Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 182
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.
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
3.
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
4.
Acta Biomater ; 180: 308-322, 2024 May.
Article in English | MEDLINE | ID: mdl-38615813

ABSTRACT

Motor functional improvement represents a paramount treatment objective in the post-spinal cord injury (SCI) recovery process. However, neuronal cell death and axonal degeneration following SCI disrupt neural signaling, impeding the motor functional recovery. In this study, we developed a multifunctional decellularized spinal cord-derived extracellular matrix (dSECM), crosslinked with glial cell-derived neurotrophic factor (GDNF), to promote differentiation of stem cells into neural-like cells and facilitate axonogenesis and remyelination. After decellularization, the immunogenic cellular components were effectively removed in dSECM, while the crucial protein components were retained which supports stem cells proliferation and differentiation. Furthermore, sustained release of GDNF from the dSECM facilitated axonogenesis and remyelination by activating the PI3K/Akt and MEK/Erk pathways. Our findings demonstrate that the dSECM-GDNF platform promotes neurogenesis, axonogenesis, and remyelination to enhance neural signaling, thereby yielding promising therapeutic effects for motor functional improvement after SCI. STATEMENT OF SIGNIFICANCE: The dSECM promotes the proliferation and differentiation of MSCs or NSCs by retaining proteins associated with positive regulation of neurogenesis and neuronal differentiation, while eliminating proteins related to negative regulation of neurogenesis. After crosslinking, GDNF can be gradually released from the platform, thereby promoting neural differentiation, axonogenesis, and remyelination to enhance neural signaling through activation of the PI3K/Akt and MEK/Erk pathways. In vivo experiments demonstrated that dSECM-GDNF/MSC@GelMA hydrogel exhibited the ability to facilitate neuronal regeneration at 4 weeks post-surgery, while promoting axonogenesis and remyelination at 8 weeks post-surgery, ultimately leading to enhanced motor functional recovery. This study elucidates the ability of neural regeneration strategy to promote motor functional recovery and provides a promising approach for designing multifunctional tissue for SCI treatment.


Subject(s)
Extracellular Matrix , Glial Cell Line-Derived Neurotrophic Factor , Neurogenesis , Rats, Sprague-Dawley , Recovery of Function , Remyelination , Spinal Cord Injuries , Animals , Spinal Cord Injuries/therapy , Spinal Cord Injuries/pathology , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Neurogenesis/drug effects , Remyelination/drug effects , Extracellular Matrix/metabolism , Recovery of Function/drug effects , Rats , Female , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism
5.
Acta Neuropathol ; 147(1): 75, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38656399

ABSTRACT

In multiple sclerosis (MS), persisting disability can occur independent of relapse activity or development of new central nervous system (CNS) inflammatory lesions, termed chronic progression. This process occurs early and it is mostly driven by cells within the CNS. One promising strategy to control progression of MS is the inhibition of the enzyme Bruton's tyrosine kinase (BTK), which is centrally involved in the activation of both B cells and myeloid cells, such as macrophages and microglia. The benefit of BTK inhibition by evobrutinib was shown as we observed reduced pro-inflammatory activation of microglia when treating chronic experimental autoimmune encephalomyelitis (EAE) or following the adoptive transfer of activated T cells. Additionally, in a model of toxic demyelination, evobrutinib-mediated BTK inhibition promoted the clearance of myelin debris by microglia, leading to an accelerated remyelination. These findings highlight that BTK inhibition has the potential to counteract underlying chronic progression of MS.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase , Encephalomyelitis, Autoimmune, Experimental , Microglia , Myelin Sheath , Piperidines , Pyrimidines , Animals , Female , Mice , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Agammaglobulinaemia Tyrosine Kinase/metabolism , Biphenyl Compounds/pharmacology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice, Inbred C57BL , Microglia/pathology , Microglia/drug effects , Microglia/metabolism , Myelin Sheath/pathology , Myelin Sheath/metabolism , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Remyelination/physiology , Remyelination/drug effects
6.
Front Immunol ; 15: 1362629, 2024.
Article in English | MEDLINE | ID: mdl-38680485

