Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Mol Cell ; 78(2): 346-358.e9, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32268123

ABSTRACT

CAG-repeat expansions in at least eight different genes cause neurodegeneration. The length of the extended polyglutamine stretches in the corresponding proteins is proportionally related to their aggregation propensity. Although these proteins are ubiquitously expressed, they predominantly cause toxicity to neurons. To understand this neuronal hypersensitivity, we generated induced pluripotent stem cell (iPSC) lines of spinocerebellar ataxia type 3 and Huntington's disease patients. iPSC generation and neuronal differentiation are unaffected by polyglutamine proteins and show no spontaneous aggregate formation. However, upon glutamate treatment, aggregates form in neurons but not in patient-derived neural progenitors. During differentiation, the chaperone network is drastically rewired, including loss of expression of the anti-amyloidogenic chaperone DNAJB6. Upregulation of DNAJB6 in neurons antagonizes glutamate-induced aggregation, while knockdown of DNAJB6 in progenitors results in spontaneous polyglutamine aggregation. Loss of DNAJB6 expression upon differentiation is confirmed in vivo, explaining why stem cells are intrinsically protected against amyloidogenesis and protein aggregates are dominantly present in neurons.


Subject(s)
Amyloidogenic Proteins/genetics , Cell Differentiation/genetics , HSP40 Heat-Shock Proteins/genetics , Molecular Chaperones/genetics , Nerve Tissue Proteins/genetics , Neural Stem Cells/metabolism , Gene Expression Regulation/genetics , Gene Knockout Techniques , Glutamic Acid/metabolism , Humans , Huntington Disease/genetics , Huntington Disease/metabolism , Huntington Disease/pathology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Machado-Joseph Disease/genetics , Machado-Joseph Disease/metabolism , Machado-Joseph Disease/pathology , Neural Stem Cells/pathology , Neurons/metabolism , Neurons/pathology , Protein Aggregates/genetics , Trinucleotide Repeat Expansion/genetics
2.
Hum Mol Genet ; 30(13): 1175-1187, 2021 06 17.
Article in English | MEDLINE | ID: mdl-33601422

ABSTRACT

Synaptic dysfunction and cognitive decline in Huntington's disease (HD) involve hyperactive A disintegrin and metalloproteinase domain-containing protein 10 (ADAM10). To identify the molecular mechanisms through which ADAM10 is associated with synaptic dysfunction in HD, we performed an immunoaffinity purification-mass spectrometry (IP-MS) study of endogenous ADAM10 in the brains of wild-type and HD mice. We found that proteins implicated in synapse organization, synaptic plasticity, and vesicle and organelles trafficking interact with ADAM10, suggesting that it may act as hub protein at the excitatory synapse. Importantly, the ADAM10 interactome is enriched in presynaptic proteins and ADAM10 co-immunoprecipitates with piccolo (PCLO), a key player in the recycling and maintenance of synaptic vesicles. In contrast, reduced ADAM10/PCLO immunoprecipitation occurs in the HD brain, with decreased density of synaptic vesicles in the reserve and docked pools at the HD presynaptic terminal. Conditional heterozygous deletion of ADAM10 in the forebrain of HD mice reduces active ADAM10 to wild-type level and normalizes ADAM10/PCLO complex formation and synaptic vesicle density and distribution. The results indicate that presynaptic ADAM10 and PCLO are a relevant component of HD pathogenesis.


Subject(s)
ADAM10 Protein/metabolism , Cytoskeletal Proteins/metabolism , Huntington Disease/metabolism , Neuropeptides/metabolism , Synaptic Vesicles/metabolism , ADAM10 Protein/genetics , Animals , Blotting, Western , Brain/metabolism , Brain/pathology , Brain/ultrastructure , Humans , Huntington Disease/genetics , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Presynaptic Terminals/metabolism , Protein Binding , Protein Interaction Maps/genetics , Proteomics/methods , Synaptic Vesicles/ultrastructure , Synaptosomes/metabolism , Tandem Mass Spectrometry/methods
3.
Brain ; 144(10): 3175-3190, 2021 11 29.
Article in English | MEDLINE | ID: mdl-33974044

