger/genetics , Oligodendroglia/metabolism , RNA Stability , Cell Differentiation
12.
Zool Res ; 45(3): 691-703, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38766750

ABSTRACT

General anesthetic agents can impact brain function through interactions with neurons and their effects on glial cells. Oligodendrocytes perform essential roles in the central nervous system, including myelin sheath formation, axonal metabolism, and neuroplasticity regulation. They are particularly vulnerable to the effects of general anesthetic agents resulting in impaired proliferation, differentiation, and apoptosis. Neurologists are increasingly interested in the effects of general anesthetic agents on oligodendrocytes. These agents not only act on the surface receptors of oligodendrocytes to elicit neuroinflammation through modulation of signaling pathways, but also disrupt metabolic processes and alter the expression of genes involved in oligodendrocyte development and function. In this review, we summarize the effects of general anesthetic agents on oligodendrocytes. We anticipate that future research will continue to explore these effects and develop strategies to decrease the incidence of adverse reactions associated with the use of general anesthetic agents.


Subject(s)
Anesthetics, General , Brain , Oligodendroglia , Oligodendroglia/drug effects , Animals , Brain/drug effects , Anesthetics, General/adverse effects , Anesthetics, General/toxicity , Neurotoxicity Syndromes/etiology , Humans
13.
Mol Biol Rep ; 51(1): 674, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38787497

ABSTRACT

BACKGROUND: Multiple sclerosis (MS) is a chronic demyelinating disease of the central nervous system (CNS) with inflammation and immune dysfunction. OBJECTIVES: We compared the remyelination and immunomodulation properties of mesenchymal stem cells (MSCs) with their conditioned medium (CM) in the cuprizone model. METHODS: Twenty-four C57BL/ 6 mice were divided into four groups. After cuprizone demyelination, MSCs and their CM were injected into the right lateral ventricle of mice. The expression level of IL-1ß, TNF-α, and BDNF genes was evaluated using the qRT-PCR. APC antibody was used to assess the oligodendrocyte population using the immunofluorescent method. The remyelination and axonal repair were studied by specific staining of the LFB and electron microscopy techniques. RESULTS: Transplantation of MSCs and CM increased the expression of the BDNF gene and decreased the expression of IL-1ß and TNF-α genes compared to the cuprizone group, and these effects in the cell group were more than CM. Furthermore, cell transplantation resulted in a significant improvement in myelination and axonal repair, which was measured by luxol fast blue and transmission electron microscope images. The cell group had a higher number of oligodendrocytes than other groups. CONCLUSIONS: According to the findings, injecting MSCs intraventricularly versus cell-conditioned medium can be a more effective approach to improving chronic demyelination in degenerative diseases like MS.


Subject(s)
Cuprizone , Demyelinating Diseases , Disease Models, Animal , Inflammation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Mice, Inbred C57BL , Animals , Mesenchymal Stem Cell Transplantation/methods , Mice , Mesenchymal Stem Cells/metabolism , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Culture Media, Conditioned/pharmacology , Inflammation/pathology , Inflammation/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics , Interleukin-1beta/metabolism , Interleukin-1beta/genetics , Oligodendroglia/metabolism , Remyelination , Multiple Sclerosis/pathology , Multiple Sclerosis/therapy , Multiple Sclerosis/metabolism , Multiple Sclerosis/chemically induced , Tumor Necrosis Factor-alpha/metabolism , Male , Myelin Sheath/metabolism
14.
Science ; 384(6696): eadk4858, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38723085

ABSTRACT

To fully understand how the human brain works, knowledge of its structure at high resolution is needed. Presented here is a computationally intensive reconstruction of the ultrastructure of a cubic millimeter of human temporal cortex that was surgically removed to gain access to an underlying epileptic focus. It contains about 57,000 cells, about 230 millimeters of blood vessels, and about 150 million synapses and comprises 1.4 petabytes. Our analysis showed that glia outnumber neurons 2:1, oligodendrocytes were the most common cell, deep layer excitatory neurons could be classified on the basis of dendritic orientation, and among thousands of weak connections to each neuron, there exist rare powerful axonal inputs of up to 50 synapses. Further studies using this resource may bring valuable insights into the mysteries of the human brain.