ABSTRACT

Introduction: Despite advances in immunomodulatory treatments of multiple sclerosis (MS), patients with non-active progressive multiple sclerosis (PMS) continue to face a significant unmet need. Demyelination, smoldering inflammation and neurodegeneration are important drivers of disability progression that are insufficiently targeted by current treatment approaches. Promising preclinical data support repurposing of metformin for treatment of PMS. The objective of this clinical trial is to evaluate whether metformin, as add-on treatment, is superior to placebo in delaying disease progression in patients with non-active PMS. Methods and analysis: MACSiMiSE-BRAIN is a multi-center two-arm, 1:1 randomized, triple-blind, placebo-controlled clinical trial, conducted at five sites in Belgium. Enrollment of 120 patients with non-active PMS is planned. Each participant will undergo a screening visit with assessment of baseline magnetic resonance imaging (MRI), clinical tests, questionnaires, and a safety laboratory assessment. Following randomization, participants will be assigned to either the treatment (metformin) or placebo group. Subsequently, they will undergo a 96-week follow-up period. The primary outcome is change in walking speed, as measured by the Timed 25-Foot Walk Test, from baseline to 96 weeks. Secondary outcome measures include change in neurological disability (Expanded Disability Status Score), information processing speed (Symbol Digit Modalities Test) and hand function (9-Hole Peg test). Annual brain MRI will be performed to assess evolution in brain volumetry and diffusion metrics. As patients may not progress in all domains, a composite outcome, the Overall Disability Response Score will be additionally evaluated as an exploratory outcome. Other exploratory outcomes will consist of paramagnetic rim lesions, the 2-minute walking test and health economic analyses as well as both patient- and caregiver-reported outcomes like the EQ-5D-5L, the Multiple Sclerosis Impact Scale and the Caregiver Strain Index. Ethics and dissemination: Clinical trial authorization from regulatory agencies [Ethical Committee and Federal Agency for Medicines and Health Products (FAMHP)] was obtained after submission to the centralized European Clinical Trial Information System. The results of this clinical trial will be disseminated at scientific conferences, in peer-reviewed publications, to patient associations and the general public. Trial registration: ClinicalTrials.gov Identifier: NCT05893225, EUCT number: 2023-503190-38-00.


Subject(s)
Brain , Metformin , Multiple Sclerosis , Adult , Female , Humans , Male , Middle Aged , Brain/diagnostic imaging , Brain/pathology , Brain/drug effects , Disease Progression , Drug Therapy, Combination , Magnetic Resonance Imaging , Metformin/therapeutic use , Multicenter Studies as Topic , Multiple Sclerosis/drug therapy , Multiple Sclerosis, Chronic Progressive/drug therapy , Randomized Controlled Trials as Topic , Remyelination/drug effects , Treatment Outcome
7.
J Neuroophthalmol ; 44(2): 143-156, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38654413

ABSTRACT

INTRODUCTION: Amelioration of disability in multiple sclerosis requires the development of complementary therapies that target neurodegeneration and promote repair. Remyelination is a promising neuroprotective strategy that may protect axons from damage and subsequent neurodegeneration. METHODS: A review of key literature plus additional targeted search of PubMed and Google Scholar was conducted. RESULTS: There has been a rapid expansion of clinical trials studying putative remyelinating candidates, but further growth of the field is limited by the lack of consensus on key aspects of trial design. We have not yet defined the ideal study population, duration of therapy, or the appropriate outcome measures to detect remyelination in humans. The varied natural history of multiple sclerosis, coupled with the short time frame of phase II clinical trials, requires that we develop and validate biomarkers of remyelination that can serve as surrogate endpoints in clinical trials. CONCLUSIONS: We propose that the visual system may be the most well-suited and validated model for the study potential remyelinating agents. In this review, we discuss the pathophysiology of demyelination and summarize the current clinical trial landscape of remyelinating agents. We present some of the challenges in the study of remyelinating agents and discuss current potential biomarkers of remyelination and repair, emphasizing both established and emerging visual outcome measures.


