Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Mol Neurobiol ; 2023 Dec 11.
Article in English | MEDLINE | ID: mdl-38079108

ABSTRACT

Environmental deprivation can have deleterious effects on adaptive myelination and oligodendroglia function. Early stage Huntington disease (HD) is characterised by white-matter myelin abnormalities in both humans and animal models. However, whether deprived environments exacerbate myelin-related pathological features of HD is not clearly understood. Here, we investigated the impact of deprivation and social isolation on ultrastructural features of myelin in the corpus callosum of the YAC128 mouse model of HD and wildtype (WT) mice using transmission electron microscopy. HD pathology on its own leads to increased representation of altered myelin features, such as thinner sheaths and compromised morphology. Interestingly, deprivation mirrors these effects in WT mice but does not greatly exacerbate the already aberrant myelin in HD mice, indicating a disease-related floor effect in the latter animals. These novel findings indicate that environmental deprivation causes abnormalities in myelin ultrastructure in the otherwise healthy corpus callosum of wild-type mice but has distinct effects on HD mice, where compromised myelin integrity is manifest from early stages of the disease.

2.
Neuromolecular Med ; 25(4): 644-649, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37684514

ABSTRACT

Transcriptional and proteomics analyses in human fragile X syndrome (FXS) neurons identified markedly reduced expression of COMT, a key enzyme involved in the metabolism of catecholamines, including dopamine, epinephrine and norepinephrine. FXS is the most common genetic cause of intellectual disability and autism spectrum disorders. COMT encodes for catechol-o-methyltransferase and its association with neuropsychiatric disorders and cognitive function has been extensively studied. We observed a significantly reduced level of COMT in in FXS human neural progenitors and neurons, as well as hippocampal neurons from Fmr1 null mice. We show that deficits in COMT were associated with an altered response in an assay of dopaminergic activity in Fmr1 null mice. These findings demonstrate that loss of FMRP downregulates COMT expression and affects dopamine signaling in FXS, and supports the notion that targeting catecholamine metabolism may be useful in regulating certain neuropsychiatric aspects of FXS.


Subject(s)
Catechol O-Methyltransferase , Fragile X Syndrome , Animals , Humans , Mice , Catechol O-Methyltransferase/genetics , Dopamine/metabolism , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , Mice, Knockout , Neurons/metabolism
3.
Epigenetics Chromatin ; 16(1): 36, 2023 Sep 28.
Article in English | MEDLINE | ID: mdl-37759327

ABSTRACT

BACKGROUND: NPM1 is a phosphoprotein highly abundant in the nucleolus. However, additional nuclear functions have been attributed to NPM1, probably through interaction with other nuclear factors. DOT1L is one interaction partner of NPM1 that catalyzes methylation of histone H3 at lysine 79 (H3K79). DOT1L, playing functional roles in several biological processes, is known for its capability to organize and regulate chromatin. For example, DOT1L modulates DNA repeats expression within peri-nucleolar heterochromatin. NPM1 also affects peri-nucleolar heterochromatin spatial organization. However, it is unclear as of yet whether NPM1 and DOT1L functionally synergize to preserve nucleoli organization and genome stability, and generally, which molecular mechanisms would be involved. RESULTS: We characterized the nuclear function of NPM1 on peri-nucleolar heterochromatin organization. We show that (i) monomeric NPM1 interacts preferentially with DOT1L in the nucleus; (ii) NPM1 acts in concert with DOT1L to maintain each other's protein homeostasis; (iii) NPM1 depletion results in H3K79me2 upregulation and differential enrichment at chromatin binding genes including Ezh2; (iv) NPM1 and DOT1L modulate DNA repeats expression and peri-nucleolar heterochromatin organization via epigenetic mechanisms dependent on H3K27me3. CONCLUSIONS: Our findings give insights into molecular mechanisms employed by NPM1 and DOT1L to regulate heterochromatin activity and structural organization around the nucleoli and shed light on one aspect of the complex role of both proteins in chromatin dynamics.