ABSTRACT

Brain cholesterol is produced mainly by astrocytes and is important for neuronal function. Its biosynthesis is severely reduced in mouse models of Huntington's disease. One possible mechanism is a diminished nuclear translocation of the transcription factor sterol regulatory element-binding protein 2 (SREBP2) and, consequently, reduced activation of SREBP2-controlled genes in the cholesterol biosynthesis pathway. Here we evaluated the efficacy of a gene therapy based on the unilateral intra-striatal injection of a recombinant adeno-associated virus 2/5 (AAV2/5) targeting astrocytes specifically and carrying the transcriptionally active N-terminal fragment of human SREBP2 (hSREBP2). Robust hSREBP2 expression in striatal glial cells in R6/2 Huntington's disease mice activated the transcription of cholesterol biosynthesis pathway genes, restored synaptic transmission, reversed dopamine receptor D2 (Drd2) transcript levels decline, cleared mutant huntingtin aggregates and attenuated behavioural deficits. We conclude that glial SREBP2 participates in Huntington's disease brain pathogenesis in vivo and that AAV-based delivery of SREBP2 to astrocytes counteracts key features of the disease.


Subject(s)
Astrocytes/metabolism , Corpus Striatum/metabolism , Gene Transfer Techniques , Genetic Therapy/methods , Huntington Disease/therapy , Sterol Regulatory Element Binding Protein 2/administration & dosage , Animals , Astrocytes/pathology , Corpus Striatum/pathology , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Huntington Disease/genetics , Huntington Disease/metabolism , Huntington Disease/pathology , Male , Mice , Mice, Inbred CBA , Mice, Transgenic , Phenotype , Sterol Regulatory Element Binding Protein 2/biosynthesis , Sterol Regulatory Element Binding Protein 2/genetics
4.
Int J Mol Sci ; 23(21)2022 Oct 29.
Article in English | MEDLINE | ID: mdl-36361956

ABSTRACT

In vitro models of corticogenesis from pluripotent stem cells (PSCs) have greatly improved our understanding of human brain development and disease. Among these, 3D cortical organoid systems are able to recapitulate some aspects of in vivo cytoarchitecture of the developing cortex. Here, we tested three cortical organoid protocols for brain regional identity, cell type specificity and neuronal maturation. Overall, all protocols gave rise to organoids that displayed a time-dependent expression of neuronal maturation genes such as those involved in the establishment of synapses and neuronal function. Comparatively, guided differentiation methods without WNT activation generated the highest degree of cortical regional identity, whereas default conditions produced the broadest range of cell types such as neurons, astrocytes and hematopoietic-lineage-derived microglia cells. These results suggest that cortical organoid models produce diverse outcomes of brain regional identity and cell type specificity and emphasize the importance of selecting the correct model for the right application.


Subject(s)
Organoids , Pluripotent Stem Cells , Humans , Pluripotent Stem Cells/metabolism , Cell Differentiation , Neurons/metabolism , Brain
5.
Neurobiol Dis ; 146: 105140, 2020 12.
Article in English | MEDLINE | ID: mdl-33065279

ABSTRACT

RUES2 cell lines represent the first collection of isogenic human embryonic stem cells (hESCs) carrying different pathological CAG lengths in the HTT gene. However, their neuronal differentiation potential has yet to be thoroughly evaluated. Here, we report that RUES2 during ventral telencephalic differentiation is biased towards medial ganglionic eminence (MGE). We also show that HD-RUES2 cells exhibit an altered MGE transcriptional signature in addition to recapitulating known HD phenotypes, with reduced expression of the neurodevelopmental regulators NEUROD1 and BDNF and increased cleavage of synaptically enriched N-cadherin. Finally, we identified the transcription factor SP1 as a common potential detrimental co-partner of muHTT by de novo motif discovery analysis on the LGE, MGE, and cortical genes differentially expressed in HD human pluripotent stem cells in our and additional datasets. Taken together, these observations suggest a broad deleterious effect of muHTT in the early phases of neuronal development that may unfold through its altered interaction with SP1.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Differentiation/physiology , Human Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Receptors, Immunologic/metabolism , Cell Differentiation/drug effects , Human Embryonic Stem Cells/pathology , Humans , Huntington Disease/genetics , Huntington Disease/metabolism , Neurogenesis/physiology , Neurons/metabolism
6.
Proc Natl Acad Sci U S A ; 114(50): E10809-E10818, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29162692