Subject(s)
Cerebral Cortex , Humans , Axons/physiology , Axons/ultrastructure , Cerebral Cortex/blood supply , Cerebral Cortex/ultrastructure , Dendrites/physiology , Neurons/ultrastructure , Oligodendroglia/ultrastructure , Synapses/physiology , Synapses/ultrastructure , Temporal Lobe/ultrastructure , Microscopy
16.
Int J Mol Sci ; 25(7)2024 Mar 23.
Article in English | MEDLINE | ID: mdl-38612430

ABSTRACT

A variety of neurological and psychiatric disorders have recently been shown to be highly associated with the abnormal development and function of oligodendrocytes (OLs) and interneurons. OLs are the myelin-forming cells in the central nervous system (CNS), while interneurons are important neural types gating the function of excitatory neurons. These two types of cells are of great significance for the establishment and function of neural circuits, and they share similar developmental origins and transcriptional architectures, and interact with each other in multiple ways during development. In this review, we compare the similarities and differences in these two cell types, providing an important reference and further revealing the pathogenesis of related brain disorders.


Subject(s)
Interneurons , Oligodendroglia , Humans , Myelin Sheath , Neurons , Brain
17.
J Neurosci Res ; 102(4): e25334, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38656648

ABSTRACT

Iron deficiency (ID) has been shown to affect central nervous system (CNS) development and induce hypomyelination. Previous work from our laboratory in a gestational ID model showed that both oligodendrocyte (OLG) and astrocyte (AST) maturation was impaired. To explore the contribution of AST iron to the myelination process, we generated an in vitro ID model by silencing divalent metal transporter 1 (DMT1) in AST (siDMT1 AST) or treating AST with Fe3+ chelator deferoxamine (DFX; DFX AST). siDMT1 AST showed no changes in proliferation but remained immature. Co-cultures of oligodendrocyte precursors cells (OPC) with siDMT1 AST and OPC cultures incubated with siDMT1 AST-conditioned media (ACM) rendered a reduction in OPC maturation. These findings correlated with a decrease in the expression of AST-secreted factors IGF-1, NRG-1, and LIF, known to promote OPC differentiation. siDMT1 AST also displayed increased mitochondrial number and reduced mitochondrial size as compared to control cells. DFX AST also remained immature and DFX AST-conditioned media also hampered OPC maturation in culture, in keeping with a decrease in the expression of AST-secreted growth factors IGF-1, NRG-1, LIF, and CNTF. DFX AST mitochondrial morphology and number showed results similar to those observed in siDMT1 AST. In sum, our results show that ID, induced through two different methods, impacts AST maturation and mitochondrial functioning, which in turn hampers OPC differentiation.


Subject(s)
Astrocytes , Cell Differentiation , Iron Deficiencies , Oligodendroglia , Astrocytes/metabolism , Astrocytes/drug effects , Oligodendroglia/metabolism , Oligodendroglia/drug effects , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Cation Transport Proteins/metabolism , Coculture Techniques , Culture Media, Conditioned/pharmacology , Rats , Oligodendrocyte Precursor Cells/drug effects , Oligodendrocyte Precursor Cells/metabolism , Deferoxamine/pharmacology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Iron/metabolism
18.
Cells ; 13(8)2024 Apr 13.
Article in English | MEDLINE | ID: mdl-38667289

ABSTRACT

Oligodendrocytes originating in the brain and spinal cord as well as in the ventral and dorsal domains of the neural tube are transcriptomically and functionally distinct. These distinctions are also reflected in the ultrastructure of the produced myelin, and the susceptibility to myelin-related disorders, which highlights the significance of the choice of patterning protocols in the differentiation of induced pluripotent stem cells (iPSCs) into oligodendrocytes. Thus, our first goal was to survey the different approaches applied to the generation of iPSC-derived oligodendrocytes in 2D culture and in organoids, as well as reflect on how these approaches pertain to the regional and spatial fate of the generated oligodendrocyte progenitors and myelinating oligodendrocytes. This knowledge is increasingly important to disease modeling and future therapeutic strategies. Our second goal was to recap the recent advances in the development of oligodendrocyte-enriched organoids, as we explore their relevance to a regional specification alongside their duration, complexity, and maturation stages of oligodendrocytes and myelin biology. Finally, we discuss the shortcomings of the existing protocols and potential future explorations.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells , Oligodendroglia , Organoids , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Organoids/cytology , Humans , Animals , Myelin Sheath/metabolism , Cell Culture Techniques/methods
19.
Neurol Neuroimmunol Neuroinflamm ; 11(3): e200230, 2024 May.
Article in English | MEDLINE | ID: mdl-38669615