Subject(s)
Heterochromatin , Histones , Chromatin , DNA , Histones/metabolism , Nucleophosmin , Histone-Lysine N-Methyltransferase/metabolism , Animals , Mice
4.
Cereb Cortex ; 33(19): 10272-10285, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37566909

ABSTRACT

The cortical plate (CP) is composed of excitatory and inhibitory neurons, the latter of which originate in the ganglionic eminences. From their origin in the ventral telencephalon, maturing postmitotic interneurons migrate during embryonic development over some distance to reach their final destination in the CP. The histone methyltransferase Disruptor of Telomeric Silencing 1-like (DOT1L) is necessary for proper CP development and layer distribution of glutamatergic neurons. However, its specific role on cortical interneuron development has not yet been explored. Here, we demonstrate that DOT1L affects interneuron development in a cell autonomous manner. Deletion of Dot1l in Nkx2.1-expressing interneuron precursor cells results in an overall reduction and altered distribution of GABAergic interneurons in the CP from postnatal day 0 onwards. We observed an altered proportion of GABAergic interneurons in the cortex, with a significant decrease in parvalbumin-expressing interneurons. Moreover, a decreased number of mitotic cells at the embryonic day E14.5 was observed upon Dot1l deletion. Altogether, our results indicate that reduced numbers of cortical interneurons upon DOT1L deletion result from premature cell cycle exit, but effects on postmitotic differentiation, maturation, and migration are likely at play as well.


Subject(s)
Histone-Lysine N-Methyltransferase , Interneurons , Parvalbumins , Telencephalon , Cell Differentiation/physiology , Interneurons/cytology , Interneurons/metabolism , Parvalbumins/genetics , Parvalbumins/metabolism , Telencephalon/cytology , Animals , Mice , Histone-Lysine N-Methyltransferase/genetics
5.
EMBO Rep ; 24(8): e56233, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37382163

ABSTRACT

Cortical neurogenesis depends on the balance between self-renewal and differentiation of apical progenitors (APs). Here, we study the epigenetic control of AP's division mode by focusing on the enzymatic activity of the histone methyltransferase DOT1L. Combining lineage tracing with single-cell RNA sequencing of clonally related cells, we show at the cellular level that DOT1L inhibition increases neurogenesis driven by a shift of APs from asymmetric self-renewing to symmetric neurogenic consumptive divisions. At the molecular level, DOT1L activity prevents AP differentiation by promoting transcription of metabolic genes. Mechanistically, DOT1L inhibition reduces activity of an EZH2/PRC2 pathway, converging on increased expression of asparagine synthetase (ASNS), a microcephaly associated gene. Overexpression of ASNS in APs phenocopies DOT1L inhibition, and also increases neuronal differentiation of APs. Our data suggest that DOT1L activity/PRC2 crosstalk controls AP lineage progression by regulating asparagine metabolism.


Subject(s)
Aspartate-Ammonia Ligase , Neural Stem Cells , Aspartate-Ammonia Ligase/metabolism , Cell Differentiation/genetics , Neural Stem Cells/metabolism , Neurogenesis/genetics
7.
Open Res Eur ; 3: 5, 2023.
Article in English | MEDLINE | ID: mdl-38343617

ABSTRACT

This article summarizes the methodology for the identification of practitioners' challenges, in the context of the H2020 funded project FIRE-IN (Fire and Rescue Innovation Network) activities. The project consisted of five thematic areas or "Thematic Working Groups", as they are called, i.e., Search and Rescue Emergency Response, Structure Fires, Landscape Fires Crisis Mitigation, Natural Hazard Mitigation and Chemical Biological Radiological Nuclear and Explosives, and three iterations, each one including the identification of capability challenges, the screening for solutions, that can potentially address these challenges, and the request for ideas regarding future innovations that will complement already existing ones and will assist in covering capability gaps. This article focuses on the natural hazard mitigation working group and tsunamis in the Mediterranean region as a case study for the 3rd and last iteration of the project. The scenario of a tsunami occurrence in the Mediterranean is the basis for the FIRE-IN 3rd cycle workshop, as an indicative example of a high impact - low probability event, which aims to identify practitioners' Future Common Capability Challenges in Europe. The current status of the tsunami hazard in Europe, national and international tsunami risk mitigation measures and procedures and operational experience from recent events are also discussed. Focus is provided on the natural hazard mitigation and tsunami related practitioners' challenges, while results from the FIRE-IN request for ideas process and the interaction between practitioners, researchers and industry are also discussed. The aim is to present practitioners' current and future capability challenges , one of the main outcomes of the FIRE-IN project, and to provide further guidelines to stakeholders of disaster management towards a safer Europe, mainly, through preparedness and adaptation for stronger and resilient societies.