ABSTRACT

Posttranslational modifications can have profound effects on the biological and biophysical properties of proteins associated with misfolding and aggregation. However, their detection and quantification in clinical samples and an understanding of the mechanisms underlying the pathological properties of misfolding- and aggregation-prone proteins remain a challenge for diagnostics and therapeutics development. We have applied an ultrasensitive immunoassay platform to develop and validate a quantitative assay for detecting a posttranslational modification (phosphorylation at residue T3) of a protein associated with polyglutamine repeat expansion, namely Huntingtin, and characterized its presence in a variety of preclinical and clinical samples. We find that T3 phosphorylation is greatly reduced in samples from Huntington's disease models and in Huntington's disease patients, and we provide evidence that bona-fide T3 phosphorylation alters Huntingtin exon 1 protein conformation and aggregation properties. These findings have significant implications for both mechanisms of disease pathogenesis and the development of therapeutics and diagnostics for Huntington's disease.


Subject(s)
Huntingtin Protein/metabolism , Huntington Disease/metabolism , Immunoassay/methods , Animals , Cells, Cultured , Exons , HEK293 Cells , Humans , Huntingtin Protein/chemistry , Huntingtin Protein/genetics , Mice , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Phosphorylation , Protein Conformation , Protein Processing, Post-Translational , Sensitivity and Specificity
7.
Mol Cell Neurosci ; 82: 46-57, 2017 07.
Article in English | MEDLINE | ID: mdl-28476540

ABSTRACT

DNA methylation (DNAm) changes are of increasing relevance to neurodegenerative disorders, including Huntington's disease (HD). We performed genome-wide screening of possible DNAm changes occurring during striatal differentiation in human induced pluripotent stem cells derived from a HD patient (HD-hiPSCs) as cellular model. We identified 240 differentially methylated regions (DMRs) at promoters in fully differentiated HD-hiPSCs. Subsequently, we focused on the methylation differences in a subcluster of genes related to Jumonji Domain Containing 3 (JMJD3), a demethylase that epigenetically regulates neuronal differentiation and activates neuronal progenitor associated genes, which are indispensable for neuronal fate acquisition. Noticeably among these genes, WD repeat-containing protein 5 (WDR5) promoter was found hypermethylated in HD-hiPSCs, resulting in a significant down-modulation in its expression and of the encoded protein. A similar WDR5 expression decrease was seen in a small series of HD-hiPSC lines characterized by different CAG length. The decrease in WDR5 expression was particularly evident in HD-hiPSCs compared to hESCs and control-hiPSCs from healthy subjects. WDR5 is a core component of the MLL/SET1 chromatin remodeling complexes essential for H3K4me3, previously reported to play an important role in stem cells self-renewal and differentiation. These results suggest the existence of epigenetic mechanisms in HD and the identification of genes, which are able to modulate HD phenotype, is important both for biomarker discovery and therapeutic interventions.