Subject(s)
Multiple Sclerosis , Remyelination , Humans , Multiple Sclerosis/physiopathology , Multiple Sclerosis/drug therapy , Remyelination/physiology , Remyelination/drug effects , Myelin Sheath
8.
Neurorehabil Neural Repair ; 38(5): 350-363, 2024 May.
Article in English | MEDLINE | ID: mdl-38491852

ABSTRACT

BACKGROUND: Yi-Qi-Tong-Luo Granules (YQTLs) is a natural compound of Traditional Chinese Medicine authorized by China Food and Drug Administration (CFDA). These granules are employed in the convalescent stage of cerebral infarction and render notable clinical efficacy. This study aims to uncover the underlying mechanisms of YQTLs on remyelination after cerebral ischemia injury. MATERIALS AND METHODS: We established cerebral ischemia model in rats using microsphere-induced multiple cerebral infarction (MCI). We evaluated the pharmacological effects of YQTLs on MCI rats, through Morri's water maze test, open field test, hematoxylin and eosin staining, and glycine silver immersion. We employed liquid chromatography mass spectrometry metabolomics to identify differential metabolites. Enzyme-linked immunosorbent assay was utilized to measure the release of neurotrophins, while immunofluorescence staining was used to assess oligodendrocyte precursor cells differences and myelin regeneration. We used Western blotting to validate the protein expression of remyelination-associated signaling pathways. RESULTS: YQTLs significantly improves cognitive function following cerebral ischemia injury. Pathological tissue staining revealed that YQTLs administration inhibits neuronal denaturation and neurofibrillary tangles. We identified 141 differential metabolites among the sham, MCI, and YQTLs-treated MCI groups. Among these metabolites, neurotransmitters were identified, and notably, gamma-aminobutyric acid (GABA) showed marked improvement in the YQTLs group. The induction of neurotrophins, such as brain-derived neurotrophic factor (BDNF) and PDGFAA, upregulation of olig2 and MBP expression, and promotion of remyelination were evident in YQTLs-treated MCI groups. Gamma-aminobutyric acid B receptors (GABABR), pERK/extracellular regulated MAP kinase, pAKT/protein kinase B, and pCREB/cAMP response element-binding were upregulated following YQTLs treatment. CONCLUSION: YQTLs enhance the binding of GABA to GABABR, thereby activating the pCREB/BDNF signaling pathway, which in turn increases the expression of downstream myelin-associated proteins and promotes remyelination and cognitive function.


Subject(s)
Brain Ischemia , Brain-Derived Neurotrophic Factor , Metabolomics , Rats, Sprague-Dawley , Remyelination , Signal Transduction , Animals , Remyelination/drug effects , Remyelination/physiology , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/drug effects , Rats , Male , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Disease Models, Animal , Drugs, Chinese Herbal/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP Response Element-Binding Protein/drug effects
9.
J Biol Chem ; 300(1): 105487, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37995941

ABSTRACT

Oligodendrocyte precursor cells are present in the adult central nervous system, and their impaired ability to differentiate into myelinating oligodendrocytes can lead to demyelination in patients with multiple sclerosis, accompanied by neurological deficits and cognitive impairment. Exosomes, small vesicles released by cells, are known to facilitate intercellular communication by carrying bioactive molecules. In this study, we utilized exosomes derived from human umbilical cord mesenchymal stem cells (HUMSCs-Exos). We performed sequencing and bioinformatics analysis of exosome-treated cells to demonstrate that HUMSCs-Exos can stimulate myelin gene expression in oigodendrocyte precursor cells. Functional investigations revealed that HUMSCs-Exos activate the Pi3k/Akt pathway and regulate the Tbr1/Wnt signaling molecules through the transfer of miR-23a-3p, promoting oligodendrocytes differentiation and enhancing the expression of myelin-related proteins. In an experimental autoimmune encephalomyelitis model, treatment with HUMSCs-Exos significantly improved neurological function and facilitated remyelination. This study provides cellular and molecular insights into the use of cell-free exosome therapy for central nervous system demyelination associated with multiple sclerosis, demonstrating its great potential for treating demyelinating and neurodegenerative diseases.


Subject(s)
Exosomes , Mesenchymal Stem Cells , MicroRNAs , Multiple Sclerosis , Remyelination , Adult , Humans , Cell Differentiation/genetics , Exosomes/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , MicroRNAs/pharmacology , MicroRNAs/therapeutic use , Multiple Sclerosis/genetics , Multiple Sclerosis/therapy , Multiple Sclerosis/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Remyelination/drug effects , Remyelination/genetics , Umbilical Cord/cytology , Umbilical Cord/metabolism , Wnt Signaling Pathway/drug effects , Signal Transduction/drug effects , Signal Transduction/genetics , T-Box Domain Proteins/metabolism , Disease Models, Animal , Cells, Cultured
10.
Ann Clin Transl Neurol ; 9(7): 1090-1094, 2022 07.
Article in English | MEDLINE | ID: mdl-35587315

ABSTRACT

Remyelination efficiency declines with advancing age in animal models, but this has been harder to demonstrate in people with multiple sclerosis. We show that bexarotene, a putatively remyelinating retinoid-X receptor agonist, shortened the visual evoked potential latency in patients with chronic optic neuropathy aged under 42 years only (with the effect diminishing by 0.45 ms per year of age); and increased the magnetization transfer ratio of deep gray matter lesions in those under 43 years only. Addressing this age-related decline in human remyelination capacity will be an important step in the development of remyelinating therapies that work across the lifespan.