8.
Brain Pathol ; 32(5): e13064, 2022 09.
Article in English | MEDLINE | ID: mdl-35285112

ABSTRACT

Ermin is an actin-binding protein found almost exclusively in the central nervous system (CNS) as a component of myelin sheaths. Although Ermin has been predicted to play a role in the formation and stability of myelin sheaths, this has not been directly examined in vivo. Here, we show that Ermin is essential for myelin sheath integrity and normal saltatory conduction. Loss of Ermin in mice caused de-compacted and fragmented myelin sheaths and led to slower conduction along with progressive neurological deficits. RNA sequencing of the corpus callosum, the largest white matter structure in the CNS, pointed to inflammatory activation in aged Ermin-deficient mice, which was corroborated by increased levels of microgliosis and astrogliosis. The inflammatory milieu and myelin abnormalities were further associated with increased susceptibility to immune-mediated demyelination insult in Ermin knockout mice. Supporting a possible role of Ermin deficiency in inflammatory white matter disorders, a rare inactivating mutation in the ERMN gene was identified in multiple sclerosis patients. Our findings demonstrate a critical role for Ermin in maintaining myelin integrity. Given its near-exclusive expression in myelinating oligodendrocytes, Ermin deficiency represents a compelling "inside-out" model of inflammatory dysmyelination and may offer a new paradigm for the development of myelin stability-targeted therapies.


Subject(s)
Demyelinating Diseases , Multiple Sclerosis , Animals , Central Nervous System/metabolism , Demyelinating Diseases/genetics , Demyelinating Diseases/metabolism , Mice , Multiple Sclerosis/metabolism , Myelin Sheath/metabolism , Oligodendroglia/metabolism
9.
J Huntingtons Dis ; 10(3): 377-384, 2021.
Article in English | MEDLINE | ID: mdl-34366364

ABSTRACT

BACKGROUND: The relative contribution of grey matter (GM) and white matter (WM) degeneration to the progressive brain atrophy in Huntington's disease (HD) has been well studied. The pathology of the spinal cord in HD is comparatively less well documented. OBJECTIVE: We aim to characterize spinal cord WM abnormalities in a mouse model of HD and evaluate whether selective removal of mutant huntingtin (mHTT) from oligodendroglia rescues these deficits. METHODS: Histological assessments were used to determine the area of GM and WM in the spinal cord of 12-month-old BACHD mice, while electron microscopy was used to analyze myelin fibers in the cervical area of the spinal cord. To investigate the impact of inactivation of mHTT in oligodendroglia on these measures, we used the previously described BACHDxNG2Cre mouse line where mHTT is specifically reduced in oligodendrocyte progenitor cells. RESULTS: We show that spinal GM and WM areas are significantly atrophied in HD mice compared to wild-type controls. We further demonstrate that specific reduction of mHTT in oligodendroglial cells rescues the atrophy of spinal cord WM, but not GM, observed in HD mice. Inactivation of mHTT in oligodendroglia had no effect on the density of oligodendroglial cells but enhanced the expression of myelin-related proteins in the spinal cord. CONCLUSION: Our findings demonstrate that the myelination abnormalities observed in brain WM structures in HD extend to the spinal cord and suggest that specific expression of mHTT in oligodendrocytes contributes to such abnormalities.