Subject(s)
Cell Differentiation/genetics , Epigenesis, Genetic/genetics , Histone-Lysine N-Methyltransferase/genetics , Huntington Disease/metabolism , Induced Pluripotent Stem Cells/cytology , Cell Line , Chromatin/metabolism , Chromatin Assembly and Disassembly/genetics , Humans , Huntington Disease/genetics , Intracellular Signaling Peptides and Proteins , Neurons/metabolism
8.
Neuropathol Appl Neurobiol ; 40(7): 899-910, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24634989

ABSTRACT

AIMS: The repressor element-1 silencing transcription factor/neurone-restrictive silencer factor (REST/NRSF) is a master regulator of neuronal gene expression. REST/NRSF functions by recruiting other cofactors to genomic loci that contain the repressor element 1/neurone restrictive silencer element (RE1/NRSE) binding motif. In brain, demonstration of REST protein presence in neurones has remained controversial. However, RE1/NRSE containing neuronal genes are actively modulated and REST dysregulation is implicated in Huntington's disease (HD). We aimed to investigate REST distribution in autopsy brain from control and HD patients. METHODS: Brain tissues from six controls and six HD cases (Vonsattel grade 3 and 4) were investigated using immunohistochemical analysis. RESULTS: REST was present in neurones and glial cells of the cortex, caudate nucleus, hippocampus and cerebellum. REST labelling was mainly cytoplasmic in neurones while preferential nuclear staining of REST was found in glial cells. We also found that REST and huntingtin (HTT) colocalize in human neurones. Low levels of cytoplasmic REST were detected in neurones of the HD cortex and caudate but no direct relationship between decreased neuronal REST expression and disease grade was observed. CONCLUSIONS: These data support the notion of REST presence in human brain neurones and glial cells and indicate the importance of developing compounds able to restore REST-regulated transcription of neuronal genes in HD.


Subject(s)
Brain/metabolism , Huntington Disease/metabolism , Neuroglia/metabolism , Neurons/metabolism , Repressor Proteins/analysis , Adult , Aged , Female , Humans , Huntingtin Protein , Male , Middle Aged , Nerve Tissue Proteins/metabolism
9.
Nat Commun ; 15(1): 6534, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095390

ABSTRACT

Huntington's disease (HD) causes selective degeneration of striatal and cortical neurons, resulting in cell mosaicism of coexisting still functional and dysfunctional cells. The impact of non-cell autonomous mechanisms between these cellular states is poorly understood. Here we generated telencephalic organoids with healthy or HD cells, grown separately or as mosaics of the two genotypes. Single-cell RNA sequencing revealed neurodevelopmental abnormalities in the ventral fate acquisition of HD organoids, confirmed by cytoarchitectural and transcriptional defects leading to fewer GABAergic neurons, while dorsal populations showed milder phenotypes mainly in maturation trajectory. Healthy cells in mosaic organoids restored HD cell identity, trajectories, synaptic density, and communication pathways upon cell-cell contact, while showing no significant alterations when grown with HD cells. These findings highlight cell-type-specific alterations in HD and beneficial non-cell autonomous effects of healthy cells, emphasizing the therapeutic potential of modulating cell-cell communication in disease progression and treatment.


Subject(s)
Huntington Disease , Organoids , Phenotype , Telencephalon , Huntington Disease/pathology , Huntington Disease/genetics , Huntington Disease/metabolism , Organoids/pathology , Organoids/metabolism , Animals , Telencephalon/pathology , Telencephalon/cytology , Telencephalon/metabolism , Humans , Mice , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Single-Cell Analysis , Cell Communication , Mosaicism , Neurons/metabolism , Neurons/pathology
10.
J Neurochem ; 127(1): 22-35, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23800350

ABSTRACT

Transcriptional dysregulation is a hallmark of Huntington's disease (HD) and one cause of this dysregulation is enhanced activity of the REST-mSIN3a-mSIN3b-CoREST-HDAC repressor complex, which silences transcription through REST binding to the RE1/NRSE silencer. Normally, huntingtin (HTT) prevents this binding, allowing expressing of REST target genes. Here, we aimed to identify HTT mimetics that disrupt REST complex formation in HD. From a structure-based virtual screening of 7 million molecules, we selected 94 compounds predicted to interfere with REST complex formation by targeting the PAH1 domain of mSIN3b. Primary screening using DiaNRSELuc8 cells revealed two classes of compounds causing a greater than two-fold increase in luciferase. In particular, quinolone-like compound 91 (C91) at a non-toxic nanomolar concentration reduced mSIN3b nuclear entry and occupancy at the RE1/NRSE within the Bdnf locus, and restored brain-derived neurotrophic factor (BDNF) protein levels in HD cells. The mRNA levels of other RE1/NRSE-regulated genes were similarly increased while non-REST-regulated genes were unaffected. C91 stimulated REST-regulated gene expression in HTT-knockdown Zebrafish and increased BDNF mRNA in the presence of mutant HTT. Thus, a combination of virtual screening and biological approaches can lead to compounds reducing REST complex formation, which may be useful in HD and in other pathological conditions.