Subject(s)
Bexarotene , Optic Nerve Diseases , Peripheral Nervous System Agents , Remyelination , Retinoid X Receptors , Age Factors , Aged , Animals , Bexarotene/pharmacology , Bexarotene/therapeutic use , Evoked Potentials, Visual/drug effects , Evoked Potentials, Visual/physiology , Humans , Multiple Sclerosis/complications , Multiple Sclerosis/drug therapy , Multiple Sclerosis/physiopathology , Optic Nerve Diseases/drug therapy , Optic Nerve Diseases/etiology , Optic Nerve Diseases/physiopathology , Peripheral Nervous System Agents/pharmacology , Peripheral Nervous System Agents/therapeutic use , Remyelination/drug effects , Remyelination/physiology , Retinoid X Receptors/administration & dosage , Retinoid X Receptors/agonists , Retinoid X Receptors/pharmacology , Retinoids/administration & dosage , Retinoids/pharmacology
11.
Article in English | MEDLINE | ID: mdl-35105686

ABSTRACT

BACKGROUND AND OBJECTIVES: Demyelinating diseases in the CNS are characterized by myelin sheath destruction or formation disorder that leads to severe neurologic dysfunction. Remission of such diseases is largely dependent on the differentiation of oligodendrocytes precursor cells (OPCs) into mature myelin-forming OLGs at the demyelinated lesions, which is defined as remyelination. We discover that baicalin (BA), a natural flavonoid, in addition to its well-known antiinflammatory effects, directly stimulates OLG maturation and CNS myelin repair. METHODS: To investigate the function of BA on CNS remyelination, we develop the complementary in vivo and in vitro models, including physiologic neonatal mouse CNS myelinogenesis model, pathologic cuprizone-induced (CPZ-induced) toxic demyelination model, and postnatal OLG maturation assay. Furthermore, molecular docking, pharmacologic regulation, and transgenic heterozygous mice were used to clarify the target and action of the mechanism of BA on myelin repair promotion. RESULTS: Administration of BA was not only merely effectively enhanced CNS myelinogenesis during postnatal development but also promoted remyelination and reversed the coordination movement disorder in the CPZ-induced toxic demyelination model. Of note, myelin-promoting effects of BA on myelination or regeneration is peroxisome proliferator-activated receptor γ (PPARγ) signaling-dependent. DISCUSSION: Our work demonstrated that BA promotes myelin production and regeneration by activating the PPARγ signal pathway and also confirmed that BA is an effective natural product for the treatment of demyelinating diseases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Demyelinating Diseases/drug therapy , Flavonoids/pharmacology , Motor Skills Disorders/drug therapy , PPAR gamma/drug effects , Remyelination/drug effects , Signal Transduction/drug effects , Animals , Animals, Newborn , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Transgenic
12.
J Neuroinflammation ; 18(1): 238, 2021 Oct 16.
Article in English | MEDLINE | ID: mdl-34656124