Subject(s)
Huntington Disease , White Matter , Animals , Huntington Disease/genetics , Mice , Myelin Sheath , Oligodendroglia , Spinal Cord
10.
Biol Psychiatry ; 88(6): 500-511, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32653109

ABSTRACT

BACKGROUND: Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by epigenetic silencing of FMR1 and loss of FMRP expression. Efforts to understand the molecular underpinnings of the disease have been largely performed in rodent or nonisogenic settings. A detailed examination of the impact of FMRP loss on cellular processes and neuronal properties in the context of isogenic human neurons remains lacking. METHODS: Using CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 to introduce indels in exon 3 of FMR1, we generated an isogenic human pluripotent stem cell model of FXS that shows complete loss of FMRP expression. We generated neuronal cultures and performed genome-wide transcriptome and proteome profiling followed by functional validation of key dysregulated processes. We further analyzed neurodevelopmental and neuronal properties, including neurite length and neuronal activity, using multielectrode arrays and patch clamp electrophysiology. RESULTS: We showed that the transcriptome and proteome profiles of isogenic FMRP-deficient neurons demonstrate perturbations in synaptic transmission, neuron differentiation, cell proliferation and ion transmembrane transporter activity pathways, and autism spectrum disorder-associated gene sets. We uncovered key deficits in FMRP-deficient cells demonstrating abnormal neural rosette formation and neural progenitor cell proliferation. We further showed that FMRP-deficient neurons exhibit a number of additional phenotypic abnormalities, including neurite outgrowth and branching deficits and impaired electrophysiological network activity. These FMRP-deficient related impairments have also been validated in additional FXS patient-derived human-induced pluripotent stem cell neural cells. CONCLUSIONS: Using isogenic human pluripotent stem cells as a model to investigate the pathophysiology of FXS in human neurons, we reveal key neural abnormalities arising from the loss of FMRP.


Subject(s)
Autism Spectrum Disorder , Fragile X Syndrome , Induced Pluripotent Stem Cells , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Humans , Neurons
12.
Neurobiol Dis ; 135: 104744, 2020 02.
Article in English | MEDLINE | ID: mdl-31931139

ABSTRACT

Structural and molecular myelination deficits represent early pathological features of Huntington disease (HD). Recent evidence from germ-free (GF) animals suggests a role for microbiota-gut-brain bidirectional communication in the regulation of myelination. In this study, we aimed to investigate the impact of microbiota on myelin plasticity and oligodendroglial population dynamics in the mixed-sex BACHD mouse model of HD. Ultrastructural analysis of myelin in the corpus callosum revealed alterations of myelin thickness in BACHD GF compared to specific-pathogen free (SPF) mice, whereas no differences were observed between wild-type (WT) groups. In contrast, myelin compaction was altered in all groups when compared to WT SPF animals. Levels of myelin-related proteins were generally reduced, and the number of mature oligodendrocytes was decreased in the prefrontal cortex under GF compared to SPF conditions, regardless of genotype. Minor differences in commensal bacteria at the family and genera levels were found in the gut microbiota of BACHD and WT animals housed in standard living conditions. Our findings indicate complex effects of a germ-free status on myelin-related characteristics, and highlight the adaptive properties of myelination as a result of environmental manipulation.


Subject(s)
Huntington Disease/microbiology , Myelin Proteins/metabolism , Myelin Sheath/pathology , White Matter/microbiology , Animals , Bacteria/isolation & purification , Corpus Callosum/metabolism , Corpus Callosum/microbiology , Disease Models, Animal , Huntington Disease/pathology , Mice, Transgenic , Myelin Sheath/metabolism , Neuronal Plasticity/physiology , Oligodendroglia/metabolism , Prefrontal Cortex/metabolism , White Matter/pathology
13.
Proc Natl Acad Sci U S A ; 116(19): 9622-9627, 2019 05 07.
Article in English | MEDLINE | ID: mdl-31015293