Subject(s)
Huntington Disease/genetics , Huntington Disease/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription, Genetic/physiology , Animals , Animals, Genetically Modified , Brain-Derived Neurotrophic Factor/metabolism , Cell Proliferation , Cells, Cultured , Chromatin Immunoprecipitation , Co-Repressor Proteins , Embryo, Nonmammalian , Enzyme-Linked Immunosorbent Assay , Humans , Huntingtin Protein , Immunohistochemistry , Luciferases/metabolism , Microinjections , Models, Molecular , Nerve Tissue Proteins/biosynthesis , Polymerase Chain Reaction , RNA, Messenger/administration & dosage , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Transcription, Genetic/genetics , Transfection , Zebrafish
11.
Neurobiol Dis ; 50: 160-70, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23089356

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder that affects muscle coordination and diminishes cognitive abilities. The genetic basis of the disease is an expansion of CAG repeats in the Huntingtin (Htt) gene. Here we aimed to generate a series of mouse neural stem (NS) cell lines that carried varying numbers of CAG repeats in the mouse Htt gene (Hdh CAG knock-in NS cells) or that had Hdh null alleles (Hdh knock-out NS cells). Towards this end, Hdh CAG knock-in mouse ES cell lines that carried an Htt gene with 20, 50, 111, or 140 CAG repeats or that were Htt null were neuralized and converted into self-renewing NS cells. The resulting NS cell lines were immunopositive for the neural stem cell markers NESTIN, SOX2, and BLBP and had similar proliferative rates and cell cycle distributions. After 14 days in vitro, wild-type NS cells gave rise to cultures composed of 70% MAP2(+) neurons and 30% GFAP(+) astrocytes. In contrast, NS cells with expanded CAG repeats underwent neuronal cell death, with only 38%±15% of the MAP2(+) cells remaining at the end of the differentiation period. Cell death was verified by increased caspase 3/7 activity on day 14 of the neuronal differentiation protocol. Interestingly, Hdh knock-out NS cells treated using the same neuronal differentiation protocol showed a dramatic increase in the number of GFAP(+) cells on day 14 (61%±20% versus 24%±10% in controls), and a massive decrease of MAP2(+) neurons (30%±11% versus 64%±17% in controls). Both Hdh CAG knock-in NS cells and Hdh knock-out NS cells showed reduced levels of Bdnf mRNA during neuronal differentiation, in agreement with data obtained previously in HD mouse models and in post-mortem brain samples from HD patients. We concluded that Hdh CAG knock-in and Hdh knock-out NS cells have potential as tools for investigating the roles of normal and mutant HTT in differentiated neurons and glial cells of the brain.


Subject(s)
Cell Differentiation/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neuroglia/cytology , Neurons/cytology , Nuclear Proteins/metabolism , Animals , Fluorescent Antibody Technique , Gene Knock-In Techniques , Gene Knockout Techniques , Huntingtin Protein , Huntington Disease/genetics , Mice , Nerve Tissue Proteins/genetics , Neuroglia/metabolism , Neurons/metabolism , Nuclear Proteins/genetics , Peptides , Reverse Transcriptase Polymerase Chain Reaction , Trinucleotide Repeats
12.
Methods Mol Biol ; 2655: 211-220, 2023.
Article in English | MEDLINE | ID: mdl-37212999

ABSTRACT

Several studies highlighted the importance of the polycomb repressive complex 2 (PRC2) already at the beginning of development. Although the crucial function of PRC2 in regulating lineage commitment and cell-fate specification has been well-established, the in vitro study of the exact mechanisms for which H3K27me3 is indispensable for proper differentiation is still challenging. In this chapter, we report a well-established and reproducible differentiation protocol to generate striatal medium spiny neurons as a tool to explore PRC2 role in brain development.