ABSTRACT

BACKGROUND: Epigenetic regulation by histone deacetylases (HDACs) in Schwann cells (SCs) after injury facilitates them to undergo de- and redifferentiation processes necessary to support various stages of nerve repair. Although de-differentiation activates the synthesis and secretion of inflammatory cytokines by SCs to initiate an immune response during nerve repair, changes in either the timing or duration of prolonged inflammation mediated by SCs can affect later processes associated with repair and regeneration. Limited studies have investigated the regulatory processes through which HDACs in SCs control inflammatory cytokines to provide a favorable environment for peripheral nerve regeneration. METHODS: We employed the HDAC inhibitor (HDACi) sodium phenylbutyrate (PBA) to address this question in an in vitro RT4 SC inflammation model and an in vivo sciatic nerve transection injury model to examine the effects of HDAC inhibition on the expression of pro-inflammatory cytokines. Furthermore, we assessed the outcomes of suppression of extended inflammation on the regenerative potential of nerves by assessing axonal regeneration, remyelination, and reinnervation. RESULTS: Significant reductions in lipopolysaccharide (LPS)-induced pro-inflammatory cytokine (tumor necrosis factor-α [TNFα]) expression and secretion were observed in vitro following PBA treatment. PBA treatment also affected the transient changes in nuclear factor κB (NFκB)-p65 phosphorylation and translocation in response to LPS induction in RT4 SCs. Similarly, PBA mediated long-term suppressive effects on HDAC3 expression and activity. PBA administration resulted in marked inhibition of pro-inflammatory cytokine secretion at the site of transection injury when compared with that in the hydrogel control group at 6-week post-injury. A conducive microenvironment for axonal regrowth and remyelination was generated by increasing expression levels of protein gene product 9.5 (PGP9.5) and myelin basic protein (MBP) in regenerating nerve tissues. PBA administration increased the relative gastrocnemius muscle weight percentage and maintained the intactness of muscle bundles when compared with those in the hydrogel control group. CONCLUSIONS: Suppressing the lengthened state of inflammation using PBA treatment favors axonal regrowth and remyelination following nerve transection injury. PBA treatment also regulates pro-inflammatory cytokine expression by inhibiting the transcriptional activation of NFκB-p65 and HDAC3 in SCs in vitro.


Subject(s)
Axons/metabolism , Histone Deacetylases/metabolism , NF-kappa B/metabolism , Nerve Regeneration/physiology , Phenylbutyrates/pharmacology , Remyelination/physiology , Animals , Axons/drug effects , Axons/pathology , Cell Line , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Humans , Inflammation/metabolism , Inflammation/pathology , Inflammation/prevention & control , Male , NF-kappa B/antagonists & inhibitors , Nerve Regeneration/drug effects , Phenylbutyrates/therapeutic use , Rats , Rats, Sprague-Dawley , Remyelination/drug effects , Schwann Cells/drug effects , Schwann Cells/metabolism , Schwann Cells/pathology , Sciatic Neuropathy , THP-1 Cells
13.
Immunopharmacol Immunotoxicol ; 43(6): 790-798, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34618622

ABSTRACT

AIM: Demyelination and subsequent remyelination are well-known mechanisms in multiple sclerosis (MS) pathology. Current research mainly focused on preventing demyelination or regulating the peripheral immune system to protect further damage to the central nervous system. However, information about another essential mechanism, remyelination, and its balance of the immune response within the central nervous system's boundaries is still limited. MATERIALS AND METHODS: In this study, we tried to demonstrate the effect of the recently introduced Janus kinase (JAK)-signal transducer and activator of transcription (STAT) inhibitor, tofacitinib, on remyelination.Demyelination was induced by 6-week cuprizone administration, followed by 2-week tofacitinib (10, 30, and 100 mg/kg) treatment. RESULTS: At the functional level, tofacitinib improved cuprizone-induced decline in motor coordination and muscle strength, which were assessed by rotarod and hanging wire tests. Tofacitinib also showed anti-inflammatory effect by alleviating the cuprizone-induced increase in the central levels of interferon-γ (IFN-γ), interleukin (IL)-6, IL-1ß, and tumor necrosis alpha (TNF-α). Furthermore, tofacitinib also suppressed the cuprizone-induced increase in matrix metalloproteinases (MMP)-9 and MMP-2 levels. Additionally, cuprizone-induced loss of myelin integrity and myelin basic protein expression was inhibited by tofacitinib. At the molecular level, we also assessed phosphorylation of STAT-3 and STAT-5, and our data indicates tofacitinib suppressed cuprizone-induced phosphorylation in those proteins. CONCLUSION: Our study highlights JAK/STAT inhibition provides beneficial effects on remyelination via inhibition of inflammatory cascade.