ABSTRACT

White matter abnormalities are a nearly universal pathological feature of neurodegenerative disorders including Huntington disease (HD). A long-held assumption is that this white matter pathology is simply a secondary outcome of the progressive neuronal loss that manifests with advancing disease. Using a mouse model of HD, here we show that white matter and myelination abnormalities are an early disease feature appearing before the manifestation of any behavioral abnormalities or neuronal loss. We further show that selective inactivation of mutant huntingtin (mHTT) in the NG2+ oligodendrocyte progenitor cell population prevented myelin abnormalities and certain behavioral deficits in HD mice. Strikingly, the improvements in behavioral outcomes were seen despite the continued expression of mHTT in nonoligodendroglial cells including neurons, astrocytes, and microglia. Using RNA-seq and ChIP-seq analyses, we implicate a pathogenic mechanism that involves enhancement of polycomb repressive complex 2 (PRC2) activity by mHTT in the intrinsic oligodendroglial dysfunction and myelination deficits observed in HD. Our findings challenge the long-held dogma regarding the etiology of white matter pathology in HD and highlight the contribution of epigenetic mechanisms to the observed intrinsic oligodendroglial dysfunction. Our results further suggest that ameliorating white matter pathology and oligodendroglial dysfunction may be beneficial for HD.


Subject(s)
Behavior, Animal , Demyelinating Diseases , Huntingtin Protein , Huntington Disease , Mutation , Oligodendroglia , Animals , Demyelinating Diseases/genetics , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Disease Models, Animal , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/genetics , Huntington Disease/metabolism , Huntington Disease/pathology , Mice , Mice, Mutant Strains , Oligodendroglia/metabolism , Oligodendroglia/pathology , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , White Matter/metabolism , White Matter/pathology
14.
Neurobiol Dis ; 127: 65-75, 2019 07.
Article in English | MEDLINE | ID: mdl-30802499

ABSTRACT

Structural and molecular myelination deficits represent early pathological features of Huntington disease (HD). Recent evidence from germ-free (GF) animals suggests a role for microbiota-gut-brain bidirectional communication in the regulation of myelination. In this study, we aimed to investigate the impact of microbiota on myelin plasticity and oligodendroglial population dynamics in the mixed-sex BACHD mouse model of HD. Ultrastructural analysis of myelin in the corpus callosum revealed alterations of myelin thickness in BACHD GF compared to specific-pathogen free (SPF) mice, whereas no differences were observed between wild-type (WT) groups. In contrast, myelin compaction was altered in all groups when compared to WT SPF animals. Levels of myelin-related proteins were generally reduced, and the number of mature oligodendrocytes was decreased in the prefrontal cortex under GF compared to SPF conditions, regardless of genotype. Minor differences in commensal bacteria at the family and genera levels were found in the gut microbiota of BACHD and WT animals housed in standard living conditions. Our findings indicate complex effects of a germ-free status on myelin-related characteristics, and highlight the adaptive properties of myelination as a result of environmental manipulation.


Subject(s)
Corpus Callosum/pathology , Gastrointestinal Microbiome/physiology , Huntington Disease/microbiology , Myelin Sheath/pathology , Neuronal Plasticity/physiology , White Matter/pathology , Animals , Disease Models, Animal , Huntington Disease/pathology , Mice
15.
Mol Neurobiol ; 56(6): 4464-4478, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30334188

ABSTRACT

Laquinimod, an immunomodulatory agent under clinical development for Huntington disease (HD), has recently been shown to confer behavioural improvements that are coupled with prevention of atrophy of the white matter (WM)-rich corpus callosum (CC) in the YAC128 HD mice. However, the nature of the WM improvements is not known yet. Here we investigated the effects of laquinimod on HD-related myelination deficits at the cellular, molecular and ultrastructural levels. We showed that laquinimod treatment improves motor learning and motor function deficits in YAC128 HD mice, and confirmed its antidepressant effect even at the lowest dose used. In addition, we demonstrated for the first time the beneficial effects of laquinimod on myelination in the posterior region of the CC where it reversed changes in myelin sheath thickness and rescued Mbp mRNA and protein deficits. Furthermore, the effect of laquinimod on myelin-related gene expression was not region-specific since the levels of the Mbp and Plp1 transcripts were also increased in the striatum. Also, we did not detect changes in immune cell densities or levels of inflammatory genes in 3-month-old YAC128 HD mice, and these were not altered with laquinimod treatment. Thus, the beneficial effects of laquinimod on HD-related myelination abnormalities in YAC128 HD mice do not appear to be dependent on its immunomodulatory activity. Altogether, our findings describe the beneficial effects of laquinimod treatment on HD-related myelination abnormalities and highlight its therapeutic potential for the treatment of WM pathology in HD patients.