Subject(s)
Polycomb Repressive Complex 2 , Polycomb Repressive Complex 2/genetics , Cell Differentiation/physiology
13.
Stem Cell Res Ther ; 14(1): 189, 2023 07 28.
Article in English | MEDLINE | ID: mdl-37507794

ABSTRACT

BACKGROUND: Huntington's disease (HD) is a motor and cognitive neurodegenerative disorder due to prominent loss of striatal medium spiny neurons (MSNs). Cell replacement using human embryonic stem cells (hESCs) derivatives may offer new therapeutic opportunities to replace degenerated neurons and repair damaged circuits. METHODS: With the aim to develop effective cell replacement for HD, we assessed the long-term therapeutic value of hESC-derived striatal progenitors by grafting the cells into the striatum of a preclinical model of HD [i.e., adult immunodeficient rats in which the striatum was lesioned by monolateral injection of quinolinic acid (QA)]. We examined the survival, maturation, self-organization and integration of the graft as well as its impact on lesion-dependent motor alterations up to 6 months post-graft. Moreover, we tested whether exposing a cohort of QA-lesioned animals to environmental enrichment (EE) could improve graft integration and function. RESULTS: Human striatal progenitors survived up to 6 months after transplantation and showed morphological and neurochemical features typical of human MSNs. Donor-derived interneurons were also detected. Grafts wired in both local and long-range striatal circuits, formed domains suggestive of distinct ganglionic eminence territories and displayed emerging striosome features. Moreover, over time grafts improved complex motor performances affected by QA. EE selectively increased cell differentiation into MSN phenotype and promoted host-to-graft connectivity. However, when combined to the graft, the EE paradigm used in this study was insufficient to produce an additive effect on task execution. CONCLUSIONS: The data support the long-term therapeutic potential of ESC-derived human striatal progenitor grafts for the replacement of degenerated striatal neurons in HD and suggest that EE can effectively accelerate the maturation and promote the integration of human striatal cells.


Subject(s)
Brain Tissue Transplantation , Human Embryonic Stem Cells , Huntington Disease , Rats , Animals , Humans , Huntington Disease/therapy , Corpus Striatum/physiology , Neurons , Disease Models, Animal
14.
Neurobiol Dis ; 48(3): 282-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22796360

ABSTRACT

Huntington disease (HD), a neurodegenerative disorder caused by an expanded CAG repeat in the HTT gene, remains without a treatment to modify the course of the illness. Lithium, a drug widely used for the treatment of bipolar disorder, has been shown to exert neuroprotective effects in a number of models of neurological disease but may have various toxic effects at conventional therapeutic doses. We examined whether NP03, a novel low-dose lithium microemulsion, would improve the disease phenotypes in the YAC128 mouse model of HD. We demonstrate that NP03 improves motor function, ameliorates the neuropathological deficits in striatal volume, neuronal counts, and DARPP-32 expression, and partially rescues testicular atrophy in YAC128 mice. These positive effects were accompanied by improvements in multiple biochemical endpoints associated with the pathogenesis of HD, including normalization of caspase-6 activation and amelioration of deficits in BDNF levels, and with no lithium-related toxicity. Our findings demonstrate that NP03 ameliorates the motor and neuropathological phenotypes in the YAC128 mouse model of HD, and represents a potential therapeutic approach for HD.