ABSTRACT

BACKGROUND AND OBJECTIVES: The aim of this study was to identify novel biomarkers for multiple sclerosis (MS) diagnosis and prognosis, addressing the critical need for specific and prognostically valuable markers in the field. METHODS: We conducted an extensive proteomic investigation, combining analysis of (1) CSF proteome from symptomatic controls, fast and slow converters after clinically isolated syndromes, and patients with relapsing-remitting MS (n = 10 per group) using label-free quantitative proteomics and (2) oligodendrocyte secretome changes under proinflammatory or proapoptotic conditions using stable isotope labeling by amino acids in cell culture. Proteins exhibiting differential abundance in both proteomic analyses were combined with other putative MS biomarkers, yielding a comprehensive list of 87 proteins that underwent quantification through parallel reaction monitoring (PRM) in a novel cohort, comprising symptomatic controls, inflammatory neurologic disease controls, and patients with MS at various disease stages (n = 10 per group). The 11 proteins that passed this qualification step were subjected to a new PRM assay within an expanded cohort comprising 158 patients with either MS at different disease stages or other inflammatory or noninflammatory neurologic disease controls. RESULTS: This study unveiled a promising biomarker signature for MS, including previously established candidates, such as chitinase 3-like protein 1, chitinase 3-like protein 2, chitotriosidase, immunoglobulin kappa chain region C, neutrophil gelatinase-associated lipocalin, and CD27. In addition, we identified novel markers, namely cat eye syndrome critical region protein 1 (adenosine deaminase 2, a therapeutic target in multiple sclerosis) and syndecan-1, a proteoglycan, also known as plasma cell surface marker CD138 and acting as chitinase 3-like protein 1 receptor implicated in inflammation and cancer signaling. CD138 exhibited good diagnostic accuracy in distinguishing MS from inflammatory neurologic disorders (area under the curve [AUC] = 0.85, CI 0.75-0.95). CD138 immunostaining was also observed in the brains of patients with MS and cultured oligodendrocyte precursor cells but was absent in astrocytes. DISCUSSION: These findings identify CD138 as a specific CSF biomarker for MS and suggest the selective activation of the chitinase 3-like protein 1/CD138 pathway within the oligodendrocyte lineage in MS. They offer promising prospects for improving MS diagnosis and prognosis by providing much-needed specificity and clinical utility. CLASSIFICATION OF EVIDENCE: This study provides Class II evidence that CD138 distinguishes multiple sclerosis from other inflammatory neurologic disorders with an AUC of 0.85 (95% CI 0.75-0.95).


Subject(s)
Biomarkers , Multiple Sclerosis, Relapsing-Remitting , Syndecan-1 , Humans , Biomarkers/cerebrospinal fluid , Adult , Female , Male , Multiple Sclerosis, Relapsing-Remitting/cerebrospinal fluid , Multiple Sclerosis, Relapsing-Remitting/diagnosis , Middle Aged , Syndecan-1/cerebrospinal fluid , Cohort Studies , Proteomics , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/diagnosis , Oligodendroglia/metabolism
20.
J Mol Neurosci ; 74(2): 40, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38594388

ABSTRACT

Astrocytes, the most prevalent cells in the central nervous system (CNS), can be transformed into neurons and oligodendrocyte progenitor cells (OPCs) using specific transcription factors and some chemicals. In this study, we present a cocktail of small molecules that target different signaling pathways to promote astrocyte conversion to OPCs. Astrocytes were transferred to an OPC medium and exposed for five days to a small molecule cocktail containing CHIR99021, Forskolin, Repsox, LDN, VPA and Thiazovivin before being preserved in the OPC medium for an additional 10 days. Once reaching the OPC morphology, induced cells underwent immunocytofluorescence evaluation for OPC markers while checked for lacking the astrocyte markers. To test the in vivo differentiation capabilities, induced OPCs were transplanted into demyelinated mice brains treated with cuprizone over 12 weeks. Two distinct lines of astrocytes demonstrated the potential of conversion to OPCs using this small molecule cocktail as verified by morphological changes and the expression of PDGFR and O4 markers as well as the terminal differentiation to oligodendrocytes expressing MBP. Following transplantation into demyelinated mice brains, induced OPCs effectively differentiated into mature oligodendrocytes. The generation of OPCs from astrocytes via a small molecule cocktail may provide a new avenue for producing required progenitors necessary for myelin repair in diseases characterized by the loss of myelin such as multiple sclerosis.


Subject(s)
Multiple Sclerosis , Oligodendrocyte Precursor Cells , Mice , Animals , Multiple Sclerosis/therapy , Multiple Sclerosis/metabolism , Astrocytes/metabolism , Oligodendrocyte Precursor Cells/metabolism , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Cell Differentiation/physiology , Disease Models, Animal , Cell Line
SELECTION OF CITATIONS
SEARCH DETAIL
...