Subject(s)
Huntington Disease/drug therapy , Myelin Sheath/pathology , Myelin Sheath/ultrastructure , Quinolones/therapeutic use , Transcription, Genetic , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Behavior, Animal , Cell Count , Corpus Callosum/drug effects , Corpus Callosum/pathology , Corpus Callosum/physiopathology , Corpus Striatum/drug effects , Corpus Striatum/pathology , Corpus Striatum/physiopathology , Cytochrome P-450 CYP1A1/metabolism , Depression/complications , Depression/drug therapy , Depression/physiopathology , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Humans , Huntington Disease/complications , Huntington Disease/genetics , Huntington Disease/physiopathology , Inflammation/genetics , Inflammation/pathology , Learning , Male , Mice, Transgenic , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Motor Activity/drug effects , Myelin Sheath/drug effects , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Oligodendroglia/pathology , Phenotype , Quinolones/pharmacology , Receptors, Aryl Hydrocarbon/metabolism , Transcription, Genetic/drug effects
16.
NMR Biomed ; 31(12): e4007, 2018 12.
Article in English | MEDLINE | ID: mdl-30260561

ABSTRACT

Recent studies suggest that neurodegenerative diseases could affect brain structure and function in disease-specific network patterns; however, how spontaneous activity affects structural covariance network (SC) is not clear. We hypothesized that hyper-excitability in Huntington disease (HD) disrupts the coordinated structural and functional connectivity, and treatment with memantine helps to reduce excitotoxicity and normalize the connectivity. MRI was conducted to measure somatosensory activation, resting-state functional-connectivity (rsFC), SC, amplitude of low frequency fluctuation (ALFF) and ALFF covariance (ALFFC) in the YAC128 mouse model of HD. We found somatosensory activation was unchanged but the subcortical ALFF was increased in HD mice, indicating subcortical but not cortical hyperactivity. The reduced sensorimotor rsFC but spared hippocampal and default mode networks in the HD mice was consistent with the more pronounced impairment in motor function compared with cognitive performance. The disease suppressed SC globally and reduced ALFFC in the basal ganglia network as well as its anti-correlation with the default mode network. By comparing these connectivity measures, we found that the originally coupled rsFC-SC relationship was impaired whereas SC-ALFFC correlation was increased by HD, suggesting disease facilitated covariation of brain volume and activity amplitude but not neural synchrony. The comparison with mono-synaptic axonal projection supports the hypothesis that rsFC, but not SC or ALFFC, is highly dependent on structural connectivity under healthy conditions. Treatment with memantine had a strong effect on normalizing the SC and reducing ALFF while slightly increasing other connectivity measures and restoring the rsFC-SC coupling, which is consistent with its effect on alleviating hyper-excitability and improving the coordinated neural growth. These results indicate that HD affects the cerebral structure-function relationship which could be partially reverted by NMDA antagonism. These connectivity measures provide unique insights into pathological and pharmaceutical effects in brain circuitry, and could be translatable biomarkers for evaluating drug effect and refining its efficacy.