Subject(s)
Brain/drug effects , Huntington Disease/drug therapy , Lithium/administration & dosage , Neuroprotective Agents/administration & dosage , Animals , Brain/pathology , Disease Models, Animal , Female , Humans , Huntington Disease/pathology , Immunoblotting , Lithium/adverse effects , Male , Mice , Motor Activity/drug effects , Neuroprotective Agents/adverse effects , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
15.
Cell Rep Methods ; 2(12): 100367, 2022 12 19.
Article in English | MEDLINE | ID: mdl-36590694

ABSTRACT

Stem cell engineering of striatal medium spiny neurons (MSNs) is a promising strategy to understand diseases affecting the striatum and for cell-replacement therapies in different neurological diseases. Protocols to generate cells from human pluripotent stem cells (PSCs) are scarce and how well they recapitulate the endogenous fetal cells remains poorly understood. We have developed a protocol that modulates cell seeding density and exposure to specific morphogens that generates authentic and functional D1- and D2-MSNs with a high degree of reproducibility in 25 days of differentiation. Single-cell RNA sequencing (scRNA-seq) shows that our cells can mimic the cell-fate acquisition steps observed in vivo in terms of cell type composition, gene expression, and signaling pathways. Finally, by modulating the midkine pathway we show that we can increase the yield of MSNs. We expect that this protocol will help decode pathogenesis factors in striatal diseases and eventually facilitate cell-replacement therapies for Huntington's disease (HD).


Subject(s)
Medium Spiny Neurons , Pluripotent Stem Cells , Humans , Reproducibility of Results , Neurogenesis , Corpus Striatum , Pluripotent Stem Cells/metabolism
16.
J Neurochem ; 116(3): 415-25, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21105876

ABSTRACT

Transcriptional dysfunction is a prominent hallmark of Huntington's disease (HD). Several transcription factors have been implicated in the aetiology of HD progression and one of the most prominent is repressor element 1 (RE1) silencing transcription factor (REST). REST is a global repressor of neuronal gene expression and in the presence of mutant Huntingtin increased nuclear REST levels lead to elevated RE1 occupancy and a concomitant increase in target gene repression, including brain-derived neurotrophic factor. It is of great interest to devise strategies to reverse transcriptional dysregulation caused by increased nuclear REST and determine the consequences in HD. Thus far, such strategies have involved RNAi or mutant REST constructs. Decoys are double-stranded oligodeoxynucleotides corresponding to the DNA-binding element of a transcription factor and act to sequester it, thereby abrogating its transcriptional activity. Here, we report the use of a novel decoy strategy to rescue REST target gene expression in a cellular model of HD. We show that delivery of the decoy in cells expressing mutant Huntingtin leads to its specific interaction with REST, a reduction in REST occupancy of RE1s and rescue of target gene expression, including Bdnf. These data point to an alternative strategy for rebalancing the transcriptional dysregulation in HD.


Subject(s)
Genetic Therapy/methods , Huntington Disease/genetics , Huntington Disease/therapy , Oligonucleotides/genetics , Oligonucleotides/pharmacology , Repressor Proteins/physiology , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cell Line , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Knock-In Techniques , Gene Silencing , Humans , Huntingtin Protein , Huntington Disease/metabolism , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oligonucleotides/therapeutic use , Repressor Proteins/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/genetics
17.
Nat Rev Neurol ; 17(6): 381-392, 2021 06.
Article in English | MEDLINE | ID: mdl-33658662

ABSTRACT

Human induced pluripotent stem cells (hiPSCs) were first generated in 2007, but the full translational potential of this valuable tool has yet to be realized. The potential applications of hiPSCs are especially relevant to neurology, as brain cells from patients are rarely available for research. hiPSCs from individuals with neuropsychiatric or neurodegenerative diseases have facilitated biological and multi-omics studies as well as large-scale screening of chemical libraries. However, researchers are struggling to improve the scalability, reproducibility and quality of this descriptive disease modelling. Addressing these limitations will be the first step towards a new era in hiPSC research - that of predictive disease modelling - involving the correlation and integration of in vitro experimental data with longitudinal clinical data. This approach is a key element of the emerging precision medicine paradigm, in which hiPSCs could become a powerful diagnostic and prognostic tool. Here, we consider the steps necessary to achieve predictive modelling of neurodegenerative disease with hiPSCs, using Huntington disease as an example.