Subject(s)
Chelating Agents/toxicity , Cuprizone/toxicity , Janus Kinase Inhibitors/pharmacology , Myelin Sheath/drug effects , Piperidines/pharmacology , Pyrimidines/pharmacology , Remyelination/drug effects , Animals , Dose-Response Relationship, Drug , Female , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Muscle Strength/drug effects , Muscle Strength/physiology , Myelin Sheath/metabolism , Myelin Sheath/pathology , Remyelination/physiology
14.
Cell Rep ; 37(1): 109695, 2021 10 05.
Article in English | MEDLINE | ID: mdl-34610310

ABSTRACT

The function of poly(ADP-ribosyl) polymerase 1 (PARP1) in myelination and remyelination of the central nervous system (CNS) remains enigmatic. Here, we report that PARP1 is an intrinsic driver for oligodendroglial development and myelination. Genetic PARP1 depletion impairs the differentiation of oligodendrocyte progenitor cells (OPCs) into oligodendrocytes and impedes CNS myelination. Mechanistically, PARP1-mediated PARylation activity is not only necessary but also sufficient for OPC differentiation. At the molecular level, we identify the RNA-binding protein Myef2 as a PARylated target, which controls OPC differentiation through the PARylation-modulated derepression of myelin protein expression. Furthermore, PARP1's enzymatic activity is necessary for oligodendrocyte and myelin regeneration after demyelination. Together, our findings suggest that PARP1-mediated PARylation activity may be a potential therapeutic target for promoting OPC differentiation and remyelination in neurological disorders characterized by arrested OPC differentiation and remyelination failure such as multiple sclerosis.


Subject(s)
Cell Differentiation , Central Nervous System/metabolism , Myelin Sheath/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly ADP Ribosylation/physiology , Animals , Cell Survival/drug effects , Central Nervous System/growth & development , Cuprizone/pharmacology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/chemically induced , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Myelin Sheath/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Oligodendrocyte Precursor Cells/cytology , Oligodendrocyte Precursor Cells/metabolism , Oligodendrocyte Transcription Factor 2/deficiency , Oligodendrocyte Transcription Factor 2/genetics , Oligodendrocyte Transcription Factor 2/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Oligodendroglia/physiology , Poly (ADP-Ribose) Polymerase-1/deficiency , Poly (ADP-Ribose) Polymerase-1/genetics , RNA/metabolism , Remyelination/drug effects , Repressor Proteins/genetics , Repressor Proteins/metabolism
15.
Article in English | MEDLINE | ID: mdl-34642237

ABSTRACT

BACKGROUND AND OBJECTIVES: To test whether low concentrations of teriflunomide (TF) could promote remyelination, we investigate the effect of TF on oligodendrocyte in culture and on remyelination in vivo in 2 demyelinating models. METHODS: The effect of TF on oligodendrocyte precursor cell (OPC) proliferation and differentiation was assessed in vitro in glial cultures derived from neonatal mice and confirmed on fluorescence-activated cell sorting-sorted adult OPCs. The levels of the 8,9-unsaturated sterols lanosterol and zymosterol were quantified in TF- and sham-treated cultures. In vivo, TF was administered orally, and remyelination was assessed both in myelin basic protein-GFP-nitroreductase (Mbp:GFP-NTR) transgenic Xenopus laevis demyelinated by metronidazole and in adult mice demyelinated by lysolecithin. RESULTS: In cultures, low concentrations of TF down to 10 nM decreased OPC proliferation and increased their differentiation, an effect that was also detected on adult OPCs. Oligodendrocyte differentiation induced by TF was abrogated by the oxidosqualene cyclase inhibitor Ro 48-8071 and was mediated by the accumulation of zymosterol. In the demyelinated tadpole, TF enhanced the regeneration of mature oligodendrocytes up to 2.5-fold. In the mouse demyelinated spinal cord, TF promoted the differentiation of newly generated oligodendrocytes by a factor of 1.7-fold and significantly increased remyelination. DISCUSSION: TF enhances zymosterol accumulation in oligodendrocytes and CNS myelin repair, a beneficial off-target effect that should be investigated in patients with multiple sclerosis.


Subject(s)
Central Nervous System Diseases/drug therapy , Cholesterol/metabolism , Crotonates/pharmacology , Demyelinating Diseases/drug therapy , Hydroxybutyrates/pharmacology , Immunosuppressive Agents/pharmacology , Nitriles/pharmacology , Oligodendrocyte Precursor Cells/drug effects , Oligodendroglia/drug effects , Remyelination/drug effects , Toluidines/pharmacology , Animals , Animals, Newborn , Cells, Cultured , Central Nervous System Diseases/metabolism , Crotonates/administration & dosage , Disease Models, Animal , Hydroxybutyrates/administration & dosage , Immunosuppressive Agents/administration & dosage , Larva , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nitriles/administration & dosage , Oligodendrocyte Precursor Cells/metabolism , Oligodendroglia/metabolism , Toluidines/administration & dosage , Xenopus laevis
16.
ACS Appl Mater Interfaces ; 13(41): 48365-48377, 2021 Oct 20.
Article in English | MEDLINE | ID: mdl-34633177