Subject(s)
Connectome , Huntington Disease/pathology , Huntington Disease/physiopathology , Magnetic Resonance Imaging , Animals , Axons/pathology , Behavior, Animal , Cognition , Disease Models, Animal , Electric Stimulation , Humans , Male , Memantine , Mice , Motor Activity , Nerve Net/physiopathology , Oxygen/blood , Rest , Somatosensory Cortex/pathology , Somatosensory Cortex/physiopathology , Structure-Activity Relationship
17.
Mol Neurodegener ; 13(1): 25, 2018 05 21.
Article in English | MEDLINE | ID: mdl-29783994

ABSTRACT

BACKGROUND: Huntington Disease (HD) is an incurable autosomal dominant neurodegenerative disorder driven by an expansion repeat giving rise to the mutant huntingtin protein (mHtt), which is known to disrupt a multitude of transcriptional pathways. Pridopidine, a small molecule in development for treatment of HD, has been shown to improve motor symptoms in HD patients. In HD animal models, pridopidine exerts neuroprotective effects and improves behavioral and motor functions. Pridopidine binds primarily to the sigma-1 receptor, (IC50 ~ 100 nM), which mediates its neuroprotective properties, such as rescue of spine density and aberrant calcium signaling in HD neuronal cultures. Pridopidine enhances brain-derived neurotrophic factor (BDNF) secretion, which is blocked by putative sigma-1 receptor antagonist NE-100, and was shown to upregulate transcription of genes in the BDNF, glucocorticoid receptor (GR), and dopamine D1 receptor (D1R) pathways in the rat striatum. The impact of different doses of pridopidine on gene expression and transcript splicing in HD across relevant brain regions was explored, utilizing the YAC128 HD mouse model, which carries the entire human mHtt gene containing 128 CAG repeats. METHODS: RNAseq was analyzed from striatum, cortex, and hippocampus of wild-type and YAC128 mice treated with vehicle, 10 mg/kg or 30 mg/kg pridopidine from the presymptomatic stage (1.5 months of age) until 11.5 months of age in which mice exhibit progressive disease phenotypes. RESULTS: The most pronounced transcriptional effect of pridopidine at both doses was observed in the striatum with minimal effects in other regions. In addition, for the first time pridopidine was found to have a dose-dependent impact on alternative exon and junction usage, a regulatory mechanism known to be impaired in HD. In the striatum of YAC128 HD mice, pridopidine treatment initiation prior to symptomatic manifestation rescues the impaired expression of the BDNF, GR, D1R and cAMP pathways. CONCLUSIONS: Pridopidine has broad effects on restoring transcriptomic disturbances in the striatum, particularly involving synaptic transmission and activating neuroprotective pathways that are disturbed in HD. Benefits of treatment initiation at early disease stages track with trends observed in the clinic.


Subject(s)
Gene Expression/drug effects , Huntington Disease , Neuroprotection/drug effects , Neuroprotective Agents/pharmacology , Piperidines/pharmacology , Animals , Brain/drug effects , Gene Expression Profiling , Humans , Mice , Mice, Transgenic , Synaptic Transmission/drug effects
18.
JCI Insight ; 2(23)2017 12 07.
Article in English | MEDLINE | ID: mdl-29212949

ABSTRACT

Pridopidine is currently under clinical development for Huntington disease (HD), with on-going studies to better characterize its therapeutic benefit and mode of action. Pridopidine was administered either prior to the appearance of disease phenotypes or in advanced stages of disease in the YAC128 mouse model of HD. In the early treatment cohort, animals received 0, 10, or 30 mg/kg pridopidine for a period of 10.5 months. In the late treatment cohort, animals were treated for 8 weeks with 0 mg/kg or an escalating dose of pridopidine (10 to 30 mg/kg over 3 weeks). Early treatment improved motor coordination and reduced anxiety- and depressive-like phenotypes in YAC128 mice, but it did not rescue striatal and corpus callosum atrophy. Late treatment, conversely, only improved depressive-like symptoms. RNA-seq analysis revealed that early pridopidine treatment reversed striatal transcriptional deficits, upregulating disease-specific genes that are known to be downregulated during HD, a finding that is experimentally confirmed herein. This suggests that pridopidine exerts beneficial effects at the transcriptional level. Taken together, our findings support continued clinical development of pridopidine for HD, particularly in the early stages of disease, and provide valuable insight into the potential therapeutic mode of action of pridopidine.