Subject(s)
Epigenesis, Genetic/genetics , Genetic Testing/trends , Induced Pluripotent Stem Cells/physiology , Neurodegenerative Diseases/diagnostic imaging , Neurodegenerative Diseases/genetics , Clinical Trials as Topic/methods , Genetic Testing/methods , Humans , Huntington Disease/diagnostic imaging , Huntington Disease/genetics , Huntington Disease/therapy , Neurodegenerative Diseases/therapy , Predictive Value of Tests
18.
EMBO Mol Med ; 13(4): e12433, 2021 04 09.
Article in English | MEDLINE | ID: mdl-33665914

ABSTRACT

MECP2 mutations cause Rett syndrome (RTT), a severe and progressive neurodevelopmental disorder mainly affecting females. Although RTT patients exhibit delayed onset of symptoms, several evidences demonstrate that MeCP2 deficiency alters early development of the brain. Indeed, during early maturation, Mecp2 null cortical neurons display widespread transcriptional changes, reduced activity, and defective morphology. It has been proposed that during brain development these elements are linked in a feed-forward cycle where neuronal activity drives transcriptional and morphological changes that further increase network maturity. We hypothesized that the enhancement of neuronal activity during early maturation might prevent the onset of RTT-typical molecular and cellular phenotypes. Accordingly, we show that the enhancement of excitability, obtained by adding to neuronal cultures Ampakine CX546, rescues transcription of several genes, neuronal morphology, and responsiveness to stimuli. Greater effects are achieved in response to earlier treatments. In vivo, short and early administration of CX546 to Mecp2 null mice prolongs lifespan, delays the disease progression, and rescues motor abilities and spatial memory, thus confirming the value for RTT of an early restoration of neuronal activity.


Subject(s)
Methyl-CpG-Binding Protein 2 , Rett Syndrome , Animals , Brain/metabolism , Female , Humans , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Mice , Neurons/metabolism , Phenotype , Rett Syndrome/genetics
19.
Science ; 372(6542)2021 05 07.
Article in English | MEDLINE | ID: mdl-33958447

ABSTRACT

Deciphering how the human striatum develops is necessary for understanding the diseases that affect this region. To decode the transcriptional modules that regulate this structure during development, we compiled a catalog of 1116 long intergenic noncoding RNAs (lincRNAs) identified de novo and then profiled 96,789 single cells from the early human fetal striatum. We found that D1 and D2 medium spiny neurons (D1- and D2-MSNs) arise from a common progenitor and that lineage commitment is established during the postmitotic transition, across a pre-MSN phase that exhibits a continuous spectrum of fate determinants. We then uncovered cell type-specific gene regulatory networks that we validated through in silico perturbation. Finally, we identified human-specific lincRNAs that contribute to the phylogenetic divergence of this structure in humans. This work delineates the cellular hierarchies governing MSN lineage commitment.


Subject(s)
Atlases as Topic , Corpus Striatum/cytology , Corpus Striatum/embryology , Neurogenesis/genetics , RNA, Long Noncoding/genetics , Single-Cell Analysis , Transcription Factors/genetics , Fetus , GABAergic Neurons/metabolism , Humans , RNA-Seq , Transcription, Genetic
20.
Stem Cell Res ; 49: 102016, 2020 12.
Article in English | MEDLINE | ID: mdl-33039807

ABSTRACT

GSX2 is a homeobox transcription factor (TF) controlling the specification of the ventral lateral ganglionic eminence and its major derivative, the corpus striatum. Medium spiny neurons (MSNs) represent the largest cell component of the striatum and they are primarily affected in Huntington disease (HD). Here, we used CRISPR technology to generate a pluripotent GSX2-reporter human embryonic stem cell (hESC) line that can be leveraged to monitor striatal differentiation in real-time and to enrich for MSN-committed progenitors.


Subject(s)
Human Embryonic Stem Cells , Cell Differentiation , Clustered Regularly Interspaced Short Palindromic Repeats , Corpus Striatum , Embryonic Stem Cells , Humans , Neurons
SELECTION OF CITATIONS
SEARCH DETAIL