ABSTRACT

It is critical to obtain an anti-inflammatory microenvironment when curing spinal cord injury (SCI). On the basis of this, we prepared Lycium barbarum oligosaccharide (LBO)-nasal mucosa-derived mesenchymal stem cells (EMSCs) fibronectin hydrogel for SCI restoration via inflammatory license effect and M2 polarization of microglias. LBO exhibited remarkable M2 polarization potential for microglia. However, EMSCs primed by LBO generated enhanced paracrine effects through the inflammatory license-like process. The observed dual function is likely based on the TNFR2 pathway. In addition, LBO-EMSC hydrogel possesses a synergistic effect on M2 polarization of microglia through the PI3K-Akt-mTOR signaling pathway. The obtained findings provide a simple approach for MSC-based therapies for SCI and shed more light on the role of TNFR2 on bidirectional regulation in tissue regeneration.


Subject(s)
Fibrin/pharmacology , Hydrogels/pharmacology , Mesenchymal Stem Cells/drug effects , Polysaccharides/pharmacology , Spinal Cord Injuries/drug therapy , Tissue Scaffolds/chemistry , Animals , Axons/drug effects , Axons/metabolism , Cell Line , Fibrin/chemistry , Humans , Hydrogels/chemistry , Inflammation/drug therapy , Inflammation/metabolism , Lycium/chemistry , Male , Mesenchymal Stem Cells/metabolism , Microglia/drug effects , Microglia/metabolism , Nasal Mucosa/cytology , Phosphatidylinositol 3-Kinases/metabolism , Polysaccharides/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Receptors, Tumor Necrosis Factor, Type II/metabolism , Recovery of Function/drug effects , Remyelination/drug effects , Signal Transduction/drug effects , Spinal Cord Injuries/metabolism , TOR Serine-Threonine Kinases/metabolism
17.
Clin Neurol Neurosurg ; 209: 106911, 2021 10.
Article in English | MEDLINE | ID: mdl-34509750

ABSTRACT

OBJECTIVE: Traumatic brain injury is a major health and socioeconomic problem and the first cause of young death worldwide. For this reason, the prevention of post-traumatic brain injury and the research of new methods for it are important today. In this study, we aimed to determine whether the use of antiepileptic drugs contributed to axonal healing after traumatic brain injury. METHODS: Thirty-six Long-Evans rats, each weighing 300-350 g, were used in this study. A total of 6 groups, including the sham, control, and 4 study groups, were determined. A 1.5 mm-sized trauma was created in the biparietal area with a blunt-tipped dissector. Carbamazepine phenytoin valproic acid and levetiracetam (phenytoin: 30 mg/kg, valproic acid: 60 mg/kg, levetiracetam: 80 mg/kg, and carbamazepine: 36 mg/kg) were intraperitoneally administered to the study groups, and the control group intraperitoneally received a physiological saline solution (15 ml/kg) twice daily for 3 days. After 72 h, hemispheres of the sacrificed subjects were taken for examination in biochemistry and histology. Glutathione, malondialdehyde, and NG2 levels in the samples were determined. RESULTS: No significant difference was found in biochemical measurements. Histopathological examination revealed that the NG2 expression was more intense in the group treated with phenytoin and levetiracetam (phenytoin was partly higher) and the amount of edema decreased. The NG2 expression increased and the edema decreased, though lower in the group treated with carbamazepine and valproic acid, compared with phenytoin and levetiracetam. An increase in the NG2 expression and edema intensity were determined in the control and sham groups. CONCLUSION: Antiepileptic drug selection after traumatic brain injury is an important medical matter. Although the patient-oriented selection is essential, the study suggests that the choice of phenytoin, levetiracetam carbamazepine, and valproic acid will, respectively, have an accelerating effect for axonal healing.