Subject(s)
Huntington Disease/drug therapy , Neuroprotective Agents/administration & dosage , Piperidines/administration & dosage , Animals , Anxiety/drug therapy , Anxiety/etiology , Behavior, Animal/drug effects , Corpus Callosum/pathology , Corpus Striatum/metabolism , Corpus Striatum/pathology , Depression/drug therapy , Depression/etiology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Evaluation, Preclinical/methods , Female , Gene Expression Regulation/drug effects , Huntington Disease/pathology , Huntington Disease/physiopathology , Huntington Disease/psychology , Male , Mice, Transgenic , Motor Activity/drug effects , Neuroprotective Agents/therapeutic use , Piperidines/therapeutic use , Secondary Prevention/methods , Transcription, Genetic/drug effects
19.
Immunity ; 47(1): 183-198.e6, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28723550

ABSTRACT

Tissue macrophages arise during embryogenesis from yolk-sac (YS) progenitors that give rise to primitive YS macrophages. Until recently, it has been impossible to isolate or derive sufficient numbers of YS-derived macrophages for further study, but data now suggest that induced pluripotent stem cells (iPSCs) can be driven to undergo a process reminiscent of YS-hematopoiesis in vitro. We asked whether iPSC-derived primitive macrophages (iMacs) can terminally differentiate into specialized macrophages with the help of growth factors and organ-specific cues. Co-culturing human or murine iMacs with iPSC-derived neurons promoted differentiation into microglia-like cells in vitro. Furthermore, murine iMacs differentiated in vivo into microglia after injection into the brain and into functional alveolar macrophages after engraftment in the lung. Finally, iPSCs from a patient with familial Mediterranean fever differentiated into iMacs with pro-inflammatory characteristics, mimicking the disease phenotype. Altogether, iMacs constitute a source of tissue-resident macrophage precursors that can be used for biological, pathophysiological, and therapeutic studies.


Subject(s)
Cell Culture Techniques/methods , Hematopoiesis , Macrophages/physiology , Neurons/physiology , Pluripotent Stem Cells/physiology , Animals , Cell Differentiation , Cells, Cultured , Embryo, Mammalian , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis
20.
Neurotherapeutics ; 14(4): 1095-1106, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28593439

ABSTRACT

The role of the T helper (Th)17 pathway has been clearly demonstrated in the onset and progression of autoimmune diseases, where interleukin (IL)-23 is a key molecule in maintaining the response mediated by Th17 cells. As a consequence, recent strategies based on blocking the interaction between IL-23 and its receptor (IL-23R), for example the anti-p19 antibody tildrakizumab, have been developed to regulate the Th17 pathway from the initial stages of the disease. Here, a soluble (s)IL-23R cDNA was cloned in expression plasmids and viral vectors. The clinical efficacy of sIL-23R was evaluated in myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis mice intravenously injected with a single dose of adeno-associated virus AAV8-sIL-23R vectors. Cytokine secretion was determined by multiplex assay, while histopathological analysis of the central nervous system was performed to study demyelination, inflammatory infiltration, and microglia and astroglia activation. We observed that administration of adeno-associated vector 8 encoding sIL-23R was associated with a significant disease improvement, including delay in the onset of the clinical signs; slower progress of the disease; interference with IL-23-mediated signal transducer and activator of transcription response by inhibiting of signal transducer and activator of transcription 3 phosphorylation; reduced demyelination and infiltration in the central nervous system; and lower astrocyte and microglia activation. Our results suggest that the use of vectors carrying sIL-23R to block the IL-23/IL-23R interaction may be a new therapeutic strategy for the treatment of multiple sclerosis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/therapy , Genetic Vectors/administration & dosage , Multiple Sclerosis/therapy , Receptors, Interleukin/metabolism , Animals , Astrocytes/metabolism , Dependovirus/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Genetic Therapy , Genetic Vectors/genetics , HEK293 Cells , Humans , Mice, Inbred C57BL , Microglia/metabolism , Myelitis/pathology , Receptors, Interleukin/genetics , Signal Transduction , Spinal Cord/pathology , Th17 Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...