Subject(s)
Anticonvulsants/therapeutic use , Axons/drug effects , Brain Injuries, Traumatic/drug therapy , Remyelination/drug effects , Animals , Anticonvulsants/administration & dosage , Axons/physiology , Carbamazepine/administration & dosage , Carbamazepine/therapeutic use , Levetiracetam/administration & dosage , Levetiracetam/therapeutic use , Phenytoin/administration & dosage , Phenytoin/therapeutic use , Rats , Rats, Long-Evans , Treatment Outcome , Valproic Acid/administration & dosage , Valproic Acid/therapeutic use
18.
Article in English | MEDLINE | ID: mdl-34376551

ABSTRACT

Neuroaxonal loss is believed to underpin the progressive disability that characterizes multiple sclerosis (MS). While focal inflammatory demyelination is a principal cause of acute axonal transection and subsequent axonal degeneration, the gradual attrition of permanently demyelinated axons may also contribute to tissue damage, particularly in the progressive phase of the disease. Therefore, remyelination is considered a putative neuroprotective strategy. In this article, we review the potential pitfalls of remyelination trials, provide a framework for their appropriate design and temper the expectations, at times unrealistic, of researchers, regulators and the pharmaceutical industry.


Subject(s)
Clinical Trials as Topic , Multiple Sclerosis/therapy , Remyelination/drug effects , Animals , Humans
19.
Front Immunol ; 12: 712241, 2021.
Article in English | MEDLINE | ID: mdl-34447379

ABSTRACT

The retinoid X receptor agonist bexarotene promotes remyelination in patients with multiple sclerosis. Murine studies have also demonstrated that RXR agonists have anti-inflammatory effects by enhancing the ability of all-trans-retinoic acid (atRA) to promote T-regulatory cell (Treg) induction and reduce Th17 differentiation in vitro. By stimulating human naïve CD4 T-cells in the presence of Treg or Th17 skewing cytokines, we show that bexarotene also tips the human Treg/Th17 axis in favor of Treg induction, but unlike murine cells this occurs independently of atRA and retinoic acid receptor signaling. Tregs induced in the presence of bexarotene express canonical markers of T-regulation and are functionally suppressive in vitro. Circulating Treg numbers did not increase in the blood of trial patients receiving bexarotene; we believe this is because Treg induction is likely to occur within tissues. These findings lend support to developing RXR agonists as treatments of autoimmune diseases, in particular multiple sclerosis.


Subject(s)
Bexarotene/pharmacology , Lymphopoiesis/drug effects , Remyelination/drug effects , Retinoid X Receptors/agonists , T-Lymphocytes, Regulatory/drug effects , Th17 Cells/drug effects , Adult , Alitretinoin/pharmacology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , Cells, Cultured , Clinical Trials as Topic , Fatty Acids, Unsaturated/pharmacology , Female , Forkhead Transcription Factors/analysis , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Middle Aged , Retinoid X Receptors/physiology , T-Lymphocytes, Regulatory/immunology , Tetrahydronaphthalenes/pharmacology , Th17 Cells/cytology
20.
Neuron ; 109(19): 3104-3118.e6, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34390652

ABSTRACT

Oligodendrocyte (OL) maturation arrest in human white matter injury contributes significantly to the failure of endogenous remyelination in multiple sclerosis (MS) and newborn brain injuries such as hypoxic ischemic encephalopathy (HIE) that cause cerebral palsy. In this study, we identify an oligodendroglial-intrinsic factor that controls OL maturation specifically in the setting of injury. We find a requirement for the ring finger protein Rnf43 not in normal development but in neonatal hypoxic injury and remyelination in the adult mammalian CNS. Rnf43, but not the related Znrf3, is potently activated by Wnt signaling in OL progenitor cells (OPCs) and marks activated OPCs in human MS and HIE. Rnf43 is required in an injury-specific context, and it promotes OPC differentiation through negative regulation of Wnt signal strength in OPCs at the level of Fzd1 receptor presentation on the cell surface. Inhibition of Fzd1 using UM206 promotes remyelination following ex vivo and in vivo demyelinating injury.


Subject(s)
Brain Injuries/genetics , Brain Injuries/pathology , Oligodendroglia/pathology , Ubiquitin-Protein Ligases/genetics , Animals , Brain Injuries/metabolism , Demyelinating Diseases/genetics , Frizzled Receptors/drug effects , Frizzled Receptors/genetics , Humans , Mice , Myelin Sheath/drug effects , Myelin Sheath/physiology , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Remyelination/drug effects , Remyelination/genetics , Stem Cells/metabolism , Stem Cells/pathology , White Matter/metabolism , White Matter/pathology , Wnt Signaling Pathway
SELECTION OF CITATIONS
SEARCH DETAIL
...